Anda di halaman 1dari 6

Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Fetal and Neonatal Medicine


journal homepage: www.elsevier.com/locate/siny

Necrotizing enterocolitis: The intestinal microbiome, metabolome and


inflammatory mediators
Josef Neua,∗, Mohan Pammib
a
Section of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
b
Section of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Necrotizing enterocolitis (NEC) is a disease of preterm infants and associated with significant mortality and
Microbiome morbidity. Although the pathogenesis of NEC is not clear, microbial dysbiosis, with a bloom of the phylum
Intestinal Proteobacteria, has been reported. Antibiotics and the use of H2 blockers, which affect the gut microbiome, are
Preterm associated with increased incidence of NEC. In association with dysbiosis, inflammatory processes are upregu-
Necrotizing enterocolitis
lated with increased Toll-like receptor signaling, leading to translocation of nuclear factor kappa-β, a tran-
scription factor that induces transcription of various pro-inflammatory cytokines and chemokines. Microbial
metabolites, short chain fatty acids including acetate and butyrate, may modulate immunity, inflammation,
intestinal integrity and regulate transcription by epigenetic mechanisms. Evaluation of microbiome and meta-
bolome may provide biomarkers for early diagnosis of NEC and microbial therapeutic approaches to correct
microbial dysbiosis.

1. Necrotizing enterocolitis outnumber human cells and genes. Host–microbe interactions on in-
testinal and respiratory mucosal surfaces play a major role in the pre-
Necrotizing enterocolitis (NEC) is a major cause of mortality in vention of infections and development of immunity. Intestinal micro-
preterm infants who survive the first few days after birth [1,2]. NEC is biota, the best studied, has a bacterial load in the magnitude of 1014
seen in 7% of very low birth weight infants (birth weight < 1500 g) and bacteria/mm3, which makes it the most densely colonized surface of the
up to 5% of admissions to the neonatal intensive care unit (NICU) [2,3]. human body. Bacteroidetes represent the most abundant phylum, fol-
NEC is associated with a mortality of 15–30% and a significant long- lowed by Firmicutes [11,12]. In healthy adults, Bacteroides, Faecali-
term neurodevelopmental morbidity but we have made little progress bacterium, and Bifidobacterium are the most prevalent genera [12].
for 60 years [3–5]. The pathogenesis of NEC is not clear but microbial The lower respiratory tract is one of the least-populated surfaces of the
dysbiosis, formula feeding and excessive inflammation have been im- human body with an estimated number of 10–100 bacteria per 1000
plicated [3]. Furthermore, a clear definition of this disease remains human cells [13]. The “core microbiota” of healthy individuals consists
elusive and it likely represents several different diseases with the final mainly of genera Pseudomonas, Streptococcus, Prevotella, Fuso-
outcome of intestinal injury or necrosis [6]. bacteria, Veillonella, Haemophilus, Neisseria, and Porphyromonas
Postmenstrual age is correlated with the development of NEC, the [14–16].
peak age being 29–31 weeks (peak 31 weeks) (based on last menstrual
period or conception) [7–9]. Fig. 1 summarizes several data sets on the 2.1. Normal development of a preterm intestinal microbiome
post-conceptual timing of NEC (Fig. 1) [10]. Prematurity is the major
risk factor, but, since not all preterm infants develop this disease, sev- Before birth, the fetus is bathed by the amniotic fluid that may not
eral other environmental factors including intestinal dysbiosis may be be sterile [17]. Rather, the fetal–maternal unit is constantly exposed to
involved. microbes and microbial metabolites that may originate from the vagina,
and mother's gastrointestinal tract including the mouth [18]. Recent
2. Microbiome and human health studies suggest that amniotic fluid contains microbes that can most
readily be detected with non-culture based techniques and that the
The microbial communities on the surface of the human body placenta harbors microbial DNA [17,18]. The fetus ingests large


Corresponding author. Section of Neonatology, Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32607, USA.
E-mail address: neuj@peds.ufl.edu (J. Neu).

https://doi.org/10.1016/j.siny.2018.08.001

1744-165X/ © 2018 Elsevier Ltd. All rights reserved.

Please cite this article as: Neu, J., Seminars in Fetal and Neonatal Medicine (2018), https://doi.org/10.1016/j.siny.2018.08.001
J. Neu, M. Pammi Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

bacterial species, less diversity and increased proportion of potential


pathogens [31,32]. The microbial dysbiosis theory of NEC is supported
by the fact that NEC cannot be produced in germ-free animals [26,33].
It is also suggested by an association between early antibiotic use and
NEC [34,35].

