AJP 2014 NEC Review

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/268822724

Pathogenesis of Necrotizing Enterocolitis : Modeling the Innate Immune


Response

Article  in  American Journal Of Pathology · November 2014


DOI: 10.1016/j.ajpath.2014.08.028

CITATIONS READS

138 593

7 authors, including:

Scott M Tanner Taylor Berryhill


Clemson University University of Alabama at Birmingham
20 PUBLICATIONS   472 CITATIONS    18 PUBLICATIONS   525 CITATIONS   

SEE PROFILE SEE PROFILE

Tamas Jilling Robin G Lorenz


University of Alabama at Birmingham Genentech
127 PUBLICATIONS   4,380 CITATIONS    148 PUBLICATIONS   7,272 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Immunoextraction & analysis of Vitamin D species from human plasma by LC-MS View project

All content following this page was uploaded by Robin G Lorenz on 01 January 2015.

The user has requested enhancement of the downloaded file.


The American Journal of Pathology, Vol. 185, No. 1, January 2015

ajp.amjpathol.org

REVIEW
Pathogenesis of Necrotizing Enterocolitis
Modeling the Innate Immune Response
Scott M. Tanner,*y Taylor F. Berryhill,* James L. Ellenburg,* Tamas Jilling,z Dava S. Cleveland,x Robin G. Lorenz,y and
Colin A. Martin*

From the Departments of Pediatric Surgery* and Pathologyy and the Division of Neonatology,z Department of Pediatrics, University of Alabama at
Birmingham, Birmingham; and the Department of Pediatric Pathology,x Children’s Hospital of Alabama, Birmingham, Alabama

Accepted for publication August 27, 2014.


Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants.
Address correspondence to Colin A. Martin,
The pathophysiology is likely secondary to innate immune responses to intestinal microbiota by
M.D., Department of Pediatric Surgery,
University of Alabama at Birmingham, 300 the premature infant’s intestinal tract, leading to inflammation and injury. This review provides an
Lowder Bldg., 1600 7th Ave., South updated summary of the components of the innate immune system involved in NEC pathogenesis.
Birmingham, AL 35233-1711; or Robin G. In addition, we evaluate the animal models that have been used to study NEC with regard to the
Lorenz, M.D., Ph.D., Department of involvement of innate immune factors and histopathological changes as compared to those seen in
Pathology, University of Alabama at infants with NEC. Finally, we discuss new approaches to studying NEC, including mathematical
Birmingham, 1825 University Blvd., SHEL models of intestinal injury and the use of humanized mice. (Am J Pathol 2015, 185: 4e16; http://
602, Birmingham, AL 35294-2182. E-mail: dx.doi.org/10.1016/j.ajpath.2014.08.028)
cmartin7@uab.edu or rlorenz@uab.edu.

Necrotizing enterocolitis (NEC) is a disorder characterized hurdles that persist are our incomplete understanding of the
by intestinal necrosis in premature infants that results in developing immune system in premature infants and our
significant morbidity and mortality.1 Approximately 7% of inability to adequately replicate these complex factors in ani-
infants with a birth weight between 500 and 1500 g develop mal models.3,4 This review summarizes the complex intestinal
NEC.1 The pathogenesis is characterized by intestinal immune system in premature infants and details what is known
inflammation that can progress to systemic infection/ about the involvement of innate immune factors in NEC, both
inflammation, multiorgan failure, and death. The bowel is in animal models and in human disease.
distended and hemorrhagic on gross inspection. On micro-
scopic examination, signs of inflammation, mucosal edema,
epithelial regeneration, bacterial overgrowth, submucosal The Neonatal Intestinal Ecosystem
gas bubbles, and ischemic transmural necrosis are seen
(Figure 1, AeE).2 The neonatal intestinal ecosystem is extremely fragile. At
Currently the pathogenesis of NEC is believed to have birth, the newborn is exposed to the external environment for
multifactorial causes, including intestinal immaturity and
microbial dysbiosis. Intestinal immaturity leads to a compro- Supported in part by grants from the Kaul Pediatric Research Institute of the
mised intestinal epithelial barrier, an underdeveloped immune Alabama Children’s Hospital Foundation (C.A.M.), NIH grant P01 DK071176
(R.G.L.), Juvenile Diabetes Research Foundation Grant #36-2008-930
defense, and altered vascular development and tone. The (R.G.L.), Crohn’s and Colitis Foundation of American Grant #26971 (R.G.L.),
compromised epithelial barrier and underdeveloped immune and the Digestive Diseases Research Development Center, University of
system, when exposed to luminal microbiota that have been Alabama at Birmingham grant P30 DK064400 (R.G.L.). Aspects of this project
shaped by formula feedings, antibiotic exposure, and Cesarean were conducted in biomedical research space that was constructed with funds
delivery, can lead to intestinal inflammation and sepsis. supported in part by NIH grant C06RR020136 (R.G.L.).
Disclosures: None declared.
Despite therapeutic success in animal model systems, there are A guest editor acted as Editor-in-Chief for this manuscript. No person at the
relatively few therapeutic strategies that have allowed for University of Alabama at Birmingham was involved in the peer review process
significantly improved outcomes in infants with NEC. Two or final disposition for this article.

Copyright ª 2015 American Society for Investigative Pathology.


Published by Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.ajpath.2014.08.028
Pathogenesis of Necrotizing Enterocolitis

Figure 1 Examples of the various grades of morphological damage in hematoxylin and eosinestained specimens. AeE: Representative samples of premature
infants with necrotizing enterocolitis. A: Age-matched control from patient with jejunal atresia. B: Mild injury with hemorrhagic necrosis of mucosa and loss of villus
tip architecture. C: Progressive injury with inflammatory infiltration of muscularis with complete villus destruction. D: Severe muscular and epithelial damage with
complete loss of mucosa. E: Perforation with transmural necrosis with complete loss of epithelial and muscular architecture. FeJ: Representative samples from
intestinal injury secondary to gavage feeding in the setting of hypothermia and hypoxia in neonatal rats. F: Intact morphology, grade 0. G: Sloughing of villus tips,
grade 1. H: Mid-villus necrosis, grade 2. I: Loss of villi, grade 3. J: Complete destruction of the mucosa, grade 4. Insets in FeJ show higher magnified portions of the
same sections, corresponding to the boxed regions. KeO: Representative images of tissue injury secondary to 60 minutes of intestinal ischemia and 90 minutes of
reperfusion in 2-week-old mice. K: Sham-operated mice (no ischemia). L: Villus tip necrosis. M: Mid-villus necrosis. N: Loss of villus architecture. O: Complete loss of
mucosal architecture. FeJ, reprinted with permission from Nature Publishing Group.28 Scale bars Z 50 mm (AeE, KeO). Original magnification, 20 (AeO, main
images, and FeJ, insets).

the first time, and the immune response must begin to to the host.8 The major components are cells (including
distinguish between self and nonself. In particular, the macrophages, neutrophils, dendritic cells (DCs), natural killer
recognition of food antigens and commensal microbiota must cells, B1 B cells, innate lymphoid cells, and gd T cells) and
be distinguished from that of potential pathogens. This pro- anatomical barriers (such as the intestinal epithelium and the
cess is even more challenging in premature infants, as they gastrointestinal mucus layer). In addition, the presence of
are typically placed on broad-spectrum antibiotics that disrupt commensal microbiota can serve as a part of the innate im-
both the timing and the diversity of the initial bacterial mune system by preventing the colonization by pathogenic
colonization of the intestinal tract.1 In addition, the immature bacteria (colonization resistance).
epithelial barrier appears to be more sensitive to the detection
of bacteria5,6 and more susceptible to bacterial translocation,7 Mucosal Innate Immunity
allowing for both an unwarranted inflammatory response to
commensals and the translocation of pathogens, both of Several major components of the human mucosal immune
which may contribute to intestinal damage. system are in place before birth. These components include
The innate immune system is the first line of defense IgMþ B cells; gd T cells found in the intraepithelial
against infections. Innate immune cells respond in a lymphocyte compartment, which are seen as early as 12 to
nonspecific manner and do not confer long-lasting immunity 15 weeks of embryonic age; and Peyer’s patches, which can

