Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Feb 1.
Published in final edited form as: Semin Perinatol. 2016 Dec 9;41(1):15–28. doi: 10.1053/j.semperi.2016.09.014

Pathogenesis of NEC: Role of the Innate and Adaptive Immune Response

Timothy L Denning 1, Amina M Bhatia 2, Andrea F Kane 3, Ravi M Patel 4, Patricia L Denning 5
PMCID: PMC5484641  NIHMSID: NIHMS835385  PMID: 27940091

Abstract

Necrotizing enterocolitis (NEC) is a devastating disease in premature infants with high case fatality and significant morbidity among survivors. Immaturity of intestinal host defenses predisposes the premature infant gut to injury. An abnormal bacterial colonization pattern with a deficiency of commensal bacteria may lead to a further breakdown of these host defense mechanisms, predisposing the infant to NEC. Here, we review the role of the innate and adaptive immune system in the pathophysiology of NEC.

Introduction

Necrotizing enterocolitis (NEC) remains a leading cause of morbidity and mortality in the neonatal intensive care unit (NICU)1-4. Because epidemiologic studies demonstrate that NEC incidence is inversely proportional to gestational age at birth5,6, Immature intestinal host defenses are thought to play a major role in its pathogenesis. These key immature defenses include intestinal barrier function, intestinal regulation of microbial colonization, regulation of intestinal circulation, and intestinal innate and adaptive immunity.

Interestingly, NEC onset has also been associated with a developmental window of susceptibility (30-32 weeks postmenstrual age)7,8. Changes in microbial colonization patterns during postnatal development may explain this apparent window of susceptibility. Recent clinical studies implicate the importance of the intestinal microbial community in regulating health and disease in the premature infant. First, increased NEC incidence has been associated with increased early empiric antibiotic use9-12. Second, administration of probiotic bacteria has been associated with decreased risk of NEC13. Finally, longitudinal stool colonization studies using molecular techniques have implicated specific changes in microbial patterns prior to NEC onset14-22.

An imbalance in the maturation of intestinal innate and adaptive immune defense mechanisms may also explain the apparent developmental window of NEC susceptibility. Adaptive immunity is often thought to regulate the innate immune system which can cause disease when allowed to respond unchecked. Neonates, especially preterm infants, are born with underdeveloped adaptive immunity. Adaptive immune defenses transferred from mom (through breast milk and placental transfer of maternal IgG) are meant to protect the newborn infant until their own adaptive immunity develops23. Maternal transfer of these adaptive immune defenses are significantly reduced in preterm infants (especially formula fed infants)24, thus placing them at greater risk for inflammatory disorders such as NEC.

In this review, we will summarize the current evidence regarding the role of the innate and adaptive immune response in the pathophysiology of NEC. Specifically, we will discuss the relative contributions of passive immunity, physical barriers protecting the gastrointestinal (GI) tract, innate immune cells, and cytokines in NEC pathogenesis.

Passive Immunity in NEC

Passive antibody transfer

The two main mechanisms of passive immunity which may act to protect the preterm infant from NEC are passive transfer of maternal antibodies in the form if IgG from the placenta or secretory IgA (sIgA) from breast milk (Table 1). Neonates are known to be born with deficiencies in both cellular and humoral immunity and this passive immunity received from the mother is meant to protect the infant from disease until its own immune system can mature25,26. Placental transfer of IgG is mediated by the FcRN receptor in the syncytiotrophoblast and maternal antibodies have been shown to protect the infant in the first 6 months of life25. Successful placental transfer of IgG is dependent upon maternal IgG levels and gestational age of the infant27. Antibody transfer begins as early as 13 weeks gestation but the greatest amount of antibody transfer occurs in the last 4 weeks of pregnancy. Preterm infants at less than 22 weeks gestation have antibody levels at < 10% maternal levels, which raises to 50% by 28-32 weeks, and continues to raise to 20-30% above maternal levels by term27. In contrast, breast milk from mothers of preterm infants have been found to have higher levels of sIgA compared to term mothers' milk28-30. Based on relative deficiency of IgG and IgA in preterm infants, several clinical trials have evaluated the effect of oral immunoglobulin administration in preterm infants31. However, the results of these trials have found no effect of oral immunoglobulin administration on risk of NEC. Of note, intestinal epithelial expression of the FcRN receptor has been demonstrated in fetuses and may play a role in additional passive immunity in the preterm infant32. FcRN expression and function in humans is reduced compared to rodents which may explain partly why rodents are relatively resistant to NEC-like injury in animal models33,34.

Table 1A. Passive Immunity Protecting the GI Tract in the Preterm Infant.

Time of Maturation Role in NEC
Placental transfer of IgG Starts at 13 weeks
Mature by term
Preterm infants with reduced IgG transfer
Deficiency may predispose to NEC
Breast milk transfer of sIgA Preterm human milk with higher levels Unclear benefit of oral IgA administration in decreasing risk of NEC
B. Additional Breast Milk Components Protecting the GI Tract in the Preterm Infant
Mechanism of Protection Role in NEC
Nutrients:
Oligosaccharides Promote growth of commensal bacteria Oligosaccharide supplementation may reduce NEC risk
Caseins Stimulate increased Paneth cell and goblet cell number and possibly function
May also reduce bacterial adherence to intestinal epithelia
Triglycerides Stimulate increased Paneth cell and goblet cell number and possibly function
May also reduce bacterial adherence to intestinal epithelia

Bioactive proteins:
Lysozyme Antibacterial, synergistic with lactoferrin
Lactoferrin Antibacterial, antifungal, antiviral
Reduces bioavailability of iron to pathogens
Lactoferrin supplementation (+/-probiotics) may reduce NEC risk
PAF-AH Inactivates PAF (key mediator of NEC)
Immunoregulatory cytokines: IL-10 Anti-inflammatory cytokine important for intestinal homeostasis
Genetic defects in IL-10R cause colitis
IL-10 supplementation in animal models is protective
Increased IL-10 in human milk associated with a decreased risk of NEC
TGF-β Involved in regulating inflammation and wound healing Low levels in human milk may predict feeding intolerance in growth restricted infants

Growth factors:
IGF family Promotes IEC proliferation; reduces IEC apoptosis IGF supplementation reduces NEC in animal models
EGF family Promotes IEC proliferation/differentiation
restitution and TJ expression
Reduces IEC autophagy
Increases mucin production
Inhibits TLR-4 signaling
Promotes anti-inflammatory macrophages
Decreased EGF associated with increased NEC risk
EGF supplementation reduces NEC in animal models
EGF supplementation in humans promotes intestinal mucosa trophic effects

Breast milk nutrients

Breast milk contains multiple additional components that help to protect the newborn infant from infectious and inflammatory diseases in the first 6-12 months of life35,36. These include antimicrobial and anti-inflammatory factors and components that also promote maturation of intestinal host defenses37. First, breast milk contains sugars, proteins, and fats that confer dual roles in nutrition and promoting intestinal homeostasis. Oligosaccharides, nondigestible sugars which promote the growth of commensal bacteria in the GI tract38-40. Oligosaccharide supplementation may reduce NEC risk in human41,42 and animal studies43. Caseins in breast milk are highly glycosylated proteins that are also thought to promote intestinal defenses by stimulating increased numbers of goblet and Paneth cells and also by promoting increased MUC2 gene expression (see more detailed discussion of importance of goblet and Paneth cells below)44. One casein subunit, κ-Casein, has also been shown to prevent attachment of bacteria to intestinal mucosal epithelia45. Triglycerides in human milk have also been shown to provide antiviral, antibacterial, and antiprotozoal activity26,46,47.

Breast milk bioactive proteins

Second, breast milk contains bioactive proteins, lactoferrin and lysozyme, with antipathogenic activity. Lysozyme can act synergistically with lactoferrin to kill gram negative bacteria48, but can independently have antibacterial effects as well49. Lactoferrin has been shown to have antibacterial, antifungal, and antiviral activity50-52, can reduce microbial activity by limiting iron availability, and be converted by gastric pepsin to lactoferricin, which disrupts gram-negative cell walls53. A recent meta-analysis reports that lactoferrin supplementation with or without probiotics may reduce the incidence of NEC and late-onset sepsis in preterm infants54. Breastmilk has also been shown to contain platelet activating factor acetylhydrolase (PAF-AH), which is thought to prevent NEC by inactivating the key pathogenic mediator PAF (see below for a more detailed discussion on PAF)55,56.

Breast milk immunoregulatory cytokines

Third, breast milk contains immunoregulatory cytokines, such as IL-10 and TGF-β57,58. Monocytes obtained from preterm infants also seem to have lower ability to produce IL-1059 and TGF-β60, potentially putting them at greater risk for inflammatory diseases. IL-10 is believed to be an anti-inflammatory cytokine critical for intestinal homeostasis61,62. Multiple animal and human studies implicate the importance of IL-10 in protecting the preterm infant from developing NEC. Both IL-10 deficient mice63,64 and human infants with IL-10 receptor genetic defects65 are at increased risk for colitis. Animal models of NEC also show that maternal milk can reduce NEC severity while increasing intestinal IL-1066. Human infants whose mothers have low levels of IL-10 are at increased risk for NEC67 and probiotics may regulate IL-10 signaling in the immature gut68. TGF-β is also thought to regulate the inflammatory response69 and promote wound healing70,71. TGF-β can also initiate local production of IgA in the gut, providing additional protection72. Levels of TGF-β in human milk may predict feeding intolerance in growth restricted infants73. Both IL-10 and TGF-β have been shown to reduce inflammatory signaling by fetal human enterocytes in vitro74.

Breast milk growth factors

Finally, breast milk contains growth factors such as epidermal growth factor (EGF) and insulin-like growth factor (IGF). IGF family members include IGF-1 and IGF-2 and have been thought to help intestinal homeostasis by promoting IEC proliferation and preventing IEC apoptosis36. Low serum IGF-1 levels in preterm infants may correlate with risk for NEC75 and IGF-1 supplementation has been shown to reduce NEC in animal models76,77. Increased IGF-1 receptors have also been detected in NEC tissue78. The EGF family members most studied are EGF and heparin-binding EGF (HB-EGF). Both are thought to be important for intestinal homeostasis and protective against NEC79. EGF is secreted by multiple cells throughout the GI tract. EGF promotes IEC proliferation and differentiation80, IEC restitution after injury81, and reduces IEC autophagy82. EGF may also act by increasing production of helpful mucus by increasing goblet cells and their production of MUC2 (for more detailed discussion on the importance of intestinal mucus layers, see below); by improving intestinal barrier function through increased tight junction protein expression (occludin and claudin)79,83; by reducing TLR-4 signaling84; and by promoting anti-inflammatory macrophages and reducing pro-inflammatory macrophages85 (for more detailed discussion of the role of TLR-4 and macrophages in NEC, see below). EGF is supplied by amniotic fluid throughout pregnancy and by colostrum in human milk. Extremely preterm human milk contains 50-80% more EGF when compared to milk from mother's with full-term infants,86 which may help to protect against NEC, but decreases over time79. Salivary EGF levels increase in preterm infants postnatally87,88 and low EGF levels in cord blood89 and preterm saliva and serum has been associated with increased risk of NEC90. EGF and HB-EGF supplementation in animal NEC models reduces NEC incidence91-103 and EGF supplementation in human neonates has been shown to have trophic effects on intestinal mucosa104.

Physical Barriers Protecting the GI Tract

The physical barriers protecting the GI tract include gastric acid, the mucus layer present throughout the GI tract, the intestinal epithelial barrier, and antimicrobial peptides (Table 2). The intestine is lined by a single layer of highly polarized epithelial cells. Four different types of cells make up the intestinal epithelial layer: hormone-secreting enteroendocrine cells, mucus-secreting goblet cells, enterocytes with absorptive and secretory functions, and antimicrobial-secreting Paneth cells (specialized secretory enterocytes located at the base of small intestinal crypts). Below, we summarize how these cells contribute the physical barriers protecting the GI tract.

Table 2. Physical Barriers Protecting the GI Tract in the Preterm Infant.