2.3. The intestinal microbial milieu prior to the diagnosis of NEC

Early studies utilizing 16S rDNA for microbiome evaluation sug-


gested that relative increases in the phylum Proteobacteria when
compared to other major microbial phyla such as Firmicutes,
Bacteroides and Negativicutes are associated with the development of
NEC [36,37]. This intriguing relationship suggests that these may be-
Fig. 1. Post-conceptual timing of necrotizing enterocolitis (NEC) diagnosis.
CGA, corrected gestational age.
come targets of microbial intervention against NEC, but true causality
[y-Axis label: ‘No. of NEC cases’]. has not yet been demonstrated and will be a requisite in the determi-
nation of whether a certain individual microbe or microbial pattern is
responsible for the disease. The microbial version of Koch's postulates
quantities, up to 150 mL/kg/d, of amniotic fluid and the fetal gastro-
needs to be fulfilled for causality to be established.
intestinal tract is exposed to large number of microbes and microbial
There are issues with 16S rRNA gene sequencing in the evaluation of
components during gestation. Several studies demonstrate that the
the human microbiome. Taxonomic resolution is limited, and strain-
meconium contains microbial DNA as well as live microbes [19,20].
level resolution and tracking are not possible. The metabolic functions
Studies suggest that the composition of these microbes in amniotic
of the microbiome cannot be assessed. In contrast, metagenomics or
fluid, meconium and placenta differ depending on gestational age of
whole genome shot-gun sequencing can provide strain-level resolution.
delivery. Meconium microbes most likely reflect the in-utero rather
In addition to understanding the microbial community structure, the
than the extrauterine environment. The microbial milieu in the meco-
metabolic potential of the microbial communities can also be assessed
nium of babies may determine the development of sepsis and NEC [19].
[38,39]. 16S rRNA gene evaluation is based on existing information of
After birth, the preterm intestinal microbiome varies with post-
conserved sequences, and unusual sequences are missed. The metage-
menstrual age and undergoes a patterned progression and increase in
nomic approach enables an unbiased evaluation of the microbial
anaerobes (around 36 weeks corrected gestational age (CGA)), in-
communities even those with unusual RNA sequences or plasmids.
dicating maturation of the microbiome [21]. An enumeration of mi-
Meta-genomic approach may also be useful in the diagnosis of a disease
crobes of stools from preterm infants using 16SrRNA sequencing, has
due to an unusual or unsuspected organism [40]. Metagenomic ap-
determined that the microbiota progresses through a succession of the
proaches are labor intensive and much more expensive than the 16S
bacterial classes from Bacilli, to Gammaproteobacteria, to Clostridia
rRNA gene sequencing methods. Metagenomic studies on NEC are
[21]. By the time the infants approach 33–36 weeks post conceptual
scarce although some small-sample studies have been published
age, anaerobes constitute a large component of the gut colonization.
[41,42]. Raveh-Sidka et al. studied the stool microbiome in preterm
infants (five with NEC and five controls) by the meta-genomic approach
2.2. Microbial dysbiosis and NEC and reported that although many microbial species were found in many
infants, the strains colonizing each infant were distinct, indicating a
The concept of NEC associated with inappropriate colonization of paucity of shared gut colonization [41]. In another meta-genomic study
the premature intestine was introduced by Claude and Walker [22]. of the early intestinal microbiome of 144 preterm and 22 term infants,
This was because “epidemics” of NEC had occurred, but no pathogen NEC and mortality were associated with uropathogenic E. coli coloni-
had been associated with NEC. The authors posited that, rather than a zation [43].
direct infection, NEC may be the result of a secondary inflammation in La Rosa et al. observed an orchestrated patterned progression of gut
response to microbial colonization or infection. Histologic sections of microbiota towards an abundance of Clostridia with increasing gesta-
intestine from infants with NEC often showed evidence of necrosis and tional age [21]. Antibiotics, feeding or mode of delivery caused abrupt
inflammation in addition to bacterial overgrowth. They suggested a shifts in microbiota but did not change this predestined progression.
cycle of microbe-induced neutrophil activation leading to inflammatory Other studies comparing gut microbiota in infants with NEC and con-
mediator release, vasoconstriction, and disruption of the intestinal trols have discussed a Proteobacteria bloom positively associated [44]
barrier that was initially caused by interaction of a “dysbiotic” in- and anaerobes (especially Negativicutes) negatively associated with
testinal microbiota and an immature intestine. They also suggested that NEC [37]. In our meta-analyses of microbiome studies in preterm in-
various factors such as breastfeeding could reduce colonization by pa- fants – NEC versus controls who did not develop NEC – we found
thogenic organisms but induce colonization by commensal organisms, gradual shifts in the relative abundances of multiple phyla at about 27
which in turn modulate inflammatory reactions and decreased in- weeks CGA, where decreased abundances of Firmicutes and Bacter-
testinal injury. Other studies have implicated microbial dysbiosis in the oidetes and increased abundances of Proteobacteria preceded the di-
gastrointestinal tract and an exaggerated inflammatory response in the agnosis of NEC [10] (Fig. 2).
development of NEC [23–25]. Immune dysregulation in association
with microbial dysbiosis has also been implicated in the pathogenesis of 2.4. Factors that alter the microbiome and may predispose to NEC
NEC. Excessive Toll-like receptor-4 (TLR4) signaling in response to li-
popolysaccharide (LPS) [26,27] and an exaggerated inflammatory re- Several studies evaluating antibiotic use in preterm infants suggest
sponse [24,25] in preterm infants have been reported. an increased odds ratio in the development of NEC, which is directly
Prior to the last decade, several studies have searched for a specific related to the days of antibiotic usage [35,45,46]. Studies by La Rosa
microbial origin of NEC using culture-based techniques [28–30]. Re- et al. and Murgas et al. suggest a relationship between antibiotic use in
cently, culture-independent techniques have been employed to evaluate preterm infants, the microbial composition of the gastrointestinal tract,
the microbial environment. Studies have reported dysbiotic microbial and the subsequent development of NEC [21,47]. Additional studies
patterns rather than a single pathogen causing NEC [10]. Compared to suggest that the acid–base environment of the gastrointestinal tract may
term infants, the intestinal microbiota in preterm infants has fewer also play a significant role in the development of NEC [48]. This may

