Вы находитесь на странице: 1из 12

FOCUS ON GUT MICROBIOTA

Microbial contact during pregnancy, intestinal


colonization and human disease
Samuli Rautava, Raakel Luoto, Seppo Salminen and Erika Isolauri
Abstract | Interaction with colonizing intestinal bacteria is essential for healthy intestinal and immunological
development in infancy. Advances in understanding early hostmicrobe interactions indicate that this early
microbial programming begins inutero and is substantially modulated by mode of birth, perinatal antibiotics
and breastfeeding. Furthermore, it has become evident that this stepwise microbial colonization process,
as well as immune and metabolic programming by the microbiota, might have a long-lasting influence
on the risk of not only gastrointestinal disease, but also allergic, autoimmune and metabolic disease, in
later life. Modulating early hostmicrobe interaction by maternal probiotic intervention during pregnancy
and breastfeeding offers a promising novel tool to reduce the risk of disease. In this Review, we describe
the current body of knowledge regarding perinatal microbial contact, initial intestinal colonization and its
association with human disease, as well as means of modulating early hostmicrobe interaction to reduce the
risk of disease in the child.
Rautava, S. etal. Nat. Rev. Gastroenterol. Hepatol. 9, 565576 (2012); published online 14 August 2012; doi:10.1038/nrgastro.2012.144

Introduction
Our understanding with regard to the influence of during pregnancy and birth, and their association
microbes on human health has gradually expanded with mode of delivery, perinatal antibiotic exposure,
from pathogens causing infectious disease to a mutu breast-milk composition and immune physiology in
ally beneficial interaction with indigenous micro the mother, fetus and infant.
organisms that contribute to normal human physiology In this Review, we describe the current body of knowl
and immune homeostasis. That gastrointestinal dis edge regarding the importance of perinatal microbial
orders, such as IBD or infectious gastroenteritis, are contact to health and disease in the gut, and the body as
associated with perturbations of gut microbiota com a whole. We also suggest that modulating the complex
position (discussed elsehwere1) is now well established. network of prenatal and postnatal metabolic, immuno
The vital role of contact with environmental and colo logical and microbiological interactions with specific
nizing bacteria in early infancy for healthy immune probiotics could provide a powerful tool to reduce risk
and metabolic maturation beyond the gastrointestinal of disease in the child.
tract has been brought to our attention during the past
two decades. Development of intestinal microbiota
Advances in elucidating early hostmicrobe interac Microbes are present in all natural environments.
tions identify two hitherto largely unknown areas of The fact that the most predominant species (such as
interest. First, groundbreaking experimental and clini Bifidobacterium spp.) present in the indigenous intes
cal studies suggest that microbial contact inutero and tinal microbiota of human infants911 are not found in
during the neonatal period, modified by mode of deliv substantial quantities in the environment suggests that Department of
Pediatrics, Turku
ery and breastfeeding, exert marked effects on immune infants acquire their commensal bacteria from other University Hospital,
and metabolic programming in the fetus and infant.2,3 humans, presumably, and most importantly, from the Kiinamyllynkatu 48,
20520 Turku, Finland
Second, reports from clinical studies indicate that early mother. Conventionally, the human fetus has been (S. Rautava).
microbial contact has a long-term influence on intes considered sterile and microbial colonization has been Department of
tinal colonization patternsthat is, microbiological thought to begin during birth and then develop under Pediatrics, University of
Turku, Kiinamyllynkatu
programming (Box1)and so, consequently, on the the influence of breastfeeding and skin-to-skin contact 48, 20520 Turku,
immune responder phenotype, metabolic status and with the mother. Contact with the enormous bacterial Finland (R.Luoto,
E.Isolauri). Functional
risk of disease later in life.48 In our opinion, the concept load of the extrauterine world during and after birth Foods Forum, University
of early origins of human disease should, therefore, be marks the beginning of massive bacterial colonization of of Turku, Itinen
revised to include the multiple interactions between mucosal surfaces, particularly the gastrointestinal tract. Pitkkatu 4A, 20520
Turku, Finland
maternal environmental and indigenous microbes Consistent with this notion, lactobacilli residing in the (S.Salminen).
birth canal need to be characterized as they might be one
Correspondence to:
Competing interests important source of intestinal microbes during the first E. Isolauri
The authors declare no competing interests. days of life.1214 erika.isolauri@utu.fi

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 565


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points 615months and harboured a more complex composi


tion of Bifidobacterium spp. than formula-fed infants.11
Contact with microbes begins inutero and proceeds in a stepwise manner
Moreover, the differences in faecal Bifidobacterium spp.
during birth and early infancy
Early hostmicrobe interaction is a crucial component of healthy immune and
composition between breastfed and formula-fed infants
metabolic programming seemed to remain after cessation of breastfeeding.10,11
Mode of delivery, prematurity, perinatal antibiotic exposure and breastfeeding
have a major influence on early microbial contact, intestinal colonization and Development of enterotypes
subsequent risk of disease Our knowledge of the human microbiome composition
Maternal probiotic supplementation during pregnancy and breastfeeding and activity has increased rapidly. Infants have been
has shown potential in reducing the risk of immune-mediated and metabolic reported to harbour a heterogeneous microbiota that
disease via modulation of early hostmicrobe interactions
is prone to compositional changes caused by environ
mental disturbances such as the use of antibiotics. 17,18
Box 1 | Definitions of key concepts in early microbial colonization Infants develop a distinctive microbiota that is more
individual and open to influence by life events than
Programming that of adults; in adults, three distinct and predomi
Early life exposures to microbiota partially determine adult phenotypes. In the
nant microbiota enterotypes have been proposed. 19
case of human disease, programming refers to the risk of developing disease
generated at an early age and is, therefore, not strictly deterministic. The
What underlies the formation of these enterotypes is
programming factors, such as contact with specific microbes, are components of unknown and the early development of enterotypes in
causal complexes and therefore nonsufficient and non-necessary causal factors. infants is not yet clear,19,20 but the somewhat stable indi
Stepwise colonization vidual gut microbiota is thought to develop according
Successive steps occur in the microbial colonization of the infant gut. Colonization to modifications by dietary and environmental factors
takes place in a stepwise succession of microbiota components and in major during infancy. A stepwise developmental trend towards
steps at birth (delivery) and early life following delivery, during breastfeeding and an overall more adult-like profile seems probable.
first contact with the environment, and finally during and after weaning. The first step comprises early colonization by mainly
Microbial contact Firmicutes, including aerobic or facultatively anaerobic
Direct interaction with a microbe or microbial component that results in a bacteria (for example, Staphylococcus, Streptococcus and
response in the host. Contact with a microbe can result in an inflammatory Enterobacteriaceae). These early colonizers are then
response with potentially detrimental consequences (as in the case of
followed by Actinobacteria as well as more anaerobic
infection) or evoke a tolerogenic immune response resulting in withholding from
inflammatory activation.
bacteria, which typically include bifidobacteria espe
cially in breastfed infants.911 The next succession step
is the increase in the Bacteroidetes phylum of bacteria
Nutrition and other anaerobes, such as Clostridia and Eubacteria,
Breastfeeding exerts a major influence on gut coloni appear after the increase in Bacteroidetes.17,18 By 1year
zation patterns in early infancy. The gut microbiota of of age, the infant microbiota is thought to begin to con
breastfed infants has been reported to be dominated by verge toward a profile that resembles that of adults, being
bifidobacteria, whereas formula-fed infants harbour established at about 23years of age.17,18 Nonetheless,
a more diverse microbiome.9 By contrast, Penders and our understanding of the dynamics of gut microbiota
colleagues15 examined the gut microbiota composition in composition is still fragmentary and it is evident that
50 breastfed and 50 formula-fed infants and found that, essential changes in physiological processes and dietary
on the species level, all infants were colonized by bifido and lifestyle exposures, which affect gut microbiota
bacteria with no differences in prevalence or the amount composition, take place beyond this age.
of bifidobacteria between the groups.15 The same investi
gators later analysed faecal samples obtained from >1,000 Preterm infants
infants at 1month of age. No differences between bifido Bacterial colonization of preterm infants is an important
bacterial colonization were detected between breastfed clinical and scientific issue as ~513% of infants are born
and formula-fed infants, but the exclusively formula-fed prematurely.21 Preterm infants are not fully developed
infants were more often colonized with Escherichia coli, to cope with the massive postnatal microbial contact
Clostridium difficile, Bacteroides spp. and lactobacilli.16 and colonization that arises after birth. Furthermore,
The authors concluded that vaginally born, full-term as survival of extremely low-birth-weight or premature
infants who were exclusively breastfed were most likely infants was rare before the advent of modern neonatal
to have the highest numbers of intestinal bifidobacteria. intensive care, our knowledge of the normal or physi
In a 2010 study, seven breastfed and seven formula-fed ological microbiota for such infants hardly exists. Factors
infants were followed until the age of 18months; faecal that can disturb normal postnatal intestinal microbial
samples obtained at >20 time points over the course of c olonizationincluding caesarean section delivery,
18months were analysed.10 During exclusive breastfeed broad-spectrum antibiotic use, parenteral nutrition and
ing, bifidobacteria dominated the gut microbiota with delayed onset of breastfeeding, and reduced skin-to-skin
a >50% share of total bacterial counts whereas, over contact (discussed below)often cluster in very low-
the same time period, only ~25% of gut microbes were birth-weight infants (<1.5kg) and premature infants.
bifidobacteria in formula-fed infants. The breastfed To date, little is known about the compositional
infants in the study received breast milk until the age of development of intestinal microbiota in preterm infants.