Physical Barrier Component Time of Maturation Role in NEC
Gastric Acid Mature secretion by 24 weeks Acid suppression associated with an increase risk of NEC
Mucus layer (Goblet Cells) Term
Premature infants with immature mucus layer
Deficiency may predispose to NEC
NEC causes reduced number & reduced production of mucins and trefoil factor
Epithelial barrier (AJC) Mature structure of AJC at 12 wks gestation (in utero)
Premature infants with increased intestinal permeability
Mature function at term
Immature barrier function may increase NEC risk
Breast milk and probiotics may reduce NEC risk by improving epithelial barrier function
Antimicrobial peptides Paneth cells detectable at 12 wks gestation with secretory capability at 13-20 wks
Premature infants with decreased Paneth cell number and secretory capability
Deficiency of Paneth cell number and function may predispose to NEC
NEC causes upregulated Paneth cell numbers but these cells are dysfunctional

Gastric acid

Gastric acid protects the GI tract by decreasing the number of viable pathogens that can pass into the distal intestine. Enteroendocrine cells and the autonomic nervous system coordinate the secretion of hydrochloric acid by parietal cells located in gastric glands within the epithelial lining. This, in turn, creates the acidic and bactericidal gastric environment. Mature gastric acid secretion seems to be present by 24 weeks gestation105-107. The importance of this acidic gastric environment to host defense is demonstrated by multiple observational studies linking the use of acid suppression by H2 antagonists to both NEC and late-onset sepsis108-111.

Mucus layer

The mucus layer lining the GI tract protects by lubricating and minimizing contact between the epithelium and commensal bacteria. The major proteins of mucus in the intestine are highly glycosylated proteins called mucins, secreted by goblet cells112,113. Goblet cells also produce secretory Immunoglobulin A (sIgA), which contribute to the function of mucus. Mucins secreted by salivary glands coat food and assist with esophageal transit114. The mucus layer in the stomach plays a role in protecting the epithelium from the harsh acidic environment114. MUC2 is the most predominant mucin in both the small and large intestine115. The single unattached layer of mucus in the small intestine works with antibacterial proteins to limit the ability of bacteria to reach the epithelium116. The mucus also moves along the small intestine with peristaltic waves, thus making it even more difficult for bacteria to approach the epithelial layer. Attached to the apical side of enterocytes in the small intestine is a separate, thin layer of mucus made up of transmembrane mucins. This layer is commonly referred to as the glycocalyx and affords protection to the intestinal epithelial cells (IECs) by means of a physical barrier and plays a role in cellular signaling117. The goblet cells in the large intestine contribute to an inner and an outer layer of mucus. The inner mucus layer is inpenetrable to larger entities, such as bacteria. The outer layer is the area where the commensal bacteria of the large intestine reside. In this way, the commensal bacteria can help in digestion of the glycans found on the mucins.118 Mucins also bind and stabilize key trophic and reparative factors (intestinal trefoil factor and epidermal growth factor, EGF) at the epithelial surface, which may aid epithelial repair119,120.

Human infants with NEC have fewer mucin-containing goblet cells121,122. The premature infant's impaired ability to secrete mucus in response to an infection, coupled with a poorly developed mucus system may contribute to the increased risk of NEC123. Reduced number of MUC2 and trefoil factor 3 goblet cells have been found in both human121,122 and rodent83 NEC, and mice with genetically aberrant MUC2 develop more severe disease124. Trefoil factor 3 supplementation may reduce NEC in animal models125.

Growth restriction may also impair intestinal barrier defenses. In rats with intrauterine growth retardation (IUGR), colonic barrier function was impaired126. This is of interest due to the possible association between NEC and IUGR127-130. The decreased function was a product of decreased colonic length, fewer goblet cells per crypt, and disruption of the normal gene expression and amount of mucin throughout the large intestine126. Decreased Paneth cell number has also been reported in murine models of IUGR and human IUGR intestinal tissue131. The combination of these differences in intestinal integrity of premature and IUGR infants compared to term infants may play a role in the possible association with NEC.

Epithelial barrier

The intestinal epithelial barrier is composed of a single layer of highly polarized intestinal epithelial cells (IECs), which creates a physical barrier regulated by the apical junction complex (AJC), consisting of tight junctions (TJ) and adherens junctions132. Tight junctions (TJs) regulate paracellular permeability and maintain separation of tissue compartments by sealing the intercellular space133,134. Three types of proteins make up TJs: occludins, claudins, and junctional adhesion molecules. The AJC starts to form as early as 10 weeks human gestation when intercellular tight junctions can be detected. However, full secretory and absorptive capabilities of the intestinal epithelia continues to occur in utero due to amniotic fluid growth and trophic factors, which induces mucosal maturation from 26 weeks to term135. Ongoing postnatal intestinal epithelial barrier maturation can also be induced by multiple factors including diet136-138, epidermal growth factor80, endogenous glucocorticoids139, and commensal bacteria140,141.

Premature infants have impaired epithelial barrier function compared to term infants142,143, which is thought to contribute to the pathogenesis of NEC2,3,144-147. The role of TJ proteins in the pathogenesis of NEC has been extensively studied in human148,149 and animal83,148,150-152 studies. Many studies have also demonstrated how cytokines induced during intestinal inflammation can further weaken intestinal barrier function153-158, leading to a vicious cycle of increased intestinal inflammation and injury. In addition, two promising biomarkers in early detection of NEC, I-FABP and claudin-3, are measures that indicate gut barrier disruption159.

IECs are also responsible for sampling intraluminal contents which instigates transcellular signaling and transcription of genes resulting in a defense response via the release of cytokines and chemokines and subsequent attraction of leukocytes. This function is mediated by multiple pattern recognition receptors (PRRs) critical for the identification of both foreign elements such as peptidoglycan, lipoproteins, viral DNA and commensal microflora. The remarkable ability of these receptors to distinguish between harmful and helpful bacteria with subsequent appropriate signaling is critical to intestinal homeostasis141. Toll-like receptors (TLRs) are the predominant type of PRR found on the apical side of IECs. Another group of PRRs that cooperate with TLRs are the intracellular Nod-like receptors (NLRs). Nod1 is expressed by IECs, and Nod2 is found in monocytes, dendritic cells, and Paneth cells117. Multiple TLRs (TLR-2, TLR-4) as well as NOD2 have been implicated in the pathogenesis of NEC in human160-164 and animal studies165-175. TLR-2 primarily senses peptidoglycan (a component of gram positive bacteria cell wall); TLR-4 primarily senses lipopolysaccharide (LPS, a component of gram negative bacteria cell wall); and TLR-9 primarily senses bacterial or viral DNA (CpG dinucleotides)176. In particular, exaggerated TLR-4 signaling and LPS are thought to play a major role in the inflammatory signaling in NEC177,178. Of note, platelet activating factor (PAF) is also an important acute mediator in the pathogenesis of NEC, which is not only a chemokine that induces inflammatory signaling but also can increase expression of TLR-4179-182. TLR-9 may play a protective role183-186.

Antimicrobial peptides

Antimicrobial peptides can be secreted into the lumen of the gut by IECs, Paneth cells, and recruited neutrophils. Antimicrobial peptides are thought to promote intestinal homeostasis by regulating the microbial population187. Traditional antimicrobial peptides are directly microbicidal (defensins (α and β), cathelicidins); other peptides regulate microbes by sequestering nutrients (e.g. iron, zinc, manganese) necessary for growth (calprotectin, REG3γ)188. For the purposes of this review, we will limit our discussion to the first group. Initially discovered in human neutrophils, defensins are small cationic peptides that kill microbes in an oxygen-independent manner189. Defensins and cathelicidins function by inserting into the membranes of a broad range of prokaryotic cells, including gram-positive and gram-negative bacteria, fungi, protozoa, spirochetes, and enveloped viruses187,190. Once inside the microbial cell membrane, they form pores allowing the passage of anions through the membrane, thus depolarizing and killing the organism191.

IECs primarily secrete β-defensins (hBD1, 2, and 3) with specific tissue distribution varying along the intestinal axis for each member of the β-defensin family191. Paneth cells secrete lysozyme, phospholipase A2, and antimicrobial peptides (defensins (α and β) and cathelicidins189,192). Paneth cells secrete α-defensins (human defensin, HD5 and HD6) in response to microbial or cholinergic stimuli, contributing to the relatively sterile and protected environment within intestinal crypts.

In vitro studies suggest that antimicrobial peptides may also contribute to host defense indirectly, by inducing host responses193. Cathelicidins and defensins may have proinflammatory properties by activating chemokine release resulting in immune cell chemotaxis and differentiation. α-defensins released into the intestinal crypt may stimulate chloride secretion from nearby enterocytes in order to flush pathogens and toxins away from sensitive stem cells194. β-defensins may promote homeostasis by promoting IEC migration, barrier function, and reducing pro-inflammatory cytokine expression195. Stool hBD2 expression has been reported as high in neonates with NEC196 and may increase in response to changes in microbiota composition associated with NEC so has been proposed as a possible biomarker for early detection197. Intestinal expression of hBD2 is high in resected NEC tissue but low in more severe cases198.

Ontogeny studies have demonstrated that Paneth cells can be detected by 12 weeks gestation and begin to produce antimicrobial defensins at 13 weeks and lysozyme at 20 weeks199,200. Significant HD5 expression can be detected at above 29 weeks201. Premature infants have been shown to have fewer Paneth cells with decreased function191,199,201,202. Multiple animal studies implicate the importance of Paneth cells in NEC pathogenesis203,204. Preterm infants with NEC have been shown to have normal4 to reduced122,205,206 numbers of Paneth cells or poorly functioning Paneth cells202,206, but infants recovering from NEC have been shown to demonstrate Paneth cell hyperplasia207.

Contribution of Innate and Adaptive Immune Cells

In addition to the physical and chemical barriers that limit unrestricted translocation of intestinal microbiota, numerous innate immune cells coordinately regulate responses that contribute to barrier fortification and host defense208,209. In the process of attempting to protect the host from real or perceived threats, however, innate immune cells can elaborate serious bystander effects that are associated with the pathogenesis of NEC including excessive intestinal damage and impaired repair process. Below, we briefly outline the current state of knowledge with regards to complex role of innate and adaptive immune cells and cytokines in regulating NEC.

Intraepithelial lymphocytes

Positioned directly between intestinal epithelial cells in both the small and large intestine are intra-epithelial lymphocytes (IEL). The two main subsets of IEL can be distinguished by expression of either αβ or γδ T cell receptors and can be further categorized into specific subsets using CD4, CD8α, and CD8β co-receptors210. γδ IEL are the pioneer T cells that colonize the intestinal epithelium during embryogenesis and the very early postnatal period when conventional αβ T cell responses are not yet fully established211,212. Given their “front-line” positioning at epithelial surfaces and expression of NK receptors, γδ IEL are poised to contribute to barrier protection and mucosal defense in response to infection and stress213. Thus, at the earliest stages of ontogeny γδ IEL are among the first intestinal-resident immune cells contributing to the maintenance of epithelial integrity.

Given the putative beneficial role for γδ IEL in the intestine early in life, these cells may fundamentally contribute to barrier defense in the preterm infant. Consistent with this hypothesis, γδ IEL were observed to be preferentially reduced in the ileum of surgical NEC patients when compared to non-NEC controls. Additionally, TCR8-deficient mice, which lack γδ IEL altogether, were more susceptible to experimental NEC-like intestinal injury149. These complementary observations from both human and experimental NEC further provided a link between loss of γδ IEL and reduction of IL-17 and RORC, the master transcription factor involved in the differentiation of IL-17 producing T cells (Th17)214. IL-17A was originally viewed as a pro-inflammatory cytokine involved in driving systemic and intestinal inflammation. However, more recent data suggests that IL-17A is involved in maintaining barrier function via regulation of tight junction proteins215-217. In addition to IL-17A, γδ IEL can afford barrier protection and repair of damaged mucosa by secretion of other factors such as epithelial growth factor218. Collectively, during the precarious developmental window in preterm infants when the intestinal epithelial barrier is functionally immature, γδ IEL appear to provide important early immune-mediated barrier protection219.