2
J. Neu, M. Pammi Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

Fig. 2. Microbial profiles in necrotizing enterocolitis (NEC).

3
J. Neu, M. Pammi Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

also relate to the types of microbe present in the gastrointestinal tract mice and TLR4 knockout mice are protected from NEC [61–64]. It is
because they respond differently to the acid–base environment in terms possible that inhibition of the TLR4 pathway may be a novel strategy in
of their growth. H2 blockers favor the proliferation of the Proteo- the treatment or prevention of NEC.
bacteria over Firmicutes in the gastrointestinal tract [49]. This re-
lationship is highly intriguing since the Proteobacteria to Firmicute 4. Metabolome in necrotizing enterocolitis
ratios also are altered in those infants who subsequently develop NEC
[10]. Animal studies in rabbits who were fed acidified milk had de- Microbiota-derived metabolites, short chain fatty acids (SCFAs),
creased gut colonization and translocation, emphasizing the protective butyrate, propionate and acetate induce IL-18 production from the in-
effects of gastric acidity on microbial colonization of the gut [50,51]. testinal epithelial cells (IECs) through activation of NOD-like family,
Human studies in which a formula was acidified resulted in a lower receptors (NLRs) [65]. Acetate produced by Bifidobacteria promotes
incidence of NEC [52]. Retrospective database reviews and secondary epithelial cell barrier function by inducing an anti-apoptotic response in
analysis of large multi-center studies show that the use of antacids, both the IECs. Microbiota-derived sphingolipids presented on CD1d by
H2 blockers and proton pump inhibitors increase the risk for the de- dendritic cells inhibit colonic invariant natural killer T-cell develop-
velopment of NEC [53,54]. Bovine lactoferrin supplementation miti- ment. Thus microbiota and their metabolites mediate immune response
gates this effect [53]. via the IECs and immune cells [66,67]. Germ-free mice have lower le-
The well-known phenomena that babies receiving their own mo- vels of SCFAs compared to conventionally housed animals [68,69].
ther's milk have a decreased incidence of NEC is likely to relate to many Butyrate also is a major fuel for colonocytes and is a major stimulus for
factors in the milk, including its macronutrient composition, the com- tight junction formation and integrity [70]. SCFAs have also been
position of polyunsaturated fatty acids, lactoferrin, various immune shown to stimulate histone acetylation of FoxP3 (forehead box P3)
cells, immunoglobulins, and microbes. The fact that human milk con- locus on naïve CD4+ T-cells, increase FoxP3 expression and promote
tains microbes that appear to be personalized for each mother's own differentiation of Tregs (anti-inflammatory effects) [65,68,71]. Faecali-
infant [55] is a fascinating phenomenon that may also relate to the bacterium prausnitzii and Eubacterium rectale/Roseburia species (Firmi-
decreased risk of NEC in babies fed their own mothers' milk. The fact cutes) are major contributors of butyrate which regulates gene ex-
that donor milk is usually pasteurized and thus devoid of cellular ma- pression by histone modifications [72]. LPS – an inflammatory marker
terial as well as live microbes could be highly germane in this regard. for cardiovascular disease – may also have a role in epigenetic regula-
Studies utilizing only donor milk in comparison to commercial formula tion of intestinal and immune cells [73].
have not overwhelmingly favored the donor human milk and the pre-
vention of NEC. Some of the studies included in meta-analyses of donor 4.1. Clinical studies of the metabolome in neonates
milk versus formula are nearly 30 years old and it is difficult to discern
the criteria utilized in these studies to diagnose NEC in these older ‘Metabolomics’ was coined in 1998 by Oliver et al. [74] and is the
studies. science of detecting small molecules, the result of metabolic pathways
from biological specimens such as plasma, serum, urine and tissues, and
3. Microbiome immunity and inflammation is the latest of the ‘omics’ technologies. Metabolomics detects the pro-
