Vous êtes sur la page 1sur 8

REVIEW

URRENT
C
OPINION

The neonatal bowel microbiome in health


and infection
Janet E. Berrington a, Christopher J. Stewart b, Stephen P. Cummings b, and
Nicholas D. Embleton a

Purpose of review
In newborns, interactions between the host and the microbiome operate synergistically, modulating host
immune function and shaping the microbiome. Next generation molecular sequencing methodologies in
tandem with modeling complex communities allow insights into the role of the microbiome in health and
disease states. Infection-related disease states in which dysbiosis is integral include late-onset sepsis (LOS)
and necrotizing enterocolitis (NEC), which still cause deaths and morbidity. Understanding microbiomic
interactions may lead to alternative prevention, monitoring or treatment strategies, and modulation of longterm health outcomes especially in the preterm population. Recent studies have advanced understanding of
the microbiome in NEC and LOS.
Recent findings
Mechanisms of hostmicrobiome interaction have been demonstrated. Patterns of microbiomic change in
association with NEC and LOS have been observed, with community changes dominated by
Proteobacteria and Firmicutes appearing to precede NEC, and very early microbiomic signatures
influencing LOS. Data on viral and fungal elements are emerging.
Summary
Greater understanding of the neonatal bowel microbiome may allow tailored clinical practice and
therapeutic intervention. Data handling and interpretation is challenging. Mechanistic studies of clinical
interventions that affect the gut microbiome are important next steps.
Keywords
health, infection, microbiome, necrotizing enterocolitis, neonatal

INTRODUCTION
The complex microbial community (microbiome)
in the human gut is viewed as a superorgan involved
in many aspects of health and disease in which the
microbiome exerts metabolic, nutritional, and
immunological effects on the host [1]. In healthy,
vaginally delivered, term newborns (HVDTNB) the
gut and adaptive immune system tolerate and
regulate this community; the microbiome in turn
sculpts gut immune and metabolic function [2]. The
establishment of the microbiome proceeds predictably, reflecting the coevolution with their host over
millennia [3,4]. However, this dynamic process is
readily disrupted, especially by events related to
prematurity [5]. Such disruption, termed dysbiosis,
is implicated in development of late-onset sepsis
(LOS) and necrotizing enterocolitis (NEC), which
are both major causes of mortality and morbidity
[6]. Therapeutic manipulation and even microbiome
transplantation are possible but the mechanistic
www.co-infectiousdiseases.com

interactions involved are complex and poorly understood. This review summarizes how advances in molecular technologies can offer mechanistic insights
into hostmicrobiome interactions and reviews
recent work suggesting a microbiome pattern representative of health and changes that precede LOS and
NEC. We review progress toward our understanding
of the functional contribution of bacterial community members, and the significance of nonbacterial organisms (fungi, viruses, and archaea). Insights
a
Newcastle Neonatal Service, Newcastle upon Tyne Hospitals NHS
Foundation Trust and Newcastle University and bFaculty of Health and
Life Sciences, University of Northumbria, Newcastle, UK

Correspondence to Janet Elizabeth Berrington, Newcastle Neonatal


Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen
Victoria Road, Newcastle, NE1 4LP, UK. Tel: +44 191 2825197; fax:
+44 191 2825048; e-mail: j.e.berrington@ncl.ac.uk
Curr Opin Infect Dis 2014, 27:236243
DOI:10.1097/QCO.0000000000000061
Volume 27  Number 3  June 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

The neonatal bowel microbiome Berrington et al.

KEY POINTS
 Very early microbe exposures and microbehost
interactions are key to the development of NEC and
LOS, and therapeutic manipulations require
underpinning with mechanistic evaluation.

DNA extraction before universal PCR primers allow


amplification of intervening hypervariable regions.
These amplicons then require differentiating into
sufficiently similar groups to be classified together
into an operational taxonomic unit (OTU). Different techniques have been used and continue to
develop (Fig. 1 [8 ]). Bacterial classification is determined by using open access sequence databases.
Methodologies for nonbacterial constituents are
less robust or well developed [9] but fungal, archaeal, protozoal, and viral microbiomic studies now
exist. The functional capacity of the microbiome is
also increasingly acknowledged: metabolomic and
proteomic techniques offer insight into functional
microbiomic changes associated with disease [10].
&

 Patterns of bacterial dysbiosis are emerging:


Proteobacteria and Firmicutes appear pivotal to
changes that predict or precede NEC or LOS
development.
 Nonbacterial elements may also be important and
require further study in carefully sampled populations
with careful exposure data.

from interventions intended to manipulate the


microbiome are considered and a brief examination
of residual research needs and priorities are outlined.