Natural killer (NK) cells and innate lymphoid cells (ILCs)

The function of natural killer (NK) cells in anti-tumor and anti-viral mediated immunity is well established220. More recently, accumulating evidence suggests a fundamental contribution of NK cells in intestinal barrier protection and regulation of inflammation. Using the DSS model of acute intestinal damage, depletion of NK cells was reported to significantly augment colonic damage, neutrophil infiltration, and proinflammatory cytokine production221. The mechanism of NK cell-mediated protection from acute barrier damage in this study was linked to expression of the NK cell inhibitory receptor NKG2A. NK cells have also been implicated in protection from chronic T cell-dependent intestinal inflammation. In the CD45RBhi model of colitis, loss of NK cells results in dramatically accelerated Th1-driven disease222. Consistent with these data, flow cytometric analyses of immune cell subsets in preterm infants established a link between a decrease in NK cells and the development of NEC223. Thus, NK cells and perhaps NK-like innate lymphoid cells224 may protect from intestinal barrier damage, promote barrier repair and decrease the risk of NEC.

Neutrophils

Despite being one of the most well studied innate immune cell populations, the role of neutrophils in NEC has remained enigmatic. Neutrophils are the most abundant= innate immune cell population among white blood cells and are normally absent from healthy peripheral tissues including the intestine. In response to intestinal damage or danger signals. However, neutrophils rapidly exit the circulation, enter affected tissues, and elaborate numerous “pro-inflammatory” effector functions including phagocytosis, production of reactive oxygen and nitrogen intermediates and ultimately killing of microbes225. While the function of neutrophils is aimed at host protection, localized tissue damage can be an unfortunate complication of neutrophil effector responses. Interestingly, neutrophils isolated from blood of preterm infants have been reported to exhibit defective phagocytic and microbicidal activities as well as impaired chemotaxis and adhesion, which could increase the risk of developing NEC226.

In the preterm infant intestine, which is developmentally immature, neutrophils may provide transient barrier protection in response to threats from potentially pathogenic bacteria or tissue damage/injury. In support of this concept, early-onset neutropenia in small-for-gestational-age infants has been shown to correlate with increased odds for developing NEC227. Additionally, depletion of neutrophils and macrophages in an experimental model of NEC induced by treating newborn mice with the virulent gram-negative pathogen Cronobacter sakazakii resulted in exacerbated disease228. Somewhat paradoxically, neutrophils are increased in intestinal tissue obtained from NEC patients229. However, whether these cells are simply “guilty-by-association” as they attempt to provide critical barrier fortification remains unclear. Consistent with these findings, inducing acute intestinal damage in mice using dextran sodium sulfate (DSS) results in neutrophil accumulation in the colon as damage to the mucosa increases in severity. This accumulation coincides with barrier repair and resolution of damage and depletion of neutrophils impairs this process230. Interestingly, one of the key mediators of neutrophil-dependent barrier repair following DSS-induced acute intestinal damage is the IL-10 family cytokine IL-22. IL-22 is a potent inducer of intestinal epithelial proliferation and mucosal healing and also leads to enhanced production of antimicrobial peptide expression by IECs231-233. Thus, it is tempting to speculate that neutrophil recruitment and neutrophil-dependent IL-22 production may help to protect the premature intestine during development.

Macrophages and dendritic cells

Like neutrophils, macrophages play important roles in host defense at barrier surfaces such as the intestine. An important dichotomy between neutrophils and macrophages, however, is that the latter reside in peripheral tissues in the steady state even very early in life. Interestingly, macrophages are already present in the fetal intestine where they are believed to contribute to maintenance of tissue homeostasis and tolerance234,235. At this stage of development, intestinal macrophages are hypo-responsive to stimulation with lipopolysaccharide (LPS). So-called “endotoxin tolerance” of intestinal macrophages is likely beneficial in establishing and maintaining a mutualistic relationship with the intestinal microbiota234.

With intestine resident macrophages being in a state of hypo-responsiveness, blood monocytes can be rapidly mobilized into the intestine in response to microbial threats and or tissue damage/stress. During the intestinal damage that accompanies NEC, blood monocytes are recruited to the damaged intestine and an acute drop in blood monocyte counts can be observed and may be useful as a biomarker for NEC in VLBW infants236. After entering the damaged intestine, blood monocytes rapidly differentiate into pro-inflammatory M1-type macrophages. These M1 macrophages isolated from human and experimental NEC exhibit high-level expression of Smad7 making them refractory to TGF-β signaling, while promoting NF-κB-mediated signaling and secretion of pro-inflammatory cytokines including IL-1β, IL-6, IL-12, and TNFα237. Macrophage products such as the proinflammatory chemokines IL-6, IL-8, and TNF-α have been found to be greatly elevated in infants with surgical NEC as compared to other preterm intestinal injury238. Activated M1 macrophages can further potentiate intestinal damage during NEC by augmenting intestinal epithelial cell apoptosis85. Thus, inhibiting the differentiation and/or effector functions of M1 macrophages has been considered as an approach to limit the dysregulated inflammatory response in the NEC intestine. A recent report has provided evidence that this may be a feasible approach by showing that heparin-binding epidermal growth factor-like growth factor (HB-EGF) can protect from experimental NEC by preventing M1 and promoting M2 polarization of macrophages85.

In addition to macrophages, dendritic cells (DCs) are another population of antigen-presenting cells that are capable of regulating intestinal immune responses239. Intestinal DCs are positioned in the muscularis mucosae and lamina propria where they can access bacterial antigens and initiate innate and adaptive immune responses240. In the steady-state, intestinal DCs promote the induction of regulatory T cells and tolerance in adults241,242, but the functions in the preterm infant intestine remain unclear. Current evidence from C. sakazakii-induced experimental NEC suggests that DC influx into the intestine during disease contributes to pathological inflammation. Using this model of NEC, the authors found that depletion of DCs in mice protected against C. sakazakii-induced intestinal damage and conversely that adoptive transfer of DCs promoted epithelial barrier disruption and the onset of NEC243. Altogether, these data suggest that controlling the activation and differentiation of intestinal macrophages and dendritic cells may hold potential for NEC therapy.

CD4+ T lymphocytes

Compared to the role of innate immune cells in NEC as described above, there exists relatively little evidence examining the role of lymphocytes in this disease process. Interestingly, a recent study showed that the intestine in mouse and human NEC contains an abundance of CD4+ T cells that are recruited in response to TLR4-mediated induction of the CCR9/CCL25 axis244. CD4+ T cells in the NEC intestine were enriched in Th17 cells, while Foxp3-expressing regulatory T cells (Tregs) were diminished, consistent with previously published data245. Th17 cells in the NEC tissue appeared to contribute to intestinal damage as blocking of IL-17 receptor or STAT3 was capable of ameliorating disease. Additionally, oral delivery of retinoic acid was sufficient to skew the polarization of CD4+ T cells away from the Th17 lineage and towards Tregs, which resulted in diminished NEC severity. These results provide an exciting new avenue for exploring the contribution of effector and regulatory T cells in the pathogenesis of NEC.

Conclusion

While it is clear that immune responses are associated with NEC, the precise role of specific innate and adaptive cells and factors in mediating protection versus contributing to pathogenesis continues to emerge. Rapid innate immune cell recruitment and cytokine production in response to barrier threats is a highly evolutionarily conserved process that is critical for host protection. However, if the threat is not efficiently and effectively neutralized, uncontrolled intestinal damage may ensue. From this standpoint, NEC may be a disease initiated, in part, due to suboptimal innate immune responses in response to dysbiotic microbiota in the preterm intestine. Following initial tissue damage, activated innate and adaptive immune cells may then accumulate in the intestine where they are associated with further tissue damage while attempting to contain invading bacteria.

figure 1. Premature infant gut in the steady state and during NEC.

figure 1

In the steady state, homeostasis is promoted by beneficial bacteria (Bifidobacteria, Lactobacillus) and breast milk components (IgA, HMO, EGF, IL-10, lactoferrin, lysozyme, TGF-β). In the preterm gut, γδ IEL are among the first intestinal-resident immune cells contributing to the maintenance of epithelial integrity via IL-17A and EGF. Natural killer (NK) cells also protect against and repair barrier damage. Neutrophils (PMN) may be important during initial colonization in the neonatal gut, providing transient barrier protection in response to threats from potentially pathogenic bacteria, via IL-22 production. Resident macrophages (Mϕ) and dendritic cells (DCs) maintain tolerance toward the intestinal microbiota via the production of IL-10, which, in combination with transforming growth factor TGF-β, induce regulatory T cells (Treg) cells. During NEC, lack of breast milk protective components and dysbiotic flora (e.g. Gammaproteobacter) may allow barrier breakdown and bacterial translocation. This leads to innate signaling via TLR-4 (in response to PAF and LPS), which in turn causes recruitment of neutrophils and monocytes into the intestine, where they, along with resident DCs drive proinflammatory cytokine production, including IL-1β, tumor necrosis factor (TNF), IL-8, and IL-12, which can promote pathogenic Th1 and Th17 responses.

Acknowledgments

Supported in part by National Institutes of Health Award R01 DK097256 (TLD), K23 HL128942 (RMP), Children's Healthcare of Atlanta Friends Grant (AFB, PLD), and Emory and Children's Center for Infections and Vaccines Pilot Grant (PLD).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting galley proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contributor Information

Timothy L. Denning, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA.

Amina M. Bhatia, Emory University School of Medicine, Atlanta, GA.

Andrea F. Kane, Neonatal Fellow, Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA

Ravi M. Patel, Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA.

Patricia L. Denning, Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta.