ducts of the metabolic pathways in an organism which may be useful in
3.1. Microbial–host interactions and innate immunity, inflammation diagnosis, prediction, prognosis or assigning disease status (biomarker
detection). Metabolomics allows identification of distinct patterns of
The relationship between microbes and the innate immune system small molecules generated during both host and microbial cellular
in the gastrointestinal tract has been reviewed [56,57]. Intestinal cells metabolism and may be useful in searching for biomarkers of micro-
including the intestinal epithelium harbor receptors to microbial com- biome patterns and dysbiosis [75–79]. Metabolite patterns are dy-
ponents, TLRs, which play a major role in innate immunity [26,58]. namic, changing with gestational age, time or disease process and at
Activation of these Toll-like receptors results in signaling cascades that any time give us a snapshot of the metabolic milieu of the organism.
induce nuclear translocation of nuclear factor kappa-β (NFKβ), a tran- The complexity and the numerous metabolites to be measured need
scription factor that induced transcription of various pro- and anti-in- sophisticated analytical techniques. Nuclear magnetic resonance (NMR)
flammatory cytokines and chemokines [25]. For example, activation of spectroscopy and mass spectrometry (MS) are the techniques most
the chemokine interleukin (IL)-8 attracts neutrophils to trigger areas of widely employed.
the intestine, where they undergo phagocytosis, and other in- Very few studies have performed on metabolomic patterns in NEC
flammatory responses which subsequently cause microvascular con- patients, compared to controls or healthy preterm infants [80–84].
striction, ischemia and injury to the intestine [59]. Certain microbes Although there is no unifying metabolomic signature in NEC, these
may also affect dendritic cells, which are of the monocyte/macrophage studies have reported interesting results. A multi-center evaluation of
lineage. These cells can traverse the intestinal epithelium, grasp anti- the gut microbiome and metabolome named ‘Mechanisms affecting the
gens from the lumen, carry them to the sub-epithelium and act as an- gut of preterm infants in enteral feeding trials (MAGPIE)’ study, funded
tigen presenting cells to undifferentiated lymphocytes in the intestinal by the UK NIHR Efficacy and Mechanistic Evaluation program, is in
lamina propria. Depending on the surface receptors that are stimulated progress [85].
by the distinct microbial patterns to which these cells are exposed, in- Morrow et al. evaluated urinary metabolome patterns in association
teraction between dendritic cells may result in the differentiation of T- with gut dysbiosis preceding NEC and found that three urinary meta-
cells into effector versus tolerogenic cell types. The precise mechanisms bolites differed in those with NEC compared to those without the dis-
of these interactions as they relate to NEC are currently poorly under- ease [81]. Alanine in the urine was directly correlated with the relative
stood. abundance of Firmicutes (P = 0.009), and was inversely correlated
In our meta-analyses of microbiome studies in NEC, Proteobacteria with the relative abundance of both Proteobacteria (P = 0.027) and
abundances increase in babies who develop NEC compared to controls Propionibacterium (P = 0.015) [81]. Histidine was not independently
[10]. LPS produced by Proteobacteria is recognized by TLR4 in the associated with microbial community characteristics, but ratio of ala-
intestine, which triggers intestinal inflammation and enterocyte injury nine to histidine was positively associated with overall NEC (Krus-
that may lead to the development of NEC in preterm infants [60]. TLR4 kal–Wallis, P = 0.001) and inversely associated with the relative
expression is higher in the intestinal tract of preterm neonates when abundance of Propionibacterium (Spearman's rho = −0.57,
compared to terms, and thus may regulate the balance between in- P = 0.002).
testinal repair and injury [26]. It is interesting to note that TLR4 mutant Wilcock et al. described metabolomic differences in serum between