METHODOLOGICAL CONSIDERATIONS
Over time the gut microbiota changes in constituent
members, relative abundance, gene expression, and
metabolic activity. Measuring this dynamic complexity poses significant challenges: analysis typically involves passed stool (rarely ileal fluid or tissue)
and is thus limited by spontaneous stooling (which
occurs less frequently when infants are ill) and
reflects alterations that are historical and dependent
on transit times. Analyzing and presenting such
complex dynamic data sets also present many
challenges.

Data handling
Next-generation sequencing techniques (Fig. 1)
generate large and complex datasets. Indeed, the
subsequent bioinformatic and statistical analysis
of these datasets have become the bottleneck for
microbial community analyses [11]. Communities
are described in terms of richness (their constituents), evenness (dominance by particular OTUs),
and diversity (a combination of richness and evenness). Ordination analyses and constrained or
redundancy analyses allow exploration of potential
mediators of structure and changes [12], approaches
that are particularly valuable when attempting to
relate microbiomic data to the patients demographic and clinical details. New methods of handling and interpreting such complex datasets are in
evolution [13 ].
&&

Molecular techniques
Culture independent analysis exploits the presence
of the ubiquitous and evolutionarily conserved 16S
ribosomal RNA (16S rRNA) gene, by which prokaryotic taxa can be differentiated. Molecular analyses
are facilitated by recent work showing that once
sampled, the stool microbiome is stable at room
temperature for several weeks [7]. Samples undergo

HOSTMICROBIOME INTERACTIONS
The neonatal period uniquely challenges the host:
pioneering organisms are encountered by nave host
gut defences, educate and modulate immune
responses, and thus regulate the microbiome development. The bacterial community structure and
host defences modulate gut endothelial cells,

First generation techniques: separation by physical means using denaturing or


temperature gradient gel electrophoresis (D/TGGE) - fingerprinting techniques
Next generation sequencing (NGS): combinations of enzymology, chemistry, highresolution optics, hardware, and software engineering. Generate enormous amounts of
data and sequence to great depth - examples: 454 pyrosequencing (Roche), HiSeq and
bench top MiSeq (Illumnia) platforms. The MiSeq was recently found to have the
highest throughput and lowest error rate of its type [8].
Third generation platforms: capable of generating reads greater than 400 base pairs,
facilitating identification to species level.

FIGURE 1. Sequencing techniques.


0951-7375 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-infectiousdiseases.com

237

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Paediatric and neonatal infections

modifying function, enhancing protection and


maintaining barrier function. Careful regulation
of pro- and anti-inflammatory effects is needed to
maintain health [14]. Recent mechanistic insights
are outlined below.

Competitive evolutionary strategies


Studies in a murine model suggest direct pathogen
control occurs through commensal bacteria controlling virulence factor expression in pathogenic bacteria, and by competition modulated through
shared carbohydrate requirements [15 ]. Environmental acidification by Bifidobacteria as a means
of controlling Escherichia coli has also been demonstrated, a mechanism of potential import for probiotic reduction of incidence of NEC [16].
&

Barrier integrity
The traditional top-down hypothesis of a breach of
barrier integrity, with breakdown in endothelial
tight junctions leading to loss of regulation of factors intrinsic to a balanced pro- and anti-inflammatory response has recently been challenged. An
alternative hypothesis, that of Paneth cell dysregulation, has been proposed that challenges many
traditional assumptions [17 ]. The antibacterial
lectin RegIII gamma has been shown to modulate
both total bacterial load and contact of luminal
bacteria with host epithelium [18].
&&

shown to induce extrathalamic Treg differentiation


[22 ,23 ].
&

&

DEFINING THE HEALTHY NEONATAL GUT


MICROBIOME
A healthy gut microbiome is the intestinal
microbial community that assists the host to maintain a healthy status under certain environmental
conditions [24]. If this could be defined for the
neonate, a comparison with disease states could
be made, allowing insights into pathogenesis or
guiding preventive or treatment options.

Term neonates
The full-term vaginally delivered breast-fed (FTVDBF)
infant is considered ideal from a microbiomic
perspective and was initially studied in 14 infants
[25]. The more comprehensive Norwegian Microflora
Study (NOMIC) enrolled 246 term infants at day 4,
10, 30, and 120 of life and showed links between early
colonization patterns and later growth patterns
[26 ]. Specifically, Bacteroides appeared protective
against obesity [27]. Three phyla Bacteroidetes,
Proteobacteria, and Firmicutes exert the greatest
effects on community dynamics [4,28]. Bifidobacterium appears specifically important to the neonate:
six species in breast milk showed properties expected
of commercial probiotics [29], and specific probiotic
formulations for preterm infants could be formulated
with this knowledge [30].
&&