References

  • 1.Heron M. Deaths: Leading Causes for 2013. Natl Vital Stat Rep. 2016;65:1–95. [PubMed] [Google Scholar]
  • 2.Lin PW, Stoll BJ. Necrotising enterocolitis. Lancet. 2006;368:1271–83. doi: 10.1016/S0140-6736(06)69525-1. [DOI] [PubMed] [Google Scholar]
  • 3.Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med. 2011;364:255–64. doi: 10.1056/NEJMra1005408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Patel RM, Kandefer S, Walsh MC, et al. Causes and timing of death in extremely premature infants from 2000 through 2011. N Engl J Med. 2015;372:331–40. doi: 10.1056/NEJMoa1403489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stoll BJ, Kanto WP, Jr, Glass RI, Nahmias AJ, Brann AW., Jr Epidemiology of necrotizing enterocolitis: a case control study. J Pediatr. 1980;96:447–51. doi: 10.1016/s0022-3476(80)80696-2. [DOI] [PubMed] [Google Scholar]
  • 6.Guthrie SO, Gordon PV, Thomas V, Thorp JA, Peabody J, Clark RH. Necrotizing enterocolitis among neonates in the United States. J Perinatol. 2003;23:278–85. doi: 10.1038/sj.jp.7210892. [DOI] [PubMed] [Google Scholar]
  • 7.Neu J. Neonatal necrotizing enterocolitis: an update. Acta Paediatr Suppl. 2005;94:100–5. doi: 10.1111/j.1651-2227.2005.tb02163.x. [DOI] [PubMed] [Google Scholar]
  • 8.Llanos AR, Moss ME, Pinzon MC, Dye T, Sinkin RA, Kendig JW. Epidemiology of neonatal necrotising enterocolitis: a population-based study. Paediatr Perinat Epidemiol. 2002;16:342–9. doi: 10.1046/j.1365-3016.2002.00445.x. [DOI] [PubMed] [Google Scholar]
  • 9.Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics. 2009;123:58–66. doi: 10.1542/peds.2007-3423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kenyon S, Boulvain M, Neilson JP. Antibiotics for preterm rupture of membranes. Cochrane Database Syst Rev. 2010:CD001058. doi: 10.1002/14651858.CD001058.pub2. [DOI] [PubMed] [Google Scholar]
  • 11.Kuppala VS, Meinzen-Derr J, Morrow AL, Schibler KR. Prolonged initial empirical antibiotic treatment is associated with adverse outcomes in premature infants. J Pediatr. 2011;159:720–5. doi: 10.1016/j.jpeds.2011.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Alexander VN, Northrup V, Bizzarro MJ. Antibiotic exposure in the newborn intensive care unit and the risk of necrotizing enterocolitis. J Pediatr. 2011;159:392–7. doi: 10.1016/j.jpeds.2011.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.AlFaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Evidence-based child health : a Cochrane review journal. 2014;9:584–671. doi: 10.1002/ebch.1976. [DOI] [PubMed] [Google Scholar]
  • 14.Warner BB, Deych E, Zhou Y, et al. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight infants: a prospective case-control study. Lancet. 2016 doi: 10.1016/S0140-6736(16)00081-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.de la Cochetiere MF, Piloquet H, des Robert C, Darmaun D, Galmiche JP, Roze JC. Early intestinal bacterial colonization and necrotizing enterocolitis in premature infants: the putative role of Clostridium. Pediatr Res. 2004;56:366–70. doi: 10.1203/01.PDR.0000134251.45878.D5. [DOI] [PubMed] [Google Scholar]
  • 16.Wang Y, Hoenig JD, Malin KJ, et al. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. ISME J. 2009;3:944–54. doi: 10.1038/ismej.2009.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Morrow AL, Lagomarcino AJ, Schibler KR, et al. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome. 2013;1:13. doi: 10.1186/2049-2618-1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mai V, Young CM, Ukhanova M, et al. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PLoS One. 2011;6:e20647. doi: 10.1371/journal.pone.0020647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Greenwood C, Morrow AL, Lagomarcino AJ, et al. Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J Pediatr. 2014;165:23–9. doi: 10.1016/j.jpeds.2014.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Torrazza RM, Ukhanova M, Wang X, et al. Intestinal microbial ecology and environmental factors affecting necrotizing enterocolitis. PLoS One. 2013;8:e83304. doi: 10.1371/journal.pone.0083304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Moles L, Gomez M, Heilig H, et al. Bacterial diversity in meconium of preterm neonates and evolution of their fecal microbiota during the first month of life. PLoS One. 2013;8:e66986. doi: 10.1371/journal.pone.0066986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Claud EC, Keegan KP, Brulc JM, et al. Bacterial community structure and functional contributions to emergence of health or necrotizing enterocolitis in preterm infants. Microbiome. 2013;1:20. doi: 10.1186/2049-2618-1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Madani G, Heiner DC. Antibody transmission from mother to fetus. Curr Opin Immunol. 1989;1:1157–64. doi: 10.1016/0952-7915(89)90009-5. [DOI] [PubMed] [Google Scholar]
  • 24.Cheng MM, Huang CF, Yang LY, et al. Development of serum IgA and IgM levels in breast-fed and formula-fed infants during the first week of life. Early Hum Dev. 2012;88:743–5. doi: 10.1016/j.earlhumdev.2012.03.005. [DOI] [PubMed] [Google Scholar]
  • 25.Niewiesk S. Maternal antibodies: clinical significance, mechanism of interference with immune responses, and possible vaccination strategies. Front Immunol. 2014;5:446. doi: 10.3389/fimmu.2014.00446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lawrence RM, Pane CA. Human breast milk: current concepts of immunology and infectious diseases. Current problems in pediatric and adolescent health care. 2007;37:7–36. doi: 10.1016/j.cppeds.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 27.Palmeira P, Quinello C, Silveira-Lessa AL, Zago CA, Carneiro-Sampaio M. IgG placental transfer in healthy and pathological pregnancies. Clin Dev Immunol. 2012;2012:985646. doi: 10.1155/2012/985646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gross SJ, Buckley RH, Wakil SS, McAllister DC, David RJ, Faix RG. Elevated IgA concentration in milk produced by mothers delivered of preterm infants. The Journal of pediatrics. 1981;99:389–93. doi: 10.1016/s0022-3476(81)80323-x. [DOI] [PubMed] [Google Scholar]
  • 29.Schlesinger L, Munoz C, Arevalo M, Arredondo S, Mendez G. Functional capacity of colostral leukocytes from women delivering prematurely. Journal of pediatric gastroenterology and nutrition. 1989;8:89–94. doi: 10.1097/00005176-198901000-00017. [DOI] [PubMed] [Google Scholar]
  • 30.Mehta R, Petrova A. Biologically active breast milk proteins in association with very preterm delivery and stage of lactation. Journal of perinatology : official journal of the California Perinatal Association. 2011;31:58–62. doi: 10.1038/jp.2010.68. [DOI] [PubMed] [Google Scholar]
  • 31.Foster JP, Seth R, Cole MJ. Oral immunoglobulin for preventing necrotizing enterocolitis in preterm and low birth weight neonates. Cochrane Database Syst Rev. 2016;4:CD001816. doi: 10.1002/14651858.CD001816.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Shah U, Dickinson BL, Blumberg RS, Simister NE, Lencer WI, Walker WA. Distribution of the IgG Fc receptor, FcRn, in the human fetal intestine. Pediatr Res. 2003;53:295–301. doi: 10.1203/01.PDR.0000047663.81816.E3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Swanson JR, Jilling T, Lu J, Landseadel JB, Marcinkiewicz M, Gordon PV. Ileal Immunoglobulin Binding by the Neonatal Fc Receptor: A Previously Unrecognized Mechanism of Protection in the Neonatal Rat Model of Necrotizing Enterocolitis? EJ Neonatol Res. 2011;1 [PMC free article] [PubMed] [Google Scholar]
  • 34.Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS. FcRn: The Architect Behind the Immune and Nonimmune Functions of IgG and Albumin. J Immunol. 2015;194:4595–603. doi: 10.4049/jimmunol.1403014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jakaitis BM, Denning PW. Human breast milk and the gastrointestinal innate immune system. Clin Perinatol. 2014;41:423–35. doi: 10.1016/j.clp.2014.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chatterton DE, Nguyen DN, Bering SB, Sangild PT. Anti-inflammatory mechanisms of bioactive milk proteins in the intestine of newborns. The international journal of biochemistry & cell biology. 2013;45:1730–47. doi: 10.1016/j.biocel.2013.04.028. [DOI] [PubMed] [Google Scholar]
  • 37.Goldman AS. The immune system of human milk: antimicrobial, antiinflammatory and immunomodulating properties. The Pediatric infectious disease journal. 1993;12:664–71. doi: 10.1097/00006454-199308000-00008. [DOI] [PubMed] [Google Scholar]
  • 38.Newburg DS. Neonatal protection by an innate immune system of human milk consisting of oligosaccharides and glycans. Journal of animal science. 2009;87:26–34. doi: 10.2527/jas.2008-1347. [DOI] [PubMed] [Google Scholar]
  • 39.Underwood MA, Gaerlan S, De Leoz ML, et al. Human milk oligosaccharides in premature infants: absorption, excretion, and influence on the intestinal microbiota. Pediatr Res. 2015;78:670–7. doi: 10.1038/pr.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Srinivasjois R, Rao S, Patole S. Prebiotic supplementation in preterm neonates: updated systematic review and meta-analysis of randomised controlled trials. Clin Nutr. 2013;32:958–65. doi: 10.1016/j.clnu.2013.05.009. [DOI] [PubMed] [Google Scholar]
  • 41.Armanian AM, Sadeghnia A, Hoseinzadeh M, et al. The Effect of Neutral Oligosaccharides on Reducing the Incidence of Necrotizing Enterocolitis in Preterm infants: A Randomized Clinical Trial. Int J Prev Med. 2014;5:1387–95. [PMC free article] [PubMed] [Google Scholar]
  • 42.Nandhini LP, Biswal N, Adhisivam B, Mandal J, Bhat BV, Mathai B. Synbiotics for decreasing incidence of necrotizing enterocolitis among preterm neonates - a randomized controlled trial. J Matern Fetal Neonatal Med. 2016;29:821–5. doi: 10.3109/14767058.2015.1019854. [DOI] [PubMed] [Google Scholar]
  • 43.Jantscher-Krenn E, Zherebtsov M, Nissan C, et al. The human milk oligosaccharide disialyllacto-N-tetraose prevents necrotising enterocolitis in neonatal rats. Gut. 2012;61:1417–25. doi: 10.1136/gutjnl-2011-301404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Plaisancie P, Claustre J, Estienne M, et al. A novel bioactive peptide from yoghurts modulates expression of the gel-forming MUC2 mucin as well as population of goblet cells and Paneth cells along the small intestine. The Journal of nutritional biochemistry. 2013;24:213–21. doi: 10.1016/j.jnutbio.2012.05.004. [DOI] [PubMed] [Google Scholar]
  • 45.Lonnerdal B. Bioactive proteins in human milk: mechanisms of action. The Journal of pediatrics. 2010;156:S26–30. doi: 10.1016/j.jpeds.2009.11.017. [DOI] [PubMed] [Google Scholar]
  • 46.Thormar H, Isaacs CE, Brown HR, Barshatzky MR, Pessolano T. Inactivation of enveloped viruses and killing of cells by fatty acids and monoglycerides. Antimicrobial agents and chemotherapy. 1987;31:27–31. doi: 10.1128/aac.31.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Newburg DS, Walker WA. Protection of the neonate by the innate immune system of developing gut and of human milk. Pediatric research. 2007;61:2–8. doi: 10.1203/01.pdr.0000250274.68571.18. [DOI] [PubMed] [Google Scholar]
  • 48.Ellison RT, 3rd, Giehl TJ. Killing of gram-negative bacteria by lactoferrin and lysozyme. The Journal of clinical investigation. 1991;88:1080–91. doi: 10.1172/JCI115407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lonnerdal B. Bioactive proteins in breast milk. Journal of paediatrics and child health. 2013;49(1):1–7. doi: 10.1111/jpc.12104. [DOI] [PubMed] [Google Scholar]
  • 50.Manzoni P, Rinaldi M, Cattani S, et al. Bovine lactoferrin supplementation for prevention of late-onset sepsis in very low-birth-weight neonates: a randomized trial. JAMA : the journal of the American Medical Association. 2009;302:1421–8. doi: 10.1001/jama.2009.1403. [DOI] [PubMed] [Google Scholar]