4
J. Neu, M. Pammi Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

preterm infants who developed NEC (five preterm Furthermore, US Food and Drug Administration-based drug standards
infants < 28 weeks GA) and those who did not (seven should be utilized if these agents are to be used for prevention of NEC.
preterm < 29 weeks GA and eight term infants) [82]. Serum samples in Whether fecal microbial transplant-like approaches such as those being
the first week of life in these preterm infants did not predict who would used in the treatment of Clostridium difficile infections may be of benefit
later develop NEC. However, when the infants were on complete in the prevention of NEC could be a basis of future investigation.
feeding, analyses revealed differences in 12 metabolites related to Transfaunation of donor milk using small amounts of the baby's own
carbohydrate, lipid metabolism and intracellular signaling, between mother's milk may also be a means to “personalize” the donor milk to
NEC and control infants. Based on the metabolites identified, an in- provide a means to protect the infant's intestinal tract from the mi-
hibition of the pro-insulin and upregulation of IL-1β were correlated crobial environment that predisposes to NEC [86].
with the development of NEC.
Stewart et al. studied a large cohort of preterm infants (641 samples Conflicts of interest
from 35 infants from a cohort of > 300 infants) and compared preterm
infants with NEC (n = 7) to those without (n = 28) by 16S rRNA gene None declared.
profiling and metabolomic analysis of stools [84]. A subset of 75
samples (n = 16 patients) were also subjected to metabolomic analysis Funding sources
by ultra-performance liquid chromatography mass spectrometry. At the
time of NEC diagnosis, five metabolites were significantly different in None.
preterm infants with NEC, two metabolites from C21-steroid hormone
biosynthesis and linoleate metabolism pathways, two metabolites from References
the linoleate metabolism pathway, and one metabolite from the leu-
kotriene metabolism and prostaglandin formation from arachidonate [1] Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med 2011;364:255–64.
pathway. Stewart et al. also reported a proteomic and metabolomic [2] Fanaroff AA, Stoll BJ, Wright LL, et al. Trends in neonatal morbidity and mortality
for very low birthweight infants. Am J Obstet Gynecol 2007;196:147. e1–8.
analysis of the serum in preterm infants who developed NEC with [3] Lin PW, Stoll BJ. Necrotising enterocolitis. Lancet 2006;368:1271–83.
controls but did not identify any protein or metabolite associated with [4] Stoll BJ, Hansen NI, Adams-Chapman I, et al. Neurodevelopmental and growth
the development of NEC [80]. impairment among extremely low-birth-weight infants with neonatal infection.
J Am Med Assoc 2004;292:2357–65.
De Magistris et al. compared six infants who developed NEC with 12 [5] Bell MJ, Ternberg JL, Feigin RD, et al. Neonatal necrotizing enterocolitis.
controls by NMR-based urinary metabolic profiles. The investigators Therapeutic decisions based upon clinical staging. Ann Surg 1978;187:1–7.
found increased urinary gluconic acid in NEC patients. Gluconic acid [6] Gordon PV, Swanson JR, MacQueen BC, Christensen RD. A critical question for NEC
researchers: can we create a consensus definition of NEC that facilitates research
can be a byproduct of calcium gluconate administration or a byproduct progress? Semin Perinatol 2017;41:7–14.
of bacterial metabolism by alternate metabolic pathways [7] Yee WH, Soraisham AS, Shah VS, Aziz K, Yoon W, Lee SK. Incidence and timing of
(Entner–Doudoroff pathway). It is possible that production of gluconic presentation of necrotizing enterocolitis in preterm infants. Pediatrics
2012;129:e298–304.
acid can favor bacteria that utilize gluconic acid but are more virulent
[8] Gonzalez-Rivera R, Culverhouse RC, Hamvas A, Tarr PI, Warner BB. The age of
[83]. necrotizing enterocolitis onset: an application of Sartwell's incubation period model.
There is no unifying hypothesis related to altered metabolism in J Perinatol 2011;31:519–23.
NEC from published studies, and the use of metabolomic techniques in [9] Gordon PV, Clark R, Swanson JR, Spitzer A. Can a national dataset generate a no-
mogram for necrotizing enterocolitis onset? J Perinatol 2014;34:732–5.
understanding NEC pathogenesis is at its infancy. Integration of all [10] Pammi M, Cope J, Tarr PI, et al. Intestinal dysbiosis in preterm infants preceding