The American Journal of Pathology - ajp.amjpathol.org 5


Tanner et al

be seen by about 30 weeks of gestation.9 This development initiate signaling pathways leading to the production of
contrasts with that of the murine mucosal system, in which AMPs, IgA, and epithelial healing factors.
the same B cells and intraepithelial lymphocytes are seen Paneth cellsdfound at the base of the intestinal cryptsd
only about 1 day before birth and macroscopic Peyer’s are yet another secretory cell type that provides protection to the
patches are not seen until at least 1 week after birth.10 These host. Paneth cells are present at around 13 weeks of gestation
differences in intestinal immune system development imply and produce AMPs, which function to disable or kill microor-
that a newborn mouse may have intestinal immaturity very ganisms that have entered the digestive tract. Enterocytes,
similar to that of a premature infantdone reason that murine macrophages, and human neutrophils can also produce AMPs,
models are widely used to study the immune mechanisms including a-defensins, b-defensins, lysozyme, and type IIA
that predispose to NEC development. secretory phospholipase A2, with a-defensins and lysozyme
being the most common in the neonatal intestine.17 Type IIA
Anatomical Barriers secretory phospholipase A2 hydrolyzes the phospholipids in
bacterial cell walls, which results in the production of arach-
Separation of the intestinal lumen from the rest of the organism idonic acid and lyso-PAF.18 As lyso-PAF is converted to the
is accomplished through a physical barrier established by the highly active inflammatory mediator PAF, type IIA secretory
intestinal epithelial cells. All intestinal epithelial cells, phospholipase A2 provides a mechanism that is at the inter-
including enterocytes, Paneth cells, and goblet cells, play an section of bactericidal and inflammatory host defenses.
important role in maintaining barrier integrity. The barrier is
maintained by the presence of tight junctions between the Luminal and Environmental Factors
epithelial cells. In humans, tight junctions are formed by 10
weeks of gestation and are composed of claudins, junctional As the newborn immune system begins its development, SIgA
adhesion molecules, and zonula occludins.11 is transferred via maternal breast milk. Additionally, growth
Goblet cells, positioned throughout the intestine, are factors, cytokines, and other immune modulators are compo-
involved in the secretion of mucin glycoproteins, which nents of human milk, including IL-6, tumor necrosis factor a,
generate the mucus layer of the intestine.12 Goblet cells can IL-10, transforming growth factor (TGF)-b, and lysozyme.19
be found as early as 9 to 10 weeks of gestation,12 and mucins Newborns acquire IgA through passive transfer via the inges-
reach adult levels by 27 weeks of gestation.13 Animal model tion of human milk after birth. The levels of IgA in human milk
studies suggest that mucus gene expression is influenced by remain high throughout the breastfeeding period and help to
bacterial colonization, that mucus protein glycosylation pat- provide protection and aid in the colonization of commensal
terns are developmentally regulated, and that glycosylation bacteria.20 Although neonatal IgA production begins at
alters interactions with bacterial pathogens; however, equiv- approximately 2 weeks after birth, it does not reach adult values
alent studies in humans have not been reported.14e16 Mucus until 2 to 6 years of age.21 Maternal IgA in milk is thought to
forms in two distinct layers; the outer, thicker layer prevents react with the microbiota present in the maternal gastrointestinal
intestinal bacteria from reaching the epithelial layer, whereas tract, and the passive transfer of these antibodies not only is
the inner layer is vital for cell signaling if a disruption oc- thought to prevent the adherence of bacteria to mucosal surfaces
curs.12,17 In addition to preventing bacterial interaction with and bacterial penetration into the mucosa but also is vital for the
the epithelium, the mucus layer also provides scaffolding for formation and composition of the newborn’s intestinal micro-
antimicrobial peptides (AMPs) and secretory IgA (SIgA), biota.19 Maternal milk has been shown to promote colonization
which are discussed in the following section. with bificobacteria.22 As NEC cases are more highly associated
with formula feeding, lower numbers of bifidobacteria and
Microbial Sensors and AMPs bacteroidetes, and higher proportions of proteobacteria and
actinobacteria, it would appear that the shaping of the intestinal
Should pathogenic or commensal bacteria penetrate the microbiota by the maternal IgA is one of the crucial factors in
mucus layer and reach the epithelial cells, pattern-recognition preventing NEC.23 Discussion of the multiple studies that have
receptors (PRRs) are able to sense the presence of the bacteria investigated the dysbiosis seen in NEC are outside of the scope
and initiate an appropriate response. The most common PRRs of this review, but those studies were recently reviewed by
are Toll-like receptors (TLRs) and nucleotide-binding oligo- Torrazza and Neu.24
merization domains (NODs), which are expressed by both IL-10 and TGF-b are anti-inflammatory cytokines involved
epithelial cells and immune cells, particularly macrophages in regulating T-cell responses and in down-regulating proin-
and DCs, throughout the intestinal compartment. The PRRs flammatory secretions from macrophages and neutrophils.
detect common bacterial structures, such as lipopolysaccha- Neonatal monocyte and T cellederived IL-10 levels are
ride (LPS), flagella, and CpG-rich DNA. Current dogma in- significantly lower than are adult levels; however, IL-10 has
dicates that abnormal TLR4 signaling in the premature been detected in human milk, suggesting an important role for
intestinal epithelium is associated with NEC, perhaps by milk-derived IL-10 in neonatal immune development.25 A
increasing platelet-activating factor (PAF) levels. However, recent study has indicated that a low blood TGF-b level is
on detection of microbial components, protective PRRs can associated with a higher risk for NEC.26

6 ajp.amjpathol.org - The American Journal of Pathology


Pathogenesis of Necrotizing Enterocolitis

Modeling NEC epithelial structures, representing a presentation similar to


that of human NEC (Figure 1, FeJ).30 This model has been
Numerous animal models are currently used for studying NEC adapted and more fully understood over the years, and it is
disease pathogenesis. Although each model has specific ad- now known that hypoxia, formula gavage, and intestinal
vantages in mimicking certain aspects of the human disease, it bacteria are required for the development of NEC-like
has limitations,3 including the fact that laboratory animal pathophysiology and histological factors.31 The mouse G/H
models lack genetic diversity, are subject to static environ- model was originally described with pups delivered by Ce-
mental conditions, and do not have innate immune and sarean section (E20-21) but has recently been simplified to
intestinal development identical to that in humans.27 However, use newborn pups gavaged with adult commensal bacte-
human tissue is also not optimal, as surgical specimens are ria.28,29 This simplified model will encourage the use of the
often necrotic and may not reflect the mechanisms that lead to plethora of available knockout and transgenic mice to further
the development of NEC. An ideal model of NEC would: i) be our understanding of NEC. In addition, the G/H model has also
based on the mediators thought to contribute to and induce has been used for linking several components of the innate
human disease, ii) take into account the developmental dif- immune system to the pathogenesis of NEC. Mucin produc-
ferences specific to the neonatal period, iii) have impaired tion, SIgA, TLR4 signaling, AMP expression, and epithelial
intestinal restitution and development of necrosis, iv) include junctional proteins have all been shown to impact the degree of
factors shown to play a role in the human disease (prematurity, intestinal injury in animals that are subjected to the G/H model
hypoxic stress, formula feeding, dysbiosis), and v) have (details in Proposed Innate Immune Mechanisms Leading to
increased severity in the ileum. As this review will demon- NEC); however, the role of innate immune cells in the path-
strate, there is no model that perfectly replicates NEC; how- ogenesis of NEC has not been investigated to date.
ever, each of the following models is a useful tool for Challenges associated with the G/H model include sig-
evaluating the specific mechanisms of the disease (Table 1): nificant variability across research groups regarding the
gavage/hypoxia (G/H) model, ischemia/reperfusion (I/R) specific age at which the protocol is started, the type of
model, PAF administration, Paneth cell depletion, xenograph formula administered, whether the formula is supplemented
model, infection model, and chemical injury models. with bacteria and/or LPS, and the modality of hypoxia
treatment. These challenges make it difficult to reproduce
G/H Model the model between laboratories and to unify the knowledge
gained in the multitude of studies. Although the piglet
Perhaps the most commonly cited animal model of NEC is model is not widely used because of costs and a lack of
the G/H model, which has been described in mice, rats, and reagents, the piglet gastrointestinal system offers the
piglets.28,29 This model links the altered vascular develop- resemblance perhaps closest to that in humans, particularly
ment and tone that are thought to be components of intestinal in the neonatal and newborn stages.32 In pigs, the hypoxia
immaturity with early exposure to cold stress and a nonmilk and gavage are not always paired because one insult (either
diet. Rats are popular for use in these studies because they are hypoxia/hypothermia or formula feedings) can result in
easier to gavage than are mouse pups and are easier and less NEC-like illness.33
expensive to maintain than are piglets. The G/H models are
characterized by sloughing epithelial cells, severe edema in I/R Model
the submucus and muscle layers, villous damage and
destruction, separation of the mucosa and lamina propria, The I/R model of intestinal injury is caused by occluding
and, ultimately, complete destruction and necrosis of all the superior mesenteric artery, thus obstructing blood flow
Table 1 Accuracy of Current Animal Models in Incorporating the Components of Human NEC
Necrotizing enterocolitis models
PAF Paneth cell Infection and
Components of an optimal animal model G/H I/R administration depletion Xenograft chemical injury
Based on predicted mediators of NEC pathogenesis Yes Yes Yes Yes Yes
Reflects developmental differences specific to the Yes
neonatal period/prematurity
Impaired intestinal restitution Yes Yes Yes
Development of necrosis Yes Yes Yes Yes Yes
Hypoxic stress Yes Yes
Temporal relationship to feeding/formula feeding Yes
Dysbiosis Yes Yes Yes Yes
More severe disease in the ileum Yes Yes
The Institutional Care and Use Committee and the institutional review board of the University of Alabama at Birmingham approved all experiments.
G/H, gavage/hypoxia; I/R, ischemia/reperfusion; NEC, necrotizing enterocolitis; PAF, platelet-activating factor.