  • 51.Manzoni P, Stolfi I, Messner H, et al. Bovine lactoferrin prevents invasive fungal infections in very low birth weight infants: a randomized controlled trial. Pediatrics. 2012;129:116–23. doi: 10.1542/peds.2011-0279. [DOI] [PubMed] [Google Scholar]
  • 52.Valenti P, Antonini G. Lactoferrin: an important host defence against microbial and viral attack. Cellular and molecular life sciences : CMLS. 2005;62:2576–87. doi: 10.1007/s00018-005-5372-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kuwata H, Yip TT, Tomita M, Hutchens TW. Direct evidence of the generation in human stomach of an antimicrobial peptide domain (lactoferricin) from ingested lactoferrin. Biochimica et biophysica acta. 1998;1429:129–41. doi: 10.1016/s0167-4838(98)00224-6. [DOI] [PubMed] [Google Scholar]
  • 54.Pammi M, Abrams SA. Oral lactoferrin for the prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev. 2015;2:CD007137. doi: 10.1002/14651858.CD007137.pub4. [DOI] [PubMed] [Google Scholar]
  • 55.Furukawa M, Narahara H, Yasuda K, Johnston JM. Presence of platelet-activating factor-acetylhydrolase in milk. Journal of lipid research. 1993;34:1603–9. [PubMed] [Google Scholar]
  • 56.Moya FR, Eguchi H, Zhao B, et al. Platelet-activating factor acetylhydrolase in term and preterm human milk: a preliminary report. J Pediatr Gastroenterol Nutr. 1994;19:236–9. doi: 10.1097/00005176-199408000-00015. [DOI] [PubMed] [Google Scholar]
  • 57.Hawkes JS, Bryan DL, James MJ, Gibson RA. Cytokines (IL-1beta, IL-6, TNF-alpha, TGF-beta1, and TGF-beta2) and prostaglandin E2 in human milk during the first three months postpartum. Pediatric research. 1999;46:194–9. doi: 10.1203/00006450-199908000-00012. [DOI] [PubMed] [Google Scholar]
  • 58.Namachivayam K, Blanco CL, Frost BL, et al. Preterm human milk contains a large pool of latent TGF-beta, which can be activated by exogenous neuraminidase. Am J Physiol Gastrointest Liver Physiol. 2013;304:G1055–65. doi: 10.1152/ajpgi.00039.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chheda S, Palkowetz KH, Garofalo R, Rassin DK, Goldman AS. Decreased interleukin-10 production by neonatal monocytes and T cells: relationship to decreased production and expression of tumor necrosis factor-alpha and its receptors. Pediatr Res. 1996;40:475–83. doi: 10.1203/00006450-199609000-00018. [DOI] [PubMed] [Google Scholar]
  • 60.Chang M, Suen Y, Lee SM, et al. Transforming growth factor-beta 1, macrophage inflammatory protein-1 alpha, and interleukin-8 gene expression is lower in stimulated human neonatal compared with adult mononuclear cells. Blood. 1994;84:118–24. [PubMed] [Google Scholar]
  • 61.Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007;8:1086–94. doi: 10.1038/ni1511. [DOI] [PubMed] [Google Scholar]
  • 62.Denning TL, Norris BA, Medina-Contreras O, et al. Functional specializations of intestinal dendritic cell and macrophage subsets that control Th17 and regulatory T cell responses are dependent on the T cell/APC ratio, source of mouse strain, and regional localization. J Immunol. 2011;187:733–47. doi: 10.4049/jimmunol.1002701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75:263–74. doi: 10.1016/0092-8674(93)80068-p. [DOI] [PubMed] [Google Scholar]
  • 64.Emami CN, Chokshi N, Wang J, et al. Role of interleukin-10 in the pathogenesis of necrotizing enterocolitis. Am J Surg. 2012;203:428–35. doi: 10.1016/j.amjsurg.2011.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Glocker EO, Kotlarz D, Boztug K, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. The New England journal of medicine. 2009;361:2033–45. doi: 10.1056/NEJMoa0907206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dvorak B, Halpern MD, Holubec H, et al. Maternal milk reduces severity of necrotizing enterocolitis and increases intestinal IL-10 in a neonatal rat model. Pediatric research. 2003;53:426–33. doi: 10.1203/01.PDR.0000050657.56817.E0. [DOI] [PubMed] [Google Scholar]
  • 67.Fituch CC, Palkowetz KH, Goldman AS, Schanler RJ. Concentrations of IL-10 in preterm human milk and in milk from mothers of infants with necrotizing enterocolitis. Acta Paediatr. 2004;93:1496–500. doi: 10.1080/08035250410022314. [DOI] [PubMed] [Google Scholar]
  • 68.Mirpuri J, Sotnikov I, Myers L, et al. Lactobacillus rhamnosus (LGG) regulates IL-10 signaling in the developing murine colon through upregulation of the IL-10R2 receptor subunit. PLoS One. 2012;7:e51955. doi: 10.1371/journal.pone.0051955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Maheshwari A, Kelly DR, Nicola T, et al. TGF-beta2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology. 2011;140:242–53. doi: 10.1053/j.gastro.2010.09.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Penttila IA. Milk-derived transforming growth factor-beta and the infant immune response. The Journal of pediatrics. 2010;156:S21–5. doi: 10.1016/j.jpeds.2009.11.016. [DOI] [PubMed] [Google Scholar]
  • 71.Yuen DE, Stratford AF. Vitamin A activation of transforming growth factor-beta1 enhances porcine ileum wound healing in vitro. Pediatr Res. 2004;55:935–9. doi: 10.1203/01.pdr.0000127023.22960.85. [DOI] [PubMed] [Google Scholar]
  • 72.Ogawa J, Sasahara A, Yoshida T, et al. Role of transforming growth factor-beta in breast milk for initiation of IgA production in newborn infants. Early human development. 2004;77:67–75. doi: 10.1016/j.earlhumdev.2004.01.005. [DOI] [PubMed] [Google Scholar]
  • 73.Frost BL, Jilling T, Lapin B, Maheshwari A, Caplan MS. Maternal breast milk transforming growth factor-beta and feeding intolerance in preterm infants. Pediatr Res. 2014;76:386–93. doi: 10.1038/pr.2014.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Claud EC, Savidge T, Walker WA. Modulation of human intestinal epithelial cell IL-8 secretion by human milk factors. Pediatr Res. 2003;53:419–25. doi: 10.1203/01.PDR.0000050141.73528.AD. [DOI] [PubMed] [Google Scholar]
  • 75.Hellstrom A, Engstrom E, Hard AL, et al. Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics. 2003;112:1016–20. doi: 10.1542/peds.112.5.1016. [DOI] [PubMed] [Google Scholar]
  • 76.Baregamian N, Rychahou PG, Hawkins HK, Evers BM, Chung DH. Phosphatidylinositol 3-kinase pathway regulates hypoxia-inducible factor-1 to protect from intestinal injury during necrotizing enterocolitis. Surgery. 2007;142:295–302. doi: 10.1016/j.surg.2007.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ozen S, Akisu M, Baka M, et al. Insulin-like growth factor attenuates apoptosis and mucosal damage in hypoxia/reoxygenation-induced intestinal injury. Biol Neonate. 2005;87:91–6. doi: 10.1159/000081897. [DOI] [PubMed] [Google Scholar]
  • 78.Baregamian N, Song J, Chung DH. Effects of oxidative stress on intestinal type I insulin-like growth factor receptor expression. Eur J Pediatr Surg. 2012;22:97–104. doi: 10.1055/s-0032-1306261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Coursodon CF, Dvorak B. Epidermal growth factor and necrotizing enterocolitis. Curr Opin Pediatr. 2012;24:160–4. doi: 10.1097/MOP.0b013e3283504ddb. [DOI] [PubMed] [Google Scholar]
  • 80.Dvorak B. Milk epidermal growth factor and gut protection. J Pediatr. 2010;156:S31–5. doi: 10.1016/j.jpeds.2009.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Su Y, Yang J, Besner GE. HB-EGF promotes intestinal restitution by affecting integrin-extracellular matrix interactions and intercellular adhesions. Growth Factors. 2013;31:39–55. doi: 10.3109/08977194.2012.755966. [DOI] [PubMed] [Google Scholar]
  • 82.Maynard AA, Dvorak K, Khailova L, et al. Epidermal growth factor reduces autophagy in intestinal epithelium and in the rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2010;299:G614–22. doi: 10.1152/ajpgi.00076.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Clark JA, Doelle SM, Halpern MD, et al. Intestinal barrier failure during experimental necrotizing enterocolitis: protective effect of EGF treatment. Am J Physiol Gastrointest Liver Physiol. 2006;291:G938–49. doi: 10.1152/ajpgi.00090.2006. [DOI] [PubMed] [Google Scholar]
  • 84.Good M, Sodhi CP, Egan CE, et al. Breast milk protects against the development of necrotizing enterocolitis through inhibition of Toll-like receptor 4 in the intestinal epithelium via activation of the epidermal growth factor receptor. Mucosal Immunol. 2015;8:1166–79. doi: 10.1038/mi.2015.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wei J, Besner GE. M1 to M2 macrophage polarization in heparin-binding epidermal growth factor-like growth factor therapy for necrotizing enterocolitis. J Surg Res. 2015;197:126–38. doi: 10.1016/j.jss.2015.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dvorak B, Fituch CC, Williams CS, Hurst NM, Schanler RJ. Increased epidermal growth factor levels in human milk of mothers with extremely premature infants. Pediatric research. 2003;54:15–9. doi: 10.1203/01.PDR.0000065729.74325.71. [DOI] [PubMed] [Google Scholar]
  • 87.Warner BB, Ryan AL, Seeger K, Leonard AC, Erwin CR, Warner BW. Ontogeny of salivary epidermal growth factor and necrotizing enterocolitis. J Pediatr. 2007;150:358–63. doi: 10.1016/j.jpeds.2006.11.059. [DOI] [PubMed] [Google Scholar]
  • 88.Gupta A, Lakhoo K, Pritchard N, Herbert M. Epidermal growth factor in neonatal saliva. Eur J Pediatr Surg. 2008;18:245–8. doi: 10.1055/s-2008-1038392. [DOI] [PubMed] [Google Scholar]
  • 89.Wahab Mohamed WA, Aseeri AM. Cord blood epidermal growth factor as a possible predictor of necrotizing enterocolitis in very low birth weight infants. J Neonatal Perinatal Med. 2013;6:257–62. doi: 10.3233/NPM-1370813. [DOI] [PubMed] [Google Scholar]
  • 90.Shin CE, Falcone RA, Jr, Stuart L, Erwin CR, Warner BW. Diminished epidermal growth factor levels in infants with necrotizing enterocolitis. J Pediatr Surg. 2000;35:173–6. doi: 10.1016/s0022-3468(00)90005-8. discussion 7. [DOI] [PubMed] [Google Scholar]
  • 91.Dvorak B, Halpern MD, Holubec H, et al. Epidermal growth factor reduces the development of necrotizing enterocolitis in a neonatal rat model. Am J Physiol Gastrointest Liver Physiol. 2002;282:G156–64. doi: 10.1152/ajpgi.00196.2001. [DOI] [PubMed] [Google Scholar]
  • 92.Dvorak B, Khailova L, Clark JA, et al. Comparison of epidermal growth factor and heparin-binding epidermal growth factor-like growth factor for prevention of experimental necrotizing enterocolitis. J Pediatr Gastroenterol Nutr. 2008;47:11–8. doi: 10.1097/MPG.0b013e3181788618. [DOI] [PubMed] [Google Scholar]
  • 93.Wei J, Zhou Y, Besner GE. Heparin-binding EGF-like growth factor and enteric neural stem cell transplantation in the prevention of experimental necrotizing enterocolitis in mice. Pediatr Res. 2015;78:29–37. doi: 10.1038/pr.2015.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Yang J, Watkins D, Chen CL, Bhushan B, Zhou Y, Besner GE. Heparin-binding epidermal growth factor-like growth factor and mesenchymal stem cells act synergistically to prevent experimental necrotizing enterocolitis. J Am Coll Surg. 2012;215:534–45. doi: 10.1016/j.jamcollsurg.2012.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chen CL, Yu X, James IO, et al. Heparin-binding EGF-like growth factor protects intestinal stem cells from injury in a rat model of necrotizing enterocolitis. Lab Invest. 2012;92:331–44. doi: 10.1038/labinvest.2011.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yu X, Radulescu A, Zorko N, Besner GE. Heparin-binding EGF-like growth factor increases intestinal microvascular blood flow in necrotizing enterocolitis. Gastroenterology. 2009;137:221–30. doi: 10.1053/j.gastro.2009.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Khailova L, Dvorak K, Arganbright KM, Williams CS, Halpern MD, Dvorak B. Changes in hepatic cell junctions structure during experimental necrotizing enterocolitis: effect of EGF treatment. Pediatr Res. 2009;66:140–4. doi: 10.1203/PDR.0b013e3181aa3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Radulescu A, Zorko NA, Yu X, Besner GE. Preclinical neonatal rat studies of heparin-binding EGF-like growth factor in protection of the intestines from necrotizing enterocolitis. Pediatr Res. 2009;65:437–42. doi: 10.1203/PDR.0b013e3181994fa0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Feng J, El-Assal ON, Besner GE. Heparin-binding EGF-like growth factor (HB-EGF) and necrotizing enterocolitis. Semin Pediatr Surg. 2005;14:167–74. doi: 10.1053/j.sempedsurg.2005.05.005. [DOI] [PubMed] [Google Scholar]