omics platforms is needed to understand the pathophysiology in NEC. necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome
2017;5:31.
[11] Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial
5. Future directions flora. Science 2005;308:1635–8.
[12] Robles Alonso V, Guarner F. Linking the gut microbiota to human health. Br J Nutr
For the prediction and diagnosis of NEC, diagnostic biomarkers in- 2013;109(Suppl 2):S21–6.
[13] Sze MA, Dimitriu PA, Hayashi S, et al. The lung tissue microbiome in chronic ob-
clude those that may not be specific for NEC such as the C-reactive structive pulmonary disease. Am J Respir Crit Care Med 2012;185:1073–80.
protein, white blood cell count, and platelet count. However, more [14] Erb-Downward JR, Thompson DL, Han MK, et al. Analysis of the lung microbiome
specific biomarkers are being developed which include tight junction in the “healthy” smoker and in COPD. PLoS One 2011;6:e16384.
[15] Charlson ES, Bittinger K, Haas AR, et al. Topographical continuity of bacterial
proteins such as claudin-3, intestinal epithelial cell proteins such as
populations in the healthy human respiratory tract. Am J Respir Crit Care Med
intestinal fatty acid binding protein (IFAB), both of which can be 2011;184:957–63.
analyzed non-invasively in the urine. More predictive biomarkers using [16] Charlson ES, Diamond JM, Bittinger K, et al. Lung-enriched organisms and aberrant
bacterial and fungal respiratory microbiota after lung transplant. Am J Respir Crit
the proteomic, metabolomic and genomic platforms are being eval-
Care Med 2012;186:536–45.
uated. A novel “electronic nose” technology is highly intriguing, but [17] DiGiulio DB. Diversity of microbes in amniotic fluid. Semin Fetal Neonatal Med
studies that critically evaluate its predictive and diagnostic potential 2012;17:2–11.
remain to be done. [18] Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta
harbors a unique microbiome. Sci Transl Med 2014;6. 237ra65.
The fact that certain microbial patterns as well as single microbes [19] Heida FH, van Zoonen A, Hulscher JBF, et al. A necrotizing enterocolitis-associated
are being found to be highly associated with NEC is intriguing. gut microbiota is present in the meconium: results of a prospective study. Clin Infect
However, the finding of an association is not adequate to directly base Dis 2016;62:863–70.
[20] Gosalbes MJ, Llop S, Valles Y, Moya A, Ballester F, Francino MP. Meconium mi-
therapeutic interventions. More rigorous studies of the causality will crobiota types dominated by lactic acid or enteric bacteria are differentially asso-
need to be done. These will need to be based on in-vitro as well as in- ciated with maternal eczema and respiratory problems in infants. Clin Exp Allergy
vivo techniques to recapitulate the disease in cell culture, and/or an- 2013;43:198–211.
[21] La Rosa PS, Warner BB, Zhou Y, et al. Patterned progression of bacterial populations
imal models. Once these are established, directed therapies using highly in the premature infant gut. Proc Natl Acad Sci USA 2014;111:12522–7.
specific antibiotics, bacteriophages, probiotics or other microbial ther- [22] Claud EC, Walker WA. Hypothesis: inappropriate colonization of the premature
apeutic techniques may be further evaluated for safety and efficacy. intestine can cause neonatal necrotizing enterocolitis. Faseb J 2001;15:1398–403.
[23] Claud EC, Walker WA. Bacterial colonization, probiotics, and necrotizing en-
Several studies suggest that a microbial therapeutic approach to terocolitis. J Clin Gastroenterol 2008;42(Suppl 2):S46–52.
NEC may be useful in the prevention of NEC. However, the most rig- [24] Nanthakumar NN, Fusunyan RD, Sanderson I, Walker WA. Inflammation in the
orous studies using probiotics to date are not supportive even though developing human intestine: a possible pathophysiologic contribution to necro-
tizing enterocolitis. Proc Natl Acad Sci USA 2000;97:6043–8.
numerous meta-analyses suggest efficacy. The fact that numerous dif-
[25] Nanthakumar N, Meng D, Goldstein AM, et al. The mechanism of excessive in-
ferent probiotics, which are very different in their character, have been testinal inflammation in necrotizing enterocolitis: an immature innate immune re-
utilized in these meta-analyses has been subject of the criticism. sponse. PLoS One 2011;6:e17776.