Host immunomodulation
Several mechanisms have recently been demonstrated through which intraluminal bacteria modify
host responses. Initial neonatal bacterial exposure
within the gut leads to activation of Toll-like receptor (TLR)-4, which has been shown to activate an
inhibitory loop through the expression of miR-146a
(a small noncoding mRNA), affecting the response
to TLR agonist [19 ]. Some Bifidobacterium bifidum
can generate a proinflammatory Th17 immune
response (involved in protection against mucosal
infection), and functional Treg lymphocytes with
plasticity for either secreting IL-17 or suppressing
activation of the immune system, depending on the
external stimuli [20]. Polysaccharide A (PSA), a
specific symbiosis factor produced by the commensal Bacteroides fragilis, uses TLR-2 receptor activation
to activate Foxp3 Treg cells to promote tolerance
in a mouse model. PSA-lacking B. fragilis cannot
promote this response, clearly demonstrating how
specific bacterial products of symbionts manipulate
host immune responses [21]. Butyrate, a short-chain
fatty acid (SCFA) produced by commensal microorganisms during starch fermentation, has also been
&&

238

www.co-infectiousdiseases.com

Preterm infants
Neonates born preterm are exposed to potentially
detrimental factors in the development of the
healthy microbiome, including prelabour rupture
of membranes, caesarean delivery, maternal and
neonatal antibiotic intervention, and lack of
breast-feeding. Previous studies, although limited,
showed reduced diversity and relative instability
[31] (Fig. 2). Recently Arboleya et al. [32 ] studied
21 preterm infants (32 weeks mean gestation) and
20 FTVDBF infants: the aim was to establish how
differently 18 microbial groups were represented,
and SCFAs were also measured reflecting microbiome metabolic activity. Increased facultative
anaerobic microorganisms including Enterobacteriaceae, Enterococcaceae, and Lactobacillus and
fewer anaerobes, including Bifidobacterium, Bacteroidetes, and Atopobium, were seen in the preterm
cohort. The authors hypothesize that this delay in
aerobic establishment may delay immune maturation in preterm neonates. Further work in an
entirely preterm cohort exploring the development
of the gut microbiota from multiple births also
&

Volume 27  Number 3  June 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

The neonatal bowel microbiome Berrington et al.

(a)
Diversity

Healthy preterm

Diseased preterm

(b)

Age

not seen in healthy controls [35]. A potential


signature seen more frequently in NEC was identified as grouping to the Enterobacteriaceae family,
but did not match any sequence at greater than
97%. Likewise, a case report of a single preterm
infant (25 weeks gestation) who developed lethal
NEC on day 48 with a dramatic increase in Proteobacteria, specifically Klebsiella, 2 days before. Proteobacteria contributed greater than 97% of phyla
compared with less than 50% before. Ileal fluid and
mucosal samples also identified the same pattern
[36 ]. Jenke [37 ] identified an increase in E. coli
preceding NEC in comparison both to earlier
samples from the same individual and to healthy
preterm infants. Fecal inflammatory markers were
also studied: S100A12 and hBD2 were seen to correlate with total bacterial counts and E. coli, linking
microbiomic changes and host immune responses.
Two potentially distinct patterns of dysbiosis were
identified by Morrow et al. [38 ] in 11 NEC infants:
an early presentation (n 4) dominated by Firmicutes, and a later presentation dominated by Proteobacteria. In our own cohort of infants less than
32 weeks gestation, distinct microbiomic differences
between NEC and/or LOS compared with healthy
infants were observed [33 ]. Detailed examination of
twins discordant for NEC demonstrated a reduction
in diversity and increasing dominance of Escherichia
(since identified as E. coli) preceding NEC [39 ].
Preterm infants, whose exposure to lipopolysaccharide (LPS) from nosocomial bacteria is modified by
neonatal intensive care practices, may be less tolerant when they do meet these bacteria and mount a
resultant excessive inflammatory response. Interestingly, a Danish group did not identify microbiomic
shifts in association with NEC using molecular
analysis, although the timing of the sampling in
relation to NEC development in this cohort is not
described, and is clearly key [40]. This group also
analyzed small intestine specimens resected surgically in infants with NEC, demonstrating technical
feasibility, but no specific profiles were identified
associated with NEC [43].
&

Diversity
Healthy term

Healthy preterm

Age

&&

&&

FIGURE 2. Schematic diagram to demonstrate the typical


patterns in diversity in (a) healthy preterm infants compared
with preterm infants diagnosed with disease and (b) healthy
term infants compared with healthy preterm infants.

&

&

found Bifidobacterium abundance to be low [33 ].


Lower levels of naturally occurring probiotic species
in preterm infants are important, given the possibility of supplementing these for the prevention of
NEC [34].

SPECIFIC INFECTIOUS DISEASES


Dysbiosis appears pivotal to both LOS and NEC
(Fig. 2). Patterns are beginning to emerge that
appear associated, but causality is not proven.