  • 100.Feng J, El-Assal ON, Besner GE. Heparin-binding epidermal growth factor-like growth factor reduces intestinal apoptosis in neonatal rats with necrotizing enterocolitis. J Pediatr Surg. 2006;41:742–7. doi: 10.1016/j.jpedsurg.2005.12.020. discussion -7. [DOI] [PubMed] [Google Scholar]
  • 101.Feng J, El-Assal ON, Besner GE. Heparin-binding epidermal growth factor-like growth factor decreases the incidence of necrotizing enterocolitis in neonatal rats. J Pediatr Surg. 2006;41:144–9. doi: 10.1016/j.jpedsurg.2005.10.018. discussion -9. [DOI] [PubMed] [Google Scholar]
  • 102.Halpern MD, Holubec H, Clark JA, et al. Epidermal growth factor reduces hepatic sequelae in experimental necrotizing enterocolitis. Biol Neonate. 2006;89:227–35. doi: 10.1159/000090015. [DOI] [PubMed] [Google Scholar]
  • 103.Halpern MD, Dominguez JA, Dvorakova K, et al. Ileal cytokine dysregulation in experimental necrotizing enterocolitis is reduced by epidermal growth factor. J Pediatr Gastroenterol Nutr. 2003;36:126–33. doi: 10.1097/00005176-200301000-00024. [DOI] [PubMed] [Google Scholar]
  • 104.Sullivan PB, Lewindon PJ, Cheng C, et al. Intestinal mucosa remodeling by recombinant human epidermal growth factor(1-48) in neonates with severe necrotizing enterocolitis. J Pediatr Surg. 2007;42:462–9. doi: 10.1016/j.jpedsurg.2006.10.039. [DOI] [PubMed] [Google Scholar]
  • 105.Boyle JT. Acid secretion from birth to adulthood. J Pediatr Gastroenterol Nutr. 2003;37(1):S12–6. doi: 10.1097/00005176-200311001-00004. [DOI] [PubMed] [Google Scholar]
  • 106.Grahnquist L, Ruuska T, Finkel Y. Early development of human gastric H,K-adenosine triphosphatase. J Pediatr Gastroenterol Nutr. 2000;30:533–7. doi: 10.1097/00005176-200005000-00013. [DOI] [PubMed] [Google Scholar]
  • 107.Kelly EJ, Newell SJ, Brownlee KG, Primrose JN, Dear PR. Gastric acid secretion in preterm infants. Early Hum Dev. 1993;35:215–20. doi: 10.1016/0378-3782(93)90108-7. [DOI] [PubMed] [Google Scholar]
  • 108.More K, Athalye-Jape G, Rao S, Patole S. Association of inhibitors of gastric acid secretion and higher incidence of necrotizing enterocolitis in preterm very low-birth-weight infants. American journal of perinatology. 2013;30:849–56. doi: 10.1055/s-0033-1333671. [DOI] [PubMed] [Google Scholar]
  • 109.Terrin G, Passariello A, De Curtis M, et al. Ranitidine is associated with infections, necrotizing enterocolitis, and fatal outcome in newborns. Pediatrics. 2012;129:e40–5. doi: 10.1542/peds.2011-0796. [DOI] [PubMed] [Google Scholar]
  • 110.Bianconi S, Gudavalli M, Sutija VG, Lopez AL, Barillas-Arias L, Ron N. Ranitidine and late-onset sepsis in the neonatal intensive care unit. Journal of perinatal medicine. 2007;35:147–50. doi: 10.1515/JPM.2007.017. [DOI] [PubMed] [Google Scholar]
  • 111.Graham PL, 3rd, Begg MD, Larson E, Della-Latta P, Allen A, Saiman L. Risk factors for late onset gram-negative sepsis in low birth weight infants hospitalized in the neonatal intensive care unit. The Pediatric infectious disease journal. 2006;25:113–7. doi: 10.1097/01.inf.0000199310.52875.10. [DOI] [PubMed] [Google Scholar]
  • 112.Laboisse C, Jarry A, Branka JE, Merlin D, Bou-Hanna C, Vallette G. Regulation of mucin exocytosis from intestinal goblet cells. Biochemical Society transactions. 1995;23:810–3. doi: 10.1042/bst0230810. [DOI] [PubMed] [Google Scholar]
  • 113.Lievin-Le Moal V, Servin AL. The front line of enteric host defense against unwelcome intrusion of harmful microorganisms: mucins, antimicrobial peptides, and microbiota. Clinical microbiology reviews. 2006;19:315–37. doi: 10.1128/CMR.19.2.315-337.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Johansson ME, Sjovall H, Hansson GC. The gastrointestinal mucus system in health and disease. Nature reviews Gastroenterology & hepatology. 2013;10:352–61. doi: 10.1038/nrgastro.2013.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Johansson ME, Ambort D, Pelaseyed T, et al. Composition and functional role of the mucus layers in the intestine. Cellular and molecular life sciences : CMLS. 2011;68:3635–41. doi: 10.1007/s00018-011-0822-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Johansson ME, Larsson JM, Hansson GC. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(1):4659–65. doi: 10.1073/pnas.1006451107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.McElroy SJ, Weitkamp JH. Innate Immunity in the Small Intestine of the Preterm Infant. Neoreviews. 2011;12:e517–e26. doi: 10.1542/neo.12-9-e517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Pelaseyed T, Bergstrom JH, Gustafsson JK, et al. The mucus and mucins of the goblet cells and enterocytes provide the first defense line of the gastrointestinal tract and interact with the immune system. Immunological Reviews. 2014;260:8–20. doi: 10.1111/imr.12182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Corfield AP, Carroll D, Myerscough N, Probert CS. Mucins in the gastrointestinal tract in health and disease. Frontiers in bioscience : a journal and virtual library. 2001;6:D1321–57. doi: 10.2741/corfield. [DOI] [PubMed] [Google Scholar]
  • 120.Louis NA, Hamilton KE, Canny G, Shekels LL, Ho SB, Colgan SP. Selective induction of mucin-3 by hypoxia in intestinal epithelia. J Cell Biochem. 2006;99:1616–27. doi: 10.1002/jcb.20947. [DOI] [PubMed] [Google Scholar]
  • 121.McElroy SJ, Prince LS, Weitkamp JH, Reese J, Slaughter JC, Polk DB. Tumor Necrosis Factor Receptor 1-Dependent Depletion of Mucus in Immature Small Intestine: A Potential Role in Neonatal Necrotizing Enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2011;301:G656–G66. doi: 10.1152/ajpgi.00550.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Schaart MW, de Bruijn AC, Bouwman DM, et al. Epithelial functions of the residual bowel after surgery for necrotising enterocolitis in human infants. J Pediatr Gastroenterol Nutr. 2009;49:31–41. doi: 10.1097/MPG.0b013e318186d341. [DOI] [PubMed] [Google Scholar]
  • 140.McElroy SJ, Prince LS, Weitkamp JH, Reese J, Slaughter JC, Polk DB. Tumor necrosis factor receptor 1-dependent depletion of mucus in immature small intestine: a potential role in neonatal necrotizing enterocolitis. American journal of physiology Gastrointestinal and liver physiology. 2011;301:G656–66. doi: 10.1152/ajpgi.00550.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Martin NA, Mount Patrick SK, Estrada TE, et al. Active transport of bile acids decreases mucin 2 in neonatal ileum: implications for development of necrotizing enterocolitis. PLoS One. 2011;6:e27191. doi: 10.1371/journal.pone.0027191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zhang BH, Yu HG, Sheng ZX, Luo HS, Yu JP. The therapeutic effect of recombinant human trefoil factor 3 on hypoxia-induced necrotizing enterocolitis in immature rat. Regul Pept. 2003;116:53–60. doi: 10.1016/s0167-0115(03)00177-0. [DOI] [PubMed] [Google Scholar]
  • 143.Fanca-Berthon P, Michel C, Pagniez A, et al. Intrauterine Growth Restriction Alters Postnatal Colonic Barrier Maturation in Rats. Pediatric Research. 2009;66:47–52. doi: 10.1203/PDR.0b013e3181a2047e. [DOI] [PubMed] [Google Scholar]
  • 144.Ree IM, Smits-Wintjens VE, Rijntjes-Jacobs EG, et al. Necrotizing enterocolitis in small-for-gestational-age neonates: a matched case-control study. Neonatology. 2014;105:74–8. doi: 10.1159/000356033. [DOI] [PubMed] [Google Scholar]
  • 145.Che L, Thymann T, Bering SB, et al. IUGR does not predispose to necrotizing enterocolitis or compromise postnatal intestinal adaptation in preterm pigs. Pediatr Res. 2010;67:54–9. doi: 10.1203/PDR.0b013e3181c1b15e. [DOI] [PubMed] [Google Scholar]
  • 146.Kliegman RM, Hack M, Jones P, Fanaroff AA. Epidemiologic study of necrotizing enterocolitis among low-birth-weight infants. Absence of identifiable risk factors. J Pediatr. 1982;100:440–4. doi: 10.1016/s0022-3476(82)80456-3. [DOI] [PubMed] [Google Scholar]
  • 147.Dong L, Zhong X, Ahmad H, et al. Intrauterine Growth Restriction Impairs Small Intestinal Mucosal Immunity in Neonatal Piglets. J Histochem Cytochem. 2014;62:510–8. doi: 10.1369/0022155414532655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Fung CM, White JR, Brown AS, et al. Intrauterine Growth Restriction Alters Mouse Intestinal Architecture during Development. PLoS One. 2016;11:e0146542. doi: 10.1371/journal.pone.0146542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Halpern MD, Denning PW. The role of intestinal epithelial barrier function in the development of NEC. Tissue Barriers. 2015;3:e1000707. doi: 10.1080/21688370.2014.1000707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Anderson JM, Van Itallie CM. Tight junctions. Current biology : CB. 2008;18:R941–3. doi: 10.1016/j.cub.2008.07.083. [DOI] [PubMed] [Google Scholar]
  • 151.Balda MS, Fallon MB, Van Itallie CM, Anderson JM. Structure, regulation, and pathophysiology of tight junctions in the gastrointestinal tract. The Yale journal of biology and medicine. 1992;65:725–35. discussion 37-40. [PMC free article] [PubMed] [Google Scholar]
  • 152.Lebenthal A, Lebenthal E. The ontogeny of the small intestinal epithelium. JPEN J Parenter Enteral Nutr. 1999;23:S3–6. doi: 10.1177/014860719902300502. [DOI] [PubMed] [Google Scholar]
  • 153.Goldman AS. Modulation of the gastrointestinal tract of infants by human milk. Interfaces and interactions. An evolutionary perspective. J Nutr. 2000;130:426S–31S. doi: 10.1093/jn/130.2.426S. [DOI] [PubMed] [Google Scholar]
  • 154.Colome G, Sierra C, Blasco J, Garcia MV, Valverde E, Sanchez E. Intestinal permeability in different feedings in infancy. Acta Paediatr. 2007;96:69–72. doi: 10.1111/j.1651-2227.2007.00030.x. [DOI] [PubMed] [Google Scholar]
  • 155.Weaver LT, Laker MF, Nelson R, Lucas A. Milk feeding and changes in intestinal permeability and morphology in the newborn. J Pediatr Gastroenterol Nutr. 1987;6:351–8. doi: 10.1097/00005176-198705000-00008. [DOI] [PubMed] [Google Scholar]
  • 156.Henning SJ. Development of the gastrointestinal tract. Proc Nutr Soc. 1986;45:39–44. doi: 10.1079/pns19860033. [DOI] [PubMed] [Google Scholar]
  • 157.Patel RM, Myers LS, Kurundkar AR, Maheshwari A, Nusrat A, Lin PW. Probiotic bacteria induce maturation of intestinal claudin 3 expression and barrier function. Am J Pathol. 2012;180:626–35. doi: 10.1016/j.ajpath.2011.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–41. doi: 10.1016/j.cell.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 123.Weaver LT, Laker MF, Nelson R. Intestinal permeability in the newborn. Arch Dis Child. 1984;59:236–41. doi: 10.1136/adc.59.3.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.van Elburg RM, Fetter WP, Bunkers CM, Heymans HS. Intestinal permeability in relation to birth weight and gestational and postnatal age. Arch Dis Child Fetal Neonatal Ed. 2003;88:F52–5. doi: 10.1136/fn.88.1.F52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Henry MC, Moss RL. Neonatal necrotizing enterocolitis. Semin Pediatr Surg. 2008;17:98–109. doi: 10.1053/j.sempedsurg.2008.02.005. [DOI] [PubMed] [Google Scholar]
  • 126.Grave GD, Nelson SA, Walker WA, et al. New therapies and preventive approaches for necrotizing enterocolitis: report of a research planning workshop. Pediatr Res. 2007;62:510–4. doi: 10.1203/PDR.0b013e318142580a. [DOI] [PubMed] [Google Scholar]