5
J. Neu, M. Pammi Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

[26] Afrazi A, Sodhi CP, Richardson W, et al. New insights into the pathogenesis and pathogenesis of necrotizing enterocolitis. Semin Pediatr Surg 2005;14. 137–4.
treatment of necrotizing enterocolitis: Toll-like receptors and beyond. Pediatr Res [57] Denning PW, Maheshwari A. Necrotizing enterocolitis: hope on the horizon. Clin
2011;69:183–8. Perinatol 2013;40. xvii–xix.
[27] Morowitz MJ, Poroyko V, Caplan M, Alverdy J, Liu DC. Redefining the role of in- [58] Caplan MS, Simon D, Jilling T. The role of PAF, TLR, and the inflammatory response
testinal microbes in the pathogenesis of necrotizing enterocolitis. Pediatrics in neonatal necrotizing enterocolitis. Semin Pediatr Surg 2005;14:145–51.
2010;125:777–85. [59] Maheshwari A, Schelonka RL, Dimmitt RA, et al. Cytokines associated with ne-
[28] Brook I. Microbiology and management of neonatal necrotizing enterocolitis. Am J crotizing enterocolitis in extremely-low-birth-weight infants. Pediatr Res
Perinatol 2008;25:111–8. 2014;76:100–8.
[29] Balamurugan R, Magne F, Balakrishnan D, et al. Faecal bifidobacteria in Indian [60] Lu P, Sodhi CP, Hackam DJ. Toll-like receptor regulation of intestinal development
neonates; the effect of asymptomatic rotavirus infection during the first month of and inflammation in the pathogenesis of necrotizing enterocolitis. Pathophysiology
life. Indian J Med Res 2010;132:721–7. 2014;21:81–93.
[30] Mancini N, Poloniato A, Ghidoli N, et al. Potential role of the detection of en- [61] Sodhi C, Richardson W, Gribar S, Hackam DJ. The development of animal models
terobacterial DNA in blood for the management of neonatal necrotizing en- for the study of necrotizing enterocolitis. Dis Models Mech 2008;1:94–8.
terocolitis. J Med Microbiol 2012;61:1465–72. [62] Sodhi CP, Neal MD, Siggers R, et al. Intestinal epithelial Toll-like receptor 4 reg-
[31] Carlisle EM, Morowitz MJ. The intestinal microbiome and necrotizing enterocolitis. ulates goblet cell development and is required for necrotizing enterocolitis in mice.
Curr Opin Pediatr 2013;25:382–7. Gastroenterology 2012;143. 708–18 e1–5.
[32] Wang Y, Hoenig JD, Malin KJ, et al. 16S rRNA gene-based analysis of fecal mi- [63] Sodhi CP, Shi XH, Richardson WM, et al. Toll-like receptor-4 inhibits enterocyte
crobiota from preterm infants with and without necrotizing enterocolitis. ISME J proliferation via impaired beta-catenin signaling in necrotizing enterocolitis.
2009;3:944–54. Gastroenterology 2010;138:185–96.
[33] Musemeche CA, Kosloske AM, Bartow SA, Umland ET. Comparative effects of [64] Jilling T, Simon D, Lu J, et al. The roles of bacteria and TLR4 in rat and murine
ischemia, bacteria, and substrate on the pathogenesis of intestinal necrosis. models of necrotizing enterocolitis. J Immunol 2006;177:3273–82.
J Pediatr Surg 1986;21:536–8. [65] Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty
[34] Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial empirical antibiotic acids, regulate colonic Treg Cell Homeostasis. Science 2013;341:569–73.
treatment is associated with increased rates of necrotizing enterocolitis and death [66] Deshmukh HS, Liu Y, Menkiti OR, et al. The microbiota regulates neutrophil
for extremely low birth weight infants. Pediatrics 2009;123:58–66. homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat
[35] Alexander VN, Northrup V, Bizzarro MJ. Antibiotic exposure in the newborn in- Med 2014;20:524–30.
tensive care unit and the risk of necrotizing enterocolitis. J Pediatr 2011;159:392–7. [67] Kabat AM, Srinivasan N, Maloy KJ. Modulation of immune development and
[36] Mai V, Torrazza RM, Ukhanova M, et al. Distortions in development of intestinal function by intestinal microbiota. Trends Immunol 2014;35:507–17.
microbiota associated with late onset sepsis in preterm infants. PLoS One [68] Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria
2013;8:e52876. promote peripheral regulatory T-cell generation. Nature 2013;504:451–5.
[37] Warner BB, Deych E, Zhou Y, et al. Gut bacteria dysbiosis and necrotising en- [69] Hoverstad T, Midtvedt T. Short-chain fatty acids in germfree mice and rats. J Nutr
terocolitis in very low birthweight infants: a prospective case–control study. Lancet 1986;116:1772–6.
2016;387:1928–36. [70] Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier
[38] Tyson GW, Chapman J, Hugenholtz P, et al. Community structure and metabolism by facilitating tight junction assembly via activation of AMP-activated protein ki-
through reconstruction of microbial genomes from the environment. Nature nase in Caco-2 cell monolayers. J Nutr 2009;139:1619–25.
2004;428:37–43. [71] Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces
[39] Gill SR, Pop M, Deboy RT, et al. Metagenomic analysis of the human distal gut the differentiation of colonic regulatory T cells. Nature 2013;504:446–50.
microbiome. Science 2006;312:1355–9. [72] Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential ben-
[40] Relman DA. Microbial genomics and infectious diseases. N Engl J Med eficial effects of butyrate in intestinal and extraintestinal diseases. Wld J
2011;365:347–57. Gastroenterol 2011;17:1519–28.