Necrotizing enterocolitis
NEC, an important neonatal disorder, is increasingly recognized as a spectrum of disease culminating in a final common pathway.
Bacterial elements
Is there a signature microbial pattern associated
with NEC? If so, does it predate NEC onset to allow
early diagnosis or modification of practice to
improve outcomes?
Recent studies addressing these questions are
shown in Table 1, highlighting populations, molecular techniques employed, and key findings. A
pyrosequencing study of nine preterm infants with
NEC sampled before NEC diagnosis and within 72 h
of diagnosis identified a bloom in Proteobacteria
and a decrease in Firmicutes between sampling,

&

Nonbacterial elements
Increasingly the role of nonbacterial organisms is
being recognized in disease etiology.
Archaea
Palmer [25] identified archaea in infants gut, but
considerably less frequently and more variably than
fungi or bacteria, appearing only transiently in the
first few weeks of life. In inflammatory bowel disease
(IBD), the presence of methanogenic archaea in the
human gut was indicative of a healthy microbiota,
with reduced methanogen presence in individuals

0951-7375 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-infectiousdiseases.com

239

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Paediatric and neonatal infections


Table 1. Recent microbiomic analysis in preterm infants with necrotizing enterocolitis or sepsis
Population gestational
age (weeks) (n) of
whom illness (n)
<32 (38) NEC (7) LOS
(13)

Technique
DGGE

Sampling
strategy

Key findings

Author [ref]

Opportunistic surveillance
study, n 99

Enterococci associated with


health

Stewart et al. [33 ]


&

Enterobacter associated with NEC


Staphylococcus associated with
LOS
<32 (18) NEC (9)

454 Flex chemistry

310 days before NEC


and within 72 h of NEC
onset

Bloom in Proteobacteria and a


reduction of Firmicutes in
NEC

Mai et al. [35]

Gammaproteobacteriacea family
signature for NEC
25 (1) NEC (1)

Pyrosequencing
(bTEFAP)

Weekly

Decreased diversity and


Proteobacteria (specifically
Klebsiella) dominant 2 days
before NEC

Tran et al. [36 ]

<27 (68) NEC (12)

qPCR

Alternate days for 4 weeks

Increase in E. coli before NEC

Jenke et al. [37 ]

&

&&

Associated increase in fecal


s100A12 and hBD2
<29 (32) NEC (11)

Pyrosequencing

Day 49 and day 1016


urine for metabolomics
(NMR)

Two patterns of dysbiosis noted


associated with NEC:
Firmicute dominant
(Staphylococcus and
Enterococcus)
Proteobacteria dominant
(Enterobacteriaceae)

Morrow et al. [38 ]


&&

(Urinary alanine increased in


group 1)
<32 (32) NEC (7) LOS
(13)

DGGE 16S rRNA


and 28S rRNA

First and weekly

Sphingomonas associated with


NEC

Stewart et al. [39 ]

<30 (163) NEC (21)

DGGE

Three samples (day 05,


10, and 30)

No differences between NEC and


non-NEC identified but no data
on sample timing relating NEC

Smith et al. [40]

Pretem (6) LOS (2), 2 well,


2 culture negative

Pyrosequencing

First, weekly, and at sepsis


evaluation

Increased Firmicutes
(Staphylococcus) in LOS

Madan et al. [41 ]

<32 (28) LOS (10)

Pyrosequencing

Weekly

Lower Bifidobacteria in cases

Mai et al. [42 ]

NEC (24)

FISH

Surgical resection
specimens

Proteobacteria (49%) Firmicutes


(30%)

Smith et al. [43]

&

&

Postulate healthy microbiome


&

DGGE, denaturing gradient gel electrophoresis; FISH, fluoresecent in-situ hybridization; LOS, late onset sepsis; NEC, necrotizing enterocolitis.

with IBD [44]: their absence from neonates may thus


be important and mark dysbiosis.
Viruses
Viruses may contribute to NEC by direct host viral
infection or by phage infection of a microbiomic
constituent. Torovirus [45] and astrovirus [46] have
been previously linked to NEC onset, but recently
there has been a resurgence of interest in cytomegalovirus (CMV). CMV was reported in association
with fulminant lethal NEC in a 25-week-gestation
infant: post-mortem inclusion bodies prompted
recognition of CMV in the intestine [36 ]. An
&

240

www.co-infectiousdiseases.com

associated bloom in Proteobacteria was noted:


CMV may have contributed to direct mucosal injury
(seen in patients with IBD and CMV) or have promoted a microbiomic shift that heralded NEC. Deep
sequencing of viral RNAs and DNAs revealed that
many viruses in the infant gut are bacteriophages,
although less diversity is seen in infants [47]. Phages
may influence microbiome composition by infecting and lysing specific bacterial hosts, allowing
another bacterial strain the opportunity to become
abundant [48]. Although identified as present in the
neonatal gut they have not been studied specifically
in the context of NEC or LOS [47].
Volume 27  Number 3  June 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

The neonatal bowel microbiome Berrington et al.