  • 127.Lin PW, Nasr TR, Stoll BJ. Necrotizing enterocolitis: recent scientific advances in pathophysiology and prevention. Semin Perinatol. 2008;32:70–82. doi: 10.1053/j.semperi.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 128.Bjarnason I. Intestinal permeability. Gut. 1994;35:S18–22. doi: 10.1136/gut.35.1_suppl.s18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bergmann KR, Liu SX, Tian R, et al. Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis. Am J Pathol. 2013;182:1595–606. doi: 10.1016/j.ajpath.2013.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Weitkamp JH, Rosen MJ, Zhao Z, et al. Small intestinal intraepithelial TCRgammadelta+ Tlymphocytes are present in the premature intestine but selectively reduced in surgical necrotizing enterocolitis. PLoS One. 2014;9:e99042. doi: 10.1371/journal.pone.0099042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rentea RM, Liedel JL, Welak SR, et al. Intestinal alkaline phosphatase administration in newborns is protective of gut barrier function in a neonatal necrotizing enterocolitis rat model. J Pediatr Surg. 2012;47:1135–42. doi: 10.1016/j.jpedsurg.2012.03.018. [DOI] [PubMed] [Google Scholar]
  • 132.Hogberg N, Stenback A, Carlsson PO, Wanders A, Lilja HE. Genes regulating tight junctions and cell adhesion are altered in early experimental necrotizing enterocolitis. J Pediatr Surg. 2013;48:2308–12. doi: 10.1016/j.jpedsurg.2013.06.027. [DOI] [PubMed] [Google Scholar]
  • 133.Shiou SR, Yu Y, Chen S, et al. Erythropoietin protects intestinal epithelial barrier function and lowers the incidence of experimental neonatal necrotizing enterocolitis. J Biol Chem. 2011;286:12123–32. doi: 10.1074/jbc.M110.154625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Prasad S, Mingrino R, Kaukinen K, et al. Inflammatory processes have differential effects on claudins 2, 3 and 4 in colonic epithelial cells. Lab Invest. 2005;85:1139–62. doi: 10.1038/labinvest.3700316. [DOI] [PubMed] [Google Scholar]
  • 135.Bruewer M, Luegering A, Kucharzik T, et al. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol. 2003;171:6164–72. doi: 10.4049/jimmunol.171.11.6164. [DOI] [PubMed] [Google Scholar]
  • 136.Lapointe TK, Buret AG. Interleukin-18 facilitates neutrophil transmigration via myosin light chain kinase-dependent disruption of occludin, without altering epithelial permeability. Am J Physiol Gastrointest Liver Physiol. 2011;302:G343–51. doi: 10.1152/ajpgi.00202.2011. [DOI] [PubMed] [Google Scholar]
  • 137.Marchiando AM, Shen L, Graham WV, et al. The epithelial barrier is maintained by in vivo tight junction expansion during pathologic intestinal epithelial shedding. Gastroenterology. 2011;140:1208–18. e1–2. doi: 10.1053/j.gastro.2011.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Fries W, Muja C, Crisafulli C, Cuzzocrea S, Mazzon E. Dynamics of enterocyte tight junctions: effect of experimental colitis and two different anti-TNF strategies. Am J Physiol Gastrointest Liver Physiol. 2008;294:G938–47. doi: 10.1152/ajpgi.00469.2007. [DOI] [PubMed] [Google Scholar]
  • 158.Halpern MD, Clark JA, Saunders TA, et al. Reduction of Experimental Necrotizing Enterocolitis with Anti-TNF-{alpha} Am J Physiol Gastrointest Liver Physiol. 2006;290:G757–64. doi: 10.1152/ajpgi.00408.2005. [DOI] [PubMed] [Google Scholar]
  • 159.Thuijls G, Derikx JP, van Wijck K, et al. Non-invasive markers for early diagnosis and determination of the severity of necrotizing enterocolitis. Ann Surg. 2010;251:1174–80. doi: 10.1097/SLA.0b013e3181d778c4. [DOI] [PubMed] [Google Scholar]
  • 160.Nanthakumar N, Meng D, Goldstein AM, et al. The mechanism of excessive intestinal inflammation in necrotizing enterocolitis: an immature innate immune response. PLoS One. 2011;6:e17776. doi: 10.1371/journal.pone.0017776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sampath V, Le M, Lane L, et al. The NFKB1 (g.-24519delATTG) variant is associated with necrotizing enterocolitis (NEC) in premature infants. J Surg Res. 2011;169:e51–7. doi: 10.1016/j.jss.2011.03.017. [DOI] [PubMed] [Google Scholar]
  • 162.Chan KY, Leung KT, Tam YH, et al. Genome-wide expression profiles of necrotizing enterocolitis versus spontaneous intestinal perforation in human intestinal tissues: dysregulation of functional pathways. Ann Surg. 2014;260:1128–37. doi: 10.1097/SLA.0000000000000374. [DOI] [PubMed] [Google Scholar]
  • 163.Ganguli K, Meng D, Rautava S, Lu L, Walker WA, Nanthakumar N. Probiotics prevent necrotizing enterocolitis by modulating enterocyte genes that regulate innate immune-mediated inflammation. Am J Physiol Gastrointest Liver Physiol. 2013;304:G132–41. doi: 10.1152/ajpgi.00142.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hartel C, Hartz A, Pagel J, et al. NOD2 Loss-of-Function Mutations and Risks of Necrotizing Enterocolitis or Focal Intestinal Perforation in Very Low-birth-weight Infants. Inflamm Bowel Dis. 2016;22:249–56. doi: 10.1097/MIB.0000000000000658. [DOI] [PubMed] [Google Scholar]
  • 165.Zhou W, Li W, Zheng XH, Rong X, Huang LG. Glutamine downregulates TLR-2 and TLR-4 expression and protects intestinal tract in preterm neonatal rats with necrotizing enterocolitis. J Pediatr Surg. 2014;49:1057–63. doi: 10.1016/j.jpedsurg.2014.02.078. [DOI] [PubMed] [Google Scholar]
  • 166.Khailova L, Mount Patrick SK, Arganbright KM, Halpern MD, Kinouchi T, Dvorak B. Bifidobacterium bifidum reduces apoptosis in the intestinal epithelium in necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2010;299:G1118–27. doi: 10.1152/ajpgi.00131.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Le Mandat Schultz A, Bonnard A, Barreau F, et al. Expression of TLR-2, TLR-4, NOD2 and pNF-kappaB in a neonatal rat model of necrotizing enterocolitis. PLoS One. 2007;2:e1102. doi: 10.1371/journal.pone.0001102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Sodhi CP, Neal MD, Siggers R, et al. Intestinal epithelial toll-like receptor 4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology. 2012;143:708–18 e5. doi: 10.1053/j.gastro.2012.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Siggers J, Sangild PT, Jensen TK, et al. Transition from parenteral to enteral nutrition induces immediate diet-dependent gut histological and immunological responses in preterm neonates. Am J Physiol Gastrointest Liver Physiol. 2011;301:G435–45. doi: 10.1152/ajpgi.00400.2010. [DOI] [PubMed] [Google Scholar]
  • 170.Lu J, Caplan MS, Li D, Jilling T. Polyunsaturated fatty acids block platelet-activating factor-induced phosphatidylinositol 3 kinase/Akt-mediated apoptosis in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2008;294:G1181–90. doi: 10.1152/ajpgi.00343.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Ostergaard MV, Cilieborg MS, Skovgaard K, Schmidt M, Sangild PT, Bering SB. Preterm Birth Reduces Nutrient Absorption With Limited Effect on Immune Gene Expression and Gut Colonization in Pigs. J Pediatr Gastroenterol Nutr. 2015;61:481–90. doi: 10.1097/MPG.0000000000000827. [DOI] [PubMed] [Google Scholar]
  • 172.Neal MD, Richardson WM, Sodhi CP, Russo A, Hackam DJ. Intestinal stem cells and their roles during mucosal injury and repair. J Surg Res. 2011;167:1–8. doi: 10.1016/j.jss.2010.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Good M, Siggers RH, Sodhi CP, et al. Amniotic fluid inhibits Toll-like receptor 4 signaling in the fetal and neonatal intestinal epithelium. Proc Natl Acad Sci U S A. 2012;109:11330–5. doi: 10.1073/pnas.1200856109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Jilling T, Simon D, Lu J, et al. The roles of bacteria and TLR4 in rat and murine models of necrotizing enterocolitis. J Immunol. 2006;177:3273–82. doi: 10.4049/jimmunol.177.5.3273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Richardson WM, Sodhi CP, Russo A, et al. Nucleotide-binding oligomerization domain-2 inhibits toll-like receptor-4 signaling in the intestinal epithelium. Gastroenterology. 2010;139:904–17. 17 e1–6. doi: 10.1053/j.gastro.2010.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol. 2010;10:131–44. doi: 10.1038/nri2707. [DOI] [PubMed] [Google Scholar]
  • 177.Hackam DJ, Good M, Sodhi CP. Mechanisms of gut barrier failure in the pathogenesis of necrotizing enterocolitis: Toll-like receptors throw the switch. Semin Pediatr Surg. 2013;22:76–82. doi: 10.1053/j.sempedsurg.2013.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Hackam DJ, Afrazi A, Good M, Sodhi CP. Innate immune signaling in the pathogenesis of necrotizing enterocolitis. Clin Dev Immunol. 2013;2013:475415. doi: 10.1155/2013/475415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Hunter CJ, De Plaen IG. Inflammatory signaling in NEC: Role of NF-kappaB, cytokines and other inflammatory mediators. Pathophysiology. 2014;21:55–65. doi: 10.1016/j.pathophys.2013.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Frost BL, Jilling T, Caplan MS. The importance of pro-inflammatory signaling in neonatal necrotizing enterocolitis. Semin Perinatol. 2008;32:100–6. doi: 10.1053/j.semperi.2008.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Soliman A, Michelsen KS, Karahashi H, et al. Platelet-activating factor induces TLR4 expression in intestinal epithelial cells: implication for the pathogenesis of necrotizing enterocolitis. PLoS One. 2010;5:e15044. doi: 10.1371/journal.pone.0015044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Gordon P, Christensen R, Weitkamp JH, Maheshwari A. Mapping the New World of Necrotizing Enterocolitis (NEC): Review and Opinion. EJ Neonatol Res. 2012;2:145–72. [PMC free article] [PubMed] [Google Scholar]
  • 183.Good M, Sodhi CP, Ozolek JA, et al. Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: evidence in mice for a role of TLR9. Am J Physiol Gastrointest Liver Physiol. 2014;306:G1021–32. doi: 10.1152/ajpgi.00452.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Arciero J, Bard Ermentrout G, Siggers R, et al. Modeling the interactions of bacteria and Toll-like receptor-mediated inflammation in necrotizing enterocolitis. J Theor Biol. 2013;321:83–99. doi: 10.1016/j.jtbi.2012.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Liu Y, Zhu L, Fatheree NY, et al. Changes in intestinal Toll-like receptors and cytokines precede histological injury in a rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2009;297:G442–50. doi: 10.1152/ajpgi.00182.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Gribar SC, Sodhi CP, Richardson WM, et al. Reciprocal expression and signaling of TLR4 and TLR9 in the pathogenesis and treatment of necrotizing enterocolitis. J Immunol. 2009;182:636–46. doi: 10.4049/jimmunol.182.1.636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Selsted ME, Ouellette AJ. Mammalian defensins in the antimicrobial immune response. Nat Immunol. 2005;6:551–7. doi: 10.1038/ni1206. [DOI] [PubMed] [Google Scholar]
  • 188.Perez-Lopez A, Behnsen J, Nuccio SP, Raffatellu M. Mucosal immunity to pathogenic intestinal bacteria. Nat Rev Immunol. 2016;16:135–48. doi: 10.1038/nri.2015.17. [DOI] [PubMed] [Google Scholar]
  • 189.Otte JM, Kiehne K, Herzig KH. Antimicrobial peptides in innate immunity of the human intestine. J Gastroenterol. 2003;38:717–26. doi: 10.1007/s00535-003-1136-5. [DOI] [PubMed] [Google Scholar]
  • 190.Fabisiak A, Murawska N, Fichna J. LL-37: Cathelicidin-related antimicrobial peptide with pleiotropic activity. Pharmacol Rep. 2016 doi: 10.1016/j.pharep.2016.03.015. [DOI] [PubMed] [Google Scholar]
  • 191.Salzman NH, Underwood MA, Bevins CL. Paneth cells, defensins, and the commensal microbiota: a hypothesis on intimate interplay at the intestinal mucosa. Seminars in immunology. 2007;19:70–83. doi: 10.1016/j.smim.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 192.Ganz T. Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol. 2003;3:710–20. doi: 10.1038/nri1180. [DOI] [PubMed] [Google Scholar]
