You are on page 1of 8

Available online at www.sciencedirect.

com

Programming infant gut microbiota: influence of dietary and


environmental factors
Tatiana Milena Marques1,2,3, Rebecca Wall1, R Paul Ross1,2,
Gerald F Fitzgerald1,3, C Anthony Ryan4 and Catherine Stanton1,2

The neonatal period is crucial for intestinal colonisation, and the However, using sequencing techniques Wang et al. [7]
composition of this ecosystem in early life is influenced by such found that 10% of species from faecal samples of infants
factors as mode of birth, environment, diet and antibiotics. The after the first two months of life were unidentifiable,
intestinal microbiota contributes to protection against whereas 30% unidentified species were observed after
pathogens, maturation of the immune system and metabolic the first year of life, highlighting the complexity of the
welfare of the host, but under some circumstances can microbiota and the importance of the development of
contribute to the pathogenesis of certain diseases. Because new and more powerful fingerprinting techniques. In a
colonisation with non-pathogenic microbiota is important for more recent study by Rajilić-Stojanović et al. [11], a
infant health and may affect health in later life, it is important to phylogenetic microarray (referred to as the human intes-
understand how the composition of this microbial organ is tinal tract chip or ‘HITChip’) was developed and applied
established and by which dietary means (e.g. supplementation for comparing the effect of ageing on the intestinal
with prebiotics/probiotics/food ingredients) it can be microbiota of young and elderly adults. Because of the
programmed in order to achieve an ecosystem that is valuable good reproducibility and the possibility for relative
for the host. quantification of microbial groups, this technique might
Addresses be a suitable tool for determining the microbial diversity
1
2
Alimentary Pharmabiotic Centre (APC), Co. Cork, Ireland of the infant gastrointestinal tract in future studies.
Teagasc, Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland Another high-throughput alternative test is the recently
3
Department of Microbiology, University College Cork, Ireland
4
Department of Paediatrics and Child Health, University College Cork,
launched GA-mapTM microarray that will enable screen-
Ireland ing of the infant gut microbiota based on sets of unique
probes that are highly specific to their target group of
Corresponding author: Stanton, Catherine bacteria. It is envisaged that by providing an ‘overall map’
(catherine.stanton@teagasc.ie)
of the enteric microbiota, this test will give valuable
information to assist in disease intervention [12].
Current Opinion in Biotechnology 2010, 21:149–156
The mutualistic interactions between the enteric micro-
This review comes from a themed issue on
Food biotechnology
biota and the human host are essential for health [13].
Edited by Dietrich Knorr and Carmen Wacher Indeed, the enteric microbiota can secrete molecules (so-
called ‘pharmabiotics’ [14]) that inhibit host pathogens,
metabolise compounds that harm the host to less toxic
0958-1669/$ – see front matter substances [13,15] and produce a range of bioactive com-
# 2010 Elsevier Ltd. All rights reserved. pounds such as conjugated linoleic acid (CLA), short chain
fatty acids (SCFA) and gamma-aminobutyric acid (GABA)
DOI 10.1016/j.copbio.2010.03.020 that may play a role in the protection from lifestyle illnesses
such as cancer, obesity and cardiovascular diseases [15].
Moreover, the microbiota contribute to biochemical path-
Introduction ways that humans cannot process because of the lack of
Microbial colonisation of the sterile infant intestine is an proper genes [16], such as fermentation of indigestible
intricate process which is influenced by many factors [1] dietary polysaccharides, metabolism of complex proteins
including mode of delivery [2], type of feeding [3] and and synthesis of vitamins [15,17]. The infant gut micro-
antibiotic therapy [4,5]. Within the first year of life, the biota can also significantly influence the maturation of the
enteric microflora is highly dynamic but microbial diver- immune system in early days of life [14,18,19]. Remark-
sity is low, and after the initial year, the microbial popu- ably, colonisation of the newborn intestine plays a key role
lation stabilises and resembles that of the adult [6]. Whilst in the development and fine-tuning of the intestinal
traditional culture-based techniques have been used in immune responses. Disruption to this process, due for
the past to determine the microbial load of the infant example to antibiotic therapy, may have long-term health
intestine, less biased DNA-based techniques including consequences, giving rise to immune-related disorders
the use of the 16s ribosomal RNA gene have recently such as eczema, allergic rhinitis and inflammatory bowel
confirmed the dominance of species of Bifidobacterium, disease (IBD) [18,20]. For example, in a study conducted
Clostridium and Bacteroides in the early microbiota [7–10]. by Wang et al., the intestinal microbial diversity of

www.sciencedirect.com Current Opinion in Biotechnology 2010, 21:149–156


150 Food biotechnology

18-month-old infants suffering from atopic eczema was Mode of delivery and type of feeding exert the most
reduced in comparison to healthy infants of the same age significant influences on the development of the micro-
[21]. biota in the infant [15,30,32]. Vaginally born infants are
initially colonised by faecal and vaginal bacteria from the
As the infant enteric microbiota is more variable in its mother, whereas infants born via caesarean section are
composition and less stable over time compared to the colonised by bacteria from the hospital environment
adult [6], the use of nutritional strategies in order to (health-care workers, air, equipment, other newborns)
shape/programme its composition to favour a more [10,28,30,31]. Newborns delivered by caesarean section
beneficial bacterial population may be a good opportunity have in general lower numbers of bifidobacteria, reduced
to avoid future health problems. Probiotics and prebiotics levels of members of the Bacteroides fragilis group and
are widely used as supplements in infant formulae and higher amounts of Clostridium difficile compared to vagin-
many studies have confirmed their efficacy in changing ally born infants [10,30,33]. Moreover, the growth of
the microbiota composition by stimulating the growth of Bacteroides, Bifidobacterium and Escherichia coli is delayed
bifidobacteria [22] and therefore helping in the treatment in infants born by caesarean section [29–31].
and prevention of certain illnesses [23,24].
Traditionally, it has been considered that the microbiota
This review will discuss the current knowledge of the of breast-fed infants are dominated by bifidobacteria and
microbial diversity in infants and the metabolic capabili- also colonised in lesser quantities by some facultative
ties that the enteric microbiota possess. Furthermore, the anaerobic bacteria such as streptococci, staphylococci,
impact of diet and dietary supplementation (with probio- enterococci, lactobacilli and enterobacteria, whilst the
tics and prebiotics) on the evolution of the microbial microbiota of formula-fed infants are more diverse and
diversity in the developing infant will be reviewed. include bacterial groups such as Bacteroides, Clostridium
and Enterobacteriaceae [10,15,33,34]. However, the effect
Development of the infant gut microbiota of diet on the composition of the infant gut microbiota,
At birth, the newborn infant gastrointestinal tract is almost more specifically in the predominance of bifidobacteria in
sterile [25–27], but is rapidly colonised in the first days of breast-fed infants is still controversial. Some reports have
life, reaching a stable population similar to that of an adult not found differences amongst the types of feeding [6,29]
when the infant is around two years old and there is the and even suggested that modern formulae are more
introduction of solid foods [6,13,15,28]. Immediately after faithful replicas of breast milk, with the addition of
birth, the newborn gut environment is colonised by facul- prebiotics such as galactooligosaccharides (GOS) and
tative anaerobic bacteria such as Enterobacteriaceae, strep- fructooligosaccharides (FOS), increasing the number of
tococci and staphylococci [10,29,30]. These first colonisers bifidobacteria and lactobacilli in the gut of formula-fed
belong to species with pathogenic potential and might be infants to a similar number found in breast-fed infants
harmful [30], however most interactions between humans [10,29,35].
and microorganisms do not result in disease [13]. Instead,
these bacteria gradually consume oxygen and produce new The gut colonisation pattern of preterm infants differs
metabolites, preparing the intestinal environment for the from that of full-term infants both temporally and quali-
establishment of a strict anaerobic bacterial population tatively [36]. The several immaturities of the preterm
dominated by bifidobacteria, Clostridium and Bacteroides, infant gut, the long time spent in the neonatal intensive
bacterial groups that may play a role in the neonatal gut care unit and the use of broad spectrum antibiotics delay
maturation [10,29,30]. Recently, the more strict hygienic the establishment of a beneficial bacterial community and
conditions during delivery, short hospital stays and practise enable the growth of potentially pathogenic bacteria
of rooming-in (where the mother, not health-care workers [33,36]. Antibiotics negatively affect the composition of
mainly handles the baby) have reduced bacterial exposure the infant gut microbiota by decreasing the numbers of
and altered the colonisation pattern with skin-derived obligate anaerobes (e.g. bifidobacteria and Bacteroides)
staphylococci been the first colonisers of the infant gut [28,33]. However, the effects differ between antibiotics
instead of faecal Enterobacteriaceae [29,30]. [10] and usually most families and genera of gut micro-
organisms return to typical levels within weeks of
The bacterial community colonising the newborn infant exposure [13].
gut is dynamic and originates from the environment,
mainly from the mother [10,13]. A broad range of factors Implications of microbiota for host health
can affect the bacterial composition of the gut including The enteric microbiota play an important role in host
mode of delivery (caesarean section or vaginally), type of health, being involved in nutritional, immunological and
feeding (exclusive breast-feeding versus formula), gesta- physiological functions. Along the epithelium, enteric
tional age, antibiotic use, hospitalisation, surrounding bacteria complement the natural defence barrier against
environment and maternal infection or illness [10,28–31] exogenous microbes, thereby preventing invasion by
(Figure 1). pathogens. In addition, the enteric microbiota have an

Current Opinion in Biotechnology 2010, 21:149–156 www.sciencedirect.com


Programming infant gut microbiota Marques et al. 151

Figure 1

Factors affecting the infant gut composition and the differences in the bacterial colonisation influenced by the delivery mode and the diet.

important role in influencing the normal structural and Because the microbiota influences the developing
functional development of the mucosal immune system immune system, variations from the normal bacterial
[37]. The molecular interactions between enteric bac- colonisation pattern through modern strategies such as
teria and the host seem to direct the development of caesarean section, formula-based diet, hygiene and the
immune responses, and in turn the immune system excessive use of antibiotics in infants, may change the
shapes the composition of the microbiota. Most of the outcome of immune development and thus potentially
information regarding the effects of the microbiota on the predispose to certain inflammatory diseases in later life.
host immune system comes from studies using germ-free Indeed, increased rates of several immune-mediated
animals. Germ-free animals show extensive defects in disorders, such as IBD, atopy, asthma and rheumatoid
the development of the gut-associated lymphoid tissue arthritis have occurred in recent years in the ‘Western’
with fewer and smaller Peyer’s patches without germinal populations. This increase may in part, be because of
centres and smaller T cell zones. Furthermore, the changes in host–microbe interactions caused by imple-
lamina propria contains essentially no immunoglobulin mentation of different antimicrobial strategies (i.e. exces-
A (IgA), plasma cells or CD4 cells, and intraepithelial sive use of antibiotics, hygiene and Western diets).
lymphocytes are also rare compared with conventional
animals [38]. Germ-free animals are therefore more Recent evidence from animal and human studies indicate
susceptible to infection compared with conventional that the composition of the gut microbiota may also be
animals. However, colonisation of animals with a single involved in several extra-intestinal disorders such as
bacterium (Bacteroides fragilis) capable of producing poly- obesity [40–42], insulin resistance [43] and non-alcoholic
saccharide A (PSA) has been shown to correct mucosal fatty liver disease (NAFLD) [44]. For example, conven-
and systemic immune defects, including correcting tionally raised mice have been shown to have 40% more
systemic T cell deficiencies and restoring the balance body fat than their germ-free counterparts and colonisation
between TH cell subsets [39]. with a normal gut microbiota induces hepatic lipogenesis

www.sciencedirect.com Current Opinion in Biotechnology 2010, 21:149–156


152 Food biotechnology

and increases lipid storage in adipocytes [42]. Moreover, it ings open up a new field of research for modulating the fatty
has been reported that differences in the gut microbiota acid composition of distal tissues [47].
during the first year of life may precede the onset of obesity
[45]. In this study, the numbers of Bifidobacterium spp. were Metabolite production by gut bacteria
higher and the numbers of Staphylococcus aureus were lower The human enteric microbiota can exert beneficial health
in children who remained at normal weight than in children effects through the production of bacterial metabolites or
who became overweight. Thus, a microbiota profile in ‘pharmabiotics’, most often small molecules which inter-
favour of a higher number of bifidobacteria and a lower act with ‘intelligent communication’ systems in the body
number of S. aureus in infancy may provide protection including those which are immune, endocrine and
against overweight and obesity development. Since anti- neuronal-based [14]. Commensal bacteria have been
biotic treatment and caesarean section result in lower shown to synthesise vitamins that are essential for human
numbers of bifidobacteria [10,33], this may lead to an survival such as vitamins K2 and B12 [17], polyunsaturated
increased risk for the development of obesity in later life. fatty acids (PUFA) such as conjugated a-linolenic acid
It has also been demonstrated that the gut microbiota does (CALA) and CLA, SCFA, neuroactive compounds such
not only affect fat quantity in the host, but also fat com- as GABA and histamine [48], PSA [49] and a variety of
position. We reported that administration of a CLA-produ- other proteins, peptides and nucleotides with immuno-
cing bifidobacterial strain in combination with linoleic acid modulatory and anti-inflammatory properties [50]. The
resulted in modulation of the fatty acid composition of the effects of some of these compounds on human health are
host, including significantly elevated concentrations of c9, briefly reviewed below and summarised in Table 1.
t11 CLA in the liver [46]. Furthermore, changes in fatty
acid composition were not only limited to CLA but SCFA are the end products of the bacterial fermentation
included changes in the content of eicosapentaenoic acid of carbohydrates in the gastrointestinal tract [16,48,51].
(EPA) and docosahexaenoic acid (DHA), alterations that SCFA are important for human metabolism since they
were coupled with an anti-inflammatory cytokine profile increase the amount of energy intake, stimulate water and
[46]. Another recent study involving a comparison of con- sodium absorption, lower luminal pH and the bioavail-
ventionally raised and germ-free mice reported that the gut ability of toxic amines [51]. In a recent study, Maslowski
microbiota have the ability to affect lens and retinal lipid et al. [52] have shown that SCFA bind to the G-protein-
composition. The distinct lipid profiles suggest that con- coupled receptor 43 (GPR43) and this interaction may
ventional mice are exposed to more oxidative stress than affect immune and inflammatory responses. Moreover,
germ-free mice, decreasing the lens lifespan. These find- butyrate is the primary energy source for colonocytes [22]

Table 1

Examples of metabolite-producing bacterial strains and their possible health effects

Metabolite Bacterial strain Health effects Reference


Vitamin K2 Bacteroides fragilis Modulation of bone mineralisation [54,56,70]
"Bone mineral density
#Fracture risk
Modulation of blood coagulation
#Risk of cardiovascular disease
#Risk of melena neonatorum
#Risk of intracranial hemorrhagic disease
Vitamin B12 Lactobacillus reuteri Stimulation of nervous system development [59,61]
Success to thrive
#Risk of neurological disorders
#Risk of haematological abnormalities
Conjugated linoleic acid (CLA) Bifidobacterium breve Modulation of the immune system [63,71]
B. longum #Carcinogenesis
#Atherosclerosis
#Inflammation
#Obesity
#Diabetes
g-Aminobutyric acid (GABA) Lactobacillus brevis Central nervous system inhibition (inhibitory neurotransmitter) [66,67]
L. paracasei "Hypotension
"Diuresis
Polysaccharide A (PSA) Bacteroides fragilis #Weight loss [36,48]
#Epithelial cell hyperplasia
Modulation of the immune system
#Cytokines levels
#Neutrophil infiltration

Current Opinion in Biotechnology 2010, 21:149–156 www.sciencedirect.com


Programming infant gut microbiota Marques et al. 153

and has been in the focus of studies because of its possible cell hyperplasia and neutrophil infiltration to the gut
action in the prevention of colon cancer [16]. [36,48].

Vitamin K2 or menaquinone is a lipophilic vitamin that can Diet and the enteric microbiota
be found in fermented foods and in the colon where it is Diet can influence the composition of the intestinal
synthesised by the gut microbiota [53,54]. It is an essen- microbiota, especially in the first days of life when the
tial cofactor for the enzyme responsible for the modifi- bacterial population is not yet established [23]. The infant
cation of specific glutamyl residues to gamma- microbiota is naturally shaped during breast-feeding but
carboxyglutamyl (Gla) residues in precursor proteins that the colonisation pattern can also be manipulated towards
possess the appropriate gamma-carboxylation recognition a more beneficial community using dietary supplement-
signal within the propeptide region [54–57] such as ation with probiotics and/or prebiotics.
vertebrate proteins involved in blood coagulation and
bone mineralisation [54]. Therefore, menaquinone is Human breast milk is considered the best nutritional
important for bone and vascular health, and deficiency option for growth and health development of newborn
has been associated with low bone mineral density, infants, since it contains a wide range of protective
increased fracture risk, increased risk of cardiovascular compounds including carbohydrates, nucleotides, fatty
diseases, melena neonatorum and intracranial hemorrha- acids, immunoglobulins, cytokines, intact immune cells
gic disorders in newborn infants [53–55]. and other immune-modulatory factors [35,51,72–74].
Moreover, breast milk seems to be a continuous source
Vitamin B12 or cobalamin is a compound required by of bacteria for the infant gut [34], with milk from healthy
humans but solely synthesised by some bacteria and mothers containing up to 109 microbes/L from different
archaea [58–60]. During foetal life and infancy, this bacterial groups [28,30]. Commensal bacteria usually
vitamin is important for rapid and normal growth and present in breast milk include staphylococci, streptococci,
for the development of the nervous system [60,61]. Its bifidobacteria and lactic acid bacteria [28,30,34]. Bacteria
deficiency in infants is related to maternal deficiency and with potential probiotic effects have been isolated from
may cause failure to thrive, delay development or human milk such as L. gasseri, L. rhamnosus, L. plantarum,
regression and lead to progressive or permanent neuro- L. fermentum, Enterococcus faecium, [74] and some bifido-
logical disorders and haematological abnormalities [60– bacterial species (B. breve, B. adolescentis and B. bifidum)
62]. [34].

CLA is a mixture of conjugated isomers of the essential Due to the positive effects of human breast milk especi-
fatty acid linoleic acid that have been reported to be ally in the maturation of the immune system of the
produced by some human strains from different bacterial newborn [31], many attempts have been made to develop
groups such as Lactobacillus, Propionibacterium, Bifidobac- artificial formula that stimulate gut colonisation similar to
terium, Pediococcus, Enterococcus and Lactococcus [63]. CLA that of breast-fed infants, with high numbers of bifido-
has been shown to exert such health properties as antic- bacteria [35,51]. Strategies used in the supplementation
arcinogenic, anti-inflammatory and immunomodulatory of the basic infant formula include the incorporation of
effects (reviewed in Bhattacharya et al. [64]). Because probiotics and/or prebiotics.
of its anti-inflammatory properties, CLA may be a poten-
tial therapeutic option for the prevention of necrotising Probiotics and prebiotics
enterocolitis in preterm infants [65]. Probiotics are defined as ‘live microorganisms’ which,
when administered in adequate amounts confer a health
GABA is a non-protein amino acid that acts as a major benefit on the host’ [75]. The most common groups of
inhibitory neurotransmitter in the central nervous system bacteria used as probiotics are bifidobacteria and lacto-
and exerts several other physiological functions such as bacilli [23]. Multiple mechanisms of probiotic action
induction of hypotension and diuresis [66,67]. GABA is have been suggested, however mechanisms are strain-
produced by various microorganisms, especially lactoba- dependent [22]. Probiotics may prevent the penetration
cilli [48] and has been used in the development of of pathogens in the human gut by increasing the pro-
functional foods such as fermented meats, pickles, cheese duction of mucin, reducing the gut permeability, releas-
[66,67] and yogurt [68]. ing antimicrobial compounds or modulating the immune
system [22]. Several studies have demonstrated that
PSA is a polysaccharide produced by the human symbio- probiotic preparations are safe and effective when admi-
tic Bacteroides fragilis [69]. Animal studies have shown that nistered to children, aiding in the treatment and preven-
PSA has several immunomodulatory activities and can tion of many types of diarrhoea [23,24,51], reducing the
ameliorate inflammatory diseases. Treatment with pur- incidence and severity of necrotising enterocolitis
ified PSA protects mice against IBD, avoiding weight loss, [24,33,76,77], alleviating symptoms of lactose malabsorp-
decreasing levels of cytokines and inhibiting epithelial tion and preventing atopic diseases and food allergies in

www.sciencedirect.com Current Opinion in Biotechnology 2010, 21:149–156


154 Food biotechnology

infants [23]. However, probiotic concentration and via- time, which raises the possibility that less than daily
bility can be affected during food processing, storage and dosing may be adequate to achieve colonisation of the
ingestion [22,23]. Therefore, the possibilities of using moderately preterm infant gut with a microbiota resem-
prebiotic as a food supplement are much wider [23]. bling that of the healthy term infant. However, more
research is necessary in order to evaluate how such a
Prebiotics are non-digestible food ingredients that selec- strategy will affect long-term health status. The incidence
tively stimulate the growth of bifidobacteria and lactoba- of childhood obesity has increased during the past years
cilli [22,23]. The two prebiotic substrates commonly and recent studies have suggested that environmental
added to infant formulae are FOS and GOS [23]. Admin- factors such as mode of delivery, type of feeding and use
istration of prebiotics is often associated with a lower of antibiotics which are known to affect the composition
faecal pH and changes in the SCFA pattern [35]. Pre- of the gut microbiota may lead to an increased risk for the
biotics increase the production of SCFA, improving the development of obesity later in life [28,42]. However,
host absorption of minerals and facilitating host metab- more well-designed clinical studies are required to
olism [22]. Moreover, by changing the composition of the confirm the impact of microbiota composition on the
gut microbiota, prebiotics play a role in the development development of such conditions as obesity.
of the newborn immune system and protection against
pathogens [22,35]. It has been demonstrated that pre- Acknowledgements
biotics can increase the number of bifidobacteria in the This work was supported in part, by Science Foundation Ireland, The
European Union (Project KBBE-211911), the Irish Ministry for Food and
gut, and are applicable in infant nutrition [22]. Agriculture, Enterprise Ireland, the Higher Education Authority and the
Health Research Board of Ireland and the Irish Government under the
Another dietary strategy is the combination of probiotics National Development Plan 2000–2006. TMM is a student funded by the
Alimentary Pharmabiotic Centre (APC).
and prebiotics, called synbiotic. Synbiotics positively
affect the host by introducing new beneficial bacteria References
to the indigenous gut population whilst ensuring its
1. O’ Toole PW, Claesson MJ: Gut microbiota: changes
growth and selectively stimulating the growth of the other throughout the lifespan from infancy to elderly. Int Dairy J 2010,
endogenous health-promoting bacteria, such as bifidobac- 20:281-291.
teria [23,51]. 2. Gronlund MM, Lehtonen OP, Eerola E, Kero P: Fecal
microflora in healthy infants born by different methods
of delivery: permanent changes in intestinal flora after
Conclusions caesarean delivery. J Pediatr Gastroenterol Nutr 1999,
Early colonisation of the infant gut is undoubtedly an 28:19-25.
important factor for the overall health of the infant and 3. Orrhage K, Nord CE: Factors controlling the bacterial
may also have effects on the health status in later life. colonization of the intestine in breast-fed infants. Acta Pediatr
1999, 430:S43-S54.
Indeed, the commensal microbiota have been implicated
4. Kalenic S, Francetic I, Polak J, Zele-Starcevic L, Bencic Z: Impact
in many diseases that occur within the gastrointestinal of ampicillin and cefuroxime on bacterial colonisation and
tract and more recently have also been shown to be infection in patients on a neonatal intensive care unit. J Hosp
involved in disorders outside the gut such as obesity, Infect 1993, 23:35-41.

diabetes and atopic allergies. Several factors have been 5. Burman LG, Berglund B, Huovinen P, Tullus K: Effect of
ampicillin versus cefuroxime on the emergence of
shown to promote a greater microbial diversity in infants beta-lactam resistance in fecal Enterobacter cloacae
including breast milk feeding, vaginal delivery and isolates from neonates. J Antimicrob Chemother 1999,
31:111-116.
decreased antibiotic use. Moreover, by programming
the infant gut microbiota using prebiotics, probiotics 6. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO:
Development of the human infant intestinal microbiota. PLoS
and/or synbiotics and thus accomplishing a more Biol 2007, 5:e177.
beneficial composition, several important diseases that
7. Wang M, Ahrne S, Antonsson M, Molin G: T-RFLP combined with
develop in early life such as necrotising enterocolitis and principal component analysis and 16S rRNA gene sequencing:
atopic eczema may be prevented. Although current data an effective strategy for comparison of fecal microbiota in
infants of different ages. J Microbiol Method 2004,
support the use of probiotics in infants, adequately pow- 59:53-69.
ered prospective studies that document changes in the 8. Hopkins MJ, Macfarlane GT, Furrie E, Fite A, Macfarlane S:
microbiota in response to diet and antibiotics and incorp- Characterisation of intestinal bacteria in infant stools using
orate long-term health status outcomes are keenly real-time PCR and northern hybridisation analyses. FEMS
Microbiol Ecol 2005, 54:77-85.
awaited. The intake limits (e.g. optimal dose, frequency,
timing of administration), variability between different 9. Sakata S, Tonooka T, Ishizeki S, Takada M, Sakamoto M,
Fukuyama M, Benno Y: Culture-independent analysis of fecal
probiotic strains and specific effects for different popu- microbiota in infants, with special reference to
lations (e.g. infants, elderly, pregnant women) are some Bifidobacterium species. FEMS Microbiol Lett 2005,
243:417-423.
aspects that have to be evaluated. For example, in a
10. Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, van
recent study by Mc Gee et al. [78], a single oral dose of den Brandt PA, Stobberingh EE: Factors influencing the
a probiotic led to colonisation of the neonatal intestinal composition of the intestinal microbiota in early infancy.
tract in low birth weight infants for an extended period of Pediatrics 2006, 118:511-521.

Current Opinion in Biotechnology 2010, 21:149–156 www.sciencedirect.com


Programming infant gut microbiota Marques et al. 155

11. Rajilic-Stojanovic M, Heilig HGHJ, Molenaar D, Kajander K, 32. Butel M-J, Suau A, Campeotto F, Magne F, Aires J, Ferraris L,
Surakka A, Smidt H, de Vos WM: Development and application Kalach N, Leroux B, Dupont C: Conditions of bifidobacterial
of the human intestinal tract chip, a phylogenetic microarray: colonization in preterm infants: a prospective analysis. J
analysis of universally conserved phylotypes in the abundant Pediatr Gastroenterol Nutr 2007, 44:577-582.
microbiota of young and elderly adults. Environ Microbiol 2009,
11:1736-1751. 33. Martin CR, Walker WA: Probiotics: role in pathophysiology and
prevention in necrotizing enterocolitis. Semin Perinatol 2008,
12. Genetic Analysis AS: New infant gut microbiota screening test 32:127-137.
provides key tool for disease development investigation.
Medial News Today 2009 http://www.medicalnewstoday.com/ 34. Martin R, Jimenez E, Heilig H, Fernandez L, Marin ML, Zoetendal EG,
articles/172575.php. Rodriguez JM: Isolation of bifidobacteria from breast milk and
assessment of the bifidobacterial population by PCR-DGGE
13. Dethlefsen L, McFall-Ngai M, Relman DA: An ecological and and qRTi-PCR. Appl Environ Microbiol 2008, 75:965-969.
evolutionary perspective on human–microbe mutualism and
disease. Nature 2007, 449:811-818. 35. Boehm G, Moro G: Structural and functional aspects of
prebiotics used in infant nutrition. J Nutr 2008, 138:1818S-1828S.
14. Shanahan F, Stanton C, Ross P, Hill C: Pharmabiotics:
Bioactives from Mining Host–Microbe–Dietary Interactions. 36. Mshvildadze M, Neu J, Mai V: Intestinal microbiota
Funct Food Rev 2009, 1:1-6. development in the premature neonate: establishment of a
lasting commensal relationship? Nutr Rev 2008, 66:658-663.
15. Wall R, Ross RP, Ryan CA, Hussey S, Murphy B, Fitzgerald GF,
Stanton C: Role of gut microbiota in early infant development. 37. Round JL, Mazmanian SK: The gut microbiota shapes intestinal
Clin Med: Pediatr 2009, 3:45-54. immune responses during health and disease. Nat Rev
Immunol 2009, 9:313-323.
16. Kovatcheva-Datchary P, Zoetendal EG, Venema K, de Vos WM,
Smidt H: Review: tools for the tract: understanding the 38. Macpherson AJ, Harris NL: Interactions between commensal
functionality of the gastrointestinal tract. Ther Adv intestinal bacteria and the immune system. Nat Rev Immunol
Gastroenterol 2009, 2:s9-s22. 2004, 4:478-485.

17. Resta SC: Effects of probiotics and commensals on intestinal 39. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL: An
epithelial physiology: implications for nutrient handling. immunomodulatory molecule of symbiotic bacteria directs
J Physiol 2009, 587:4169-4174. maturation of the host immune system. Cell 2005, 122:107-118.

18. Kelly D, King T, Aminov R: Importance of microbial colonization 40. Ley RE, Turnbaugh PJ, Klein S, Gordon JI: Microbial ecology:
of the gut in early life to the development of immunity. Mutat human gut microbes associated with obesity. Nature 2006,
Res 2007, 622:58-69. 444:1022-1023.

19. Gottrand F: Long-chain polyunsaturated fatty acids influence 41. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER,
the immune system of infants. J Nutr 2008, 138:1807S-1812. Gordon JI: An obesity-associated gut microbiome with
increased capacity for energy harvest. Nature 2006, 444:1027-
20. Conroy ME, Shi HN, Walker WA: The long-term health effects of 1131.
neonatal microbial flora. Curr Opin Allergy Clin Immunol 2009,
9:197-201. 42. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A,
Semenkovich CF, Gordon JI: The gut microbiota as an
21. Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, environmental factor that regulates fat storage. Proc Natl Acad
Strachan DP, Martricardi PM, Aberg N, Perkin MR, Tripodi S et al.: Sci USA 2004, 101:15718-15723.
Reduced diversity in the early fecal microbiota of infants with
atopic eczema. J Allergy Clin Immunol 2008, 121:129-134. 43. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM,
Burcelin RM: Changes in gut microbiota control metabolic
22. Saulnier DMA, Spinler JK, Gibson GR, Versalovic J: Mechanisms endotoxemia-induced inflammation in high-fat diet induced
of probiosis and prebiosis: considerations for enhanced obesity and diabetes in mice. Diabetes 2008, 57:1470-1481.
functional foods. Curr Opin Biotechnol 2009, 20:135-141.
44. Dumas M-E, Barton RH, Toye A, Cloarec O, Blancher C,
23. Parracho H, McCartney AL, Gibson GR: Probiotics and Rothwell A, Fearnside J, Tatoud R, Blanc VR, Lindon JC et al.:
prebiotics in infant nutrition. Proc Nutr Soc 2007, Metabolic profiling reveals a contribution of gut microbiota to
66:405-411. fatty liver phenotype in insulin-resistant mice. Proc Natl Acad
Sci 2006, 103:12511-12516.
24. O’Hara A, Shanahan F: Mechanisms of action of probiotics in
intestinal diseases. TheScientificWorldJOURNAL 2007, 7:31-46. 45. Kalliomaki M, Carmen Collado M, Salminen S, Isolauri E: Early
differences in fecal microbiota composition in children may
25. Jiménez E, Marı́n ML, Martı́n R, Odriozola JM, Olivares M, Xaus J, predict overweight. Am J Clin Nutr 2008, 87:534-538.
Fernández L, Rodrı́guez JM: Is meconium from healthy
newborns actually sterile? Res Microbiol 2008, 159:187-193. 46. Wall R, Ross RP, Shanahan F, O’Mahony L, O’Mahony C,
Coakley M, Hart O, Lawlor P, Quigley EM, Kiely B et al.: Metabolic
26. Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E: activity of the enteric microbiota influences the fatty acid
Bifidobacterium and Lactobacillus DNA in the human composition of murine and porcine liver and adipose tissues.
placenta. Lett Appl Microbiol 2009, 48:8-12. Am J Clin Nutr 2009, 89:1393-1401.
27. DiGiulio DB, Romero R, Amogan HP, Kusanovic JP, Bik EM, 47. Oresic M, Seppänen-Laakso T, Yetukuri L, Bäckhed F,
Gotsch F, Kim CJ, Erez O, Edwin S, Relman DA: Microbial Hänninen V: Gut microbiota affects lens and retinal lipid
prevalence, diversity and abundance in amniotic fluid during composition. Exp Eye Res 2009, 89:604-607.
preterm labor: a molecular and culture-based investigation.
PLoS ONE 2008, 3:e3056. 48. Forsythe P, Sudo N, Dinan T, Taylor VH, Bienenstock J: Mood and
gut feelings. Brain, Behav, Immun 2010, 24:9–16.
28. Reinhardt C, Reigstad CS, Bäckhed F: Intestinal microbiota
during infancy and its implications for obesity. J Pediatr 49. Mazmanian SK, Round JL, Kasper DL: A microbial symbiosis
Gastroenterol Nutr 2009, 48:249-256. factor prevents intestinal inflammatory disease. Nature 2008,
453:620-625.
29. Adlerberth I, Wold A: Establishment of the gut microbiota in
Western infants. Acta Paediatr 2009, 98:229-238. 50. Shanahan F, Kiely B: The gut microbiota and disease — an
inner repository for drug discovery. Drug Discov Today: Ther
30. Morelli L: Postnatal development of intestinal microflora as Strat 2007, 4:195-200.
influenced by infant nutrition. J Nutr 2008, 138:1791S-1795S.
51. Puccio G, Cajozzo C, Meli F, Rochat F, Grathwohl D, Steenhout P:
31. Biasucci G, Benenati B, Morelli L, Bessi E, Boehm G: Cesarean Clinical evaluation of a new starter formula for infants
delivery may affect the early biodiversity of intestinal bacteria. containing live Bifidobacterium longum BL999 and prebiotics.
J Nutr 2008, 138:1796S-1800S. Nutrition 2007, 23:1-8.

www.sciencedirect.com Current Opinion in Biotechnology 2010, 21:149–156


156 Food biotechnology

52. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, 66. Huang J, Mei L-H, Wu H, Lin D-Q: Biosynthesis of g-
Schilter HC, Rolph MS, Mackay F, Artis D et al.: Regulation of aminobutyric acid (GABA) using immobilized whole cells of
inflammatory responses by gut microbiota and Lactobacillus brevis. World J Microbiol Biotechnol 2007,
chemoattractant receptor GPR43. Nature 2009, 23:865-871.
461:1282-1286.
67. Komatsuzaki N, Nakamura T, Kimura T, Shima J:
53. Greer FR: Vitamin K the basics — What’s new? Early Hum Dev Characterization of glutamate decarboxylase from a high
2010, in press. doi:10.1016/j.earlhumdev.2010.01.015. gamma-aminobutyric acid (GABA)-producer, Lactobacillus
paracasei. Biosci, Biotechnol, Biochem 2008, 72:278-285.
54. Schurgers LJ, Teunissen KJF, Hamulyak K, Knapen MHJ, Vik H,
Vermeer C: Vitamin K-containing dietary supplements: 68. Park K-B, Oh S-H: Production of yogurt with enhanced levels of
comparison of synthetic vitamin K1 and natto-derived gamma-aminobutyric acid and valuable nutrients using lactic
menaquinone-7. Blood 2007, 109:3279-3283. acid bacteria and germinated soybean extract. Bioresour
Technol 2007, 98:1675-1679.
55. Hojo K, Watanabe R, Mori T, Taketomo N: Quantitative
measurement of tetrahydromenaquinone-9 in cheese 69. Mazmanian SK, Kasper DL: The love–hate relationship between
fermented by propionibacteria. J Dairy Sci 2007, bacterial polysaccharides and the host immune system. Nat
90:4078-4083. Rev Immunol 2006, 6:849-858.
56. van Summeren MJH, Braam LAJLM, Lilien MR, Schurgers LJ, 70. Parker P, Jones G, Smith S: Mixed cultures of food-grade
Kuis W, Vermeer C: The effect of menaquinone-7 (vitamin K2) probiotic bacteria and enteric bacteria demonstrate both
supplementation on osteocalcin carboxylation in healthy synergism and inhibition of menaquinone production. J Food
prepubertal children. Br J Nutr 2009, 102:1171-1178. Sci 2003, 68:2325-2330.
57. Kaneda M, Zhang D, Bhattacharjee R, Nakahama K-I, Arii S, 71. Barrett E, Ross RP, Fitzgerald GF, Stanton C: Rapid screening
Morita I: Vitamin K2 suppresses malignancy of HuH7 method for analysing the conjugated linoleic acid production
hepatoma cells via inhibition of connexin 43. Cancer Lett 2008, capabilities of bacterial cultures. Appl Environ Microbiol 2007,
263:53-60. 73:2333-2337.
58. Martens JH, Barg H, Warren MJ, Jahn D: Microbial production of 72. Singhal A, Macfarlane G, Macfarlane S, Lanigan J, Kennedy K,
vitamin B12. Appl Microbiol Biotechnol 2002, 58:275-285. Elias-Jones A, Stephenson T, Dudek P, Lucas A: Dietary
nucleotides and fecal microbiota in formula-fed infants: a
59. Santos F, Wegkamp A, de Vos WM, Smid EJ, Hugenholtz J: High- randomized controlled trial. Am J Clin Nutr 2008, 87:1785-1792.
level folate production in fermented foods by the B12 producer
Lactobacillus reuteri JCM1112. Appl Environ Microbiol 2008, 73. M’Rabet L, Vos AP, Boehm G, Garssen J: Breast-feeding and its
74:3291-3294. role in early development of the immune system in infants:
consequences for health later in life. J Nutr 2008,
60. Hay G, Johnston C, Whitelaw A, Trygg K, Refsum H: Folate and 138:1782S-1790S.
cobalamin status in relation to breastfeeding and weaning in
healthy infants. Am J Clin Nutr 2008, 88:105-114. 74. Martı́n R, Heilig GHJ, Zoetendal EG, Smidt H, Rodrı́guez JM:
Diversity of the Lactobacillus group in breast milk and vagina
61. Dror DK, Allen LH: Effect of vitamin B12 deficiency on of healthy women and potential role in the colonization of the
neurodevelopment in infants: current knowledge and possible infant gut. J Appl Microbiol 2007, 103:2638-2644.
mechanisms. Nutr Rev 2008, 66:250-255.
75. FAO/WHO: Report on Joint FAO/WHO Expert Consultation on
62. Bjorke-Monsen A-L, Torsvik I, Saetran H, Markestad T, Evaluation of Health and Nutritional Properties of Probiotics in
Ueland PM: Common metabolic profile in infants indicating Food including Powder Milk with Live Lactic Acid Bacteria. 2001
impaired cobalamin status responds to cobalamin ftp://ftp.fao.org/docrep/fao/009/a0512e/a0512e00.pdf.
supplementation. Pediatrics 2008, 122:83-91.
76. Deshpande G, Rao S, Patole S: Probiotics for prevention of
63. Ross RP, Mills S, Hill C, Fitzgerald GF, Stanton C: Specific necrotising enterocolitis in preterm neonates with very low
metabolite production by gut microbiota as a basis for birthweight: a systematic review of randomised controlled
probiotic function. Int Dairy J 2010, 20:269-276. trials. Lancet 2007, 369:1614-1620.
64. Bhattacharya A, Banu J, Rahman M, Causey J, Fernandes G: 77. AlFaleh KM, Bassler D: Probiotics for prevention of necrotizing
Biological effects of conjugated linoleic acids in health and enterocolitis in preterm infants. Cochrane Database Syst Rev
disease. J Nutr Biochem 2006, 17:789-810. 2008, Issue 1.: Art. No.: CD005496.
65. Rosberg-Cody E, Ross RP, Hussey S, Ryan CA, Murphy BP, 78. Mc Gee R, O’Connor PM, Russell D, Dempsey G, Ryan CA, Ross
Fitzgerald GF, Devery R, Stanton C: Mining the microbiota of the RP, Stanton C: Prolonged faecal excretion following a single
neonatal gastrointestinal tract for conjugated linoleic dose of probiotic in low birth weight infants. Acta Paediatr
acid-producing bifidobacteria. Appl Environ Microbiol 2004, 2010, in press.
70:4635-4641.

Current Opinion in Biotechnology 2010, 21:149–156 www.sciencedirect.com

You might also like