566 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON GUT MICROBIOTA

An early study has indicated that there is delayed colo intestinal epithelial cells with bacterial TLR ligands has
nization by Bifidobacterium spp. as compared with been observed to initially activate proinflammatory
colonization in full-term infants. 22 Although not all pathways, but after continuous exposure lead to a state
subsequent studies have corroborated this finding,23 of hyporesponsiveness.37 The importance of gut microbes
it seems that the premature gut is not yet fully devel and TLR signalling to the establishment and mainte
oped for optimal bifidobacterial colonization.24 A 2011 nance of immune tolerance is further demonstrated in a
study assessing faecal microbiota by novel pyrosequenc report that indicates that food allergy can be induced in
ing techniques in serial samples obtained from 10 very mice whose gut microbiota has been reduced using anti
low-birth-weight preterm infants revealed that gut colo biotics.38 Moreover, similar susceptibility to sensitization
nization in these preterm infants is a dynamic process to food allergens has been observed in animals in which
involving a large number of bacterial species, of which signalling from intestinal microbes is lacking as a result
~20% are currently not classified.25 Nonetheless, several of deficient TLR4 function.38
investigators have reported that the gut microbiota in The wide range of intestinal physiology affected
preterm infants has low diversity when compared with by microbial colonization is demonstrated by a study
that from full-term infants.26,27 Even lower diversity has in which monocolonization of germ-free mice with
been detected in preterm infants who develop necrotiz Bacteroides thetaiotaomicron, a prominent member
ing enterocolitis, a devastating immunoinflammatory of the normal mouse gut microbiota, leads to major
intestinal process often resulting in death or severe changes in the expression of genes involved in nutri
impairment.26,28 Despite extensive studies of samples ent absorption, mucosal barrier function, metabolism,
obtained before the onset of necrotizing enterocolitis, angiogenesis and maturation in intestinal epithelial
however, no single organism or distinct bacterial colo cells.39 The indigenous gut microbiota has come to be
nization patterns that would be causative or predictive seen as a key organ involved in host energy homeo
of necrotizing enterocolitis have been identified, even stasis, affecting the harvest and storage of energy from
though organisms such as Clostridium perfringens or the diet.40 Pioneering studies using experimental animal
members of Enterobacteriaceae have been implicated.29,30 models have provided evidence of metabolic activity of
the gut microbiota that facilitates the extraction of calo
Immunological and metabolic maturation ries from ingested dietary substances and their storage
Interaction with colonizing microbes is essential for in host adipose tissue for later use.40,41 Following obser
healthy immune maturation and development of tolerance. vations of characteristic alterations in the gut microbiota
Our core understanding of early hostmicrobe interactions composition of obese compared with lean mice,42 the
is mainly derived from innovative studies conducted using transferable nature of the obese phenotype by trans
germ-free or gnotobiotic animals. These findings should, plantation of obese microbiota to germ-free mice has
however, be interpreted with some caution, bearing in been recognized. 43 Collectively, these experimental
mind the limits of these reductionist approaches when data suggest that early interaction with indigenous
considering the complex interactions between microbes, microbes is essential for healthy immunological and
the host and dietary and other environmental factors in the metabolic programming.
infants gastrointestinal tract.
As early as 1997, Sudo and colleagues31 showed that Microbial programming of host health
germ-free animals fail to develop immune tolerance, and Epidemiological and intervention studies have high
that tolerance is achieved after monocolonization with lighted analogous microbial influences in diverse clini
the commensal Bifidobacterium infantis in the neonatal cal conditions. Allergic disease and obesity, as well as
period, but not if B.infantis was introduced at a later chronic inflammatory diseases such as coeliac disease
age (after the neonatal period). Unfortunately, whether and IBD, share common environmental risk factors and
the effect is specific to this bacterium is not certain as, immunological mechanisms, and frequently coexist in
to our knowledge, data regarding other commensal the same individual (reviewed in detail elsewhere44,45).
bacteria introduced in the same experimental design Consequently, early microbial contact has been sug
have not been reported. Early microbial colonization gested to be involved in the initiation and perpetua
has subsequently been demonstrated to have persistent tion of aberrant immune activation and responsiveness
effects on host immune phenotype.32 Advances in experi central to the pathogenesis of allergic, autoimmune and
mental research suggest that innate immune receptors metabolic disease. Failure to maintain immune tolerance
such as Toll-like receptors (TLRs) are key mediators of towards indigenous intestinal microbes is a key element
the communication between the host and colonizing of IBD.46 To take an example, caesarean section delivery
microbes that leads to tolerance.33 Transient immune seems to increase the risk of coeliac disease,47,48 type1
activation and subsequent immune maturation medi diabetes mellitus49,50 and asthma,51,52 as compared with
ated by TLR-dependent mechanisms can be induced in vaginal delivery. These conditions, generally classified as
germ-free animals by colonization with the single spe diseases associated with excessive or aberrant Thelper
cific commensal microbial strain, Bacteroides fragilis, or (TH)1 and TH2 immune responsiveness, again, have been
even by administration of a specific commensal surface associated with deviations in the compositional develop
molecule such as the B.fragilis surface polysaccharide ment of gut microbiota,1,44,5355 typified as a less diverse
PSA.3436 In a parallel manner, stimulation of human microbiota specifically lacking bifidobacterial species

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 567


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

during infancy in individuals who later become over


weight.6 Moreover, the indigenous microbiomes of obese
Transfer of individuals are enriched in gene categories involved
bacteria in carbohydrate and lipid metabolism, 42,56 and some
experimental data indicate that gut microbes directly
influence energy harvest and storage by fermenting non
Placenta and digestible carbohydrates, activating digestive enzymes
amniotic fluid
Mode of and modulating adipocyte function (for example, by
delivery inducing the expression of fasting-induced adipocyte
factor).40 Finally, gut microbiota composition has been
suggested to be an instrumental component in initiating
the low-grade inflammatory state and, as a consequence,
insulin resistance associated with metabolic diseases par
ticularly among individuals predisposed to obesity.58,59
Hence, although the key cause of overweight in humans
is indubitably an imbalance between energy intake and
expenditure, it would seem that a combination of dietary,
environmental and genetic factors work in concert to
define the individual microbial composition, which
could favour either an obese or lean phenotype. Taken
together, it is evident that the nutritional status, immune
Breast milk
response and mode of delivery collectively interact with
each other, and all these elements are in turn influenced
by the gut microbiota (Figure1).

Early sources of microbial contact


Maternal gut During the past decade, it has become evident that
environmental influences during fetal development can
have long-lasting health consequences. Prenatal pro
gramming of human physiology as a result of nutritional
deficiency and intrauterine growth restriction has been
Infant gut extensively investigated. In parallel, research advances
suggest that the intimate interaction between the host
Figure 1 | The gut microbiota programmes host healthfrom mother to infant. The
fetus comes into contact with microbes originating in the maternal gut through the
and environmental and indigenous microbes begins
placenta and amniotic fluid. A massive inoculum of maternal bacteria is inutero. Maternal immune cells have been reported to
encountered during vaginal delivery, which is also associated with increased cross the placenta and modulate immune responses in
maternal intestinal permeability and translocation of gut bacteria into breast milk. the human fetus.60 In addition, maternal immunological
Breastfeeding not only modulates neonatal bacterial colonization and immune and metabolic state during pregnancy can lead to inflam
maturation, but is a direct source of maternal bacteria. After birth, skin-to-skin matory immune activation in the placenta.61 This state
contact and nursing ensures direct transfer of maternal bacteria to the infant to also modulates the initial microbial inoculum for the
enhance healthy immune and metabolic maturation.
newborn and breast-milk composition, thereby affect
ing the stepwise microbial colonization in early infancy.62
with simultaneous increases in specific clostridia.1,5456 The complex interactions between microbes, inflamma
Immunoinflammatory diseases mediated by pathological tory responses, dietary factors and immunological and
TH1 and TH2 responses thus seem to have common roots metabolic programming that influence the risk of disease
in early life, with perturbations in the intestinal microbial could thus partially have their origins in fetomaternal
colonization process leading to failure to establish and hostmicrobe interactions (Figure1).
maintain tolerance to environmental, dietary, microbial
or self antigens. Microbial contact during pregnancy
In a similar fashion, alterations in gut microbiota From the point of view of evolutionary biology, the
composition have been detected between obese and lean dogma of the microbiologically and immunologically
individuals.42,43,56 Obesity is associated with alterations naive fetal life seems bizarre, as mammals have evolved
in the relative amounts of Bacteroidetes and Firmicutes, in a world already inhabited by an enormous variety of
and animal studies have demonstrated that a 20% microbes. Perhaps more appropriately, it can be argued
increase in Firmicutes and a corresponding decrease in that the intrauterine environment, which is protected
Bacteroidetes were associated with an increased energy by maternal barriers against pathogenic invaders, offers
harvest.57 Early microbial colonization patterns might the embryo and fetus a controlled environment in which
not only be associated with, but also have a causal role, there is limited and selective exposure to microbes
in the development of obesity and metabolic disease, during the vulnerable periods of organogenesis and
as alterations in gut microbiota have been detected early development.