Fungial
LaTuga et al. [47] extended microbiomic analysis
beyond the bacterial elements in their study of
11 infants less than 1000 g in their first month, five
of whom had coagulase-negative staphylococci
infection and two of whom developed NEC (but
only one within the sampling window). They identified fungal species including several Candida
species despite nystatin prophylaxis, a finding confirmed by Stewart et al. [49 ] who identified 12 fungal
species (all nonviable) in their cohort who received
fluconazole prophylaxis. The role of fungi in NEC
pathogenesis requires further elucidation.
&

INFECTION
There are many parallels between LOS and NEC. Gut
dysbiosis with a preponderance of Proteobacteria
and Firmicutes abundance has been noted in
neonates with LOS. Altered barrier and immune
function and imbalance between pro- and antiinflammatory responses have also been implicated.
Mai et al. [42 ] analyzed 10 preterminfants with LOS
and 18 preterm controls, showing association
between microbiomic signatures in the two weeks
before diagnosis of LOS but interestingly not at
diagnosis. A high-resolution study of six preterm
infants showed differences in microbial patterns
in the first weeks of life in infants at high risk
&

of sepsis: less diversity and a preponderance of


staphylococci [41 ]. Archaea, viruses, and fungi in
the gut may also influence LOS, but as with NEC,
studies to date do not adequately analyze these
domains in the pathogenesis of sepsis.
&

THERAPEUTIC INTERVENTIONS
Interventions that affect the microbiome affect
health: prolonged early postnatal antibiotics
increase the risk of NEC [50], oral antibiotics appear
to reduce it [51], probiotics can reduce NEC [52],
lactoferrin appears to reduce LOS and possibly NEC
[53,54]; however, mechanistic understanding for
these findings is lacking.
However, clinical interventions could theoretically be manipulated and tailored to engineer a gut
microbiota reflective of an FTVDBF infant. Breast
milk contains complex immune-protective and
growth factors, bioactive immune-modulatory cells
and other immunonutrients including amino acids,
fatty acids, lysozyme, lactoferrin, minerals, and
metals such as zinc, and prebiotic oligosaccharides
as well as live bacteria. It seems unlikely that artificial supplements could ever replicate the complexity of own mothers breast milk. Manufacturing
complex combination supplements is attractive,
but a better understanding is needed before this
would be feasible. Few clinical trials in neonates

Is dysbiosis of the microbiome cause or effect of NEC and LOS?


What is the contribution of the host genetic background to
alterations to the microbiome in disease states?
Can aspects of clinical care be manipulated to engineer a gut
microbiota reflective of a FTVDBF infant?
Can the host immune response be selectively enhanced to promote
the growth of organisms seen in health states rather than in disease
pathophysiology?
Can probiotic use, dose, strain, and duration be realistically
optimized given the number of options and babies available to
study?
What is the role of prebiotics either instead of probiotics or in
combination (synbiotics)?
Can intelligent use of antibiotics help harness the microbiome to
promote health?
What role do non-bacterial members have individually and in
community interactions with other microorganisms?
How are vast amount of data best presented, analysed and made
publicly available? Can data from different studies be meaningfully
meta-analysed or visually presented?

FIGURE 3. Key unanswered questions and challenges FTVDBF, full-term vaginally delivered breast-fed; LOS, late-onset sepsis;
NEC, necrotizing enterocolitis.
0951-7375 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-infectiousdiseases.com

241

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Paediatric and neonatal infections

have attempted mechanistic evaluations: few probiotic studies assess the microbiome it is hypothesized but unproven that probiotics result in a
reduction in the growth of potential pathogens
including enterobacteria, enterococci, and clostridia
[55]. A serendipitous increase in Lactobacillus spp. in
preterm infants was associated with 1% lactulose
added to feeds, potentially attributable to its prebiotic effects [56]. Lactoferrin, a molecule with
many mechanistic effects on the microbiome, has
not been subject to detailed exploration of the
effects on the preterm infant microbiome [57].

CONCLUSION
Understanding the neonatal microbiome may hold
the key to preventing LOS and NEC. Although
patterns are emerging, the causal pathway is
unclear. Where interventions do reduce morbidities (e.g., probiotics), the mechanisms are poorly
understood, and key questions remain unanswered
(Fig. 3). Mechanistic evaluation in the context of
controlled clinical trials would help elucidate this,
and may provide insight that allows improved prevention, diagnosis, and treatment. The inclusion of
microbiomic assessment, including bacterial and
nonbacterial elements and noninvasive sample
collection, in future clinical trials of microbiomic
modulation deserves further attention. This could
include trials of feeding, probiotics, and enteral
or immunonutrient supplements. Key unknowns
about host response should also be addressed. Little
is known about gut immune tissue responses to NEC
in humans, yet many infants have resection of gut
as part of their treatment, and immunohistochemistry techniques now allow elucidation of these
aspects.
Acknowledgements
None.
Conflicts of interest
There are no conflicts of interest.