[41] Raveh-Sadka T, Thomas BC, Singh A, et al. Gut bacteria are rarely shared by co- [73] Angrisano T, Pero R, Peluso S, et al. LPS-induced IL-8 activation in human intestinal
hospitalized premature infants, regardless of necrotizing enterocolitis development. epithelial cells is accompanied by specific histone H3 acetylation and methylation
eLife 2015;4. changes. BMC Microbiol 2010;10:172.
[42] Morowitz MJ, Denef VJ, Costello EK, et al. Strain-resolved community genomic [74] Oliver SG, Winson MK, Kell DB, Baganz F. Systematic functional analysis of the
analysis of gut microbial colonization in a premature infant. Proc Natl Acad Sci USA yeast genome. Trends Biotechnol 1998;16:373–8.
2011;108:1128–33. [75] Tuohy KM, Gougoulias C, Shen Q, Walton G, Fava F, Ramnani P. Studying the
[43] Ward DV, Scholz M, Zolfo M, et al. Metagenomic sequencing with strain-level re- human gut microbiota in the trans-omics era – focus on metagenomics and meta-
solution implicates uropathogenic E. coli in necrotizing enterocolitis and mortality bonomics. Curr Pharmaceut Des 2009;15:1415–27.
in preterm infants. Cell Rep 2016;14:2912–24. [76] Swann JR, Tuohy KM, Lindfors P, et al. Variation in antibiotic-induced microbial
[44] Torrazza RM, Neu J. The altered gut microbiome and necrotizing enterocolitis. Clin recolonization impacts on the host metabolic phenotypes of rats. J Proteome Res
Perinatol 2013;40:93–108. 2011;10:3590–603.
[45] Wojkowska-Mach J, Rozanska A, Borszewska-Kornacka M, et al. Necrotising en- [77] Ponnusamy K, Choi JN, Kim J, Lee SY, Lee CH. Microbial community and meta-
terocolitis in preterm infants: epidemiology and antibiotic consumption in the bolomic comparison of irritable bowel syndrome faeces. J Med Microbiol
Polish neonatology network neonatal intensive care units in 2009. PLoS One 2011;60:817–27.
2014;9:e92865. [78] Dai ZL, Zhang J, Wu G, Zhu WY. Utilization of amino acids by bacteria from the pig
[46] Cotten CM. Adverse consequences of neonatal antibiotic exposure. Curr Opin small intestine. Amino Acids 2010;39:1201–15.
Pediatr 2016;28:141–9. [79] Cevallos-Cevallos JM, Danyluk MD, Reyes-De-Corcuera JI. GC-MS based metabo-
[47] Torrazza RM, Ukhanova M, Wang X, et al. Intestinal microbial ecology and en- lomics for rapid simultaneous detection of Escherichia coli O157:H7, Salmonella
vironmental factors affecting necrotizing enterocolitis. PLoS One 2013;8:e83304. Typhimurium, Salmonella Muenchen, and Salmonella Hartford in ground beef and
[48] Terrin G, Passariello A, De Curtis M, et al. Ranitidine is associated with infections, chicken. J Food Sci 2011;76:M238–46.
necrotizing enterocolitis, and fatal outcome in newborns. Pediatrics [80] Stewart CJ, Nelson A, Treumann A, et al. Metabolomic and proteomic analysis of
2012;129:e40–5. serum from preterm infants with necrotising entercolitis and late-onset sepsis.
[49] Gupta RW, Tran L, Norori J, et al. Histamine-2 receptor blockers alter the fecal Pediatr Res 2016;79:425–31.
microbiota in premature infants. J Pediatr Gastroenterol Nutr 2013;56:397–400. [81] Morrow AL, Lagomarcino AJ, Schibler KR, et al. Early microbial and metabolomic
[50] Mehall JR, Northrop R, Saltzman DA, Jackson RJ, Smith SD. Acidification of for- signatures predict later onset of necrotizing enterocolitis in preterm infants.
mula reduces bacterial translocation and gut colonization in a neonatal rabbit Microbiome 2013;1:13.
model. J Pediatr Surg 2001;36:56–62. [82] Wilcock A, Begley P, Stevens A, Whatmore A, Victor S. The metabolomics of ne-
[51] Boneti C, Habib CM, Keller JE, et al. Probiotic acidified formula in an animal model crotising enterocolitis in preterm babies: an exploratory study. J Matern Fetal
reduces pulmonary and gastric bacterial load. J Pediatr Surg 2009;44:530–3. Neonatal Med 2016;29:758–62.
[52] Carrion V, Egan EA. Prevention of neonatal necrotizing enterocolitis. J Pediatr [83] De Magistris A, Corbu S, Cesare Marincola F, et al. NMR-based metabolomics
Gastroenterol Nutr 1990;11:317–23. analysis of urinary changes in neonatal necrotizing enterocolitis. 11th International
[53] Manzoni P, Garcia Sanchez R, Meyer M, et al. Exposure to gastric acid inhibitors Workshop on Neonatology; Cagliari, Italy, October 26th–31st, 2015. Abstract 34, p.
increases the risk of infection in preterm very low birth weight infants but con- 37–38.
comitant administration of lactoferrin counteracts this effect. J Pediatr 2018;193. [84] Stewart CJ, Embleton ND, Marrs EC, et al. Temporal bacterial and metabolic de-
62–7 e1. velopment of the preterm gut reveals specific signatures in health and disease.
[54] Romaine A, Ye D, Ao Z, et al. Safety of histamine-2 receptor blockers in hospitalized Microbiome 2016;4:67.
VLBW infants. Early Hum Dev 2016;99:27–30. [85] Embleton ND, Berrington JE, Dorling J, et al. Mechanisms affecting the gut of
[55] Hunt KM, Foster JA, Forney LJ, et al. Characterization of the diversity and temporal preterm infants in enteral feeding trials. Front Nutr 2017;4:14.
stability of bacterial communities in human milk. PLoS One 2011;6:e21313. [86] Cacho NT, Harrison NA, Parker LA, et al. Personalization of the microbiota of donor
[56] Neu J, Chen M, Beierle E. Intestinal innate immunity: how does it relate to the human milk with mother's own milk. Front Microbiol 2017;8:1470.

Anda mungkin juga menyukai