568 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON GUT MICROBIOTA

It has gradually become evident that maternal micro or ruling out contamination by maternal bacteria is not
bial exposure during pregnancy can have implications possible. Interestingly, however, live bacteria have also
for infant health. Data from epidemiological studies been detected in 25% of placentas after caesarean section
indicate that maternal exposure to farm animals, which delivery necessitated by pre-eclampsia at 2328weeks of
presumably results in higher maternal microbial contact gestation.64 In line with this observation, bacteria have
during pregnancy, protects the infant from develop been cultured in the chorionamnion of 15% of women
ing eczema, an immune-mediated skin condition.2 after caesarean section without rupture of membranes.74
Moreover, decreased occurrence of eczema is associated Thus, data exist to suggest that traces of microbes are
with altered expression of TLR genes at birth, which detectable in the fetoplacental unit from healthy, term
suggests that maternal microbial exposure modulates pregnancies.7578 DNA from intestinal bacteria such as
fetal immune physiology and points to a critical role bifidobacteria and lactobacilli has been found in term
for innate immune function in the fetoplacental unit.2 placentas after both vaginal and caesarean section deliv
In line with this notion, maternal mucosal contact with ery.75 In a similar fashion, microbial DNA is present at
Acinetobacter lwoffiiF78, a bacterium isolated from term in fetal membranes without signs of inflamma
cowsheds, modulates TLR gene expression in the pla tion.76 Our data confirm these findings by demonstrating
centa and decreases the risk of developing asthma in the the presence of bacterial DNAmost often belonging
offspring in an experimental mouse model.3 to the common gut bacteria Lactobacillus spp. and
Bifidobacterium spp.in all placentas and 43% amniotic
Microbial contact inutero fluid samples obtained after 29 sterile elective caesarean
In addition to the indirect modulation of fetoplacental section deliveries at term without signs of infection,
immune responses through maternal exposure to bac rupture of membranes or onset of labour.77 The pres
teria, accumulating evidence points to direct intra ence of bacterial DNA in infant meconium suggesting
uterine contact with microbes or microbial components. prenatal origin has also been reported.78
Bacteria are often present in the nonpregnant uterus.63
Live bacteria thought to originate both from the skin Prenatal immune effects
and the gastrointestinal tract have also been discov At which point or how bacteria gain entry into the
ered in nearly half of placentas obtained after extremely amniotic cavity is not currently known, but entry could
premature delivery at 2328 weeks of gestation. 64 take place by ascension from the birth canal, haemato
Chorionamnionitis is a major cause of premature birth, geneously through the placenta or by retrograde spread
accounting for ~2540% premature deliveries.21 Other via the Fallopian tubes.21 Data indicate that bacterial
known risk factors for preterm birth include mater DNA is frequently detected in the amniotic fluid as early
nal IBD, bacterial vaginosis and periodontitis, 21,65 all as midtrimester and is not always associated with adverse
of which are associated with perturbations of mucosal outcome or inflammation.79 The consequences of this
microbiota and inflammation. Even though periodon asymptomatic prenatal microbial contact are only now
tal disease associated with premature delivery has not starting to be elucidated.
been observed to be related to intra-amniotic bacterial Bacteria in the placenta have been reported to induce
translocation or inflammation,66 some data indicate that a systemic immune response in neonates after preterm
streptococci, Porphyromonas gingivalis or Fusobacterium delivery.80 Moreover, the response induced seems to be
nucleatum detetcted in amniotic fluid might have species specific with, for example, lactobacilli resulting
oral origins.67,68 in a reduced inflammatory response in the infant.80 We
Bacteria in the placenta and amniotic membranes have observed that DNA from nonpathogenic bacteria
can lead to premature birth, fetal demise and severe detected in the placenta is associated with alterations
neonatal and maternal infection. Invasion of the amni in TLR-related innate immune gene expression in the
otic cavity by bacteria such as Ureaplasma urealyticum fetal gut.77 Maternal obesity, on the other hand, has
or Mycoplasma hominis markedly increases the risk of been observed to modulate innate microbial pattern-
premature rupture of membranes and preterm birth.69 recognition receptor gene expression in the placenta in
Furthermore, vertical transfer of U.urealyticum fre an experimental sheep model81 and is known to be asso
quently occurs70 and has been associated with increased ciated with increased inflammatory mediator expression
risk of bronchopulmonary dysplasia71 and necrotizing in the placenta.61 Collectively, these emerging new data
enterocolitis in preterm infants.72 The presence of bac suggest that manipulation of maternal microbial expo
teria in the placenta has also been related to an increased sure during pregnancy might provide a novel means to
risk of cerebral palsy in extremely preterm infants.73 modulate early immune development and to reduce the
These clinically important associations mean that risk of immunoinflammatory or metabolic disease both
research efforts to understand bacterial presence in the in the mother and the child (Figure2).
placenta and amniotic fluid have mostly concentrated
on women with imminent preterm delivery caused by Antibiotics in the perinatal period
preterm premature rupture of membranes or preterm The use of antimicrobial agents is known to cause per
labour. Of note, these conditions can be caused by, or turbations in indigenous microbiota, particularly those
result in, bacterial translocation in the amniotic sack in the gut. Prenatal maternal antibiotics are used to
and, therefore, determining the direction of causality treat infections such as chorionamnionitis and also

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 569


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Delivery Extrauterine enviroment


Maternal factors Mode of delivery: Breastfeeding:
Maternal nutrition Microbiological factors Vaginal Exclusive breastfeeding
Metabolic state Enviromental and Assisted vaginal Formula feeding
Immune state indigenous microbes Caesarian section Microbes
Immune
and
Genotype Fetal life Birth Early infancy
metabolic
phenotype
Early programming
Figure 2 | Early programming of immune and metabolic phenotype. During pregnancy and early infancy, early programming
of immune and metabolic phenotypes occurs as a result of environmental and indigenous microbes that are modulated by
maternal state, mode of delivery and feeding practices. The intensity of the bold arrow reflects the window of opportunity to
modify the programming process to reduce the risk of disease in later life.