The American Journal of Pathology - ajp.amjpathol.org 7


Tanner et al

to the small bowel for a short time, followed by a period of operations in young mice. However, it implies a central
reperfusion.34 In the intestine, the initial ischemia results in role of Paneth cells in disease initiation, a hypothesis that
damage to the epithelial cells, whereas the subsequent has recently been proposed but not proven.44 In adults, it is
reperfusion injury causes significant damage to the rest of clear that Paneth cell dysfunction, secondary to genetic risk
the mucus layer.35 Histologically, this model is typically factors, results in dysbiosis and plays a role in intestinal
evaluated based on the extent of necrosis in the intestine, inflammation; therefore, the evaluation of NEC patients for
ranging from necrosis of the villus tip to transmural similar genetic defects might yield support for the role of
necrosis penetrating the muscle layer (Figure 1, KeO).36 Paneth cells in NEC.45
This model attempts to replicate the ischemia that is
thought to occur before NEC; however, it is more accu- Xenograft Model of Human Intestinal Development
rately classified as a model of intestinal injury rather than as
a true model of NEC. The I/R model is particularly useful The xenograft model provides a means of recapitulating the
for investigating the role of free-radical damage in intestinal developmental changes in innate immunity and intestinal
injury, as high levels of oxidative damage are induced after development, both of which may have a role in NEC
reperfusion (mimicking the potential reperfusion after an pathophysiology. Human fetal intestine is implanted s.c.
ischemic event in newborn humans). The primary strength into an immunodeficient mouse host, the host is allowed to
of the I/R model is that it leads to defined and reproducible heal, and the fetal intestinal tissue matures for a designated
mucosal injury and barrier dysfunction. Both TLR4 and period. The xenograft model allows for the assessment of
junctional protein expression have been shown to impact ontogenic changes at points in human intestinal develop-
intestinal injury in the I/R model.37 ment that correspond to the second and third trimesters of
pregnancy.46e48 Although this is a potentially accurate
PAF Administration model of human intestinal development, it does not mirror
NEC pathogenesis as other models do.
The administration of PAF, combined with bacterial LPS,
has been reported to initiate NEC-like pathologies in both Infection and Chemical Injury Models
mice and rats.38,39 PAF has been identified as a proin-
flammatory phospholipid, causing vascular permeability, A mouse model of NEC based on Cronobacter sakazakii
enhanced leukocyte degranulation, adhesion, and chemo- infection was recently reported.49,50 The justification of
taxis, as well as the production of tumor necrosis factor a, using this specific pathogen is the relatively common
IL-6, and IL-8. PAF is primarily produced by innate im- contamination of various formula preparations by this bac-
mune cells, such as neutrophils, basophils, and monocytes, teria.51 This model involves the recruitment of innate im-
as well as endothelial cells and platelets. To induce NEC- mune cells (DCs, macrophages, and neutrophils) and has
like necrosis, PAF and LPS are injected directly into the been used for showing that the depletion of DCs in newborn
mesenteric vascular circulation. In rats, the hemorrhagic mice protects against C. sakazakiieinduced NEC, whereas
lesions of necrosis observed in the jejunum and ileum are the depletion of neutrophils and macrophages exacerbates
similar to those observed in human NEC. On microscopic inflammation. Macrophages also may have a role in a
examination, severe cases demonstrate transmural necrosis, chemically induced model of NEC.52 This model is based
with loss of mucosal architecture and sloughing of epithe- on the clinical observation of numerous macrophages and
lial cells.40 few neutrophils in tissue samples from NEC patients.
MohanKumar et al52 hypothesized that this observation was
Paneth Cell Depletion a reflection of the underdeveloped innate mucosal immune
system, and they designed a model that used trinitrobenzene
Dithizone, when administered i.v. to rats, leads to cell death sulfonic acideinduced inflammation for comparing mucosal
in 25% to 30% of Paneth cells.41 This technique was used injury in newborn versus adult mice. The neonatal mouse
in conjunction with the administration of enteric pathogens has a macrophage-predominate infiltrate similar to that in
(Escherichia coli, Klebsiella pneumonia) to induce NEC- human NEC. These two new models may serve as tools
like pathology in one study.42 That study revealed gross useful for the study of innate immune cells in NEC, which
necrotic lesions as well as damage to the intestinal tissue, to date have not been adequately investigated.
including mucosal edema, loss of villous integrity, areas of
transmural necrosis, and intramural air, all of which are Proposed Innate Immune Mechanisms Leading
observed in human patients. It is interesting to note that
when this treatment was performed in mice before mature
to NEC
Paneth cell development, no disease was observed.43 The Dysbiosis and Barrier Dysfunction
Paneth cell depletion model is useful as it is simpler
to administer dithizone and an enteric pathogen than it A recent study demonstrated a lack of diversity in fecal
is to perform feeding gavages or ischemia-reperfusion bacteria from patients who developed NEC compared to

8 ajp.amjpathol.org - The American Journal of Pathology


Pathogenesis of Necrotizing Enterocolitis

that from unaffected patients.53 A further study importance of dysbiosis and the innate immune compo-
described an increase in proteobacteria and a decrease nents that shape the microbiota in NEC.84
in firmicutes in NEC patients relative to healthy
neonatal controls.54 These studies clearly demonstrate a TLRs
difference in intestinal microbiota in NEC patients.
Because these alterations were detected at the initial One potential initiating factor of NEC is the response of
colonization phase in the patients, it is possible that this epithelial and immune cells to the microbiota that colonize
dysbiosis may contribute to the development of NEC infants after birth. The expression of TLR4, a sensor of the
and is not just a reflection of the disease state. Several Gram-negative bacterial component LPS, is increased in
factors may result in a bacterial dysbiosis after birth, human NEC as well as fetal intestine, and this increase
including Cesarean instead of vaginal delivery, feeding appears to be dependent on microbial colonization
of breast milk instead of formula, and broad-spectrum (Table 2).37,47 PAF has also been shown to induce TLR4
antibiotic exposure. expression in intestinal epithelial cells, and PAF levels are
It is becoming increasing clear that dysbiosis can drasti- elevated in newborns with advanced NEC, whereas levels
cally impact intestinal immune defenses by altering of acetylhydrolase (the PAF-inactivation enzyme) are
apoptotic and inflammatory signaling, barrier function, and decreased in NEC patients.68 Mouse G/H studies have
bacterial detection mechanisms in the intestine.55 Increased demonstrated that the changes in TLR4 expression precede
intestinal permeability and epithelial damage, coupled with histological injury and that epithelial expression of TLR4
deficient epithelial healing, have been identified in NEC, is required for NEC development.5,60,61 Also, treatment
although it is unclear whether these aspects precede or result with polyunsaturated fats to reduce TLR4 expression or
from NEC. Alterations in the tight junctions and the mucus with amniotic fluid to inhibit TLR4 signaling reduces the
layer would provide a mechanism for dysbiotic bacteria to severity of G/H-induced NEC, and the absence of TLR4
reach and cross the epithelial barrier and initiate altered protects from disease.5,64,65 TLR4 signaling during bacte-
immune responses. Conversely, full development of a rial colonization results in inhibited proliferation, migra-
functional mucosal immune system requires the normal tion, and survival and increased endoplasmic reticulum
bacterial colonization that occurs after birth, as newborn stress and apoptosis of enterocytes in the intestine.62,63
pups raised by dams on broad-spectrum antibiotics have These findings suggest that TLR4 activation leads to
increased bacterial translocation, decreased numbers of impaired enterocyte repair and migration, therefore inhib-
CD4þ T cells in mesenteric lymph nodes, and altered iting the effective repair of the intestinal epithelium and
cytokine production (a pattern that is similar to that in in- subsequently resulting in increased bacterial translocation,
fants with NEC).17,37,56 inflammation, and NEC.
Several G/H studies have demonstrated a loss of mucin Recently, Yazji et al85 described a novel role for TLR4 on
2, which might allow for increased bacterial translocation endothelial cells in the regulation of intestinal perfusion.
from the lumen (Table 2).59,73 In addition, rat pups sub- Intestinal ischemia has been thought to be a potential trigger
jected to G/H and subsequently treated with epidermal of NEC; this study shows the importance of innate immune
growth factor (EGF) showed an amelioration of experi- recognition outside the intestinal mucosa in the develop-
mental NEC that was correlated with increases in mucin 2, ment of NEC. The role of TLR4 in NEC has also recently
occludin, and claudin-3 and decreased epithelial cell been linked to other PRRs, including TLR9 and NOD2. The
apoptosis.69,73 These studies demonstrate the importance administration of bacterial CpG DNA (the ligand of TLR9)
of the epithelial barrier and imply its role in preventing or the activation of NOD2 inhibited TLR4 signaling and
bacterial translocation that may be crucial for the pre- greatly reduced the incidence of murine NEC.66,67 The
vention of NEC. dysbiosis seen in infants predisposed to NEC may
Also controlling microbial colonization in the intestines contribute to the disease through differential ligation of
of neonates are AMPs, several of which are produced by TLR4 versus TLR9/NOD2.
Paneth cells. Studies have demonstrated that AMP secre- Using the xenograft model of intestinal development,
tions are altered in infants with NEC; specifically, lyso- studies indicate that immature xenograft and NEC intestinal
zyme is decreased, whereas the a human defensins 5 and 6 epithelial cells have increased mRNA expression of TLR2,
are increased.57,58 Human NEC samples have been re- TLR4, myeloid differentiation primary response 88 (MyD88),
ported to have both an increase and a decrease in Paneth NF-kB, and IL-8 and decreased expression of the TLR and
cell numbers relative to control samples.17,57 Because NF-kB negative regulators, toll-interacting protein, and single
Paneth cell numbers increase during gestation and are Ig IL-1erelated receptor compared with that in mature con-
regulated by inflammatory mediators, these discrepancies trols.47 Additionally, reports describe that the treatment of
may be attributed to differences in gestational age at birth immature xenografts with media containing probiotic factors
or they might reflect the heterogeneity of samples. As the led to the down-regulation of TLR2 and TLR4 as well as
dithizone model certainly mimics some of these observa- the up-regulation of toll-interacting protein and single Ig
tions, it should be useful for further studies on the IL-1erelated receptor comparable to those in mature