  • 193.Lin PWaN, Andrew S. Innate Immunity and Epithelial Biology: Special Considerations in the Neonatal Gut. In: Neu J, editor. Gastroenterology and Nutrition: Neonatal Questions and Controversies. Philadelphia: Saunders; 2008. pp. 51–72. [Google Scholar]
  • 194.Lin PW, Simon PO, Jr, Gewirtz AT, et al. Paneth cell cryptdins act in vitro as apical paracrine regulators of the innate inflammatory response. J Biol Chem. 2004;279:19902–7. doi: 10.1074/jbc.M311821200. [DOI] [PubMed] [Google Scholar]
  • 195.Sheng Q, Lv Z, Cai W, et al. Human beta-defensin-3 promotes intestinal epithelial cell migration and reduces the development of necrotizing enterocolitis in a neonatal rat model. Pediatr Res. 2014;76:269–79. doi: 10.1038/pr.2014.93. [DOI] [PubMed] [Google Scholar]
  • 196.Campeotto F, Baldassarre M, Laforgia N, et al. Fecal expression of human beta-defensin-2 following birth. Neonatology. 2010;98:365–9. doi: 10.1159/000315872. [DOI] [PubMed] [Google Scholar]
  • 197.Jenke AC, Postberg J, Mariel B, et al. S100A12 and hBD2 correlate with the composition of the fecal microflora in ELBW infants and expansion of E. coli is associated with NEC. Biomed Res Int. 2013;2013:150372. doi: 10.1155/2013/150372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Jenke AC, Zilbauer M, Postberg J, Wirth S. Human beta-defensin 2 expression in ELBW infants with severe necrotizing enterocolitis. Pediatr Res. 2012;72:513–20. doi: 10.1038/pr.2012.110. [DOI] [PubMed] [Google Scholar]
  • 199.Mallow EB, Harris A, Salzman N, et al. Human enteric defensins. Gene structure and developmental expression. J Biol Chem. 1996;271:4038–45. doi: 10.1074/jbc.271.8.4038. [DOI] [PubMed] [Google Scholar]
  • 200.Rumbo M, Schiffrin EJ. Ontogeny of intestinal epithelium immune functions: developmental and environmental regulation. Cell Mol Life Sci. 2005;62:1288–96. doi: 10.1007/s00018-005-5033-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Heida FH, Beyduz G, Bulthuis ML, et al. Paneth cells in the developing gut: when do they arise and when are they immune competent? Pediatr Res. 2016 doi: 10.1038/pr.2016.67. [DOI] [PubMed] [Google Scholar]
  • 202.Salzman NH, Polin RA, Harris MC, et al. Enteric defensin expression in necrotizing enterocolitis. Pediatr Res. 1998;44:20–6. doi: 10.1203/00006450-199807000-00003. [DOI] [PubMed] [Google Scholar]
  • 203.McElroy SJ, Castle SL, Bernard JK, et al. The ErbB4 ligand neuregulin-4 protects against experimental necrotizing enterocolitis. Am J Pathol. 2014;184:2768–78. doi: 10.1016/j.ajpath.2014.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Underwood MA, Kananurak A, Coursodon CF, et al. Bifidobacterium bifidum in a rat model of necrotizing enterocolitis: antimicrobial peptide and protein responses. Pediatr Res. 2012;71:546–51. doi: 10.1038/pr.2012.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.McElroy SJ, Underwood MA, Sherman MP. Paneth cells and necrotizing enterocolitis: a novel hypothesis for disease pathogenesis. Neonatology. 2013;103:10–20. doi: 10.1159/000342340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Coutinho HB, da Mota HC, Coutinho VB, et al. Absence of lysozyme (muramidase) in the intestinal Paneth cells of newborn infants with necrotising enterocolitis. J Clin Pathol. 1998;51:512–4. doi: 10.1136/jcp.51.7.512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Puiman PJ, Burger-Van Paassen N, Schaart MW, et al. Paneth cell hyperplasia and metaplasia in necrotizing enterocolitis. Pediatr Res. 2011;69:217–23. doi: 10.1203/PDR.0b013e3182092a9a. [DOI] [PubMed] [Google Scholar]
  • 208.Sperandio B, Fischer N, Sansonetti PJ. Mucosal physical and chemical innate barriers: Lessons from microbial evasion strategies. Seminars in immunology. 2015;27:111–8. doi: 10.1016/j.smim.2015.03.011. [DOI] [PubMed] [Google Scholar]
  • 209.Kinnebrew MA, Pamer EG. Innate immune signaling in defense against intestinal microbes. Immunol Rev. 2012;245:113–31. doi: 10.1111/j.1600-065X.2011.01081.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Cheroutre H. Starting at the beginning: new perspectives on the biology of mucosal T cells. Annu Rev Immunol. 2004;22:217–46. doi: 10.1146/annurev.immunol.22.012703.104522. [DOI] [PubMed] [Google Scholar]
  • 211.Cheroutre H, Lambolez F. The thymus chapter in the life of gut-specific intra epithelial lymphocytes. Curr Opin Immunol. 2008;20:185–91. doi: 10.1016/j.coi.2008.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Cheroutre H. IELs: enforcing law and order in the court of the intestinal epithelium. Immunol Rev. 2005;206:114–31. doi: 10.1111/j.0105-2896.2005.00284.x. [DOI] [PubMed] [Google Scholar]
  • 213.Hayday A, Tigelaar R. Immunoregulation in the tissues by gammadelta T cells. Nat Rev Immunol. 2003;3:233–42. doi: 10.1038/nri1030. [DOI] [PubMed] [Google Scholar]
  • 214.Ivanov II, McKenzie BS, Zhou L, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–33. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 215.Maxwell JR, Zhang Y, Brown WA, et al. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity. 2015;43:739–50. doi: 10.1016/j.immuni.2015.08.019. [DOI] [PubMed] [Google Scholar]
  • 216.Lee JS, Tato CM, Joyce-Shaikh B, et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity. 2015;43:727–38. doi: 10.1016/j.immuni.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Symons A, Budelsky AL, Towne JE. Are Th17 cells in the gut pathogenic or protective? Mucosal Immunol. 2012;5:4–6. doi: 10.1038/mi.2011.51. [DOI] [PubMed] [Google Scholar]
  • 218.Ramirez K, Witherden DA, Havran WL. All hands on DE(T)C: Epithelial-resident gammadelta T cells respond to tissue injury. Cell Immunol. 2015;296:57–61. doi: 10.1016/j.cellimm.2015.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Vantourout P, Hayday A. Six-of-the-best: unique contributions of gammadelta T cells to immunology. Nat Rev Immunol. 2013;13:88–100. doi: 10.1038/nri3384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9:503–10. doi: 10.1038/ni1582. [DOI] [PubMed] [Google Scholar]
  • 221.Hall LJ, Murphy CT, Quinlan A, et al. Natural killer cells protect mice from DSS-induced colitis by regulating neutrophil function via the NKG2A receptor. Mucosal Immunol. 2013;6:1016–26. doi: 10.1038/mi.2012.140. [DOI] [PubMed] [Google Scholar]
  • 222.Fort MM, Leach MW, Rennick DM. A role for NK cells as regulators of CD4+ T cells in a transfer model of colitis. J Immunol. 1998;161:3256–61. [PubMed] [Google Scholar]
  • 223.Bochennek K, Fryns E, Wittekindt B, et al. Immune cell subsets at birth may help to predict risk of late-onset sepsis and necrotizing enterocolitis in preterm infants. Early Hum Dev. 2016;93:9–16. doi: 10.1016/j.earlhumdev.2015.10.018. [DOI] [PubMed] [Google Scholar]
  • 224.Tait Wojno ED, Artis D. Innate lymphoid cells: balancing immunity, inflammation, and tissue repair in the intestine. Cell Host Microbe. 2012;12:445–57. doi: 10.1016/j.chom.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Fournier BM, Parkos CA. The role of neutrophils during intestinal inflammation. Mucosal Immunol. 2012;5:354–66. doi: 10.1038/mi.2012.24. [DOI] [PubMed] [Google Scholar]
  • 226.Tanner SM, Berryhill TF, Ellenburg JL, et al. Pathogenesis of necrotizing enterocolitis: modeling the innate immune response. Am J Pathol. 2015;185:4–16. doi: 10.1016/j.ajpath.2014.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Christensen RD, Yoder BA, Baer VL, Snow GL, Butler A. Early-Onset Neutropenia in Small-for-Gestational-Age Infants. Pediatrics. 2015;136:e1259–67. doi: 10.1542/peds.2015-1638. [DOI] [PubMed] [Google Scholar]
  • 228.Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Role of neutrophils and macrophages in the pathogenesis of necrotizing enterocolitis caused by Cronobacter sakazakii. J Surg Res. 2012;172:18–28. doi: 10.1016/j.jss.2011.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.MohanKumar K, Kaza N, Jagadeeswaran R, et al. Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model. Am J Physiol Gastrointest Liver Physiol. 2012;303:G93–102. doi: 10.1152/ajpgi.00016.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Zindl CL, Lai JF, Lee YK, et al. IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis. Proc Natl Acad Sci U S A. 2013;110:12768–73. doi: 10.1073/pnas.1300318110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Parks OB, Pociask DA, Hodzic Z, Kolls JK, Good M. Interleukin-22 Signaling in the Regulation of Intestinal Health and Disease. Front Cell Dev Biol. 2015;3:85. doi: 10.3389/fcell.2015.00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Dudakov JA, Hanash AM, van den Brink MR. Interleukin-22: immunobiology and pathology. Annu Rev Immunol. 2015;33:747–85. doi: 10.1146/annurev-immunol-032414-112123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Mizoguchi A. Healing of intestinal inflammation by IL-22. Inflamm Bowel Dis. 2012;18:1777–84. doi: 10.1002/ibd.22929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Renz H, Brandtzaeg P, Hornef M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nat Rev Immunol. 2012;12:9–23. doi: 10.1038/nri3112. [DOI] [PubMed] [Google Scholar]
  • 235.Lotz M, Gutle D, Walther S, Menard S, Bogdan C, Hornef MW. Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells. J Exp Med. 2006;203:973–84. doi: 10.1084/jem.20050625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Remon J, Kampanatkosol R, Kaul RR, Muraskas JK, Christensen RD, Maheshwari A. Acute drop in blood monocyte count differentiates NEC from other causes of feeding intolerance. J Perinatol. 2014;34:549–54. doi: 10.1038/jp.2014.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.MohanKumar K, Namachivayam K, Chapalamadugu KC, et al. Smad7 interrupts TGF-beta signaling in intestinal macrophages and promotes inflammatory activation of these cells during necrotizing enterocolitis. Pediatr Res. 2016 doi: 10.1038/pr.2016.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Bhatia AM, Stoll BJ, Cismowski MJ, Hamrick SE. Cytokine levels in the preterm infant with neonatal intestinal injury. Am J Perinatol. 2014;31:489–96. doi: 10.1055/s-0033-1353437. [DOI] [PubMed] [Google Scholar]
  • 239.Gross M, Salame TM, Jung S. Guardians of the Gut - Murine Intestinal Macrophages and Dendritic Cells. Front Immunol. 2015;6:254. doi: 10.3389/fimmu.2015.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Bogunovic M, Mortha A, Muller PA, Merad M. Mononuclear phagocyte diversity in the intestine. Immunol Res. 2012;54:37–49. doi: 10.1007/s12026-012-8323-5. [DOI] [PubMed] [Google Scholar]
  • 241.Starcevic V. Contrasting patterns in the relationship between hypochondriasis and narcissism. Br J Med Psychol. 1989;62(Pt 4):311–23. doi: 10.1111/j.2044-8341.1989.tb02841.x. [DOI] [PubMed] [Google Scholar]
  • 242.Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat Rev Immunol. 2008;8:435–46. doi: 10.1038/nri2335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Recruitment of dendritic cells is responsible for intestinal epithelial damage in the pathogenesis of necrotizing enterocolitis by Cronobacter sakazakii. J Immunol. 2011;186:7067–79. doi: 10.4049/jimmunol.1100108. [DOI] [PubMed] [Google Scholar]
  • 244.Egan CE, Sodhi CP, Good M, et al. Toll-like receptor 4-mediated lymphocyte influx induces neonatal necrotizing enterocolitis. J Clin Invest. 2015 doi: 10.1172/JCI83356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Weitkamp JH, Koyama T, Rock MT, et al. Necrotising enterocolitis is characterised by disrupted immune regulation and diminished mucosal regulatory (FOXP3)/effector (CD4, CD8) T cell ratios. Gut. 2013;62:73–82. doi: 10.1136/gutjnl-2011-301551. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES