ArticlePDF AvailableLiterature Review

Gut Microbiota, Probiotics, and Human Health

Authors:

Abstract and Figures

The review is devoted to the problems of microbiota and the ways of it correction employing beneficial life bacteria- probiotics. It covers the issues related to the functioning of human microbiota and its importance for the health, individual variability of microbial content, functioning of the probiotics in the human organism and the history of probiotic studies with particular focus on the microbiological investigations in the USSR. The article discusses the safety issues related to probiotics and the problems with probiotic therapy, trying to explain the reasons for the side effects caused by probiotics. The necessity of personified selection of the probiotic strain or individual microbial therapy autoprobiotics is also discussed.
Content may be subject to copyright.
Gut Microbiota, Probiotics, and Human Health
Alexander SUVOROV
1
1
Department of Molecular Microbiology, Institute for Experimental Medicine, Acad. Pavlov Street 12, Saint-Petersburg 197376, Russia
Received November 30, 2012; Accepted April 2, 2013
The review is devoted to the problems of microbiota and the ways of it correction employing benecial life bacteria-
probiotics. It covers the issues related to the functioning of human microbiota and its importance for the health,
individual variability of microbial content, functioning of the probiotics in the human organism and the history of
probiotic studies with particular focus on the microbiological investigations in the USSR. The article discusses the
safety issues related to probiotics and the problems with probiotic therapy, trying to explain the reasons for the side
effects caused by probiotics. The necessity of personied selection of the probiotic strain or individual microbial
therapy autoprobiotics is also discussed.
Key words: microbiota, probiotic, enterotype
INTRODUCTION
The entire concept of the human organism being
located at the top of the evolutionary tree is deeply rooted
in the brain of many people due to traditional, cultural or
religious modes of thinking. This concept was reanalyzed
deeply due to the recent ndings of the damages caused
by the modern civilization to the outer environment and
general public health. Serious ecological catastrophes,
global warming, nuclear waste contamination and
chemical leaks are accompanied by the appearance of
novel important bacterial or viral pathogens, spread of
antibiotic resistance strains and the dramatic increase
in cancer or cardiovascular diseases. All these exo- and
endoecological changes lead to novel modes of thinking
and seeing of the human being as a complex organism
tightly bound to its outer world and its endoecology.
Human civilization witnessed the negative effects of
its own behavior long ago: extensive animal breeding
in the Sahara and destructive and deadly epidemics of
the middle ages in cities with poor sanitary conditions
are the small examples of the importance of equilibrium
between the “outer” nature and “inner” bacterial world.
However, the impact of modern technologies on the
surrounding world and human health surpass all the
previously noticed negative effects. The role of bacteria
as factors inuencing human health has never been fully
understood but was always intuitively acknowledged
by human habits and tradition. Many of the social
restrictions regarding food and diets were and are based
on negative effects of bacterial food contamination or
inability to store certain products properly. In other cases
the benecial health effects of fermented food products
were noticed ages ago and were sometimes considered
sacred. The current review is devoted to the role of the
microbiota in maintaining health and application of
health benecial bacteria in medical practice.
HUMAN MICROBIOTA AS SEEN NOW
The concept of the human microbiota and its role in
human health underwent signicant changes in the eyes of
the scientic community, physicians and common people.
The former attitude of microorganisms as something
alien to humans or even dangerous changed into the
understanding that bacteria (more correct would be the
term “microbiota,” including viruses, bacteria, archaea
and some eukaryotes) are normal and even necessary for
proper functioning of the human organism, populating the
entire body with large a prevalence of microbes in such
loci as the gut, skin, mouth and urogenital system. The
gut is the human organ the most populated by bacteria,
the number of which exceeds by at least by two orders
of magnitude the total number of human body cells [1,
2]. This understanding gradually allowed change the
entire concept of the indigenous microbiota as a vitally
important part of the body and its role in the maintenance
of human health. At present with the advent of new
sequencing technologies and the joint effort of American
and European microbiota analysis programs (Human
Microbiome Project - www.hmpdacc.org and MetaHIT
Corresponding author. Mailing address: Alexander Suvorov, De-
partment of Molecular Microbiology, Institute for Experimental
Medicine, Acad. Pavlov Street 12, Saint-Petersburg 197376, Rus-
sia. E-mail: alexander_suvorov1@hotmail.com
Review
Bioscience of Microbiota, Food and Health Vol. 32 (3), 8191, 2013
A. Suvorov
82
- www.metahit.eu) the composition and the major
dominant bacterial phyla, representing human microbiota
were identied in contrast to the previous studies based
on classical bacteriology [1]. It is established that
bacterial content of human gut microbiota is composed
mainly from Firmicutes, Bacteriodetes, Actinobacteria,
Proteobacteria, Fusobacteria and Archaea with
predominance of Firmicutes and Bacteriodetes [3, 4].
Indigenous gut microbiota tend to form a complex
multispecies biolm covering entire mucus layer with
only few bacterial species reaching the very gut epithelium
[5]. Composition of human microbiota depends on
the diet preferences of the host but also depend on the
individual peculiarities of the host genetics and his/her
innate immune system. Individual microbial content
seems to be stable during the life span remaining as it
was established quite early in life [6]. Interestingly even
the neonates seem to differ by the predominance of either
Bacteroidetes or Bidobacteriaceae [6]. These individual
features change gradually during life, switching from
bidobacteria being predominant in the breast-feeding
period to the dominance of Bacteriodetes and Firmicutes
in the later stages of life [7]. These discoveries agree with
the nding that the normal microbiota in adults, being
highly individual, has a signicant degree of stability and
tends to recover after temporary dysbiotic conditions.
[8]. The signicant amount of data on the microbiota
sequencing followed by bioinformatic analysis allowed
generation of a concept of enterotypes. According to
the suggestion of Arumugam et al. [9], the human gut
microbiome can be partitioned into three enterotypes:
one with the prevalence of Bacteroides, another with
Prevotella and a third that is almost completely afliated
with phylum Firmicutes with slightly higher levels
of Ruminococcus. This distribution was found to be
independent from the diet preferences, body mass index,
race or gender. It implied a host-controlled microbiota
composition. Almost instantly, this concept of the magic
“three” was challenged by other studies, in which the
existence of two or four enterotypes was found [10, 11].
This fairly articial bioinformatics- based approach of
enterotyping humans, boosted dramatically the research
in the eld because it provided the scientic community
for the rst time with a simple and easily accessible tool
for the analysis of the results of studies of microbiota.
It is already clear that these relatively stable microbiota
compositions (two, three or four) are providing similar
solutions for the organism of the human host at the level
of the metobolome.
The functional role of gut microbiota as an additional
vitally important para-/meta-organ is almost impossible
to overestimate. The gut microbiome participates in
almost all metabolisms of incoming nutrients, is involved
in vitamin synthesis, in cholesterol catabolism, shapes
numerous immune reactions related to the innate and
adaptive immunity, and modulates the relationship of the
human being with pathogenic microorganisms [12, 13].
Indigenous bacteria hydrolyse exogenous and
endogenous substrates. Mucins enable them to obtain
an uninterrupted supply of carbon and energy despite
differences in the human diet. In return bacteria produce
short chain fatty acids (such as butyrate), amines, phenols,
indols, and gases [14]. Even the development of immune
system or the brain depends on the host microbiota [15,
16]. It is also established that many gastrointestinal and
somatic diseases develop as result of microbiota changes
(dysbiosis) inicted by the stress, intoxication, radiation
or antibiotic treatment. Dysbiosis, dened as deregulation
of the normal homeostasis of the intestinal microbiota,
is involved in the pathogenesis of various diseases
including (but not limited to) antibiotic-associated
diarrhea (AAD), Clostridium difcile-associated disease
(CDAD), inammatory bowel disease (IBD), acquired
immune deciency syndrome (AIDS) and obesity [17].
Dysbiotic conditions depending on the degree of the
microbiota disturbances either disappear themselves
or transform into different pathologies, which require
specic microbial (probiotic) treatments.
HISTORY OF PROBIOTICS AND
“RUSSIAN CONNECTION”
Most likely, the rst reason why humans started
selecting certain bacterial stocks for their use was the need
for food preservation. When the access to the food was
sporadic, the ability to preserve the aliments in fermented
form was the only way to prevent hunger. Fermented
milk or meats in the form of cheeses or different kinds of
processed meat (Spanish Jamon Serrano as an example)
were able to preserve the nutritious properties of food for
several months. That was vitally important for farmers
and shepherds, allowing them to make distant journeys
and enhancing dissemination of humankind around
the Earth. Natural selection of the best strains allowed
choosing the best strains and those that were most
advantageous regarding the prevention of food spoilage
and preservation of the nutritional food properties.
During the evolution of human societies, some direct
healing properties of lactic acid bacteria (LAB) strains
were selected based on their health benets. Yogurts,
kers, matsoni, kumis, airan and many other fermented
milk products became known and were sometimes
GUT MICROBIOTA, PROBIOTICS, AND HUMAN HEALTH
83
thought to posses mystical powers because of their health
benets and life-extending properties. At the end of the
19th century, Nobel prize winner Ilia Metchnikoff was the
rst to study LAB scientically. Metchnikoff noticed the
correlation between the longevity of Bulgarian shepherds
and their yogurt diet. In the results of his studies he was
the rst to suggest that humans could live signicantly
longer and healthier if they consume benecial bacteria
[18]. This simple idea happened to be quite sound. In
order to nd the bacteria thriving in yogurts, Metchnikoff
isolated several strains of lactobacilli, which he called
Lactobacillus bulgaricus. He proved that it is possible to
make eatable fermented milk products using pure cultures
of L. bulgaricus. According to Metchnikoffs hypothesis,
lactobacilli were eliminating pathogenic toxin- producing
bacteria from the colon - what he considered the main
reason for life shortening. His collaborators at the Pasteur
Institute were also the rst to perform experiments on
germ-free animals, starting gnotobiology as a new branch
of biological science. Metchnikoff was not only the rst
to study bacteria in fermented milk; he also promoted
production of the rst bacterial drug, Lactobacillin, which
was manufactured in Saint Petersburg starting 1912. That
was long before Nissle in 1917 suggested his Escherichia.
coli product wrongfully cited as the rst probiotic [http://
www.probiotics-help.com/mutaor.html].
Metchnikoffs studies were later overshadowed by the
development of antibacterial drugs after the discovery of
antibiotics, and Soviet Union remained the only country
in which scientists continued selecting and analyzing
health benecial strains and certifying them as drugs sold
in pharmacies (Table 1).
Studies of several brilliant Soviet scientists such as
Tsiklinskaia P., Peretz L., Ugolev A., Kiselev P. and
Shenderof B. made a signicant impact in understanding
of the action of health-promoting bacteria in the human
organism and in launch of production of several strains
of health benecial bacteria, belonging to the species of
lactobacilli, enterococci, bidobacteria and E.coli, on an
industrial scale [19–22].
Products containing LAB approved as “drugs” with the
commercial names Lactobacterin, Bidumbacterin and
Colibacterin are still on the market of Russian Federation.
For example, Bidumbacterin a drug containing
bidobacteria was designed in 1966, and industrial
production of it started in 1972 [23]. “Lactobacterin”
(probiotic drug containing Lactobacillus plantarum
strain 8P-A3) production also was stared in early 70s.
The term probiotic meening food or drugs containing life
health benecial bacteria, appeared in world literature
much later, in the 80s, after the revival of interest in these
benecial bacteria [14]. Around that time, a signicant
Table 1. Probiotic drugs and food products produced and distributed in Russian Federation
Species included Name of the product Type of product Company
Bidobacterium bidum No.1 or
Bidobacterium bidum 791
Bidumbacterin
Bidumbacterin forte
Freeze dried powder 10
8
CFU/ml, 10
7
CFU/g
Biomed Metchnikoff JSC, FSUC
“SIC “Microgen”, Patrner LTD
Bidobacterium bidum No.1 +
Lysozym
Bilis Freeze dried powder, 10
6
CFU/ml Ferment, LTD
Lactobacillus plantarum or
Lactobacillus fermentum
Lactobacterin Freeze dried powder, 10
7
CFU/ml, in 10
ml asks, tablets, vaginal suppositories
Biomed Metchnikoff JSC, FSUC
“SIC “Microgen”IM-Bio
Enterococcus faecium L3 Laminolact Bon-bons with contact dried bacteria 10
6
CFU/g in 200g boxes
Avena, LTD
Lactobacillus acidophilus Acilact Vaginal suppositories 10
7
CFU/ml Lekko, LTD
Bacillus cereus IP 5832 Bactisubtil Freeze dried powder, 10
9
CFU/g in
capsules
Aventis Pharma International,
France
Lactobacillus acidophilus D-76,
D-75
Vitaor Freeze dried powder, 10
7
State Institute of Fine pure
Biochemicals
Escherichia coli М-17 Colibacterin Freeze dried powder, 10
7
CFU/ml, in 10
ml asks
FSUC “SIC “Microgen”
Lactobacillus acidophilus,
Bidobacterium infantis,
Enterococcus faecium
Linex Freeze dried powder, 1.2 × 10
7
CFU/g in
capsules
Sandoz, Lec, Slovenia
Bidobacterium bidum bidum
No.1 and Е. coli М-17
Bicol 10
7
CFU/ml, 10
7
CFU/ml in 10 ml asks Biomed Metchnikoff JSC, FSUC
“SIC “Microgen”
Bidobacterium longum
Enterococcus faecium SF68
Biform 10
7
CFU/ml, 10
7
CFU/ml, in capsules Ferrosan, Denmark
A. Suvorov
84
amount of studies has been already accomplished in
the USSR regarding the selection of probiotic strains,
their antagonistic activities, vitamin production and
specic inuence on the intestinal microbiota. The main
health benets of intestinal bacteria such as antagonistic
activities, vitamin production, enzymatic activities and
immunomodulation were postulated by Leonid Peretz
already in 1955 [19].
PROBIOTICS AND THEIR FUNCTIONS IN
THE HOST
Use of probiotics as health benecial products or
ingredients containing live bacteria is huge, and there is a
constantly growing number of different functional foods
and pharmaceuticals.
Most of the commonly used probiotic strains belong to
the group of LAB and bidobacteria. LAB include several
different genera including Streptococcus, Staphylococcus,
Lactococcus, Pediococcus, Lactobacillus, Enterococcus,
Leuconostoc and some others. LAB had acquired the
ability to recognize several sugars, such as for instance
xylose, cellobiose, ribose, arabinose, glucose, and fructose
before they developed the ability to ferment lactose
to lactate. They rstly colonized fruit and vegetable
ecological niches, and later cheese, wine, and especially
milk, which reected their preference for habitats rich
in lactose [24]. Starting with Metchnikoff, studies of
LAB and their use as probiotics have predominantly
focused on the genus of Lactobacillus. Enterococcus-
based probiotics are well represented in the post-Soviet
and Eastern European market and are less common in
Western Europe and the United States. For example,
the Enterococcus-containing drugs Linex and Biform
are comprise more than 80% of the Russian market for
probiotics (www.gidrm.ru/includes/mktng/marketing ).
Among the other probiotic strains, one should mention
bidobacteria as the dominant microbiota in breast-fed
children which are also prominent as components of both:
probiotic drugs and food products. Other probiotics on
the market belong to different species of bacilli, E.coli,
saccharomyces and some clostridial strains [25, 26].
At present time, a large number of relevant clinical
studies with probiotics have been performed and even
analyzed employing meta-analysis. Some of these
studies aimed at treatment of gastrointestinal diseases
Table 2. Some probiotic strains used in clinical practice
Probiotic strain (preparation) Disease References
VSL#3 (Streptococcus thermophilus Ulcerative colitis [59–61]
Bidobacterium breve
Bidobacterium longum
Bidobacterium infantis
Lactobacillus acidophilus
Lactobacillus plantarum
Lactobacillus casei
Lactobacillus bulgaricus)
Escherichia coli Nissle Ulcerative colitis [62]
Lactobacillus GG Ulcerative colitis [63]
VSL#3 Pouchitis [64]
Lactobacillus GG Crohn’s disease [65, 66]
Saccharomyces boulardii Crohn’s disease [67]
Lactobacillus GG Irritable bowel syndrome [68]
Bidobacterium animalis DN-173 010 Irritable bowel syndrome [69]
Bidobacterium infantis 35624 Irritable bowel syndrome [70]
Escherichia coli (DSM17252) Irritable bowel syndrome [71]
Lactobacillus plantarum MF1298 Irritable bowel syndrome [27]
Lactobacillus plantarum 299v Irritable bowel syndrome [72]
Lactobacillus reuteri ATCC 55730 Irritable bowel syndrome [73, 74]
Bidobacterium bidum CECT 7366 Lactobacillus spp H. pylori infection [75, 76]
Enterococcus faecium L3 H. pylori infection [77, 78]
Clostridium butyricum H. pylori infection [79]
GUT MICROBIOTA, PROBIOTICS, AND HUMAN HEALTH
85
such as irritable bowel syndrome, Crohn’s disease,
pouchitis and ulcerative colitis, are listed in Table 2. The
positive outcomes of probiotic treatment in most of the
studies reect the effectiveness of probiotics in clinical
practice. However, the results of treatments employing
different or even the very same probiotic strain vary
from study to study. For example, in the case of irritable
bowel syndrome (IBS) treatment together with studies
demonstrating positive effects of probiotic therapy, some
studies showed no differences compared with the control
or even the aggravation of pathologies [27–30]. In a
recent study on patients with IBS, intake of L. plantarum
MF 1298 was associated with a signicant aggravation of
symptoms, but neither intake of L. plantarum MF 1298
nor symptoms were associated with the composition of
the fecal microbiota [27]. What was most striking in
this respect was results of a clinical study of patients
with acute pancreatitis, in which 16% of patients in the
probiotics group died, compared with 6% in the control
group [31].
This discrepancy in the results of clinical studies
reects the fact that the probiotic bacteria (sometimes
poorly studied) administered to the individual patients
with their own unique microbiota might interact with the
host tissues or their own microbiota in different ways.
Medical doctors and scientists who made decisions
regarding the clinical studies in many cases neglected the
endoecological aspects of introduction of bacteria into the
gut of patients. These possible side effects of microbial
therapy, which have been proved as effective in most
of the studies, are also postulated by Matsushima and
Takagi in the editorial titled “Is it effective?” to “How to
use it?”: the era has changed in probiotics and functional
food products against Helicobacter pylori infection
[32]. However, accurate prediction of the functioning of
probiotics in the gut is impossible without understanding
the physiology of probiotic strains and the mode of their
interactions with the host.
MECHANISMS OF PROBIOTIC ACTION
In numerous reviews describing the use of probiotics,
several features of the strains included into the
preparations were mentioned. Probiotics should be of
human or animal origins depending on their intended
uses. They should have the ability to survive in sufcient
numbers as well as to pass through the gut (bile and acid
tolerant), be safe for consumption, and be adhesive to the
intestinal mucosa. They should exert an antagonistic effect
against pathogens, and interfere with the translocation of
the pathogenic bacteria and modulate the immune system
[14, 27–30, 33]. However, none of the probiotic strains
meet these criteria in full or the studies showing this are
not convincing. First, the relevance of the probiotic strain
to the host is often questionable due to the fact that most of
the historically selected LAB probiotic strains including
Metchnikoff Lactobacillus delbrueckii subsp. bulgaricus
most likely originated from the cattle microbiota. Three
things regarding probiotic functions are most obvious:
antagonistic potential, the inuence of the probiotics on
the process of digestion and immunomodulation.
Antagonistic activity of most probiotic strains can
be studied outside the host, allowing evaluation of the
range of the affected opportunistic/pathogenic bacteria.
Different mechanisms of antibacterial action are involved,
but synthesis of organic acids and antimicrobial peptides
(bacteriocins) are the most common weapons of bacterial
wars for colonization locus and for nutrients. Expression
of many bacteriocins of lactobacilli, enterococci or
bidobacteria is strictly regulated by the complex
genetic regulatory systems involving three-component
signaling and pheromone activation by the quorum
sensing mechanism [34–36]. The majority of bacteriocin-
producing strains generate peptides inhibiting growth
of a narrow range of bacteria with similar colonization
preferences; however, some probiotics such as L.
plantarum 8P-A3 or E. faecium L3 synthesize multiple
bacteriocins with extremely high inhibitory activities
against gram-positive and gram-negative pathogens [35,
36].
Similar effects were determined in studies with
the other bacteriocins, isolated from LAB [37, 38].
Appearance of probiotics in the gut induces noticeable
metabolic effects on the organism such as lowering of the
cholesterol level, vitamin production, diabetes or obesity
[33, 39–41]. However, it is usually difcult to distinguish
the effects of relatively small amounts of bacteria being
introduced into the total microbiome. These reactions are
better monitored in gnotobiotic animals or animals with
articially induced dysbiosis [42]. On the other hand, a
healthy microbiota is usually resistant to colonization by
external microorganisms [43]. Objective evaluation of
the immunomodulatory functions of probiotics presents
similar problems because the tests are usually performed
either on the organisms with established microbiota or
gnotobionts known to have a defective innate immune
system. Both these models have their weaknesses. It
has been established that probiotics do inuence the
innate and adaptive immune functions involving toll-
like receptors (TLRs) and their downstream systems
including NF-κB, JAKSTAT, MAPK, and SAPK/JNK
pathways. These reactions are followed by interleukin
A. Suvorov
86
and defensin differential expression, which can vary
depending on the type of probiotic used. For example,
the most common reactions to probiotic lactobacilli
or enterococci are downregulation of NF-κB and IL-8
expression and induction of IL-10 [16, 44–47]. However,
these effects are very strain dependant. Different strains
belonging to the same species can modulate the immune
response quite differently by helper T (Th1/Th2) cell
polarization.
Another probiotic feature, which has been under
intensive investigation lately, is their inuence on
epithelium integrity. Probiotics belonging to different
species can inuence protein expression in tight junctions
blocking the process of bacterial translocation [48]. These
effects were more visible in the case when the microbiota
of the experimental animals was in an articially induced
dysbiotic condition [48–50].
PROBIOTICS AND SAFETY
Many scientists and especially physicians active in this
eld are considering only lactobacilli or bidobacteria as
safe probiotics meeting generally regarded as safe (GRAS)
criteria. They are completely ignoring the fact that many
probiotics including the GRAS strains bear putative
pathogenicity factors and mobile genetic elements in
their genomes. On the other hand the strains with a long
history of being successfully used as probiotics belonging
to such species as E.coli, enterococci or Bacillus subtilis
are regarded as potentially hazardous. However, this point
of view has nothing to do with microbial ecology or with
common sense and in reality harms the entire concept
of the clinical usage of probiotics. Bacteria being highly
plastic and adaptive to different environments do not
“respect any human moral values” or do not particularly
target the humans. The only thing they can do and will do
is propagate in the presence of appropriate nutrients and
in certain environments. Many strains of Lactobacillus
salivarius used in several probiotic preparations in reality
express a brinogen-binding protein encoded by the gene
CCUG_2371. The presence of this virulence factor in the
strain can cause platelet aggregation facilitating a septic
infection [51]. The most used and studied probiotic
strain, Lactobacillus rhamnosus GG, carries vancomicin
resistance genes and 5 timidly called “genomic islands”
(in other organisms they are named pathogenicity islands
or the PAI) with several bacteriophages and genes for 3
surface expressed LPXTG-like pilins (spaCBA) and a
pilin-dedicated sortase [52]. These genomic ndings are
considered an explanation of the probiotic features of the
strain [52]. However, the very same genetic features in
other species such as enterococci are considered virulence
factors. This is a good example of a pseudoscientic
approach with double standards that has propagated
under the pressure of large industrial corporations selling
certain types of probiotics. On the other hand this mode
of thinking reects a natural desire to follow the pattern
of commonly accepted stereotypes.
AUTOPROBIOTICS AND FECAL
TRANSPLANTATION
It is of general agreement that at least some health
benets of probiotics occur as result of the interactions
of the probiotic strains or strain composition with the
host microbiota. It also established that the benecial
effects of probiotic are most evident under dysbiotic
conditions and are not seen in the healthy microbiota.
Other solutions for restoring the microbiota back to
normal are fecal transplantation or autoprobiotic therapy.
Fecal transplantation is a medical procedure based on the
replacement of the host microbiota with the microbiota
of a donor. This procedure had been evaluated in several
clinical studies on patients with inammatory bowel
disease (IBD) or for the treatment of Clostridium difcile
infection [53, 54]. Besides being fairly unhealthy way
to introduce bacterial biomass (through the nose or the
rectum), this approach has Achilles’ heels such as the
donor microbiota, which may carry opportunistic bacteria
able to cause problems in the treated patient. In our
previous study of healthy individuals, about 50% of the
indigenous enterococci carried several putative virulence
factors in their genome [55]. Also, the enterococci are
clearly not the most dangerous bacteria in the gut.
Another approach is based on the indigenous bacteria
used for restoring the normal microbiota in the case
of a dysbiotic condition [20]. This approach, named
as autoprobiotic technology, can be based on LAB or
bidobacteria previously stored in cryobanks, isolation of
individual strains from the microbiota and returning the
bacteria back into the gut after propagating them outside
the organism, allowing analysis of each individual strain
and return of it to the host. Usually it takes a week to
prepare autoprobiotic yogurt for the patient. In our
clinical studies of patients with IBS, ulcerative colitis
and pneumonia autoprobiotics introduced to patients by
employing a randomized placebo-controlled approach
provided signicant positive effects as judged from the
majority of clinical parameters and life quality [56].
GUT MICROBIOTA, PROBIOTICS, AND HUMAN HEALTH
87
CONCLUSIONS
Contemporary science is collecting more and more
data regarding the human microbiota, which functions as
an important “organ” tightly bound to the other organs
of the body. Previous dogmas of clinical microbiology,
which were trying to divide the microbial world into
hazardous and benecial microorganisms, are questioned
by the new genomic and metabolomic data. The
contemporary crisis of pharmacology being unable to
produce and bring new antibiotics into the market [57]
is giving human race a chance to see the problem of
human health from the level of microecology, moving
away from the simple eradication strategy. The emotional
appeal of Blaser, “Stop the killing of benecial bacteria,”
needs to attract more attention from the scientic and
medical community [58]. It is obvious that the tight
systemic links between the microbiota and the cells of
the human body are highly individualized and need to
be restored when the microbiota changes due to various
reasons, with antibiotic treatments being number one.
Dysbiotic conditions lay underneath many infectious and
somatic diseases of our contemporaries. It is obvious that
microbial therapy should be much better implemented
in the arsenal of medical doctors; however, a signicant
amount of studies needs to be done before this kind of
therapy will become really common.
Despite the great number of different probiotics
on the world market and permanently growing sales
of probiotics, there is no agreement in the scientic
community regarding their mode of functioning and
interpretation of the results of the clinical studies. The
main reason for this is simply based on the lack of the
relevant studies and extremely complex microbiota of
each individual. There is no common agreement on the
expected features or the composition of probiotic strains.
Only several things about probiotics are obvious: we want
them to pass alive to the target locus of the organism,
interact with the host microbiota and the host immune
system and they should not cause an infection.
On the other hand there are a lot of things they are
supposed to do: they supposedly must deplete a number
of opportunistic bacteria, somehow modulate the immune
system, most likely consume internal nutrients and
produce their own metabolites, strengthen the epithelial
barriers, colonize sites in the organism or disappear from
the host. There is no agreement: regarding the issues of
the preferred period of colonization, ability of probiotics
to adhere to the host epithelium, afliation of probiotics
to the indigenous human microbiota, and the features
regarding the safety of the probiotic strains. Most of these
issues of scientic disagreement are being minor but at
rst glance require clarication. For example, the ability
to colonize the epithelium in bacteria is often correlated
with the presence of the adhesins, which are considered
virulence factors on bacterial surfaces. Thus the presence
of the adhesions or mbriae on the surface of the probiotic
bacteria can be judged differently.
There is no agreement regarding the preferred time of
colonization and very limited data on monitoring the fate
of probiotic strains inside the organism. The preferred
dosage of probiotic bacteria is not clear too. Most likely,
the optimal amount of consumed probiotic bacteria is
strain specic and depends on the survival of the probiotic
bacteria in the host.
It is unclear what is better: one probiotic strain or a
multistrain composition. The interrelationship between
the strains of such probiotic compositions is the mostly
poorly studied. In any case, the more alien strains are
introduced into the gut, the more chances there are that
one of the members of the consortiums will cause an
unpredicted reaction.
In this respect the idea using indigenous strains as
probiotics looks quite attractive. Autoprobiotic strains
have better chances relative to probiotics to colonize the
host and thus normalize the host microbiota. However,
autoprobiotics as medical therapies require further study.
In spite of the obstacles and the problems with
microbial therapy stated in the present overview, the
body of evidence concerning the use of probiotics in
medicine is substantial, and better solutions for returning
the individual microbiota back to normal are not on the
horizon.
ACKNOWLEDGEMENTS
I am thankful to my colleagues for constructive advise
regarding this review.
Major help was provided by Thomas Haertle from INRA,
France, who kindly edited the text and gave many important
critical remarks. I am also thankful to my Russian colleagues
Ekaterina Kisseleva from the Institute of Experimental
Medicine, Galina Alechina from Avena Ltd and Vetchaslav
Melnikov from ITSC for the constructive criticism and
some suggestions. Some experimental data sited in the
review were collected in the Department of Molecular
microbiology, Institute Experimental Medicine Russia. This
work was supported by RFBR grant 10-04-00750a.
REFERENCES
1. Tannock G., editor. 1995. Normal Microora. Chapman
& Hall, London.
2. Zoetendal EG, Vaughan E, de Vos W. 2006. A microbial
A. Suvorov
88
world within us MicroReview. Mol Microbiol 59:
1639–1650. [Medline] [CrossRef]
3. Available at http://mbio.asm.org/content/3/5/e00376-
12.long (accessed 2012-10-23)
4. Van den Abbeele P, Grootaert C, Marzorati M,
Possemiers S, Verstraete W, Ge’rard P, Rabot S, Bruneau
A, El Aidy S, Derrien M, Zoetendal E, Kleerebezem M,
Smidt H, Van de Wiele T. 2010. Microbial community
development in a dynamic gut model is reproducible,
colon region specic, and selective for Bacteroidetes
and Clostridium cluster IX. Appl Environ Microbiol 76:
5237–5246.
5. Swidsinski A, Loening-Baucke V, Lochs H, Hale
L. 2005. Spatial organization of bacterial ora in
normal and inamed intestine: a uorescence in situ
hybridization study in mice. World J Gastroenterol 11:
1131–1140. [Medline]
6. Jost T, Lacroix C, Braegger C, Chassard C. 2012. New
insights in gut microbiota establishment in healthy
breast fed neonates. PLoS One 7:e44595. Available
at: http://www.ncbi.nlm.nih.gov/pubmed/22957008
(accessed 2012-08-30).
7. Biagi E, Nylund L, Candela M, Ostan R, Bucci L, Pini
E, Nikkı J, Monti D, Satokari R, Franceschi C, Brigidi
P, De Vos W. 2010. Through Ageing, and Beyond: Gut
Microbiota and Inammatory Status in Seniors and
Centenarians PLoS ONE 5:e10667. Available at: http://
www.ncbi.nlm.nih.gov/pmc/articles/PMC2871786/
(accessed 2010-05-17)
8. Budding AE, Grasman M, Lin F, Bogaards J,
Soeltan-Kaersenhout D, Vandenbroucke-Grauls C,
van Bodegraven A, Savelkoul P. 2010. IS-pro: high-
throughput molecular ngerprinting of the intestinal
microbiota. FASEB J 24: 4556–4564. [Medline]
[CrossRef]
9. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada
T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM,
Bertalan M, Borruel N, Casellas F, Fernandez L, Gautier
L, Hansen T, Hattori M, Hayashi T, Kleerebezem
M, Kurokawa K, Leclerc M, Levenez F, Manichanh
C, Nielsen HB, Nielsen T, Pons N, Poulain J, Qin J,
Sicheritz-Ponten T, Tims S, Torrents D, Ugarte E,
Zoetendal EG, Wang J, Guarner F, Pedersen O, de Vos
WM, Brunak S, Doré J, MetaHIT Consortium, Antolín
M, Artiguenave F, Blottiere HM, Almeida M, Brechot C,
Cara C, Chervaux C, Cultrone A, Delorme C, Denariaz
G, Dervyn R, Foerstner KU, Friss C, van de Guchte M,
Guedon E, Haimet F, Huber W, van Hylckama-Vlieg J,
Jamet A, Juste C, Kaci G, Knol J, Lakhdari O, Layec
S, Le Roux K, Maguin E, Mérieux A, Melo Minardi
R, M’rini C, Muller J, Oozeer R, Parkhill J, Renault
P, Rescigno M, Sanchez N, Sunagawa S, Torrejon A,
Turner K, Vandemeulebrouck G, Varela E, Winogradsky
Y, Zeller G, Weissenbach J, Ehrlich SD, Bork P. 2011.
Enterotypes of the human gut microbiome. Nature 473:
174–180.
10. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen Y,
Keilbaugh S, Bewtra M, Knights D, Walters WA, Knight
R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel
L, Li H, Bushman F, Lewis J. 2011. Linking long-term
dietary patterns with gut microbial enterotypes. Science
334: 105–108. [Medline] [CrossRef]
11. Holmes I, Harris K, Quince C. 2012. Dirichlet
multinomial mixtures: generative models for microbial
metagenomics. PLoS One 7(2):e30126. Available
at: http://www.ncbi.nlm.nih.gov/pubmed/22319561
(accessed 2012-02-3)
12. Pessione E. 2012. Lactic acid bacteria contribution to
gut microbiota complexity: lights and shadows. Front
Cell Infect Microbiol 2: 86. [Medline] [CrossRef]
13. Kumar M, Nagpal R, Kumar R, Hemalatha R, Verma
V, Kumar A, Chakraborty C, Singh B, Marotta F, Jain
S, Yadav H. 2012. Cholesterol-lowering probiotics as
potential biotherapeutics for metabolic diseases. Exp
Diabetes Res 2012: 902917. [Medline] [CrossRef]
14. Tannock GW. 2010. The bowel microbiota and
inammatory bowel diseases. Int J Inam 5:954051.
Available at: http://www.ncbi.nlm.nih.gov/pmc/articles/
PMC3004003/ (accessed 2010-08-5)
15. Manco M. 2012. Gut microbiota and developmental
programming of the brain: from evidence in behavioral
endophenotypes to novel perspective in obesity. Front
Cell Infect Microbiol 2: 109. [Medline] [CrossRef]
16. Hooper LV, Littman D, Macpherson A. 2012.
Interactions between the microbiota and the immune
system. Science 336: 1268–1273. [Medline] [CrossRef]
17. Malo MS, Alam S, Mostafa G, Zeller S, Johnson P,
Mohammad N, Chen K, Moss A, Ramasamy S, Faruqui
A, Hodin S, Malo P, Ebrahimi F, Biswas B, Narisawa
S, Millán J, Warren H, Kaplan J, Kitts C, Hohmann
E, Hodin R. 2010. Intestinal alkaline phosphatase
preserves the normal homeostasis of gut microbiota.
Gut 59: 1476–1484. [Medline] [CrossRef]
18. Metchnikoff E. 1908. The prolongation of the life.
Optimistic study. G.P. Putnam’s sons, New York and
London.
19. Peretz L. 1955. Role of the normal microbiota for the
organism of human, Medgis, Moscow (in Russian).
20. Shenderov B., editor. 2011. Probiotics and functional
foods, in Food Engineering, Eolss Publishers, Oxford.
21. Ugolev AM, Ivashkin V. 1992. Theory of universal
functional blocks and fundamental biomedical
problems. Klin Med (Mosk) 70: 8–14 (in Russian).
[Medline]
22. Kiselev PN, Shutko T. 1968. On mechanisms of the
cellular self-defence against the action of microbial
toxins. Tsitologiia 10: 1068–1073 (in Russian).
[Medline]
23. Aleshkin V, Amerhanova A, Pospelova V, Afanasiev
S, Shenderov B. 2008. History, present situation,
and prospects of probiotic research conducted in the
G.N. Gabrichevsky Institute for Epidemiology and
GUT MICROBIOTA, PROBIOTICS, AND HUMAN HEALTH
89
Microbiology. Microbial Ecology in Health and Disease
20: 113–115. [CrossRef]
24. Carr FJ, Chill D, Maida N. 2002. The lactic acid
bacteria: a literature survey. Crit Rev Microbiol 28:
281–370. [Medline] [CrossRef]
25. Hempel S, Newberry S, Maher A, Wang Z, Miles J,
Shanman R, Johnsen B, Shekelle P. 2012. Probiotics for
the prevention and treatment of antibiotic-associated
diarrhea: a systematic review and meta-analysis. JAMA
307: 1959–1969. [Medline] [CrossRef]
26. Woo TD, Oka K, Takahashi M, Hojo F, Osaki T, Hanawa
T, Kurata S, Yonezawa H, Kamiya S. 2011. Inhibition
of the cytotoxic effect of Clostridium difcile in vitro
by Clostridium butyricum MIYAIRI 588 strain. J Med
Microbiol 60: 1617–1625. [Medline] [CrossRef]
27. Ritchie M, Romanuk T. 2012. A Meta-Analysis of
probiotic efcacy for gastrointestinal diseases. PLoS
One. 7: e34938. Available at: http://www.ncbi.nlm.nih.
gov/pubmed/22529959 (accessed 2012-04-18)
28. Farup PG, Jacobsen M, Ligaarden SC, Rudi K. 2012.
Probiotics, symptoms, and gut microbiota: what are
the relations? A randomized controlled trial in subjects
with irritable bowel syndrome. Gastroenterol Res Pract
2012:214102. Available at: http://www.ncbi.nlm.nih.
gov/pmc/articles/PMC3415104/ (accessed 2012-07-31)
29. Lee BJ, Bak Y. 2011. Irritable bowel syndrome, gut
microbiota and probiotics. J Neurogastroenterol Motil
17: 252–266. [Medline] [CrossRef]
30. McFarland LV, Dublin S. 2008. Meta-analysis of
probiotics for the treatment of irritable bowel syndrome.
World J Gastroenterol 14: 2650–2661. [Medline]
[CrossRef]
31. Besselink MG, van Santvoort H, Buskens E, Boermeester
M, van Goor H, Timmerman H, Nieuwenhuijs V, Bollen
T, van Ramshorst B, Witteman B, Rosman C, Ploeg R,
Brink M, Schaapherder A, Dejong C, Wahab P, van
Laarhoven C, van der Harst E, van Eijck C, Cuesta M,
Akkermans L, Gooszen H, Dutch Acute Pancreatitis
Study Group 2008. Probiotic prophylaxis in predicted
severe acute pancreatitis: a randomised, double-
blind, placebo-controlled trial. Lancet 371: 651–659.
[Medline] [CrossRef]
32. Matsushima M, Takagi AJ. 2012. “Is it effective?” to
“How to use it?”: the era has changed in probiotics
and functional food products against Helicobacter
pylori infection. J Gastroenterol Hepatol 27: 851–853.
[Medline] [CrossRef]
33. Wallace TC, Guarner F, Madsen K, Cabana M, Gibson
G, Hentges E, Sanders ME. 2011. Human gut microbiota
and its relationship to health and disease. Nutr Rev 69:
392–403. [Medline] [CrossRef]
34. Dobson A, Cotter P, Ross RP, Hill C. 2012. Bacteriocin
production: a probiotic trait? Appl Environ Microbiol
78: 1–6. [Medline] [CrossRef]
35. Tsapieva A, Duplik N, Suvorov A. 2011. Structure of
plantaricin locus of Lactobacillus plantarum 8P-A3.
Benef Microbes 2: 255–261. [Medline] [CrossRef]
36. Yermolenko E, Kolobov A, Chernysh A, Suvorov
A. 2011. Inuence of synthetic peptide inductors
on antibacterial activity of enterococci. Benecial
Microbes 1: 253–257.
37. Batdorj B, Dalgalarrondo M, Choiset Y, Pedroche
J, Métro F, Prévost H, Chobert JM, Haertlé T. 2006.
Purication and characterization of two bacteriocins
produced by lactic acid bacteria isolated from
Mongolian airag. J Appl Microbiol 101: 837–848.
[Medline] [CrossRef]
38. Atanassova M, Choiset Y, Dalgalarrondo M, Chobert
JM, Dousset X, Ivanova I, Haertlé T. 2003. Isolation and
partial biochemical characterization of a proteinaceous
anti-yeast compound produced by Lactobacillus
paracasei subsp. paracasei strain M3 from Bulgarian
yellow cheese. Int J Food Microbiol 87: 63–73.
[Medline] [CrossRef]
39. Machado MV, Cortez-Pinto H. 2012. Gut microbiota
and nonalcoholic fatty liver disease. Ann Hepatol 11:
440–449. [Medline]
40. Nakamura Y, Omaye S. 2012. Metabolic diseases and
pro- and prebiotics: Mechanistic insights. Nutr Metab
(Lond) 19:60 Available at: http://www.ncbi.nlm.nih.
gov/pmc/articles/PMC3464869/ (accessed 2012-06-19)
41. Cani PD, Delzenne N. 2009. Interplay between obesity
and associated metabolic disorders: new insights into
the gut microbiota. Curr Opin Pharmacol 9: 737–743.
[Medline] [CrossRef]
42. Macho Fernandez E, Valenti V, Rockel C, Hermann C,
Pot B, Boneca IG, Grangette C. 2011. Antiinammatory
capacity of selected lactobacilli in experimental colitis
is driven by NOD2-mediated recognition of a specic
peptidoglycan derived muropeptide. Gut 60: 1050–
1059. [Medline] [CrossRef]
43. Mangalat N, Liu Y, Fatheree NY, Ferris MJ, Van
Arsdall MR, Chen Z, Rahbar MH, Gleason WA, Norori
J, Tran DQ, Rhoads JM. 2012. Safety and tolerability
of Lactobacillus reuteri DSM 17938 and Effects on
biomarkers in healthy adults: results from a randomized
masked trial. PLoS One 7:e43910. Available at: http://
www.ncbi.nlm.nih.gov/pmc/articles/PMC3435331/
(accessed 2012-09-6)
44. Petrof E, Claud E, Sun J, Abramova T, Guo Y, Waypa
T, He S, Nakagawa Y, Chang E. 2009. Bacteria-free
solution derived from Lactobacillus plantarum inhibits
multiple NFkappaB pathways and inhibits proteasome
function. Inamm Bowel Dis 15: 1537–1547.
[CrossRef]
45. Schlee M, Harder J, Köten B, Stange E, Wehkamp J,
Fellermann K. 2008. Probiotic lactobacilli and VSL#3
induce enterocyte β-defensin 2. Clin Exp Immunol 151:
528–535. [Medline] [CrossRef]
46. Yoon SS, Sun J. 2011. Probiotics, nuclear receptor
signaling, and anti-inammatory pathways.
Gastroenterol Res Pract 2011: 971–938. [Medline]
A. Suvorov
90
47. Tarasova E, Yermolenko E, Donets V, Sundukova Z,
Bochkareva A, Borschev I, Suvorova M, Ilyasov I,
Simanenkov V, Suvorov A. 2010. The inuence of
probiotic enetrococci on the microbiota and cytokines
expression in rats with dysbiosis induced by antibiotics.
Benecial Microbes 1: 265–270. [Medline] [CrossRef]
48. Khailova L, Dvorak K, Arganbright KM, Halpern MD,
Kinouchi T, Yajima M, Dvorak B. 2009. Bidobacterium
bidum improves intestinal integrity in a rat model of
necrotizing enterocolitis. Am J Physiol Gastrointest
Liver Physiol 297: G940–G949. [Medline] [CrossRef]
49. Mennigen R, Nolte K, Rijcken E, Utech M, Loeer
B, Senninger N, Brewer M. 2009. Probiotic mixture
VSL#3 protects the epithelial barrier by maintaining
tight junction protein expression and preventing
apoptosis in a murine model of colitis. Am J Physiol
296: 1140–1149.
50. Ukena S, Singh A, Dringenberg U, Engelhardt R, Seidler
U, Hansen W, Bleich A, Bruder D, Franzke A, Rogler
G, Suerbaum S, Buer J, Gunzer F, Westendorf AM.
2007. Probiotic Escherichia coli Nissle 1917 inhibits
leaky gut by enhancing mucosal integrity,” PLoS ONE,
2: IDe1308. Available at: http://www.ncbi.nlm.nih.gov/
pmc/articles/PMC2110898/ (accessed 2007-12-12)
51. Collins J, van Pijkeren J, Svensson L, Claesson
M, Sturme M, Li Y, Cooney J, van Sinderen D,
Walker AW, Parkhill J, Shannon O, O’Toole P. 2012.
Fibrinogen-binding and platelet-aggregation activities
of a Lactobacillus salivarius septicaemia isolate are
mediated by a novel brinogen-binding protein. Mol
Microbiol 85: 862–877. [Medline] [CrossRef]
52. Kankainen M, Paulin L, Tynkkynen S, von Ossowski
I, Reunanen J, Partanen P, Satokari R, Vesterlund
S, Hendrickx AP, Lebeer S, De Keersmaecker SC,
Vanderleyden J, Hämäläinen T, Laukkanen S, Salovuori
N, Ritari J, Alatalo E, Korpela R, Mattila-Sandholm
T, Lassig A, Hatakka K, Kinnunen KT, Karjalainen H,
Saxelin M, Laakso K, Surakka A, Palva A, Salusjärvi
T, Auvinen P, de Vos WM. 2009. Comparative genomic
analysis of Lactobacillus rhamnosus GG reveals pili
containing a human- mucus binding protein. Proc
Natl Acad Sci USA 106: 17193–17198. [Medline]
[CrossRef]
53. Damman CJ, Miller SI, Surawicz CM, Zisman TL.
2012. The microbiome and inammatory bowel
disease: is there a therapeutic role for fecal microbiota
transplantation? Am J Gastroenterol 107: 1452–1459.
[Medline] [CrossRef]
54. Brandt LJ. 2012. Fecal transplantation for the treatment
of Clostridium difcile infection. Gastroenterol Hepatol
N Y 8: 191–194. [Medline]
55. Bondarenko V, Suvorov A., editors. 2007. Symbiotic
enterococci and the problem of enterococcal infection,
Sandoz, Moscow (in Russian).
56. Suvorov A, Simanenkov V, Gromova L, Kolodjieva V,
Tsapieva A, Chernish A, Solovieva O, Ermolenko E.
2011. Enterococci as probiotics or autoprobiotics. In
Prebiotics and probiotics potential for human health,
Ivanova I (ed), Paisi Hilendarski, Soa, pp. 104–112.
57. Projan SJ. 2003. Why is big Pharma getting out of
antibacterial drug discovery? Curr Opin Microbiol 6:
427–430. [Medline] [CrossRef]
58. Blaser M. 2011. Antibiotic overuse: Stop the killing of
benecial bacteria. Nature 476: 393–394. [Medline]
[CrossRef]
59. Tursi A, Brandimarte G, Papa A, Giglio A, Elisei W,
Giorgetti GM, Forti G, Morini S, Hassan C, Pistoia MA,
Modeo ME, Rodino’ S, D’Amico T, Sebkova L, Sacca’
N, Di Giulio E, Luzza F, Imeneo M, Larussa T, Di Rosa
S, Annese V, Danese S, Gasbarrini A. 2010. Treatment
of relapsing mild-to-moderate ulcerative colitis with
the probiotic VSL#3 as adjunctive to a standard
pharmaceutical treatment: a double-blind, randomized,
placebo-controlled study. Am J Gastroenterol 105:
2218–2227. [Medline] [CrossRef]
60. Miele E, Pascarella F, Giannetti E, Quaglietta L,
Baldassano RN, Staiano A. 2009. Effect of a probiotic
preparation (VSL#3) on induction and maintenance
of remission in children with ulcerative colitis. Am J
Gastroenterol 104: 437–443. [Medline] [CrossRef]
61. Bibiloni R, Fedorak R, Tannock G, Madsen K,
Gionchetti P, Campieri M, De Simone C, Sartor R. 2005.
VSL#3 probiotic-mixture induces remission in patients
with active ulcerative colitis. Am J Gastroenterol 100:
1539–1546. [Medline] [CrossRef]
62. Adam B, Liebregts T, Holtmann G. 2006. Maintaining
remission of ulcerative colitis with the probiotic
Escherichia coli Nissle 1917 is as effective as with
standard mesalazine. Z Gastroenterol 44: 267–269.
[Medline] [CrossRef]
63. Zocco MA, dal Verme L, Cremonini F, Piscaglia A,
Nista E, Candelli M, Novi M, Rigante D, Cazzato IA,
Ojetti V, Armuzzi A, Gasbarrini G, Gasbarrini A. 2006.
Efcacy of Lactobacillus GG in maintaining remission
of ulcerative colitis. Aliment Pharmacol Ther 23:
1567–1574. [Medline] [CrossRef]
64. Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber
S, Talbot IC, Nicholls RJ, Gionchetti P, Campieri M,
Kamm MA. 2004. Once daily high dose probiotic
therapy (VSL#3) for maintaining remission in recurrent
or refractory pouchitis. Gut 53: 108–114. [Medline]
[CrossRef]
65. Prantera C, Scribano M, Falasco G, Andreoli A, Luzi
C. 2002. Ineffectiveness of probiotics in preventing
recurrence after curative resection for Crohn’s disease:
a randomized controlled trial with Lactobacillus GG.
Gut 51: 405–409. [Medline] [CrossRef]
66. Bousvaros A, Guandalini S, Baldassano RN, Botelho C,
Evans J, Ferry GD, Goldin B, Hartigan L, Kugathasan
S, Levy J, Murray KF, Oliva-Hemker M, Rosh JR, Tolia
V, Zholudev A, Vanderhoof JA, Hibberd PL. 2005. A
randomized, double-blind trial of Lactobacillus GG
GUT MICROBIOTA, PROBIOTICS, AND HUMAN HEALTH
91
versus placebo in addition to standard maintenance
therapy for children with Crohn’s disease inammatory
bowel diseases. Inamm Bowel Dis 11: 833–839.
[Medline] [CrossRef]
67. Garcia Vilela E, Ferrari M, Torres H, Guerra Pinto A,
Carolina Carneiro Aguirre A, Paiva Martins F, Marcos
Andrade Goulart E, Sales Da Cunha A. 2008. Inuence of
Saccharomyces boulardii on the intestinal permeability
of patients with Crohn’s disease in remission. Scand J
Gastroenterol 43: 842–848. [Medline] [CrossRef]
68. Bausserman M, Michail S. 2005. The use of
Lactobacillus GG in irritable bowel syndrome in
children: a double-blind randomized control trial. J
Pediatr 147: 197–201. [Medline] [CrossRef]
69. Guyonnet D, Chassany O, Ducrotte P, Picard C, Mouret
M, Mercier CH, Matuchansky C. 2007. Effect of a
fermented milk containing Bidobacterium animalis
DN-173 010 on the health-related quality of life and
symptoms in irritable bowel syndrome in adults in
primary care: a multicentre, randomized, double blind,
controlled trial. Aliment Pharmacol Ther 26: 475–486.
[Medline] [CrossRef]
70. Whorwell PJ, Altringer L, Morel J, Bond Y,
Charbonneau D, O’Mahony L, Kiely B, Shanahan
F, Quigley EM. 2006. Efcacy of an encapsulated
probiotic Bidobacterium infantis 35624 in women
with irritable bowel syndrome. Am J Gastroenterol 101:
1581–1590. [Medline] [CrossRef]
71. Enck P, Zimmermann K, Menke G, Klosterhalfen S.
2009. Randomized controlled treatment trial of irritable
bowel syndrome with a probiotic E. coli preparation
(DSM17252) compared to placebo. Z Gastroenterol 47:
209–214. [Medline] [CrossRef]
72. Sen S, Mullan M, Parker T, Woolner J, Tarry S, Hunter
J. 2002. Effect of Lactobacillus plantarum 299v on
colonic fermentation and symptoms of irritable bowel
syndrome. Dig Dis Sci 47: 2615–2620. [Medline]
[CrossRef]
73. Niv E, Naftali T, Hallak R, Vaisman N. 2005. The
efcacy of Lactobacillus reuteri ATCC 55730 in the
treatment of patients with irritable bowel syndrome - a
double blind, placebo-controlled, randomized study.
Clin Nutr 24: 925–931. [Medline] [CrossRef]
74. Drouault-Holowacz S, Bieuvelet S, Burckel A,
Cazaubiel M, Dray X, Marteau P. 2008. A double blind
randomized controlled trial of a probiotic combination
in 100 patients with irritable bowel syndrome.
Gastroenterol Clin Biol 32: 147–152. [Medline]
[CrossRef]
75. Chenoll E, Casinos B, Bataller E, Astals P, Echevarría
J, Iglesias JR, Balbarie P, Ramón D, Genovés S.
2011. Novel probiotic Bidobacterium bidum CECT
7366 strain active against the pathogenic bacterium
Helicobacter pylori. Appl Environ Microbiol 77:
1335–1343. [Medline] [CrossRef]
76. García CA, Henríquez A, Retamal R, Pineda C,
Delgado Sen C, González C. 2009. Probiotic properties
of Lactobacillus spp isolated from gastric biopsies
of Helicobacter pylori infected and non-infected
individuals. Rev Med Chil 137: 369–376. [Medline]
77. Zakharova NV. 2006. Methods for increasing the
efcacy and safety of Helicobacter pylori eradication
regimens. Eksp Klin Gastroenterol 4: 59–66 (in
Russian). [Medline]
78. Baryshnikova NV, Suvorov A, Tkachenko E, Uspenskiĭ
IU. 2009. The role of genetic features of Helicobacter
pylori in pathogenesis of digestive system diseases:
from theory to practice. Eksp Klin Gastroenterol 1:
12–19 (in Russian). [Medline]
79. Imase K, Takahashi M, Tanaka A, Tokunaga K, Sugano
H, Tanaka M, Ishida H, Kamiya S, Takahashi S.
2008. Efcacy of Clostridium butyricum preparation
concomitantly with Helicobacter pylori eradication
therapy in relation to changes in the intestinal
microbiota. Microbiol Immunol 52: 156–161. [Medline]
[CrossRef]
... The application of autoprobiotics as a personalized functional food product (PFFP) represents an innovative therapeutic approach with several advantages over the use of commercial probiotics. Key advantages of employing indigenous non-pathogenic strains of lactobacilli, bifidobacteria, and enterococci include biological compatibility with other components of the host microbiota, adaptation to the body's living conditions, minimal immune system burden, and positive effects on digestion [30]. ...
... Enterococcal strains have been used as probiotics in many countries. Examples include Linex (LEK, Slovenia), Bifiform (Ferrosan, Denmark), Symbioflor 1 (Symbio Pharm, Germany), and Laminolakt (Avena, Russia) [30,31]. Enterococci being the common part of indigenous human microbiota, numerous fermented food products and probiotics presently are often positioned as health-threatening bacterial pathogens. ...
... For the first time, we used aut biotics for CRC therapy. They are characterized by greater safety (autoprobiotics c stored in the patient's body for a longer time without entering into conflict with th crobiota, they are protected by the immune system with immunological tolerance they adapt to the conditions of coexistence as part of a consortium of microorganism habitual living conditions) [30]. Previously, we conducted a study on the effect of probiotic strains of E. faecium, bifidobacteria, lactobacilli, and their mixtures on the m of experimental antibiotic-associated intestinal dysbiosis in Wistar rats. ...
Article
Full-text available
Despite great advances in the treatment of oncological diseases, the development of medical technologies to prevent or reduce complications of therapy, in particular, those associated with surgery and the introduction of antibiotics, remains relevant. The aim of this study is to evaluate the effectiveness of the use of autoprobiotics based on indigenous non-pathogenic strains of Enterococcus faecium and Enterococcus hirae as a personalized functional food product (PFFP) in the complex therapy of colorectal cancer (CRC) in the early postoperative period. A total of 36 patients diagnosed with CRC were enrolled in the study. Study group A comprised 24 CRC patients who received autoprobiotic therapy in the early postoperative period, while the control group C included 12 CRC patients without autoprobiotic therapy. Prior to surgery and between days 14 and 16 post-surgery, comprehensive evaluations were conducted on all patients, encompassing the following: stool and gastroenterological complaints analysis, examination of the gut microbiota (bacteriological study, quantitative polymerase chain reaction, metagenome analysis), and analysis of interleukins in the serum. Results: The use of autoprobiotics led to a decrease in dyspeptic complaints after surgery. It was also associated with the absence of postoperative complications, did not cause any side effects, and led to a decrease in the level of pro-inflammatory cytokines (IL-6 and IL-18) in the blood serum. The use of autoprobiotics led to positive changes in the structure of escherichia and enterococci populations, the elimination of Parvomonas micra and Fusobacterium nucleatum, and a decrease in the quantitative content of Clostridium perfringens and Akkermansia muciniphila. Metagenomic analysis (16S rRNA) revealed an increase in alpha diversity. Conclusion: The introduction of autoprobiotics in the postoperative period is a highly effective and safe approach in the complex treatment of CRC. Future studies will allow the discovery of additional fine mechanisms of autoprobiotic therapy and its impact on the digestive, immune, endocrine, and neural systems.
... The count of viable probiotic microorganisms in synbiotics must be 6-8 Log CFU/mL to improve the balance in gut microbiota which will improve the symptoms of various kinds of metabolic disorders such as diabetes mellitus (DM), cardiovascular diseases (CVD), irritating bowel disease (IBD), neurodegenerative disease (ND) [31]. Probiotics must specifically be securely supplied to the intestine, where the microbiota are found and affect human health [54]. It has been difficult to safely deliver probiotics across the gastric acidic and enzymatic barriers to the intestine. ...
... Experimental studies utilizing autoprobiotics have demonstrated that in rats with antibiotic-associated dysbiosis, the administration of different indigenous strains of bacteria (such as bifidobacteria, enterococci, or a bacterial mixture) resulted in a rapid restoration of the microbiota compared to untreated animals [86,90]. Several clinical studies have already shown the effectiveness of autoprobiotics based on indigenous strains of Lactobacillus spp. in restoring and stabilizing the levels of key representatives of the normal intestinal microbiota (such as Bifidobacterium spp., Lactobacillus spp., and E. coli) in dysbiotic disorders caused by antibiotic usage [91][92][93], as well as in infection and inflammatory diseases [86,90,94]. The efficacy of monotherapy using autoprobiotics based on indigenous non-pathogenic enterococci has been demonstrated in the treatment of chronic gastritis associated with H. pylori: The eradication rate was 80%, and there was 100% relief of symptoms after a 20-d autoprobiotic treatment [95]. ...
Article
Full-text available
The article discusses various approaches for probiotic treatment of Helicobacter pylori (H. pylori) infection: Probiotics as an adjuvant treatment in the standard eradication therapy; probiotic strains as a monotherapy; and autoprobiotics as a monotherapy. Autoprobiotics refer to indigenous bifidobacteria, lactobacilli, or enterococci isolated from a specific individual, intended to restore his/her microbiota and improve his/her health. The potential mechanisms of probiotic action against H. pylori include correction of the gut microbiota, immunological effects (enhancement of humoral and cellular immunity, and reduction of oxidative stress), direct antagonistic effects against H. pylori (such as colonization resistance and bacteriocin synthesis), and stimulation of local immunological protection (strengthening of the mucous protective barrier and reduction of gastric mucosa inflammation). The incorporation of probiotics into comprehensive eradication therapy shows promise in optimizing the treatment of H. pylori infection. Probiotics can enhance the eradication rates of H. pylori, reduce the occurrence and severity of side effects, and improve patient compliance. Probiotic or autoprobiotic monotherapy can be considered as an alternative treatment approach in cases of allergic reactions and insufficient effectiveness of antibiotics. We recommend including probiotics as adjunctive medications in anti-H. pylori regimens. However, further randomized multicenter studies are necessary to investigate the effects of probiotics and autoprobiotics against H. pylori, in order to gain a better understanding of their mechanisms of action.
... Probiotic microbial strains, i.e., different preparations of Lactobacilli and Bifidobacteria are widely used in order to prevent and treat a number of intestinal disorders [32]. Given the uncertain therapeutic effect and safety profile of classical commercially provided probiotics in the period of cytopenia and immunodeficiency after HSCT, until sufficient immune reconstruction, the routine probiotics therapy is not currently applied in immunocompromised patients, and further studies are required [33]. ...
... Before LABs were widely recognized as probiotics, which are well known as beneficial microorganisms for human health, [22][23][24] a great variety of LAB strains had been commercially provided not only as dairy products but also as bacterial drugs. [25][26][27] The purpose of taking probiotics is mainly to increase healthy intestinal bacteria by modifying the gut microbiota, resulting in health-promoting effects. 28,29) Al-though the mechanisms of those effects in many species have been gradually or partially clarified, such as the production of bacteriocins 24) and short-chain fatty acids, 30) there could be still many unknown parts. ...
Article
In the present study, we have obtained a temperature-sensitive replication mutant in the Escherichia (E.) coli–lactic acid bacterium (LAB) shuttle vector pLES003-b carrying erythromycin-resistance gene by error-prone PCR technique. Among 858 clones obtained in the construction of the random mutation libraries of pLES003-b in the ori and repA regions, three clones could grow normally at 28 °C but not at 42 °C. One of the clones was designated as pLES003-b TS1. The sequencing analysis of pLES003-b TS1 revealed that the plasmid has four substitution mutations (376G > A, 435A > T, 914C > A, and 1996T > A) and one insertional mutation (1806_1807insA). Among those mutations, substitution mutation 914C > A, which leads to a CGC-to-AGC codon change at position 44 of the RepA protein (arginine-to-serine substitution mutation: R44S in RepA), was predicted to be a cause of temperature sensitivity. Therefore, the C-to-A substitution was introduced into the repA gene in pLES003-b using a site-directed mutagenesis method, and the resultant plasmid was electroporated into a Lactobacillus (L.) plantarum cell. The resultant transformant cannot grow at 42 °C in the presence of erythromycin, which is used as a selective marker, indicating that the R44S point mutation in the RepA protein may be crucial for temperature sensitivity. Furthermore, we have developed a new plasmid as an efficient genetic engineering tool for random insertional mutagenesis in LABs using a combination of transposon Tn10 and the temperature-sensitive replication system in pLES003-b. The resultant plasmid vector, which was designated pLES-Tn10-TS1, would be useful for genetic analysis of the functional molecule in lactic acid bacterial strains. Fullsize Image
... Autoprobiotic technology can be applied to modulate gut microbiota using selected indigenous probiotic bacteria isolated from a healthy donor. The isolated bacteria are stored in cryobanks and returned to the host if dysbiotic conditions occur [39]. ...
Chapter
Full-text available
The gut microbiota is being recognized as a factor with a significant influence on host physiology, health maintenance, and disease prevention. Distinct alterations of the gut microbiota are correlated with several chronic diseases. Currently, gut microbiota can be modulated by diet, probiotics, prebiotics, postbiotics, pharmabiotics, and fecal microbiota transplantation. An effective strategy in gut microbiota modulation is needed for the prevention and supportive treatment of chronic diseases. New and more effective approaches toward gut microbiota modulation are emerging, namely personalization and targeted modulation. The composition of novel products and treatments based on the individual gut microbiome, metabolome, strain specificity, and clinical data analysis can reveal and address specific changes to the diversity, composition, and function of gut microbiota. These analyses enable the development of personalized and targeted gut microbiota modulation, by the application of beneficial microorganisms, their consortia, their metabolites, and their effective combination.
... Bifidobacterium longum (BB536), which is the first human-residential bifidobacterial to receive Food and Drug Administration's Global Substance Registration System classification, reduced the frequency of gastrointestinal toxicity and the detection of Clostridium spp. in feces (12). These favorable effects may be owed to the influence of probiotics on the intestinal epithelial integrity, immune system, bacterial flora, and other factors (13). The growth and activity of probiotics are stimulated by prebiotics, which are non-digestible food ingredients (14). ...
Article
Objective High-dose chemotherapy with autologous hematopoietic stem cell transplantation (auto-HSCT) is an effective treatment option for relapsed and refractory aggressive malignant lymphoma. However, patients frequently experience treatment-induced gastrointestinal symptoms. Synbiotics, including live microorganisms and nondigestible food ingredients, reportedly ameliorate chemotherapy-induced mucosal damage. In this study, we assessed the efficacy and safety of synbiotics in patients undergoing auto-HSCT. Methods This randomized, double-blinded study included patients with malignant lymphoma eligible for auto-HSCT. The patients were randomly assigned to either a synbiotic group receiving Bifidobacterium longum (BB536) and guar gum or a placebo group receiving a placebo containing dextrin. The supplements were administered twice daily from the start of conditioning chemotherapy up to 28 days after auto-HSCT. The primary endpoint was the duration of total parenteral nutrition (TPN). Results In total, 12 patients were included and randomized. The median duration of TPN was 15 (range, 12-33) days in the synbiotic group and 17.5 (range, 0-32) days in the placebo group. The median duration of grade ≥3 diarrhea was shorter in the synbiotic group than in then placebo group (2.5 vs. 6.5 days), as was the duration of hospital stay (31.5 vs. 43 days). The oral intake and quality of life regarding diarrhea and anorexia improved in the synbiotic group after engraftment. Synbiotic infections, including bacteremia, were not observed. Conclusion Synbiotics may reduce gastrointestinal toxicity, thereby reducing nutritional problems and improving the quality of life of patients undergoing auto-HSCT, without severe adverse events.
Article
Full-text available
Galacto-N-biose (GNB) is an important core structure of glycan of mucin glycoproteins in the gastrointestinal (GI) mucosa. Because certain beneficial bacteria inhabiting the GI tract, such as bifidobacteria and lactic acid bacteria, harbor highly specialized GNB metabolic capabilities, GNB is considered a promising prebiotic for nourishing and manipulating beneficial bacteria in the GI tract. However, the precise interactions between GNB and beneficial bacteria and their accompanying health-promoting effects remain elusive. First, we evaluated the proliferative tendency of beneficial bacteria and their production of beneficial metabolites using gut bacterial strains. By comparing the use of GNB, glucose, and inulin as carbon sources, we found that GNB enhanced acetate production in Lacticaseibacillus casei, Lacticaseibacillus rhamnosus, Lactobacillus gasseri, and Lactobacillus johnsonii. The ability of GNB to promote acetate production was also confirmed by RNA-seq analysis, which indicated the upregulation of gene clusters that catalyze the deacetylation of N-acetylgalactosamine-6P and biosynthesize acetyl-CoA from pyruvate, both of which result in acetate production. To explore the in vivo effect of GNB in promoting acetate production, antibiotic-treated BALB/cA mice were administered with GNB with L. rhamnosus, resulting in a fecal acetate content that was 2.7-fold higher than that in mice administered with only L. rhamnosus. Moreover, 2 days after the last administration, a 3.7-fold higher amount of L. rhamnosus was detected in feces administered with GNB with L. rhamnosus than in feces administered with only L. rhamnosus. These findings strongly suggest the prebiotic potential of GNB in enhancing L. rhamnosus colonization and converting L. rhamnosus into higher acetate producers in the GI tract. IMPORTANCE Specific members of lactic acid bacteria, which are commonly used as probiotics, possess therapeutic properties that are vital for human health enhancement by producing immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. The long residence time of probiotic lactic acid bacteria in the GI tract prolongs their beneficial health effects. Moreover, the colonization property is also desirable for the application of probiotics in mucosal vaccination to provoke a local immune response. In this study, we found that GNB could enhance the beneficial properties of intestinal lactic acid bacteria that inhabit the human GI tract, stimulating acetate production and promoting intestinal colonization. Our findings provide a rationale for the addition of GNB to lactic acid bacteria-based functional foods. This has also led to the development of therapeutics supported by more rational prebiotic and probiotic selection, leading to an improved healthy lifestyle for humans.
Article
Full-text available
One hypothesis for the etiology of inflammatory bowel disease is that an altered or pathogenic microbiota causes inflammation in a genetically susceptible individual. Understanding the microbiota's role in the pathogenesis of the disease could lead to new IBD treatments aimed at shifting the bacteria in the gut back to eubiosis. Probiotics have some efficacy in the treatment of ulcerative colitis (UC), but our current repertoire is limited in potency. Fecal microbiota therapy (FMT) is an emerging treatment for several gastrointestinal and metabolic disorders. It has demonstrated efficacy in treating refractory Clostridium difficile infection, and there are case reports of FMT successfully treating UC. Further clinical studies are justified, and could be complemented by mouse models of fecal transplantation, in which variables can be controlled and manipulated.
Article
Full-text available
There are few carefully-designed studies investigating the safety of individual probiotics approved under Investigational New Drug policies. The primary aim of this prospective, double-blind placebo-controlled trial was to investigate if daily treatment of adults with Lactobacillus reuteri DSM 17938 (LR) for 2 months is safe and well-tolerated. Our secondary aim was to determine if LR treatment has immune effects as determined by regulatory T cell percentages, expression of toll-like receptors (TLR)-2 and -4 on circulating peripheral blood mononuclear cells (PMBCs), cytokine expression by stimulated PBMC, and intestinal inflammation as measured by fecal calprotectin. Forty healthy adults were randomized to a daily dose of 5×10(8) CFUs of LR (n = 30) or placebo (n = 10) for 2 months. Participants completed a daily diary card and had 7 clinic visits during treatment and observation. There were no severe adverse events (SAEs) and no significant differences in adverse events (AEs). There were no differences in PBMC subclasses, TLRs, or cytokine expression after treatment. The probiotic-treated group had a significantly higher fecal calprotectin level than the placebo group after 2 months of treatment: 50 µg/g (IQR 24-127 µg/g) vs. 17 µg/g (IQR 11-26 µg/g), p = 0.03, although values remained in the normal clinical range (0-162.9 µg/g). LR vials retained >10(8) CFUs viable organisms/ml. LR is safe and well tolerated in adults, without significant changes in immunologic markers. There was a small but significant increase in fecal calprotectin, perhaps indicating some element of immune recognition at the intestinal level. Clinical Trials.gov NCT00922727.
Article
Full-text available
The establishment of a pioneer gut microbiota is increasingly recognized as a crucial stage in neonatal development influencing health throughout life. While current knowledge is mainly based on either culture or molecular analysis of feces, we opted for a comprehensive approach complementing culture with state-of-the-art molecular methods. The bacterial composition in feces from seven healthy vaginally-delivered, breast-fed neonates was analyzed at days 4-6, 9-14 and 25-30 postnatal, using culture, 16S rRNA gene sequencing of isolates, quantitative PCR and pyrosequencing. Anaerobes outnumbered facultative anaerobes in all seven neonates within the first days of life, owing to high levels of Bifidobacterium and unexpectedly also Bacteroides, which were inversely correlated. Four neonates harbored maternal Bacteroides levels, comprising typical adult species, throughout the neonatal period, while in three only subdominant levels were detected. In contrast, the major adult-type butyrate-producing anaerobic populations, Roseburia and Faecalibacterium, remained undetectable during the neonatal period. The presence of Bacteroidetes as pioneer bacteria in the majority of neonates studied demonstrates that adult-type strict anaerobes may reach adult-like population densities within the first week of life. Consequently the switch from facultative to strict anaerobes may occur earlier than previously assumed in breast-fed neonates, and the establishment of the major butyrate-producing populations may be limited by other factors than the absence of anaerobic conditions. The impact of breast milk components on the timing of establishment of anaerobic pioneer bacteria, as well as opportunistic pathogens should be further studied in regard to priming of the gut-associated immune system and consequences on later health.
Article
Aim: To study the role of intestinal flora in inflammatory bowel disease (IBD). Methods: The spatial organization of intestinal flora was investigated in normal mice and in two models of murine colitis using fluorescence in situ hybridization. Results: The murine small intestine was nearly bacteria-free. The normal colonic flora was organized in three distinct compartments (crypt, interlaced, and fecal), each with different bacterial compositions. Crypt bacteria were present in the cecum and proximal colon. The fecal compartment was composed of homogeneously mixed bacterial groups that directly contacted the colonic wall in the cecum but were separated from the proximal colonic wall by a dense interlaced layer. Beginning in the middle colon, a mucus gap of growing thickness physically separated all intestinal bacteria from contact with the epithelium. Colonic inflammation was accompanied with a depletion of bacteria within the fecal compartment, a reduced surface area in which feces had direct contact with the colonic wall, increased thickness and spread of the mucus gap, and massive increases of bacterial concentrations in the crypt and interlaced compartments. Adhesive and infiltrative bacteria were observed in inflamed colon only, with dominant Bacteroides species. Conclusion: The proximal and distal colons are functionally different organs with respect to the intestinal flora, representing a bioreactor and a segregation device. The highly organized structure of the colonic flora, its specific arrangement in different colonic segments, and its specialized response to inflammatory stimuli indicate that the intestinal flora is an innate part of host immunity that is under complex control.
Article
In this article the comparative analysis of various methods of Helicobacter pylori verification is shown. Also algorithm of optimization of helicobacteriosis diagnostic, developed on the basis of a comparative estimation of results of various diagnostic techniques is presented. According to this algorithm, for increase of accuracy of helicobacteriosis diagnostic it is recommended to use at least two, and it is better three diagnostic methods: the combination of fast urease test or Helic-test with a histologic method (biopsy material from a stomach corpus) or polymerase chain reaction (ureC gene detection) is preferable. For an efficiency estimation of eradication therapy we recommend "Helic-test" as an effective noninvasive method that is especially important for children.
Article
Background: Ten to 15% of patients with pouchitis experience refractory or recurrent disease. The aim of this study was to evaluate the effectiveness of a single daily high dose probiotic preparation (VSL#3) in maintaining antibiotic induced remission, and quality of life (QOL), for one year in such patients. Methods: Patients with pouchitis at least twice in the previous year or requiring continuous antibiotics, associated with a pouchitis disease activity index (PDAI) ⩾7 (0 = perfect; 18 = worst), in whom remission was induced by four weeks of combined metronidazole and ciprofloxacin, were randomised to receive VSL#3 6 g or placebo once daily for one year or until relapse. Symptomatic, endoscopic, and histological evaluations were made before, and two and 12 months after randomisation or at the time of relapse. Remission was defined as a clinical PDAI ⩽2 and endoscopic PDAI ⩽1. Relapse was defined as an increased clinical PDAI score ⩾2 and increased endoscopic PDAI score ⩾3. QOL was assessed using the inflammatory bowel disease questionnaire (IBDQ). Results: Thirty six patients were randomised: 20 to VSL#3 and 16 to placebo. Remission was maintained at one year in 17 patients (85%) on VSL#3 and in one patient (6%) on placebo (p<0.0001). The IBDQ score remained high in the VSL#3 group (p = 0.3) but deteriorated in the placebo group (p = 0.0005). Conclusion: The once daily high dose probiotic VSL#3 is effective in maintaining antibiotic introduced remission for at least a year in patients with recurrent or refractory pouchitis. This is associated with a high level of quality of life.
Article
The G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology has made a major contribution to the development of probiotic preparations in the Russian Federation. As long ago as the 1960s the researchers at the institute began to conduct investigations on the selection of probiotic strains, development of the probiotic preparations of different formulations and functions and technologies for their industrial production, studies of clinical efficacy and the mechanisms of their action at the cell and molecular levels. At the present time the scientific research and biotechnological activities in this area continue to progress: the scientists at the institute have created a new generation of probiotic preparations, determined new areas for their application, and acquired new knowledge of human microecology.