The American Journal of Pathology - ajp.amjpathol.org 9


Tanner et al

Table 2 Changes in Innate Immune Mechanisms Observed in Animal Models of Necrotizing Enterocolitis and Related Observations in
Human Studies
General alteration Specific physiological changes Species Model Links to human disease
Dysbiosis/barrier Altered Gram-negative bacterial Mouse G/H Changes in microbiota24,53
dysfunction populations24,28,29
Altered bacterial populations49,50 Mouse Infection
Paneth cell depletion41,43 Rat Dithizone Altered Paneth cell numbers17,57
Decreased antimicrobial activity41,43 Rat Dithizone Decreased lysozyme and
a-defensin58
Decreased mucin 259 Rat G/H Decreased Goblet cells2
Altered microbial Increased epithelial TLR4 expression5,60,61 Rat G/H Increased epithelial TLR447
sensing Mouse
Increased TLR2, TLR4, myeloid Rat Xenograft
differentiation primary response 88, NF-
kB, and IL-8; decreased toll interacting
protein and single Ig IL-1e
related receptor47
Increased epithelial damage after TLR4 Mouse G/H
signaling62,63
Decreased TLR4 after polyunsaturated fat Mouse G/H
treatment64
Decreased TLR4 after amniotic fluid Mouse G/H
treatment (via EGF)65
Decreased TLR4 after TLR9 and NOD2 Mouse G/H
stimulation66,67
PAF induction of TLR468 Rat PAF/LPS Elevated PAF in NEC patients68
EGF Decreased epithelial cell apoptosis/ Rat G/H Low EGF levels in NEC patients71
autophagy after EGF treatment69,70
Decreased NEC after HB-EGF treatment72 Rat G/H
Increased mucin 2, occludin, claudin-3 after Rat G/H
EGF treatment69,73
External modulators Increased intestinal damage on PAF/LPS Rat PAF/LPS Increased PAF levels68
administration38,39 Mouse Decreased acetylhydrolase levels68
Increased disease in IL-10/ mice74 Mouse G/H Differences in IL-10 in breast milk,
IL-10 administration ameliorates Rat G/H serum75,76
disease77,78
Decreased disease on increased IL-10 Mouse PAF/LPS
signaling induced by Lactobacillus
rhamnosus GG79
IGF-1 protects intestinal epithelium from Rat Cell line Low IGF levels in NEC patients81
oxidative stress80
Decreased TGF-b signaling leads to Mouse PAF/LPS Low TGF-b levels in NEC patients and
increased disease40 preterm milk82
Innate immune cells Depletion of neutrophils and macrophages Mouse Infection
increases disease50
Increased macrophage infiltrate52 Mouse Chemical injury Macrophage predominant
infiltrate52
Depletion of DCs protects from disease49 Mouse Infection Decreased plasmacytoid DC function
in preterm infants83
Physiological changes indicated in italics have been reported to be protective.
DC, dendritic cells; EGF, epidermal growth factor; G/H, gavage/hypoxia; HB-EGF, heparin bound-epidermal growth factor; IGF, insulin growth factor; LPS,
lipopolysaccharide; NEC, necrotizing enterocolitis; NOD, nucleotide-binding oligomerization domains; PAF, platelet-activating factor; TGF, transforming
growth factor; TLR, toll-like receptors.

xenografts.48 These data suggest that differences in the Together, these data highlight the importance of proper
maturation of the TLR-induced NF-kB signaling pathway may bacterial detection by the intestinal epithelial barrier
be a part of the mechanism of NEC development, and that (particularly through TLR4 and TLR9) and their subsequent
these pathways may be controlled by interaction with probiotic signaling cascades to induce a proper immune response.
microorganisms. When this sensing is disrupted through antibiotic treatment,

10 ajp.amjpathol.org - The American Journal of Pathology


Pathogenesis of Necrotizing Enterocolitis

or occurs prematurely in neonatal infants, the physical lumen and breast milk by transcytosis through the epithelial
barrier is breached and bacteria are exposed to the infant’s cells. During this process, it acquires a secretory component
immune system. In contrast to the increased epithelial that protects the IgA antibodies from degradation by the
expression of TLR4 in premature infants, it has been re- gastric acids and enzymes of the digestive system. There-
ported that infants born at <28 weeks of gestation have very fore, SIgA has properties that are profoundly different from
low levels of innate immune receptors (CD14, TLR2, those of the serum IgA used so far in studies. Further in-
TLR4) on their core blood leukocytes and have severely vestigations are necessary to elucidate the role of SIgA in
impaired leukocyte inflammatory responses to bacteria.86 protection from NEC, as none of the current animal models
This impaired innate immune function likely leads to an have evaluated this aspect of innate immunity in the disease.
inability to control infection due to the impaired barrier, and Studies involving IL-10 have revealed slightly more in-
subsequently aberrant immune activation results, leading to formation and do suggest an important role of IL-10 in
NEC. However, further studies are needed to specifically protection from NEC. It has been reported that the per-
investigate the contributions of PRRs on leukocytes in the centage of mothers with undetectable IL-10 in their milk
development of NEC. was significantly greater in mothers of infants who devel-
oped NEC.25 In addition, levels of SIgA and IL-10 in
EGF human milk are lower after very preterm delivery (<30
weeks of gestation) compared with preterm (30 to 37 weeks
Studies in both animal models and human patients have of gestation) and term (38 to 42 weeks of gestation) de-
identified EGF as a potential biomarker for NEC develop- livery.75 Conversely, a recent study measured increased
ment and as a future treatment of NEC. EGF is found in the levels of IL-10 in the serum of NEC patients relative to
amniotic fluid and is produced by the salivary glands, healthy controls.76 This increase could be explained by a
Brunner’s glands in the duodenum, Paneth cells, and mac- lack of IL-10 in the maternal milk, requiring increased IL-10
rophages. EGF is important for cell survival (reduced auto- production by the newborn in an attempt of the young im-
phagy), division, and migration in the intestine.70,71 Human mune system to control the disease; however, IL-10 pro-
studies have revealed that decreased cord blood EGF levels duction by the infant may be too delayed to effectively
are predictive of NEC in veryelow-birth-weight infants and control the immune response. The G/H model has been used
that heparin-binding EGF (HB-EGF) treatment in rat pups for investigating the role of IL-10 in the development of
subjected to the G/H NEC model protects the pups from NEC. IL-10/ mice develop more severe histological as-
injury (Table 2).72,87 In addition, in the G/H model, the pects, characterized by increased epithelial cell apoptosis
administration of mesenchymal stem cells along with HB- and decreased organization of tight junction proteins.74
EGF results in a synergism that reduces injury and im- Additionally, the administration of IL-10 ameliorates dis-
proves survival.88 These studies demonstrate that impaired ease, and ileal IL-10 levels are increased in rats fed with rat
epithelial healing secondary to reduced EGF/HB-EGF may milk versus a cow-milk substitute.77,78 The PAF/LPS mouse
be a major component in the pathogenesis of NEC. model shows that the probiotic Lactobacillus rhamnosus
GG protects mice from NEC by increasing IL-10 receptor
External Modulators transcription, thereby increasing the signaling abilities of
both maternal milkederived and endogenous IL-10.
Full-term infants acquire several immunomodulatory com- Increased IL-10 signaling in these studies resulted in
ponents from the breast milk of their mothers, including lower levels of colonic production of tumor necrosis factor
PAF acetylhydrolase; growth factors such as EGF, HB- a and the neutrophil chemoattractant macrophage inflam-
EGF, insulin-like growth factor (IGF), and TGF-b; and matory protein 2, indicating that IL-10 signaling in the
immune factors such as SIgA and IL-10 (Table 2).89 The context of NEC leads to decreased inflammatory cytokine
immaturity of a preterm infant’s immune system may make production, and the modulation of IL-10 may be possible
the presence of these molecules even more vital for pre- through the administration of probiotics.79
venting intestinal damage during the first weeks of life. A low IGF level has been reported to be associated with
Due to the nearly absent levels of IgA in newborn infants NEC.81 IGF-1 can protect intestinal epithelial cells from
and the importance of IgA in regulating the commensal oxidative stresseinduced apoptosis and promote the devel-
microbiota, it has been suggested that a lack of IgA may be opment and cytotoxic activity of natural killer cells, whereas
involved in NEC, particularly in formula-fed infants.17 To IGF-2 induces TGF-b release.80,91,92 NEC is associated with
address this issue, several human studies have attempted to decreased tissue levels of TGF-b, and in mice, the disrup-
supplement IgA levels, as well as other immunoglobulins, in tion of TGF-b signaling results in more severe NEC-like
newborns; however, there has not been a striking mucosal injury in the PAF/LPS model.40 As TGF-b nor-
improvement in outcomes.90 In interpreting these data, it mally suppresses macrophage inflammatory responses in the
should be noted that in all studies in which formula was developing intestine, its absence (from either a lack of
supplemented with immunoglobulins, serum immunoglob- breastfeeding or decreased IGF levels) can clearly impact
ulins and not SIgA was used. IgA arrives in the intestinal the function of the innate immune system in premature