REFERENCES AND RECOMMENDED


READING
Papers of particular interest, published within the annual period of review, have
been highlighted as:
&
of special interest
&& of outstanding interest
1. Kinross JM, Darzi AW, Nicholson JK. Gut microbiome-host interactions in
health and disease. Genome Med 2011; 3:14.
2. Wardwell LH, Huttenhower C, Garrett WS. Current concepts of the intestinal
microbiota and the pathogenesis of infection. Curr Infect Dis Rep 2011;
13:2834.
3. Dethlefsen L, McFall-Ngai M, Relman DA. An ecological and evolutionary
perspective on human-microbe mutualism and disease. Nature 2007; 449:
811818.

242

www.co-infectiousdiseases.com

4. Eggesb M, Moen B, Peddada S, et al. Development of gut microbiota in


infants not exposed to medical interventions. Acta Pathol Microbiol Immunol
Scand 2011; 119:1735.
5. Dominguez-Bello MG, Blaser MJ, Ley RE, et al. Development of the human
gastrointestinal microbiota and insights from high-throughput sequencing.
Gastroenterology 2011; 140:17131719.
6. Berrington JE, Hearn RI, Bythell M, et al. Deaths in preterm infants: changing
pathology over 2 decades. J Pediatr 2012; 160:4953.
7. Lauber CL, Zhou N, Gordon JI, et al. Effect of storage conditions on the
assessment of bacterial community structure in soil and human-associated
samples. FEMS Microbiol Lett 2010; 307:8086.
8. Loman NJ, Misra RV, Dallman TJ, et al. Performance comparison of benchtop
&
high-throughput sequencing platforms. Nature Biotechnol 2012; 30:434
439.
Comparison of three platforms the 454 GS Junior (Roche), MiSeq (Illumina), and
Ion Torrent PGM (Life Technologies). Comparison of read length and indel errors
favored MiSeq.
9. Amend SA, Seifert KA, Bruns TD. Quantifying microbial communities with 454
pyrosequencing: does read abundance count? Mol Ecol 2010; 19:5555
5556.
10. Mussap M, Noto A, Cibecchini F, Fanos V. The importance of biomarkers in
neonatology. Semin Fetal Neonatal Med 2013; 18:5664.
11. Mardis ER. The $1,000 genome, the $100,000 analysis? Genome Med
2010; 2:84.
12. Rushton SP, Shirley MDF, Sheridan EA, et al. The transmission of nosocomial
pathogens in an intensive care unit: a space-time clustering and structural
equation modelling approach. Epidemiol Infect 2010; 138:915926.
13. Langille MGI, Zaneveld J, Caporaso JG, et al. Predictive functional profiling of
&&
microbial communities using 16S rRNA marker gene sequences. Nature
Biotechnol 2013; 31:814821.
A novel computational approach to predict the function of a metagenome.
PICRUST (phylogenetic investigation of communities by reconstruction of unobserved states) uses algorithmic methods to predict gene presence and compose a
metagenome. The authors identify the potential to use PICRUST in communities
where only marker genes are available.
14. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune
responses during health and disease. Nat Rev Immunol 2009; 9:313323.
15. Kamada N, Kim YG, Sham HP, et al. Regulated virulence controls the ability of
&
a pathogen to compete with the gut microbiota. Science 2012; 336:1325
1329.
In a germ-free murine model, the authors showed that virulence genes are not
required for colonization of Citrobacter rodentium in germ-free mice, but control
relocation to the lumen in later infection, where commensal bacteria outcompete
the Citrobacter for shared carbohydrate requirement.
16. Fukuda S, Toh H, Hase K, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011; 469:543547.
17. McElroy SJ, Underwood MA, Sherman MP. Paneth cells and necrotizing
&&
enterocolitis: a novel hypothesis for disease pathogenesis. Neonatology
2013; 103:1020.
Excellent challenging article, seeking fresh perspective and interpretation on the
traditional hypotheses of pathogenesis of NEC. The authors postulate a primary
role for the Paneth cells injury in the NEC pathway in contradiction to the bacterially
originated hypotheses.
18. Vaishnava S, Yamamoto M, Severson KM, et al. The antibacterial lectin
RegIIIgamma promotes the spatial segregation of microbiota and host in
the intestine. Science (New York) 2011; 334:255258.
19. Tourner E, Chassin C. Neonatal immune adaptation of the gut and its role
&&
during infections. Clin Dev Immunol 2013; 2013:270301.
Excellent overview of the complexities of interactions between the gut and host
immune function and bacterial elements from this group with extensive own
research into microRNA, iRAK, and TLR-4 pathways in mouse and neonatal
models of mucosal injury.
20. Lopez P, Gonzalez-Rodrguez I, Gueimonde M, et al. Immune response to
Bifidobacterium bifidum strains support Treg/Th17 plasticity. PloS one 2011;
6:e24776.
21. Round JL, Lee SM, Li J, et al. The Toll-like receptor 2 pathway establishes
colonization by a commensal of the human microbiota. Science 2011;
332:974977.
22. Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate
&
induces the differentiation of colonic regulatory T cells. Nature 2013;
504:446450.
Study helping establish that microbial metabolites induce peripheral Treg cells: in a
murine study, colonization with chloroform-resistant bacteria fed a high-fiber diet
increased the number of peripheral TReg cells when compared to germ-free mice.
Fecal metabolites in these mice showed higher levels of short-chain fatty acids,
such as butyrate and propionate.
23. Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal
&
bacteria promote peripheral regulatory T-cell generation. Nature 2013;
504:451455.
Butyrate-deficient fecal murine extracts were shown to induce less FOXP3
expression in CD4 T cells than nonbutyrate deficient mice. Feeding butyrate
to mice led to higher Treg expression in the colon. The data suggest that
manipulation of colonic immune function may be possible by either direct bacterial
manipulation or supplementation of bacterial products.