prophylactically to prevent infections in the neonate. Due to its unique protein and oligosaccharide compo
After birth, a large proportion of neonates are subjected nents, breast milk modulates the neonatal gut microbiota
to antibiotic treatment for suspected sepsis. Whilst these composition by promoting the growth of bifidobacteria.88
practices are life-saving in the case of disease, exposure Breast milk might also be a source of colonizing bacte
to antibiotics in the perinatal period could also disturb ria to the newborn infant (Table1).89,90 In addition to
healthy hostmicrobe interactions. In line with this providing a distinct inoculum for the stepwise micro
notion, broad-spectrum maternal perinatal antibiotic bial colonization process through physical contact with
therapy has been associated with an increased risk of maternal vaginal and intestinal bacteria, vaginal delivery
necrotizing enterocolitis 82,83 and cerebral palsy in the is associated with stress and hormonal changes during
infant.84 Postnatally, prolonged empiric antibiotic admin labour, which influence maternal gut permeability
istration for suspected sepsis when infection was not later and neonatal immune physiology.91 Interestingly, the
confirmed also reportedly increases the risk of death and importance of delivery mode on both gut microbiota
necrotizing enterocolitis in the preterm infant,85,86 but the composition and infant immune responses is detectable
effects on long-tem outcomes have, to our knowledge, throughout the first year of life.92,93
not been reported. Antibiotic use in the first year of life We have observed that the microbial composition of
has also been associated with increased risk of IBD in human breast milk is different after vaginal or caesar
childhood.87 These potentially detrimental effects of peri ean section delivery.94 Milk samples from mothers who
natal antibiotic use highlight the importance of microbial had undergone elective, but not nonelective, caesarian
contact during birth and in the neonatal period. section contained a different and less diverse bacte
rial community than milk samples from mothers who
Breast milk modulates hostmicrobe crosstalk had undergone vaginal delivery. Mothers giving birth
During pregnancy, the fetus swallows only amniotic by elective caesarean section had a marked composi
fluid, which passes through the gastrointestinal tract, tional shift in the microbiotapresenting decreased
whereas nutrition is provided parenterally by the mother levels of Leuconostocaceae and increased levels of
via the placenta and umbilical cord. The newborn infant Carnobacteriaceae, among otherscompared with
starts to feed during the first hours of life and the naive those who had a vaginal delivery.94 These observations
gut has to rapidly adjust to nutrient uptake and contact might be explained by a hypothesis according to which
with a myriad of foreign antigens whilst simultaneously the lactating mother introduces her gut microbiota to the
being colonized by microorganisms. From a physiologi newborn infant. This theory has been proposed on the
cal and evolutionary point of view, breast milk serves basis of observations indicating that there is increased
as the sole source of nutrition during the first months bacterial translocation in the maternal gut during preg
of life and could also be a source of colonizing bacteria nancy and lactation, resulting in the presence of bacteria
for the infant gut. In addition, breast milk provides the within dendritic cells in the mammary gland in experi
infant with important protection against disease during mental animals.95 In humans, maternal peripheral blood
the vulnerable neonatal period. The beneficial health mononuclear cells and cells in breast milk have also
effects of breastfeeding range from a reduced risk of been shown to contain bacteria and bacterial DNA.95 In
necrotizing enterocolitis and infections of the gastro accordance with this hypothesis, the number and type
intestinal and respiratory tracts in infancy to improved of bifidobacteria in maternal faeces and breast milk cor
cognitive development and decreased occurrence of relate with those in infant faeces,90,96 and breast milk and
coeliac disease, type2 diabetes mellitus, obesity, hyper infant faeces consistently share strains of staphylococci,
cholesterolaemia and asthma in later life.88 Whereas bifidobacteria and lactobacilli.97 The link between the
protection from infectious disease during breastfeed maternal gut and the mammary gland is also suggested
ing can be explained by passive immunoprotection by by a study showing that maternal consumption of the
molecules such as IgA antibodies and oligosaccharides probiotic Lactobacillus reuteri is associated with recovery
in breast milk, the long-term health effects suggest active of live L.reuteri in breast milk.98
programming of infant physiology, which could be medi Bioactive molecules in breast milk could substan
ated by modulation of infant immune responses and gut tially modulate interactions between the host immune
microbiota composition.88 system and the developing gut microbiota. For instance,

570 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON GUT MICROBIOTA

Table 1 | Characterization of bacteria present in breast milk


Type of bacteria, species or class Characterization methods Reference
Culture method Molecular method
Streptococcus spp., Staphylococcus spp., Bacillus Yes NA Bjrkstn etal. (1980)137
spp., Escherichia coli and Enterobacter spp.
Staphylococcus epidermidis, Enterococcus faecalis, Yes PCR, DGGE Martin etal. (2003)138
Streptococcus mitis, Propionibacterium acnes and
Staphylococcus lugdunensis
Staphylococcus aureus, Enterococcus fecalis, Yes, cultured PCR and/or RFLP Heikkil & Saris (2003)139
Streptococcus mitis, Lactobacillus rhamnosus, in MRS broth
L.crispatus and Leuconostoc mesentroides
Lactococcus lactis Yes PCR Beasley & Saris (2004)140
Lactic acid bacteria, Lactococcus lactis, NA Identification by PCRDGGE Martn etal. (2007)141
Leuconostoc citreum, Pseudomonas spp. and
Propionibacterium spp.
Bifidobacterium longum, B.breve, B.adolescentis NA qPCR Gueimonde etal. (2007)89
and B.animalis
Staphylococcus epidermidis, Enterococcus spp., Yes Identification by PCRDGGE Delgado etal. (2008)142
Streptococcusmitis and other Streptococcus spp.,
Corynebacterium spp., Klebsiellaoxytoca and
unculturable bacteria
Staphylococcus epidermidis, Enterococcus faecalis, Yes Genetic profiling Jimnez etal. (2008)143
Streptococcus mitis, Propionibacterium acnes and
Staphylococcus lugdunensis
Bifidobacterium breve, B.longum, B.adolescentis, NA qPCR Martn etal. (2009)144
B.bifidum and B.dentium
Carnobacterium spp., Veillonella spp., Leptotrichia NA Pyrosequencing and qPCR Hunt etal. (2011)145
spp., Prevotella spp., Lactobacillus spp. and
Bifidobacterium spp.
Staphylococcus spp., Lactobacillus spp. and Yes PCR, RAPD, PFGE, and/or Martn etal. (2012)97
Bifidobacterium spp. MLST genotyping
Staphylococcus spp., Streptococcus spp., Weisella NA Pyrosequencing and qPCR Cabrera Rubio etal.
spp., Leuconostoc spp., Veillonella spp., (2012)94
Leptotrichia spp. and Prevotella spp.
Abbreviations: DGGE, denaturing gradient gel electrophoresis; MLST, multilocus sequence type; NA, not applicable; PFGE, pulsed field gel electrophoresis;
qPCR, quantitative PCR; RAPD, random amplified polymeric DNA; RFLP, restriction fragment length polymorphism.

transforming growth factor (TGF)2, one of the most Successful reprogramming by probiotics
comprehensively investigated immunomodulatory The association between early microbial contact and
factors in breast milk, is known to be necessary for subsequent development of disease has spawned inter
breast-milk-induced immune tolerance.99 Experimental est in the modulation of early hostmicrobe interaction
studies indicate that TGF2 at a concentration cor as a means to reduce the risk of disease. The potentially
responding to that in breast milk considerably alters harmful effects of caesarean sections (discussed earlier)
inflammatory responses, and that exposure to TGF2 have prompted caution when choosing the optimal mode
induces immune maturation in the immature human of delivery. Data from well-conducted clinical studies
gut.100,101 The interactions between breast milk TGF2, have brought to attention the adverse effects of perinatal
gut microbes and infant immune development are or prolonged neonatal empirical antibiotic administra
further demonstrated by the observation that mater tion.8287 In addition to these potential changes in clinical
nal consumption of the probiotic L.rhamnosusGG practice, novel interventions aiming to modulate early
during pregnancy and breastfeeding increases the con hostmicrobe interactions have been introduced and
centration of TGF2 in breast milk and is associated assessed in clinical trials.
with a reduced risk of atopic eczema in the infant.102 Prebiotics are nondigestible food ingredients, the
On the other hand, administration of the probiotic purpose of which is to selectively promote the growth of
Bifidobacterium breve to preterm infants modulates indigenous intestinal bacteria thought to be beneficial
their responsiveness to TGF, although the clinical to health. Accordingly, consumption of prebiotic oligo
effects of this response are unknown. 103 On the basis saccharides reportedly increases the amount of faecal
of these collective data, we hypothesize that breast bifidobacteria in infants.104107 Published data regard
milk might function as a vehicle to introduce maternal ing clinical efficacy of prebiotics in infancy are still
gut microbiota to the infant in a tolerogenic immune sparse. Administration of a combination of prebiotic
milieu (Figure3). galacto-oligosaccharides and fructo-oligosaccharides

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 571


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Maternal gut Breast milk within the context of modulating early microbial contact
relates to the use of specific probiotics. Even though this
Microbes TGF-2 approach is still experimental and the long-term effects
of modulating early microbial contactsome of which
might not be beneficialremain largely unknown,
specific probiotics can be considered a paradigmatic
example of an intervention aiming to modulate host
microbe interaction. Perinatal probiotic intervention has
Increased been well tolerated and might influence early microbiota
responsiveness composition and activity, but no interference with the
long-term composition or quantity of gut microbiota has
yet been reported. Although permanent establishment or
colonization by probiotics has not been observed,117120
Colonization Maturation Tolerance Inflammation the clinical health effects of probiotic intervention have
been shown to be long-lasting, consistent with the notion
of perinatal microbial programming.121123
Infant gut Data suggesting that specific probiotics administered
in early infancy might enhance healthy immune matura
Figure 3 | Interactions between indigenous microbes and TGF2 in breast milk.
TGF2 is essential for breast-milk-induced tolerance towards environmental tion and reduce the risk of eczema in high-risk infants
antigens, attenuates excessive inflammatory responses in the neonatal gut and are accumulating,115,121128 even though not all studies
induces gut maturation and production of IgA antibodies. These processes are have generated positive results.129131 On the basis of
also dependent on the indigenous gut microbiota. According to a novel hypothesis, published reports, it seems that probiotic intervention is
maternal gut bacteria are actively transported into the mammary gland and most effective if commenced prenatally.115,121,124128 One
secreted in breast milk and are thus transferred to the infant in a tolerogenic putative mechanism for the protective effect postnatally
immune milieu with TGF2. Moreover, maternal supplementation with probiotic
has been suggested to be increased concentration of
lactobacilli is associated with increased levels of TGF2 in the breast milk and
TGF2 in breast milk of women receiving probiotics.102
bifidobacteria in the infant gut optimize responsiveness to breast milk TGF2.
Abbreviation: TGF, transforming growth factor. These observations emphasize both prenatal and post
natal interaction between maternal gut microbes and
infant immune system.
(90% galacto-oligosaccharides and 10% fructo- The discovery of the importance of prenatal micro
oligosaccharides as defined in the infant formula direc bial contact to fetal and infant development opens up
tive in Europe) in infancy has been shown to modulate intriguing new possibilities for probiotic intervention
antibody production and reduce the incidence of aller via the pregnant woman. In line with this notion, we
gic outcomes such as eczema.108,109 Subsequently, a larger have observed altered innate immune gene expression
multicentre trial of >800 infants demonstrated that oligo in the placenta and fetal gut after prenatal maternal
saccharide-enriched infant formula might reduce the risk probiotic intervention.77 Perhaps more importantly, we
of eczema in low-risk infants.110 However, oligofructose- have reported that maternal supplementation with the
enriched infant cereal has failed to decrease the incidence combination of the probiotics L.rhamnosusGG and
of diarrhoeal disease in a developing country setting 111 Bifidobacterium lactisBb12 from the first trimester of
and both inulin and fructo-oligosaccharides have been pregnancy to the end of exclusive breastfeeding reduces
deemed to have no influence on the risk of antibiotic- the risk of atopic sensitization in high-risk infants.131
associated diarrhoea in children.112 It might be argued Surprisingly, a statistically significant reduction of ges
that prebiotics only are effective if an appropriate com tational diabetes mellitus was observed in women receiv
position of bifidobacteria, which is amenable for specific ing probiotics as compared with those on placebo in the
prebiotic enhancement, resides in the target gut. same study. 132 Furthermore, probiotic supplementa
The justification for the synbiotic approach, therefore, tion resulted in consistently improved plasma glucose
lies in providing both the beneficial microbial stimulus concentrations and insulin sensitivity not only during
(probiotics) and their growth factors (prebiotics), thus pregnancy, but also 12months postpartum among meta
mimicking the net effect of breastfeeding. Our current bolically healthy women.133 The same study has provided
knowledge of the optimal prebiotic component for each clinical evidence that probiotic consumption reduces the
specific probiotic is fragmentary at best. Nonetheless, risk of maternal central adiposity over the 6month post
published data are available indicating that supplemen partum period.134 Maternal obesity and glucose intoler
tation with synbiotics is associated with reduced inci ance are known risk factors for overweight and obesity in
dence acute diarrhoeal disease,113,114 whereas data on the the offspring and, consistent with this evidence, we have
effects of synbiotic combinations on the risk of eczema demonstrated that perinatal probiotic intervention mod
have been conflicting.115,116 Dissecting the contribution of erates excessive weight gain especially among children
the prebiotic and probiotic elements or their synergistic who later became overweight during the first years of
influence in these studies is difficult. life, the effect being most pronounced at 4years of age.135
The most substantial body of clinical evidence to date To our knowledge, no published trials have assessed
regarding a reduction in the risk of disease in humans the efficacy of probiotics in the primary prevention of

572 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON GUT MICROBIOTA

gastrointestinal disease (such as IBD or coeliac disease) Emerging understanding of the importance of micro
in infants. bial contact during the fragile periods of fetal life,
delivery and infancy to healthy immune and metabolic
Conclusions programming creates new opportunities to improve
Our life-long coexistence with microbes seems to infant health and reduce the risk of disease in later life.
begin earlier than conventionally thought (Figure1). The notion of perinatal origins of human disease sug
Microbial contact during embryogenesis and fetal life is gests an early window of opportunity (Figure2) to influ
selectively buffered by maternal protective mechanisms. ence disease risk and necessitates prenatal and perinatal
Nonetheless, environmental and maternal indigenous intervention to achieve the desired effects. Consistent
microbes, together with maternal nutritional and meta with this hypothesis, accumulating evidence indicates
bolic state, exert direct and indirect effects on the fetus, that modulation of early microbial contact during preg
which might be reflected in later health and disease. nancy and breastfeeding with maternal probiotic sup
Vaginal delivery seems not only to provide substantial plementation could be effective in reducing the risk of
bacterial exposure, but also primes healthy hostmicrobe immunoinflammatory and metabolic disease.
interactions and immune development with hitherto
poorly understood effects on infant immunology and
microbial composition of breast milk. Establishment
Review criteria
of the indigenous microbiota in the perinatal period
and early infancy might be considered the last phase of This Review article was created to give the reader a
comprehensive, but critical, view on the current state-of-
organogenesis, affecting not only gastrointestinal, but
the-art on hostmicrobe interaction at the critical stage
also systemic, physiology. Similarly, breastfeeding can
of maturation including prenatal, perinatal and early
be viewed as an extension of intrauterine maternal guid postnatal microbial exposure. The literature selection
ance and protection that facilitates survival and healthy was based on the personal experience of the authors
programming in the microbial world during early life. representing the different areas of the topic: immunology,
The various components and functions of breast milk microbiology, food science, clinical paediatrics and
underscore the complex interactions between contact neonatology. The expertise of the authors derives from
with microbes, diet and immune maturation, including pioneer research work in the field, as well as expert
activities with probiotics and health messages related to
establishment and maintenance of tolerance towards
functional foods.
dietary antigens and indigenous microbiota.

1. DuPont, A.W. & DuPont, H.L. The intestinal infants by using molecular identification and human infant intestinal microbiota. PLoS Biol. 5,
microbiota and chronic disorders of the gut. Nat. detection methods. J.Pediatr. Gastroenterol. e177 (2007).
Rev. Gastroenterol. Hepatol. 16, 523531 Nutr. 30, 6167 (2000). 18. Koenig, J.E. etal. Succession of microbial
(2011). 10. Roger, L.C., Costabile, A., Holland, D.T., consortia in the developing infant gut
2. Roduit, C. etal. Prenatal animal contact and Hoyles,L. & McCartney, A.L. Examination of microbiome. Proc. Natl Acad. Sci. USA 108
gene expression of innate immunity receptors at faecal Bifidobacterium populations in breast- and (Suppl. 1), 45784585 (2011).
birth are associated with atopic dermatitis. formula-fed infants during the first 18months of 19. Arumugam, M. etal. Enterotypes of the human
J.Allergy Clin. Immunol. 127, 179185 (2011). life. Microbiology 156, 33293341 (2010). gut microbiome. Nature 473, 174180 (2011).
3. Conrad, M.L. etal. Maternal TLR signalling is 11. Roger, L.C. & McCartney, A.L. Longitudinal 20. Wu, G.D. etal. Linking long-term dietary patterns
required for prenatal asthma protection by the investigation of the faecal microbiota of healthy with gut microbial enterotypes. Science 334,
nonpathogenic microbe Acinetobacter lwoffii full-term infants using fluorescence insitu 105108 (2011).
F78. J.Exp. Med. 206, 28692877 (2009). hybridization and denaturing gradient gel 21. Goldenberg, R.L., Culhane, J.F., Iams, J.D. &
4. Kalliomki, M. etal. Disticnt patterns of electrophoresis. Microbiology 156, 33173328 Romero, R. Epidemiology and causes of preterm
neonatal gut microflora in infants in whom (2010). birth. Lancet 371, 7584 (2008).
allergy was and was not developing. J.Allergy 12. Matsumiya, Y., Kato, N., Watanabe, K. & Kato, H. 22. Stark, P.L. & Lee, A. The bacterial colonization of
Clin. Immunol. 107, 129134 (2001). Molecular epidemiological study of vertical the large bowel of pre-term low birth weight
5. Bjrkstn, B., Sepp, E., Julge K., Voor, T. & transmission of vaginal Lactobacillus species neonates. J.Hyg. (Lond). 89, 5967 (1982).
Mikelsaar, M. Allergy development and the from mothers to newborn infants in Japanese, by 23. Bjrkstrm, M.V. etal. Intestinal flora in very
intestinal microflora during the first year of life. arbitrarily primed polymerase chain reaction. low-birth weight infants. Acta Paediatr. 98,
J.Allergy Clin. Immunol. 108, 516520 (2001). J.Infect. Chemother. 8, 4349 (2002). 17621767 (2009).
6. Kalliomki, M., Collado, M.C., Salminen, S. & 13. Dominguez-Bello, M.G. etal. Delivery mode 24. Butel, M.J. etal. Conditions of bifidobacterial
Isolauri, E. Early differences in fecal microbiota shapes the acquisition and structure of the initial colonization in preterm infants: a prospective
composition in children may predict overweight. microbiota across multiple body habitats in analysis. J.Pediatr. Gastroenterol. Nutr. 44,
Am. J.Clin. Nutr. 87, 534538 (2008). newborns. Proc. Natl Acad. Sci. USA 107, 577582 (2007).
7. Johansson, M.A., Sjgren, Y.M., Persson,J.O., 1197111975 (2010). 25. Chang, J.Y., Shin, S.M., Chun, J., Lee, J.H. &
Nilsson, C. & Sverremark-Ekstrm, E. Early 14. Gajer, P. etal. Temporal dynamics of the human Seo, J.K. Pyrosequencing-based molecular
colonization with a group of Lactobacilli vaginal microbiota. Sci. Transl. Med. 4, 132ra52 monitoring of the intestinal bacterial colonization
decreases the risk for allergy at five years of age (2012). in preterm infants. J.Pediatr. Gastroenterol. Nutr.
despite allergic heredity. PLoS ONE 6, e23031 15. Penders, J. etal. Quantification of 53, 512519 (2011).
(2011). Bifidobacterium spp., Escherichia coli and 26. Wang, Y. etal. 16S rRNA gene-based analysis of
8. Ajslev, T., Andersen, C.S., Gamborg, M., Clostridium difficile in faecal samples of breast- fecal microbiota from preterm infants with and
Srensen, T.I. & Jess, T. Childhood overweight fed and formula-fed infants by real-time PCR. without necrotizing enterocolitis. ISME J. 3,
after establishment of the gut microbiota: the FEMS Microbiol. Lett. 243, 141147 (2005). 944954 (2009).
role of delivery mode, pre-pregnancy weight and 16. Penders, J. etal. Factors influencing the 27. Roug, C. etal. Investigation of the intestinal
early administration of antibiotics. Int. J.Obes. composition of the intestinal microbiota in early microbiota in preterm infants using different
(Lond). 35, 522529 (2011). infancy. Pediatrics 118, 511521 (2006). methods. Anaerobe 16, 362370 (2010).
9. Harmsen, H.J. etal. Analysis of intestinal flora 17. Palmer, C., Bik, E.M., DiGiulio, D.B., 28. Neu, J. & Walker, W.A. Necrotizing enterocolitis.
development in breast-fed and formula-fed Relman,D.A. & Brown, P.O. Development of the N.Engl. J.Med. 364, 255264 (2011).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 573


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

29. de la Cochetiere, M.F. etal. Early intestinal 47. Decker, E. etal. Cesarean delivery is associated 66. Goepfert, A.R. etal. Periodontal disease and
bacterial colonization and necrotizing with celiac disease but not inflammatory bowel upper genital tract inflammation in early
enterocolitis in premature infants: the putative disease in children. Pediatrics 125, 14331440 spontaneous preterm birth. Obstet. Gynecol.
role of Clostridium. Pediatr. Res. 56, 366370 (2010). 104, 777783 (2004).
(2004). 48. Mrild, K., Stephansson, O., Montgomery, S., 67. Bearfield, C., Davenport, E.S.,
30. Mai, V. etal. Fecal microbiota in premature Murray, J.A. & Ludvigsson, J.F. Pregnancy Sivapathasundaram, V. & Allaker, R.P. Possible
infants prior to necrotizing enterocolitis. PLoS outcome and risk of celiac disease in offspring: association between amniotic fluid micro-
ONE 6, e20647 (2011). a nationwide case-control study. organism infection and microflora in the mouth.
31. Sudo, N. etal. The requirement of intestinal Gastroenterology 142, 3945 (2012). BJOG 109, 527533 (2002).
bacterial flora for the development of an IgE 49. Algert, C.S., McElduff, A., Morris, J.M. & 68. Len, R. etal. Detection of Porphyromonas
production system fully susceptible to oral Roberts, C.L. Perinatal risk factors for early gingivalis in the amniotic fluid in pregnant
tolerance induction. J.Immunol. 159, 739745 onset of Type1 diabetes in a 20002005 birth women with a diagnosis of threatened
(1997). cohort. Diabet. Med. 26, 11931197 (2009). premature labor. J.Periodontol. 78, 12491255
32. Olszak, T. etal. Microbial exposure during early 50. Bonifacio, E., Warncke, K., Winkler, C., (2007).
life has persistent effects on natural killer Tcell Wallner,M. & Ziegler, A.G. Cesarean section and 69. DiGiulio, D.B. Diversity of microbes in amniotic
function. Science 336, 489493 (2012). interferon-induced helicase gene polymorphisms fluid. Semin. Fetal Neonatal Med. 17, 211
33. Round, J.L. etal. The Toll-like receptor 2 combine to increase childhood type1 diabetes (2012).
pathway establishes colonization by a risk. Diabetes 60, 33003306 (2011). 70. Snchez, P.J. & Regan, J.A. Vertical
commensal of the human microbiota. Science 51. Kero, J. etal. Mode of delivery and asthmais transmission of Ureaplasma urealyticum from
332, 974977 (2011). there a connection? Pediatr. Res. 52, 611 mothers to preterm infants. Pediatr. Infect. Dis.
34. Mazmanian, S.K., Liu, C.H., Tzianabos, A.O. & (2002). J. 9, 398401 (1990).
Kasper, D.L. An immunomodulatory molecule of 52. Roduit, C. etal. Asthma at 8years of age in 71. Snchez, P.J. & Regan, J.A. Ureaplasma
symbiotic bacteria directs maturation of the children born by caesarean section. Thorax 64, urealyticum colonization and chronic lung
host immune system. Cell 122, 107118 107113 (2009). disease in low birth weight infants. Pediatr.
(2005). 53. Nistal, E. etal. Differences of small intestinal Infect. Dis. J. 7, 542546 (1988).
35. Ruiz, P.A., Hoffmann, M., Szcesny, S., Blaut, M. bacteria populations in adults and children with/ 72. Okogbule-Wonodi, A.C. etal. Necrotizing
& Haller, D. Innate mechanisms for without celiac disease: effect of age, gluten diet, enterocolitis is associated with Ureaplasma
Bifidobacterium lactis to activate transient pro- and disease. Inflamm. Bowel Dis. 18, 649656 colonization in preterm infants. Pediatr. Res. 69,
inflammatory host responses in intestinal (2012). 442447 (2011).
epithelial cells after the colonization of germ- 54. Giongo, A. etal. Toward defining the autoimmune 73. Leviton, A. etal. Microbiologic and histologic
free rats. Immunology 115, 441450 (2005). microbiome for type1 diabetes. ISME J. 5, characteristics of the extremely preterm infants
36. Wang, Q. etal. A bacterial carbohydrate links 8291 (2011). placenta predict white matter damage and later
innate and adaptive responses through Toll-like 55. Russell, S.L. etal. Early life antibiotic-driven cerebral palsy. the ELGAN study. Pediatr. Res.
receptor2. J.Exp. Med. 203, 28532863 changes in microbiota enhance susceptibility to 67, 95101 (2010).
(2006). allergic asthma. EMBO Rep. 13, 440447 74. Goldenberg, R.L., Hauth, J.C. & Andrews, W.W.
37. Otte, J.M., Cario, E. & Podolsky, D.K. (2012). Intrauterine infection and preterm delivery.
Mechanisms of cross hyporesponsiveness to 56. Turnbaugh, P.J. etal. A core gut microbiome in N.Engl. J.Med. 342, 15001507 (2000).
Toll-like receptor bacterial ligands in intestinal obese and lean twins. Nature 457, 480484 75. Satokari, R., Grnroos, T., Laitinen, K.,
epithelial cells. Gastroenterology 126, (2009). Salminen,S. & Isolauri, E. Bifidobacterium and
10541070 (2004). 57. Jumpertz, R.etal. Energy-balance studies reveal Lactobacillus DNA in the human placenta. Lett.
38. Bashir, M.E., Louie, S., Shi, H.N. & Nagler- associations between gut microbes, caloric load, Appl. Microbiol. 48, 812 (2009).
Anderson, C. Toll-like receptor 4 signaling by and nutrient absorption in humans. Am. J.Clin. 76. Steel, J.H. etal. Bacteria and inflammatory
intestinal microbes influences susceptibility to Nutr. 94, 5865 (2011). cells in fetal membranes do not always cause
food allergy. J.Immunol. 172, 69786987 58. Cani, P.D. etal. Metabolic endotoxemia initiates preterm labor. Pediatr. Res. 57, 404411
(2004). obesity and insulin resistance. Diabetes 56, (2005).
39. Hooper, L.V. etal. Molecular analysis of 17611772 (2007). 77. Rautava, S., Collado, M.C., Salminen, S. &
commensal host-microbial relationships in the 59. Membrez, M. etal. Gut microbiota modulation Isolauri, E. Probiotics modulate hostmicrobe
intestine. Science 291, 881884 (2001). with norfloxacin and ampicillin enhances glucose interaction in the placenta and fetal guta
40. Bckhed, F. etal. The gut microbiota as an tolerance in mice. FASEB J. 22, 24162426 randomized, double-blind, placebo-controlled
environmental factor that regulates fat storage. (2008). trial. Neonatology 102, 178184 (2005).
Proc. Natl Acad. Sci. USA 101, 1571815723 60. Mold, J.E. etal. Maternal alloantigens promote 78. Mshvildadze, M. etal. Intestinal microbial
(2004). the development of tolerogenic fetal regulatory ecology in premature infants assessed with
41. Bckhed, F., Manchester, J.K., Tcells inutero. Science 322, 15621565 nonculturebased techniques. J.Pediatr. 156,
Semenkovich,C.F. & Gordon, J.I. Mechanisms (2008). 2025 (2010).
underlying the resistance to diet-induced 61. Roberts, K.A. etal. Placental structure and 79. Markenson, G.R., Adams, L.A., Hoffman, D.E.
obesity in germ-free mice. Proc. Natl Acad. Sci. inflammation in pregnancies associated with & Reece, M.T. Prevalence of Mycoplasma
USA 104, 979984 (2007). obesity. Placenta 32, 247254 (2011). bacteria in amniotic fluid at the time of genetic
42. Ley, R.E. etal. Obesity alters gut microbial 62. Collado, M.C., Isolauri, E., Laitinen, K. & amniocentesis using the polymerase chain
ecology. Proc. Natl Acad. Sci. USA 102, Salminen, S. Effect of mothers weight on reaction. J.Reprod. Med. 48, 775779 (2003).
1107011075 (2005). infants microbiota acquisition, composition, and 80. Fichorovna, R.N. etal. Maternal microbe-
43. Turnbaugh, P.J. etal. An obesity-associated gut activity during early infancy: a prospective specific modulation of inflammatory response in
microbiome with increased capacity for energy follow-up study initiated in early pregnancy. Am. J. extremely lowgestationalage newborns. MBio
harvest. Nature 444, 10271031 (2006). Clin. Nutr. 92, 10231030 (2010). 2, e00280-10 (2011).
44. Rautava, S., Kalliomki, M. & Isolauri, E. New 63. Andrews, W.W. etal. Endometrial microbial 81. Zhu, M.J., Du, M., Nathanielsz, P.W. &
therapeutic strategy for combating the colonization and plasma cell endometritis after Ford,S.P. Maternal obesity up-regulates
increasing burden of allergic disease: spontaneous or indicated preterm versus term inflammatory signalling pathways and enhances
ProbioticsA Nutrition, Allergy, Mucosal delivery. Am. J.Obstet. Gynecol. 193, 739745 cytokine expression in the mid-gestation sheep
Immunology and Intestinal Microbiota (NAMI) (2005). placenta. Placenta 31, 387391 (2010).
Research Group report. J.Allergy Clin. Immunol. 64. Onderdonk, A.B., Delaney, M.L., DuBois, A.M., 82. Kenyon, S.L., Taylor, D.J., Tarnow-Mordi, W. &
116, 3137 (2005). Allred, E.N. & Leviton, A. Detection of bacteria in ORACLE Collaborative Group. Broad-spectrum
45. Isolauri, E., Kalliomki, M., Rautava, S., placental tissues obtained from extremely low antibiotics for preterm, prelabour rupture of
Salminen, S. & Laitinen, K. Obesityextending gestational age neonates. Am. J.Obstet. Gynecol. fetal membranes: the ORACLE I randomised
the hygiene hypothesis. Nestle Nutr. Workshop 198, 17 (2008). trial. ORACLE Collaborative Group. Lancet 357,
Ser. Pediatr. Program. 64, 7585 (2009). 65. Bengtson, M.B. etal. Relationships between 979988 (2001).
46. Abraham, C. & Medzhitov, R. Interactions inflammatory bowel disease and perinatal 83. Weintraub, A.S. etal. Antenatal antibiotic
between the host innate immune system and factors: both maternal and paternal disease are exposure in preterm infants with necrotizing
microbes in inflammatory bowel disease. related to preterm birth of offspring. Inflamm. enterocolitis. J.Perinatol. http://dx.doi.org/
Gastroenterology 140, 17291737 (2011). Bowel Dis. 16, 847855 (2010). 10.1038/jp.2011.180.

574 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON GUT MICROBIOTA

84. Kenyon, S. etal. Childhood outcomes after epithelial cells and inhibits inflammatory 118. Rinne, M., Kalliomki, M., Salminen, S. &
prescription of antibiotics to pregnant women cytokine responses induced via the NFB Isolauri, E. Probiotic intervention in the first
with spontaneous preterm labour: 7year pathway. J.Pediatr. Gastroenterol. Nutr. 54, months of life: short-term effects on
follow-up of the ORACLE II trial. Lancet 372, 630638 (2012). gastrointestinal symptoms and long-term effects
13191327 (2008). 102. Rautava, S., Kalliomki, M. & Isolauri, E. on gut microbiota. J.Pediatr. Gastroenterol. Nutr.
85. Cotten, C.M. etal. Prolonged duration of initial Probiotics during pregnancy and breast-feeding 43, 200205 (2006).
empirical antibiotic treatment is associated with might confer immunomodulatory protection 119. Gueimonde, M., Kalliomki, M., Isolauri, E. &
increased rates of necrotizing enterocolitis and against atopic disease in the infant. J.Allergy Salminen, S. Probiotic intervention in
death for extremely low birth weight infants. Clin. Immunol. 109, 11911121 (2002). neonateswill permanent colonization ensue?
Pediatrics 123, 5866 (2009). 103. Fujii, T. etal. Bifidobacterium breve enhances J.Pediatr. Gastroenterol. Nutr. 42, 604606
86. Kuppala, V.S., Meinzen-Derr, J., Morrow, A.L. & transforming growth factor 1 signaling by (2006).
Schibler, K.R. Prolonged initial empirical regulating Smad7 expression in preterm infants. 120. Grzeskowiak, . etal. The impact of perinatal
antibiotic treatment is associated with adverse J.Pediatr. Gastroenterol. Nutr. 43, 8388 (2006). probiotic intervention on gut microbiota: double-
outcomes in premature infants. J.Pediatr. 159, 104. Moro, G.E. etal. Effects of a new mixture of blind placebo-controlled trials in Finland and
720725 (2011). prebiotics on faecal flora and stools in term Germany. Anaerobe 18, 713 (2012).
87. Shaw, S.Y., Blanchard, J.F. & Bernstein, C.N. infants. Acta Paediatr. Suppl. 91, 7791 (2003). 121. Kalliomki, M. etal. Probiotics in primary
Association between the use of antibiotics in the 105. Veereman-Wauters, G. etal. Physiological and prevention of atopic disease: a randomised
first year of life and pediatric inflammatory bowel bifidogenic effects of prebiotic supplements in placebo-controlled trial. Lancet 357, 10761079
disease. Am. J.Gastroenterol. 105, 26872692 infant formulae. J.Pediatr. Gastroenterol. Nutr. (2001).
(2010). 52, 763771 (2011). 122. Kalliomki, M., Salminen, S., Poussa, T.,
88. Rautava, S. & Walker, W.A. Academy of 106. Salvini, F. etal. A specific prebiotic mixture Arvilommi, H. & Isolauri, E. Probiotics and
Breastfeeding Medicine founders lecture 2008: added to starting infant formula has long-lasting prevention of atopic disease: 4year follow-up of
breastfeedingan extrauterine link between bifidogenic effects. J.Nutr. 141, 13351339 a randomised placebo-controlled trial. Lancet
mother and child. Breastfeed. Med. 4, 310 (2011). 361, 18691871 (2003).
(2010). 107. Holscher, H.D. etal. Effects of prebiotic- 123. Kalliomki, M., Salminen, S., Poussa, T. &
89. Gueimonde, M., Laitinen, K., Salminen, S. & containing infant formula on gastrointestinal Isolauri, E. Probiotics during the first 7years of
Isolauri, E. Breast milk: a source of bifidobacteria tolerance and fecal microbiota in a randomized life: a cumulative risk reduction of eczema in a
for infant gut development and maturation? controlled trial. JPEN J.Parenter. Enteral Nutr. 36, randomized, placebo-controlled trial. J.Allergy
Neonatology 92, 6466 (2007). 95S105S (2012). Clin. Immunol. 119, 10191021 (2007).
90. Grnlund, M.M. etal. Maternal breast-milk and 108. van Hoffen, E. etal. A specific mixture of short- 124. Abrahamsson, T.R. etal. Probiotics in prevention
intestinal bifidobacteria guide the compositional chain galacto-oligosaccharides and long-chain of IgE-associated eczema: a double-blind,
development of the Bifidobacterium microbiota in fructo-oligosaccharides induces a beneficial randomized, placebo-controlled trial. J.Allergy
infants at risk of allergic disease. Clin. Exp. immunoglobulin profile in infants at high risk for Clin. Immunol. 119, 11741180 (2007).
Allergy 37, 17641772 (2007). allergy. Allergy 64, 484487 (2009). 125. Wickens, K. etal. A differential effect of 2
91. Yektaei-Karin, E. etal. The stress of birth 109. Arslanoglu, S. etal. Early dietary intervention probiotics in the prevention of eczema and atopy:
enhances invitro spontaneous and IL8induced with a mixture of prebiotic oligosaccharides a double-blind, randomized, placebo-controlled
neutrophil chemotaxis in the human newborn. reduces the incidence of allergic manifestations trial. J.Allergy Clin. Immunol. 122, 788794
Pediatr. Allergy Immunol. 18, 643651 (2007). and infections during the first two years of life. (2008).
92. Grnlund, M.M. etal. Mode of delivery directs J.Nutr. 138, 10911095 (2008). 126. Niers, L. etal. The effects of selected probiotic
the phagocyte functions of infants for the first 110. Grber, C. etal. Reduced occurrence of early strains on the development of eczema (the
6months of life. Clin. Exp. Immunol. 116, atopic dermatitis because of immunoactive PandA study). Allergy 64, 13491358 (2009).
521526 (1999). prebiotics among lowatopyrisk infants. J.Allergy 127. Dotterud, C.K., Storr, O., Johnsen, R. & Oien, T.
93. Huurre, A. etal. Mode of deliveryeffects on gut Clin. Immunol. 126, 791797 (2010). Probiotics in pregnant women to prevent allergic
microbiota and humoral immunity. Neonatology 111. Duggan, C. etal. Oligofructose-supplemented disease: a randomized, double-blind trial. Br. J.
93, 236240 (2008). infant cereal: 2 randomized, blinded community- Dermatol. 163, 616623 (2010).
94. Cabrera-Rubio, R. etal. The human milk based trials in Peruvian infants. Am. J.Clin. Nutr. 128. Kim, J.Y. etal. Effect of probiotic mix
microbiome changes over lactation and is 77, 937942 (2003). (Bifidobacterium bifidum, Bifidobacterium lactis,
shaped by maternal weight and mode of delivery. 112. Szajewska, H. etal. Inulin and fructo- Lactobacillus acidophilus) in the primary
Am. J.Clin. Nutr. http://dx.doi.org/10.3945/ oligosaccharides for the prevention of antibiotic- prevention of eczema: a double-blind,
ajcn.112.037382. associated diarrhea in children: report by the randomized, placebo-controlled trial. Pediatr.
95. Perez, P.F. etal. Bacterial imprinting of the ESPGHAN working group on probiotics and Allergy Immunol. 21, 386393 (2010).
neonatal immune system: lessons from maternal prebiotics. J.Pediatr. Gastroenterol. Nutr. 54, 129. Taylor, A.L., Dunstan, J.A. & Prescott, S.L.
cells? Pediatrics 119, 724732 (2007). 828829 (2012). Probiotic supplementation for the first 6months
96. Grnlund, M.M., Grzeskowiak, ., Isolauri, E. & 113. Vandenplas, Y., De Hert, S.G. & PROBIOTICAL- of life fails to reduce the risk of atopic dermatitis
Salminen, S. Influence of mothers intestinal study group. Randomised clinical trial: the and increases the risk of allergen sensitization in
microbiota on gut colonization in the infant. Gut symbiotic food supplement Probiotical vs. high-risk children: a randomized controlled trial.
Microbes 2, 227233 (2011). placebo for acute gastroenteritis in children. J.Allergy Clin. Immunol. 119, 184191 (2007).
97. Martn, V. etal. Sharing of bacterial strains Aliment. Pharmacol. Ther. 34, 862867 (2011). 130. Kopp, M.V., Hennemuth, I., Heinzmann, A. &
between breast milk and infant feces. J.Hum. 114. Passariello, A. etal. Randomised clinical trial: Urbanek, R. Randomized, double-blind, placebo-
Lact. 28, 3644 (2012). efficacy of a new symbiotic formulation controlled trial of probiotics for primary
98. Abrahamsson, T.R., Sinkiewicz, G., Jakobsson,T., containing Lactobacillus paracasei B21060 plus prevention: no clinical effects of Lactobacillus GG
Fredrikson, M. & Bjrkstn, B. Probiotic arabinogalactan and xilooligosaccharides in supplementation. Pediatrics 121, 850856
lactobacilli in breast milk and infant stool in children with acute diarrhea. Aliment. Pharmacol. (2008).
relation to oral intake during the first year of life. Ther. 35, 782788 (2012). 131. Soh, S.E. etal. Probiotic supplementation in the
J.Pediatr. Gastroenterol. Nutr. 49, 349354 115. Kukkonen, K. etal. Probiotics and prebiotic first 6months of life in at risk Asian infants
(2009). galacto-oligosaccharides in the prevention of effects on eczema and atopic sensitization at
99. Verhasselt, V. etal. Breast milk-mediated transfer allergic diseases: a randomized, double-blind, the age of 1year. Clin. Exp. Allergy 39, 571578
of an antigen induces tolerance and protection placebo-controlled trial. J.Allergy Clin. Immunol. (2009).
from allergic asthma. Nat. Med. 14, 170175 119, 192198 (2007). 132. Huurre, A., Laitinen, K., Rautava, S.,
(2008). 116. Van der Aa, L.B. etal. Effect of a new symbiotic Korkeamki,M. & Isolauri, E. Impact of maternal
100. Rautava, S. etal. Breast milk-transforming growth mixture on atopic dermatitis in infants: atopy and probiotic supplementation during
factor2 specifically attenuates IL1-induced a randomized-controlled trial. Clin. Exp. Allergy pregnancy on infant sensitization: a double-blind
inflammatory responses in the immature human 40, 795804 (2010). placebo-controlled study. Clin. Exp. Allergy 38,
intestine via an SMAD6- and ERK-dependent 117. Schultz, M., Gttl, C., Young, R.J., Iwen, P. & 13421348 (2008).
mechanism. Neonatology 99, 192201 (2011). Vanderhoof, J.A. Administration of oral probiotic 133. Luoto, R., Laitinen, K., Nermes, M. & Isolauri, E.
101. Rautava, S., Lu, L., Nanthakumar, N.N., Dubert- bacteria to pregnant women causes temporary Impact of maternal probiotic-supplemented
Ferrandon, A. & Walker, W.A. TGF2 induces infantile colonization. J.Pediatr. Gastroenterol. dietary counselling on pregnancy outcome and
maturation of immature human intestinal Nutr. 38, 293297 (2004). prenatal and postnatal growth: a double-blind,

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 9 | OCTOBER 2012 | 575


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

placebo-controlled study. Br. J.Nutr. 103, 137. Bjrkstn, B. etal. Collecting and banking lactational infectious mastitis. BMCInfect. Dis.
17921799 (2010). human milk: to heat or not to heat? Br. Med. J. 8, 51 (2008).
134. Laitinen, K., Poussa, T. & Isolauri, E. Nutrition, 281, 765769 (1980). 143. Jimnez, E. etal. Assessment of the bacterial
Allergy, Mucosal Immunology and Intestinal 138. Martn, R. etal. Human milk is a source of lactic diversity of human colostrum and screening of
Microbiota Group. Probiotics and dietary acid bacteria for the infant gut. J.Pediatr. 143, staphylococcal and enterococcal populations for
counselling contribute to glucose regulation 754758 (2003). potential virulence factors. Res. Microbiol. 159,
during and after pregnancy: a randomised 139. Heikkil, M.P. & Saris, P.E. Inhibition of 595601 (2008).
controlled trial. Br. J.Nutr. 101, 16791687 Staphylococcus aureus by the commensal 144. Martn, R. etal. Isolation of bifidobacteria from
(2009). bacteria of human milk. J.Appl. Microbiol. 95, breast milk and assessment of the
135. Ilmonen, J., Isolauri, E., Poussa, T. & Laitinen, K. 471478 (2003). bifidobacterial population by PCR-denaturing
Impact of dietary counselling and probiotic 140. Beasley, S.S. & Saris, P.E. Nisin-producing gradient gel electrophoresis and quantitative
intervention on maternal anthropometric Lactococcus lactis strains isolated from human real-time PCR. Appl. Environ. Microbiol. 75,
measurements during and after pregnancy: milk. Appl. Environ. Microbiol. 70, 50515053 965969 (2009).
a randomized placebo-controlled trial. Clin. Nutr. (2004). 145. Hunt, K.M. etal. Characterization of the
30, 156164 (2011). 141. Martn, R. etal. Cultivation-independent diversity and temporal stability of bacterial
136. Luoto, R., Kalliomki, M., Laitinen, K. & assessment of the bacterial diversity of breast communities in human milk. PLoS ONE 6,
Isolauri,E. The impact of perinatal probiotic milk among healthy women. Res. Microbiol. 158, e21313 (2011).
intervention on the development of overweight 3137 (2007).
and obesity: follow-up study from birth to 142. Delgado, S., Arroyo, R., Martn, R. & Author contributions
10years. Int. J.Obes. (Lond). 34, 15311537 Rodrguez,J.M. PCR-DGGE assessment of the All authors contributed equally to all aspects of the
(2010). bacterial diversity of breast milk in women with manuscript.

576 | OCTOBER 2012 | VOLUME 9  www.nature.com/nrgastro


2012 Macmillan Publishers Limited. All rights reserved

Вам также может понравиться