The American Journal of Pathology - ajp.amjpathol.org 11


Tanner et al

Prematurity
Figure 2 The innate immune response in
Formula Feeding necrotizing enterocolitis (NEC. Top: hematoxylin
and eosin stain of normal neonatal human small
Antibiotics bowel. Middle: Mechanistic diagram of the fac-
tors that have been associated with the increased
Delivery Method incidence of human NEC and the potential innate
immune cells and molecules involved in the dis-
ease. In this diagram, prematurity, antibiotic use,
Dysbiosis method of delivery (vaginal versus Cesarean), and
Proteobacteria formal feeding can result in dysbiosis, altered
Firmicutes barrier function, and altered growth factors and
cytokines. The dashed arrows represent a recip-
rocal vicious cycle between dysbiosis and barrier
function, and dysbiosis and altered growth fac-
Barrier Dysfunction Altered Growth tors and cytokines. All of these factors can lead to
TLR4 Factors and Cytokines increased bacterial translocation, decreased
Permeability Acetylhydrolase enterocyte migration and to increased inflamma-
Epithelial damage EGF/HB-EGF tion and necrosis, as evidenced in the hematox-
Epithelial healing IL-10 ylin and eosin panel (bottom), showing severe
Mucus layer SIgA
Altered AMP function muscular and epithelial damage with complete
IGF
TGF-β loss of mucosa. EGF, epidermal growth factor; HB-
EGF, heparin bound-epidermal growth factor; IGF,
insulin-like growth factor; SIgA, secretory immu-
Bacterial Translocation noglobulin A; TLR4, Toll-like receptor 4; and TGF,
Enterocyte Migration transforming growth factor.
Inflammation/Necrosis

infants. Of note, recent data indicate that low blood TGF-b these murine models, reports show that numerous macro-
levels are associated with a higher risk for NEC.26 phages are seen in NEC lesions and that macrophages from
the human preterm intestine produce inflammatory cyto-
Innate Immune Cells kines, whereas macrophages from full-term neonates and
adults do not.40,52 These macrophages may play a role in
Both of the newly described chemical and infection models disease, as they can be suppressed by the regulatory cyto-
of NEC have suggested that there may be a defective kine TGF-b2, which is decreased in both patients with NEC
response by innate immune cells, leading to disease and in preterm milk.40,82 In addition, the trinitrobenzene
(Table 2).49,50 The data suggest a destructive role for DCs in sulfonic acideinduced NEC model is driven by pro-
coordinating a response to an NEC-like disease and a pro- inflammatory macrophages. These differences between the
tective role for neutrophils and macrophages. Further infection and chemical models, as well as human disease,
investigation of the phenotypes of macrophages will reveal show the difficulty in modeling a disease with an unknown
whether they are predominantly of the IL-10esecreting M2 etiology and that each model must be fully investigated to
phenotype, and study of the subsets of DCs will determine determine its strengths and weaknesses.
whether there may be defects in DC subtypes that are known More studies of other innate immune cells are still needed
to control T-cell homeostasis.93,94 However, in contrast to in both human disease and animal models. One recent

12 ajp.amjpathol.org - The American Journal of Pathology


Pathogenesis of Necrotizing Enterocolitis

publication looking at plasmacytoid DCs reported a pre- depends on a complex combination of intestinal immaturity,
mature morphology and decreased functional capacity, inflammation, injury repair, and microbial communities.102
whereas another study reported diminished levels of natural Several mathematical models of NEC have been devel-
killer cell activity in the peripheral blood of preterm in- oped, including an effort to combine independent areas of
fants.83,95 To our knowledge, there are no reports regarding NEC research, including the inflammatory response in NEC,
the presence or function of B1 B cells (which produce the dynamics of epithelial healing, the protective factors
natural serum antibody and T-independent antibodies) or the supplied by breastfeeding, and the role of pathogenic bacte-
newly described innate lymphoid cells in the term versus the ria.103 However, these models have yet to translate effec-
premature neonate. Because these innate cells are present in tively into clinical practice and must be further optimized.
neonatal murine models and are involved in intestinal ho-
meostasis, they may also play an important role in NEC and
should be studied further.96
Future Perspectives
Our understanding of NEC has increased slowly during
Future NEC Models recent years. Despite this increase in knowledge, NEC
diagnosis and treatment remain challenging, indicating that
Humanized Mouse Models
the scientific and medical communities are far from opti-
One potential avenue that has not been explored in NEC mally understanding NEC pathogenesis. The animal models
research to date is the utilization of humanized mouse described herein have proven to be invaluable resources in
models. Due to species-specific differences in protein achieving our current level of understanding; however, the
structure and expression and differences in various ligand difficulty in replicating all aspects of the human disease, the
specificities between species,97 humanized mouse models lack of spontaneous disease development, and the lack of
may provide a bridge to further enhance NEC research. data on the involvement of the innate immune system are
Humanized TLR4 mice have recently been developed98 and drawbacks common to all models (Table 1). While a shift in
may prove to be a useful model in future NEC studies. In microbiota, breakdown in the intestinal barrier, altered
addition, the recently developed ability to colonize germ- sensing of and response to bacterial components, and dis-
free mice with human fecal microbiota is a new opportu- rupted immune factors and cells may all be involved in
nity for developing models of the disease that are based on NEC, it is difficult to say which, if any, is the primary insult
the fecal communities of NEC and control patients, and leading to disease (Figure 2). It is clear that an NEC model
these models may result in a dramatic new understanding of that more accurately replicates human disease will be
the disease while avoiding the current confounding factors required for effectively studying the disease and for
of the differing microbial communities of each institution’s improving patient outcomes.
animal facility.99
References
Novel Transgenic Technologies
1. Neu J, Walker WA: Necrotizing enterocolitis. N Engl J Med 2011,
One of the main reasons scientists that gravitate toward the 364:255e264
mouse as a model organism is the proficiency of generating 2. Ballance WA, Dahms BB, Shenker N, Kliegman RM: Pathology of
neonatal necrotizing enterocolitis: a ten-year experience. J Pediatr
genetically modified animals. The availability of this useful 1990, 117:S6eS13
tool often overrides legitimate concerns regarding devel- 3. Lu P, Sodhi CP, Jia H, Shaffiey S, Good M, Branca MF, Hackam DJ:
opmental, anatomical, and physiological differences Animal models of gastrointestinal and liver diseases. Animal models
between mice and humans. The availability of mice with of necrotizing enterocolitis: pathophysiology, translational relevance,
cell- or tissue-specific knockouts of receptors for IgA, and challenges. Am J Physiol Gastrointest Liver Physiol 2014, 306:
G917eG928
TGF-b, IGF, and IL-10 make the urgency for an appropriate 4. Dowling DJ, Levy O: Ontogeny of early life immunity. Trends
mouse model of NEC crucial for testing the importance of Immunol 2014, 35:299e310
these molecules in the disease. However, the increasing 5. Jilling T, Simon D, Lu J, Meng FJ, Li D, Schy R, Thomson RB,
availability of genetically modified pigs, as well as newly Soliman A, Arditi M, Caplan MS: The roles of bacteria and TLR4 in
described enterocolitis models in zebrafish, may enhance rat and murine models of necrotizing enterocolitis. J Immunol 2006,
177:3273e3282
interest in using these novel models for further investigation 6. Leaphart CL, Cavallo J, Gribar SC, Cetin S, Li J, Branca MF,
of NEC.100,101 Dubowski TD, Sodhi CP, Hackam DJ: A critical role for TLR4 in the
pathogenesis of necrotizing enterocolitis by modulating intestinal
injury and repair. J Immunol 2007, 179:4808e4820
Mathematical Modeling of NEC
7. Udall JN, Pang K, Fritze L, Kleinman R, Walker WA: Development
of gastrointestinal mucosal barrier. I. The effect of age on intestinal
Mathematical modeling is developing as a method useful for permeability to macromolecules. Pediatr Res 1981, 15:241e244
uncovering the intricacies of human diseases. A mathematic 8. Kayama H, Nishimura J, Takeda K: Regulation of intestinal ho-
model of NEC is appropriate because the severity of NEC meostasis by innate immune cells. Immune Netw 2013, 13:227e234

The American Journal of Pathology - ajp.amjpathol.org 13


Tanner et al

9. Strunk T, Currie A, Richmond P, Simmer K, Burgner D: Innate 31. Caplan MS, Hedlund E, Adler L, Hsueh W: Role of asphyxia and
immunity in human newborn infants: prematurity means more than feeding in a neonatal rat model of necrotizing enterocolitis. Pediatr
immaturity. J Matern Fetal Neonatal Med 2011, 24:25e31 Pathol 1994, 14:1017e1028
10. McCracken VJ, Lorenz RG: The gastrointestinal ecosystem: a pre- 32. Sangild PT, Thymann T, Schmidt M, Stoll B, Burrin DG,
carious alliance among epithelium, immunity and microbiota. Cell Buddington RK: Invited Review: the preterm pig as a model in pe-
Microbiol 2001, 3:1e11 diatric gastroenterology. J Anim Sci 2013, 91:4713e4729
11. Utech M, Brüwer M, Nusrat A: Tight junctions and cell-cell in- 33. Sangild PT, Siggers RH, Schmidt M, Elnif J, Bjornvad CR, Thymann T,
teractions. Methods Mol Biol 2006, 341:185e195 Grondahl ML, Hansen AK, Jensen SK, Boye M, Moelbak L,
12. Kim YS, Ho SB: Intestinal goblet cells and mucins in health and Buddington RK, Weström BR, Holst JJ, Burrin DG: Diet- and
disease: recent insights and progress. Curr Gastroenterol Rep 2010, colonization-dependent intestinal dysfunction predisposes to necrotizing
12:319e330 enterocolitis in preterm pigs. Gastroenterology 2006, 130:1776e1792
13. Buisine MP, Devisme L, Savidge TC, Gespach C, Gosselin B, 34. Musemeche CA, Baker JL, Feddersen RM: A model of intestinal
Porchet N, Aubert JP: Mucin gene expression in human embryonic ischemia in the neonatal rat utilizing superior mesenteric artery oc-
and fetal intestine. Gut 1998, 43:519e524 clusion and intraluminal platelet-activating factor. J Surg Res 1995,
14. Chu SH, Walker WA: Developmental changes in the activities of 58:724e727
sialyl- and fucosyltransferases in rat small intestine. Biochim Biophys 35. Mallick IH, Yang W, Winslet MC, Seifalian AM: Ischemia-reperfu-
Acta 1986, 883:496e500 sion injury of the intestine and protective strategies against injury.
15. Dai D, Nanthkumar NN, Newburg DS, Walker WA: Role of oligo- Dig Dis Sci 2004, 49:1359e1377
saccharides and glycoconjugates in intestinal host defense. J Pediatr 36. Clark DA, Fornabaio DM, McNeill H, Mullane KM, Caravella SJ,
Gastroenterol Nutr 2000, 30(Suppl 2):S23eS33 Miller MJ: Contribution of oxygen-derived free radicals to experi-
16. Bergström A, Kristensen MB, Bahl MI, Metzdorff SB, Fink LN, mental necrotizing enterocolitis. Am J Pathol 1988, 130:537e542
Frøkiaer H, Licht TR: Nature of bacterial colonization influences 37. Dimmitt RA, Staley EM, Chuang G, Tanner SM, Soltau TD,
transcription of mucin genes in mice during the first week of life. Lorenz RG: Role of postnatal acquisition of the intestinal microbiome
BMC Res Notes 2012, 5:402 in the early development of immune function. J Pediatr Gastroenterol
17. McElroy SJ, Weitkamp JH: Innate Immunity in the Small Intestine of Nutr 2010, 51:262e273
the Preterm Infant. Neoreviews 2011, 12:e517ee526 38. Sun X, Caplan MS, Liu Y, Hsueh W: Endotoxin-resistant mice are
18. Gallo RL, Hooper LV: Epithelial antimicrobial defence of the skin protected from PAF-induced bowel injury and death. Role of TNF,
and intestine. Nat Rev Immunol 2012, 12:503e516 complement activation, and endogenous PAF production. Dig Dis Sci
19. Goldman AS: Modulation of the gastrointestinal tract of infants by 1995, 40:495e502
human milk. Interfaces and interactions. An evolutionary perspective. 39. Gonzalez-Crussi F, Hsueh W: Experimental model of ischemic bowel
J Nutr 2000, 130(Suppl 2S):426Se431S necrosis. The role of platelet-activating factor and endotoxin. Am J
20. Peitersen B, Bohn L, Andersen H: Quantitative determination of Pathol 1983, 112:127e135
immunoglobulins, lysozyme, and certain electrolytes in breast milk 40. Maheshwari A, Kelly DR, Nicola T, Ambalavanan N, Jain SK,
during the entire period of lactation, during a 24-hour period, and in Murphy-Ullrich J, Athar M, Shimamura M, Bhandari V,
milk from the individual mammary gland. Acta Paediatr Scand 1975, Aprahamian C, Dimmitt RA, Serra R, Ohls RK: TGF-b2 suppresses
64:709e717 macrophage cytokine production and mucosal inflammatory re-
21. Burgio GR, Lanzavecchia A, Plebani A, Jayakar S, Ugazio AG: sponses in the developing intestine. Gastroenterology 2011, 140:
Ontogeny of secretory immunity: levels of secretory IgA and natural 242e253
antibodies in saliva. Pediatr Res 1980, 14:1111e1114 41. Sawada M, Takahashi K, Sawada S, Midorikawa O: Selective killing
22. Sharma R, Tepas JJ 3rd: Microecology, intestinal epithelial barrier of Paneth cells by intravenous administration of dithizone in rats. Int J
and necrotizing enterocolitis. Pediatr Surg Int 2010, 26:11e21 Exp Pathol 1991, 72:407e421
23. Torrazza RM, Ukhanova M, Wang X, Sharma R, Hudak ML, Neu J, 42. Sherman MP, Bennett SH, Hwang FF, Sherman J, Bevins CL: Paneth
Mai V: Intestinal microbial ecology and environmental factors cells and antibacterial host defense in neonatal small intestine. Infect
affecting necrotizing enterocolitis. PLoS One 2013, 8:e83304 Immun 2005, 73:6143e6146
24. Torrazza RM, Neu J: The altered gut microbiome and necrotizing 43. Zhang C, Sherman MP, Prince LS, Bader D, Weitkamp JH,
enterocolitis. Clin Perinatol 2013, 40:93e108 Slaughter JC, McElroy SJ: Paneth cell ablation in the presence of
25. Fituch CC, Palkowetz KH, Goldman AS, Schanler RJ: Concentrations Klebsiella pneumoniae induces necrotizing enterocolitis (NEC)-like
of IL-10 in preterm human milk and in milk from mothers of infants injury in the small intestine of immature mice. Dis Model Mech 2012,
with necrotizing enterocolitis. Acta Paediatr 2004, 93:1496e1500 5:522e532
26. Maheshwari A, Schelonka RL, Dimmitt RA, Carlo WA, Munoz- 44. McElroy SJ, Underwood MA, Sherman MP: Paneth cells and
Hernandez B, Das A, McDonald SA, Thorsen P, Skogstrand K, necrotizing enterocolitis: a novel hypothesis for disease pathogenesis.
Hougaard DM, Higgins RD; Eunice Kennedy Shriver National Neonatology 2013, 103:10e20
Institute of Child Health and Human Development Neonatal Research 45. Adolph TE, Tomczak MF, Niederreiter L, Ko HJ, Böck J, Martinez-
Network: Cytokines associated with necrotizing enterocolitis in Naves E, Glickman JN, Tschurtschenthaler M, Hartwig J, Hosomi S,
extremely-low-birth-weight infants. Pediatr Res 2014, 76:100e108 Flak MB, Cusick JL, Kohno K, Iwawaki T, Billmann-Born S,
27. van der Worp HB, Howells DW, Sena ES, Porritt MJ, Rewell S, Raine T, Bharti R, Lucius R, Kweon MN, Marciniak SJ, Choi A,
O’Collins V, Macleod MR: Can animal models of disease reliably Hagen SJ, Schreiber S, Rosenstiel P, Kaser A, Blumberg RS: Paneth
inform human studies? PLoS Med 2010, 7:e1000245 cells as a site of origin for intestinal inflammation. Nature 2013, 503:
28. Sodhi C, Richardson W, Gribar S, Hackam DJ: The development of 272e276
animal models for the study of necrotizing enterocolitis. Dis Model 46. Nanthakumar NN, Young C, Ko JS, Meng D, Chen J, Buie T,
Mech 2008, 1:94e98 Walker WA: Glucocorticoid responsiveness in developing human
29. Tian R, Liu SX, Williams C, Soltau TD, Dimmitt R, Zheng X, De intestine: possible role in prevention of necrotizing enterocolitis. Am
Plaen IG: Characterization of a necrotizing enterocolitis model in J Physiol Gastrointest Liver Physiol 2005, 288:G85eG92
newborn mice. Int J Clin Exp Med 2010, 3:293e302 47. Nanthakumar N, Meng D, Goldstein AM, Zhu W, Lu L, Uauy R,
30. Jilling T, Lu J, Jackson M, Caplan MS: Intestinal epithelial apoptosis Llanos A, Claud EC, Walker WA: The mechanism of excessive in-
initiates gross bowel necrosis in an experimental rat model of testinal inflammation in necrotizing enterocolitis: an immature innate
neonatal necrotizing enterocolitis. Pediatr Res 2004, 55:622e629 immune response. PLoS One 2011, 6:e17776

14 ajp.amjpathol.org - The American Journal of Pathology


Pathogenesis of Necrotizing Enterocolitis

48. Ganguli K, Meng D, Rautava S, Lu L, Walker WA, Nanthakumar N: 63. Afrazi A, Branca MF, Sodhi CP, Good M, Yamaguchi Y, Egan CE,
Probiotics prevent necrotizing enterocolitis by modulating enterocyte Lu P, Jia H, Shaffiey S, Lin J, Ma C, Vincent G, Prindle T Jr,
genes that regulate innate immune-mediated inflammation. Am J Weyandt S, Neal MD, Ozolek JA, Wiersch J, Tschurtschenthaler M,
Physiol Gastrointest Liver Physiol 2013, 304:G132eG141 Shiota C, Gittes GK, Billiar TR, Mollen K, Kaser A, Blumberg R,
49. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV: Recruit- Hackam DJ: Toll-like receptor 4-mediated endoplasmic reticulum
ment of dendritic cells is responsible for intestinal epithelial damage stress in intestinal crypts induces necrotizing enterocolitis. J Biol
in the pathogenesis of necrotizing enterocolitis by Cronobacter Chem 2014, 289:9584e9599
sakazakii. J Immunol 2011, 186:7067e7079 64. Lu J, Jilling T, Li D, Caplan MS: Polyunsaturated fatty acid sup-
50. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV: Role of plementation alters proinflammatory gene expression and reduces the
neutrophils and macrophages in the pathogenesis of necrotizing incidence of necrotizing enterocolitis in a neonatal rat model. Pediatr
enterocolitis caused by Cronobacter sakazakii. J Surg Res 2012, 172: Res 2007, 61:427e432
18e28 65. Good M, Siggers RH, Sodhi CP, Afrazi A, Alkhudari F, Egan CE,
51. Agostoni C, Axelsson I, Goulet O, Koletzko B, Michaelsen KF, Neal MD, Yazji I, Jia H, Lin J, Branca MF, Ma C, Prindle T, Grant Z,
Puntis JW, Rigo J, Shamir R, Szajewska H, Turck D, Vandenplas Y, Shah S, Slagle D 2nd, Paredes J, Ozolek J, Gittes GK, Hackam DJ:
Weaver LT; ESPGHAN Committee on Nutrition: Preparation and Amniotic fluid inhibits Toll-like receptor 4 signaling in the fetal and
handling of powdered infant formula: a commentary by the neonatal intestinal epithelium. Proc Natl Acad Sci U S A 2012, 109:
ESPGHAN Committee on Nutrition. J Pediatr Gastroenterol Nutr 11330e11335
2004, 39:320e322 66. Gribar SC, Sodhi CP, Richardson WM, Anand RJ, Gittes GK,
52. MohanKumar K, Kaza N, Jagadeeswaran R, Garzon SA, Bansal A, Branca MF, Jakub A, Shi XH, Shah S, Ozolek JA, Hackam DJ:
Kurundkar AR, Namachivayam K, Remon JI, Bandepalli CR, Reciprocal expression and signaling of TLR4 and TLR9 in the
Feng X, Weitkamp JH, Maheshwari A: Gut mucosal injury in neo- pathogenesis and treatment of necrotizing enterocolitis. J Immunol
nates is marked by macrophage infiltration in contrast to pleomorphic 2009, 182:636e646
infiltrates in adult: evidence from an animal model. Am J Physiol 67. Richardson WM, Sodhi CP, Russo A, Siggers RH, Afrazi A,
Gastrointest Liver Physiol 2012, 303:G93eG102 Gribar SC, Neal MD, Dai S, Prindle T Jr, Branca M, Ma C, Ozolek J,
53. Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, Hackam DJ: Nucleotide-binding oligomerization domain-2 inhibits
Antonopoulos DA, Chang EB, Claud EC: 16S rRNA gene-based toll-like receptor-4 signaling in the intestinal epithelium. Gastroen-
analysis of fecal microbiota from preterm infants with and without terology 2010, 139:904e917. 17 e1-6
necrotizing enterocolitis. ISME J 2009, 3:944e954 68. Soliman A, Michelsen KS, Karahashi H, Lu J, Meng FJ, Qu X,
54. Mai V, Young CM, Ukhanova M, Wang X, Sun Y, Casella G, Crother TR, Rabizadeh S, Chen S, Caplan MS, Arditi M, Jilling T:
Theriaque D, Li N, Sharma R, Hudak M, Neu J: Fecal microbiota in Platelet-activating factor induces TLR4 expression in intestinal
premature infants prior to necrotizing enterocolitis. PLoS One 2011, epithelial cells: implication for the pathogenesis of necrotizing
6:e20647 enterocolitis. PLoS One 2010, 5:e15044
55. Bergmann KR, Liu SX, Tian R, Kushnir A, Turner JR, Li HL, 69. Clark JA, Lane RH, Maclennan NK, Holubec H, Dvorakova K,
Chou PM, Weber CR, De Plaen IG: Bifidobacteria stabilize claudins Halpern MD, Williams CS, Payne CM, Dvorak B: Epidermal growth
at tight junctions and prevent intestinal barrier dysfunction in mouse factor reduces intestinal apoptosis in an experimental model of
necrotizing enterocolitis. Am J Pathol 2013, 182:1595e1606 necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol
56. Vieten D, Corfield A, Carroll D, Ramani P, Spicer R: Impaired 2005, 288:G755eG762
mucosal regeneration in neonatal necrotising enterocolitis. Pediatr 70. Maynard AA, Dvorak K, Khailova L, Dobrenen H, Arganbright KM,
Surg Int 2005, 21:153e160 Halpern MD, Kurundkar AR, Maheshwari A, Dvorak B: Epidermal
57. Salzman NH, Polin RA, Harris MC, Ruchelli E, Hebra A, Zirin- growth factor reduces autophagy in intestinal epithelium and in the rat
Butler S, Jawad A, Martin Porter E, Bevins CL: Enteric defensin model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver
expression in necrotizing enterocolitis. Pediatr Res 1998, 44:20e26 Physiol 2010, 299:G614eG622
58. Coutinho HB, da Mota HC, Coutinho VB, Robalinho TI, Furtado AF, 71. Coursodon CF, Dvorak B: Epidermal growth factor and necrotizing
Walker E, King G, Mahida YR, Sewell HF, Wakelin D: Absence of enterocolitis. Curr Opin Pediatr 2012, 24:160e164
lysozyme (muramidase) in the intestinal Paneth cells of newborn 72. Chen CL, Yu X, James IO, Zhang HY, Yang J, Radulescu A,
infants with necrotising enterocolitis. J Clin Pathol 1998, 51: Zhou Y, Besner GE: Heparin-binding EGF-like growth factor pro-
512e514 tects intestinal stem cells from injury in a rat model of necrotizing
59. McElroy SJ, Prince LS, Weitkamp JH, Reese J, Slaughter JC, enterocolitis. Lab Invest 2012, 92:331e344
Polk DB: Tumor necrosis factor receptor 1-dependent depletion of 73. Clark JA, Doelle SM, Halpern MD, Saunders TA, Holubec H,
mucus in immature small intestine: a potential role in neonatal Dvorak K, Boitano SA, Dvorak B: Intestinal barrier failure during
necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol experimental necrotizing enterocolitis: protective effect of EGF treat-
2011, 301:G656eG666 ment. Am J Physiol Gastrointest Liver Physiol 2006, 291:G938eG949
60. Sodhi CP, Neal MD, Siggers R, Sho S, Ma C, Branca MF, 74. Oztürk H, Dokucu AI, Ogun C, Büyükbayram H: Protective effects of
Prindle T Jr, Russo AM, Afrazi A, Good M, Brower-Sinning R, recombinant human interleukin-10 on intestines of hypoxia-induced
Firek B, Morowitz MJ, Ozolek JA, Gittes GK, Billiar TR, necrotizing enterocolitis in immature rats. J Pediatr Surg 2002, 37:
Hackam DJ: Intestinal epithelial Toll-like receptor 4 regulates goblet 1330e1333
cell development and is required for necrotizing enterocolitis in mice. 75. Castellote C, Casillas R, Ramírez-Santana C, Pérez-Cano FJ,
Gastroenterology 2012, 143:708e718.e1-5 Castell M, Moretones MG, López-Sabater MC, Franch A: Premature
61. Liu Y, Zhu L, Fatheree NY, Liu X, Pacheco SE, Tatevian N, delivery influences the immunological composition of colostrum and
Rhoads JM: Changes in intestinal Toll-like receptors and cytokines transitional and mature human milk. J Nutr 2011, 141:1181e1187
precede histological injury in a rat model of necrotizing enterocolitis. 76. Benkoe T, Baumann S, Weninger M, Pones M, Reck C, Rebhandl W,
Am J Physiol Gastrointest Liver Physiol 2009, 297:G442eG450 Oehler R: Comprehensive evaluation of 11 cytokines in premature
62. Sodhi CP, Shi XH, Richardson WM, Grant ZS, Shapiro RA, infants with surgical necrotizing enterocolitis. PLoS One 2013, 8:
Prindle T Jr, Branca M, Russo A, Gribar SC, Ma C, Hackam DJ: Toll- e58720
like receptor-4 inhibits enterocyte proliferation via impaired beta- 77. Emami CN, Chokshi N, Wang J, Hunter C, Guner Y, Goth K,
catenin signaling in necrotizing enterocolitis. Gastroenterology Wang L, Grishin A, Ford HR: Role of interleukin-10 in the patho-
2010, 138:185e196 genesis of necrotizing enterocolitis. Am J Surg 2012, 203:428e435

The American Journal of Pathology - ajp.amjpathol.org 15


Tanner et al

78. Dvorak B, Halpern MD, Holubec H, Dvorakova K, Dominguez JA, 90. Foster J, Cole M: Oral immunoglobulin for preventing necrotizing
Williams CS, Meza YG, Kozakova H, McCuskey RS: Maternal milk enterocolitis in preterm and low birth-weight neonates. Cochrane
reduces severity of necrotizing enterocolitis and increases intestinal Database Syst Rev 2004:CD001816
IL-10 in a neonatal rat model. Pediatr Res 2003, 53:426e433 91. Ni F, Sun R, Fu B, Wang F, Guo C, Tian Z, Wei H: IGF-1 promotes
79. Mirpuri J, Sotnikov I, Myers L, Denning TL, Yarovinsky F, the development and cytotoxic activity of human NK cells. Nat
Parkos CA, Denning PW, Louis NA: Lactobacillus rhamnosus (LGG) Commun 2013, 4:1479
regulates IL-10 signaling in the developing murine colon through 92. Yang G, Geng XR, Song JP, Wu Y, Yan H, Zhan Z, Yang L, He W,
upregulation of the IL-10R2 receptor subunit. PLoS One 2012, 7: Liu ZQ, Qiu S, Liu Z, Yang PC: Insulin-like growth factor 2 enhances
e51955 regulatory T-cell functions and suppresses food allergy in an exper-
80. Baregamian N, Song J, Jeschke MG, Evers BM, Chung DH: IGF-1 imental model. J Allergy Clin Immunol 2014, 133:1702e1708
protects intestinal epithelial cells from oxidative stress-induced 93. Liu YC, Zou XB, Chai YF, Yao YM: Macrophage polarization in
apoptosis. J Surg Res 2006, 136:31e37 inflammatory diseases. Int J Biol Sci 2014, 10:520e529
81. Capoluongo E, Ameglio F, Zuppi C: Insulin-like growth factor-I and 94. Welty NE, Staley C, Ghilardi N, Sadowsky MJ, Igyártó BZ,
complications of prematurity: a focus on bronchopulmonary Kaplan DH: Intestinal lamina propria dendritic cells maintain T cell
dysplasia. Clin Chem Lab Med 2008, 46:1061e1066 homeostasis but do not affect commensalism. J Exp Med 2013, 210:
82. Namachivayam K, Blanco CL, Frost BL, Reeves AA, Jagadeeswaran R, 2011e2024
MohanKumar K, Safarulla A, Mandal P, Garzon SA, Raj JU, 95. Merrill JD, Sigaroudinia M, Kohl S: Characterization of natural killer
Maheshwari A: Preterm human milk contains a large pool of latent TGF- and antibody-dependent cellular cytotoxicity of preterm infants
b, which can be activated by exogenous neuraminidase. Am J Physiol against human immunodeficiency virus-infected cells. Pediatr Res
Gastrointest Liver Physiol 2013, 304:G1055eG1065 1996, 40:498e503
83. Schüller SS, Sadeghi K, Wisgrill L, Dangl A, Diesner SC, Prusa AR, 96. Mortha A, Chudnovskiy A, Hashimoto D, Bogunovic M, Spencer SP,
Klebermasz-Schrehof K, Greber-Platzer S, Neumüller J, Helmer H, Belkaid Y, Merad M: Microbiota-dependent crosstalk between mac-
Husslein P, Pollak A, Spittler A, Förster-Waldl E: Preterm neonates rophages and ILC3 promotes intestinal homeostasis. Science 2014,
display altered plasmacytoid dendritic cell function and morphology. 343:1249288
J Leukoc Biol 2013, 93:781e788 97. Scheer N, Roland Wolf C: Xenobiotic receptor humanized mice and
84. Brook I: Microbiology and management of neonatal necrotizing their utility. Drug Metab Rev 2013, 45:110e121
enterocolitis. Am J Perinatol 2008, 25:111e118 98. Hajjar AM, Ernst RK, Fortuno ES 3rd, Brasfield AS, Yam CS,
85. Yazji I, Sodhi CP, Lee EK, Good M, Egan CE, Afrazi A, Neal MD, Newlon LA, Kollmann TR, Miller SI, Wilson CB: Humanized
Jia H, Lin J, Ma C, Branca MF, Prindle T, Richardson WM, Ozolek J, TLR4/MD-2 mice reveal LPS recognition differentially impacts
Billiar TR, Binion DG, Gladwin MT, Hackam DJ: Endothelial TLR4 susceptibility to Yersinia pestis and Salmonella enterica. PLoS Pathog
activation impairs intestinal microcirculatory perfusion in necrotizing 2012, 8:e1002963
enterocolitis via eNOS-NO-nitrite signaling. Proc Natl Acad Sci U S 99. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI:
A 2013, 110:9451e9456 The effect of diet on the human gut microbiome: a metagenomic
86. Tissières P, Ochoda A, Dunn-Siegrist I, Drifte G, Morales M, analysis in humanized gnotobiotic mice. Sci Transl Med 2009, 1:
Pfister R, Berner M, Pugin J: Innate immune deficiency of extremely 6ra14
premature neonates can be reversed by interferon-g. PLoS One 2012, 100. Fan N, Lai L: Genetically modified pig models for human diseases. J
7:e32863 Genet Genomics 2013, 40:67e73
87. Wahab Mohamed WA, Aseeri AM: Cord blood epidermal growth 101. Yang Y, Tomkovich S, Jobin C: Could a swimming creature inform
factor as a possible predictor of necrotizing enterocolitis in very low us on intestinal diseases? Lessons from zebrafish. Inflamm Bowel Dis
birth weight infants. J Neonatal Perinatal Med 2013, 6:257e262 2014, 20:956e966
88. Yang J, Watkins D, Chen CL, Bhushan B, Zhou Y, Besner GE: 102. Arciero JC, Ermentrout GB, Upperman JS, Vodovotz Y, Rubin JE:
Heparin-binding epidermal growth factor-like growth factor and Using a mathematical model to analyze the role of probiotics and
mesenchymal stem cells act synergistically to prevent experimental inflammation in necrotizing enterocolitis. PLoS One 2010, 5:e10066
necrotizing enterocolitis. J Am Coll Surg 2012, 215:534e545 103. Barber J, Tronzo M, Harold Horvat C, Clermont G, Upperman J,
89. Chatterton DE, Nguyen DN, Bering SB, Sangild PT: Anti-inflam- Vodovotz Y, Yotov I: A three-dimensional mathematical and
matory mechanisms of bioactive milk proteins in the intestine of computational model of necrotizing enterocolitis. J Theor Biol 2013,
newborns. Int J Biochem Cell Biol 2013, 45:1730e1747 322:17e32

16 ajp.amjpathol.org - The American Journal of Pathology


View publication stats

You might also like