Volume 27  Number 3  June 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

The neonatal bowel microbiome Berrington et al.


24. Echarri PP, Gracia CM, Berruezo GR, et al. Assessment of intestinal microbiota of full-term breast-fed infants from two different geographical locations.
Early Hum Dev 2011; 87:511513.
25. Palmer C, Bik EM, DiGiulio DB, et al. Development of the human infant
intestinal microbiota. PLoS Biol 2007; 5:15561573.
26. White R, Bjrnholt JV, Baird DD, et al. Novel developmental analyses identify
&&
longitudinal patterns of early gut microbiota that affect infant growth. PLoS
Comput Biol 2013; 9:e1003042.
Large longitudinal study of weight change in association with microbiomic patterns
over time facilitated by a novel statistical approach, modeling detection, or lack of
detection of 22 species on days 4,10, 30, and 120. The authors suggest that the
presence of some species (staphylococci and Escherichia) at critical time points is
associated with normal growth, and that Bacteroides presence was protective
against obesity.
27. Musso G, Gambino R, Cassader M. Obesity, diabetes, and gut microbiota:
the hygiene hypothesis expanded? Diabetes Care 2010; 33:22772284.
28. Trosvik P, Stenseth NC, Rudi K. Convergent temporal dynamics of the human
infant gut microbiota. ISME J 2010; 4:151158.
29. Arboleya S, Ruas-Madiedo P, Margolles A, et al. Characterization and in vitro
properties of potentially probiotic Bifidobacterium strains isolated from
breast-milk. Int J Food Microbiol 2011; 149:2836.
30. Arboleya S, Gonzalez S, Salazar N, et al. Development of probiotic products
for nutritional requirements of specific human populations. Eng Life Sci 2012;
12:368376.
31. Westerbeek EAM, Van den Berg A, Lafeber HN, et al. The intestinal bacterial
colonisation in preterm infants: a review of the literature. Clin Nutr 2006;
25:361368.
32. Arboleya S, Binetti A, Salazar N, et al. Establishment and development of
&
intestinal microbiota in preterm neonates. FEMS Microbiol Ecol 2012;
79:763772.
Twenty FTVDBF infants compared with 21 preterm (mean gestational age 32.7) by
denaturing gradient gel electrophoresis (DGGE) sampled four times over their first
90 days. Importantly also incorporated a measure of function, with SCFA measurement. Clear differences were seen betweem term and preterm populations
despite significant interindividual variability.
33. Stewart C, Marrs E, Magorrian S, et al. The preterm gut microbiota: changes
&
associated with necrotizing enterocolitis and infection. Acta Paediatr 2012;
101:11211127.
Extensively sampled cohort of significantly preterm infants (<32 weeks) including
detailed exposure data and cases of NEC and LOS. Analysis by standard culture
and DGGE for comparison.
34. Hickey L, Jacobs SE, Garland SM. Probiotics in neonatology. J Paediatr Child
Health 2012; 48:777783.
35. Mai V, Young CM, Ukhanova M, et al. Fecal microbiota in premature infants
prior to necrotizing enterocolitis. PLoS One 2011; 6:e20647.
36. Tran L, Ferris M, Norori J, et al. Necrotizing enterocolitis and cytomegalovirus
&
infection in a premature infant. Pediatrics 2013; 131:e318e322.
Case report of a very preterm infant with devastating NEC in whom post-mortem
samples suggested CMV infection, and stool had already been analyzed with
16SrRNA.
37. Jenke AC, Postberg J, Mariel B, et al. S100A12 and hBD2 correlate with the
&&
composition of the fecal microflora in ELBW infants and expansion of E. coli is
associated with NEC. BioMed Res Int 2013; i:150372.
This study correlates inflammatory markers in the stool with microbial shifts in
association with NEC, moving forward the hypothesis that microbial shifts are
accompanied by functional changes that impact on neonatal health.
38. Morrow AL, Lagomarcino AJ, Schibler KR, et al. Early microbial and meta&&
bolomic signatures predict later onset of necrotizing enterocolitis in preterm
infants. Biomed Central 2013; 1:13.
First study to evaluate metabolomics as well as microbiomic data. Although small
numbers of NEC cases were included, the authors postulate two different
signatures for NEC in association with two different timings of NEC onset,
and raise the possibility of a urinary biomarker being established for early
diagnosis.

39. Stewart CJ, Marrs ECL, Nelson A, et al. Development of the preterm gut
microbiome in twins at risk of necrotising enterocolitis and sepsis. PLoS ONE
2013; 8:e73465.
Detailed comparitor of longitudinal sampling in twin cohort. Contains detailed
microbiomic data from a pair of twins discordant for NEC development and with
detailed exposure data.
40. Smith S, Bode S, Skov T, et al. Investigation of the early intestinal microflora in
premature infants with/without necrotizing enterocolitis using two different
methods. Ped Res 2012; 71:115120.
41. Madan JC, Salari RC, Saxena D, et al. Gut microbial colonisation in premature
&
neonates predicts neonatal sepsis. Arch Dis Child Fetal Neonatal Ed 2012;
97:F456F462.
In only six preterm infants, patterns of microbiome predictive of LOS, systemic
inflammatory response syndrome, and remaining healthy are postulated.
42. Mai V, Torrazza RM, Ukhanova M, et al. Distortions in development of intestinal
&
microbiota associated with late onset sepsis in preterm infants. PLoS One
2013; 8:e52876.
Changes in microbial constitution were shown to precede a diagnosis of LOS in
preterm infants, but not be present at the point of diagnosis.
43. Smith S, Bode S, Petersen B, et al. Community analysis of bacteria colonising
intestinal tissue of neonates with necrotising enterocilitis. BMC Microbiol
2011; 11:73.
44. Scanlan PD, Shanahan F, Marchesi JR. Human methanogen diversity and
incidence in healthy and diseased colonic groups using mcrA gene analysis.
BMC Microbiol 2008; 8:79.
45. Lodha A, de Silva N, Petric M, et al. Human torovirus: a new virus associated
with neonatal necrotizing enterocolitis. Acta Paediatr 2005; 94:10851088.
46. Bagci S, Eis-Hubinger A. Detection of astrovirus in premature infants with
necrotizing enterocolitis. Pediatr Infect Dis J 2008; 27:347350.
47. LaTuga MS, Ellis JC, Cotton CM, et al. Beyond bacteria: a study of the enteric
microbial consortium in extremely low birth weight infants. PLoS One 2011;
6:e27858.
48. Thingstad T, Lignell R. Theoretical models for the control of bacterial growth
rate, abundance, diversity and carbon demand. Aquat Microb Ecol 1997;
13:1927.
49. Stewart C, Nelson A, Scribbins D, et al. Bacterial and fungal viability in the
&
preterm gut: NEC and sepsis. Arch Dis Child Fetal Neonatal Ed 2013;
98:F298F303.
Article analyzing both fungal and viable elements of the microbiota in a large preterm
cohort with complex exposome including to prophylactic antifungal treatment.
Establishment of an effect on the bacterial community from antifungal receipt.
50. Cotton MC, Taylor S, Stoll B, et al. Prolonged duration of initial empiric
antibiotic treatment is associated with increased rates of necrotising enterocolitis and death for extremely low birth weight infants. Pediatrics 2009;
123:5866.
51. Bury RG, Tudehope D. Enteral antibiotics for preventing necrotizing enterocolitic in low birthweight or preterm infants. Cochrane Database Syst Rev
2007; CD000405.
52. Embleton N, Berrington JE. Probiotics reduce the risk of necrotising enterocolitis (NEC) in preterm infants. Evid Based Med 2013; 18:219220.
53. Manzoni P, Rinaldi M, Cattani S, et al. Bovine lactoferrin supplementation for
prevention of late-onset sepsis in very low-birth-weight neonates: a randomized trial. JAMA 2009; 302:14211428.
54. Venkatesh MP, Abrams S. Oral lactoferrin for the prevention of sepsis and
necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev
2010; 5:CD007137.
55. Deshpande G, Rao S, Patole S, et al. Probiotics for prevention of necrotising
enterocolitis in preterm neonates with very low birthweight: a systematic
review of randomised controlled trials. Lancet 2007; 369:16141620.
56. Riskin A, Hochwald O, Bader D, et al. The effects of lactulose supplementation to enteral feedings in premature infants: a pilot study. J Pediatr 2010;
156:209214.
57. Embleton ND, Berrington JE, McGuire W. Lactoferrin: antimicrobial activity and
therapeutic potential. Semin Fetal Neonatal Med 2013. [Epub ahead of print]
&

0951-7375 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-infectiousdiseases.com

243

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Vous aimerez peut-être aussi