Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-16T22:13:08.973Z Has data issue: false hasContentIssue false

The immunological landscape in necrotising enterocolitis

Published online by Cambridge University Press:  24 June 2016

Steven X. Cho
Affiliation:
Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia Department of Paediatrics, Monash University, Melbourne, Australia
Philip J. Berger
Affiliation:
Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia Department of Paediatrics, Monash University, Melbourne, Australia
Claudia A. Nold-Petry
Affiliation:
Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia Department of Paediatrics, Monash University, Melbourne, Australia
Marcel F. Nold*
Affiliation:
Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia Department of Paediatrics, Monash University, Melbourne, Australia
*
*Corresponding author: Marcel F. Nold, Hudson Institute of Medical Research, 27-31 Wright St., Melbourne, Victoria, Australia. E-mail: marcel.nold@monash.edu
Rights & Permissions [Opens in a new window]

Abstract

Necrotising enterocolitis (NEC) is an uncommon, but devastating intestinal inflammatory disease that predominantly affects preterm infants. NEC is sometimes dubbed the spectre of neonatal intensive care units, as its onset is insidiously non-specific, and once the disease manifests, the damage inflicted on the baby's intestine is already disastrous. Subsequent sepsis and multi-organ failure entail a mortality of up to 65%. Development of effective treatments for NEC has stagnated, largely because of our lack of understanding of NEC pathogenesis. It is clear, however, that NEC is driven by a profoundly dysregulated immune system. NEC is associated with local increases in pro-inflammatory mediators, e.g. Toll-like receptor (TLR) 4, nuclear factor-κB, tumour necrosis factor, platelet-activating factor (PAF), interleukin (IL)-18, interferon-gamma, IL-6, IL-8 and IL-1β. Deficiencies in counter-regulatory mechanisms, including IL-1 receptor antagonist (IL-1Ra), TLR9, PAF-acetylhydrolase, transforming growth factor beta (TGF-β)1&2, IL-10 and regulatory T cells likely facilitate a pro-inflammatory milieu in the NEC-afflicted intestine. There is insufficient evidence to conclude a predominance of an adaptive Th1-, Th2- or Th17-response in the disease. Our understanding of the accompanying regulation of systemic immunity remains poor; however, IL-1Ra, IL-6, IL-8 and TGF-β1 show promise as biomarkers. Here, we chart the emerging immunological landscape that underpins NEC by reviewing the involvement and potential clinical implications of innate and adaptive immune mediators and their regulation in NEC.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © Cambridge University Press 2016

Introduction

Necrotising enterocolitis (NEC) is a serious gastrointestinal disease that most commonly afflicts infants born prematurely. Although infrequent, NEC is a major cause of morbidity and mortality in neonatal intensive care units (NICUs). In older children, NEC occurs most commonly in association with cyanotic heart disease or major cardiac surgery (Ref. Reference Al Tawil1). NEC is a multifactorial disease whose pathogenesis remains poorly understood despite decades of research. However, risk factors for NEC have been identified, namely prematurity, formula feeding, hypoxic–ischaemic injury and abnormal bacterial colonisation. Yet, no single risk factor is essential, and the mechanisms by which each precipitates NEC are largely unknown. Nonetheless, evidence is mounting that formula feeding, hypoxia–ischemia, and dysbiosis lead to inflammation, and that immaturity of the immune system in preterm babies – although itself poorly characterised – is one of the pivotal pathogenic factors in NEC. Here, we review current knowledge on inflammation and immunity in NEC and highlight frontiers emerging in this field.

Epidemiology, staging criteria and disease outcomes

Death of extremely premature infants from most causes has decreased across the period from 2000 to 2011, whereas the incidence of death from NEC has increased (Ref. Reference Patel2). Thus, NEC is now the most common cause of death between days 15 and 60 (Ref. Reference Patel2). The overall incidence of NEC is 1–3 per 1000 live births (Ref. Reference Henry and Moss3), but reaches 11% in very low birth weight infants (VLBW, <1500 g) (Ref. Reference Stoll4). NEC-associated mortality has changed little over the past 50 years, ranging from 20 to 30% in confirmed cases (Ref. Reference Fitzgibbons5). Approximately 20–50% of NEC infants require surgery; mortality then rises to about 65% (Refs Reference Stoll4, Reference Blakely6, 7).

Treatment options for NEC infants are limited to bowel rest, antibiotics and supportive therapy, e.g. blood pressure management (Ref. Reference Hall, Eaton and Pierro8). Decisions on such treatment or escalation to surgery are aided by Bell's staging criteria (Refs Reference Bell9, Reference Walsh and Kliegman10) (Fig. 1). The clinical presentation of stage I NEC is largely non-specific, which explains why diagnosing NEC early is difficult. It is for this reason, and because NEC often manifests rapidly and quickly wreaks intestinal and systemic havoc that many neonatologists perceive NEC as an ever-looming spectre in NICUs.

Figure 1. Modified Bell's staging criteria for necrotising enterocolitis, adapted from (Ref. Reference Walsh and Kliegman10).

Short-term consequences of NEC include severe multisystem morbidity, leading to extended hospitalisation with all its financial and social burdens (Ref. Reference Bisquera, Cooper and Berseth11). The cost of surgically managed NEC is enormous at approximately US$200,000 per survivor in excess of the per-baby cost of routine neonatal intensive care (Refs Reference Bisquera, Cooper and Berseth11, Reference Ganapathy, Hay and Kim12).

In childhood, prior history of NEC is an independent risk factor for bowel-related chronic conditions such as diarrhoea and constipation (Ref. Reference Pike13). Similarly, neurodevelopmental issues often persist into later life and may include epilepsy, attention deficit hyperactivity disorder, cerebral palsy, deafness, blindness and compromised mental and psychomotor functions (Refs Reference Pike13, Reference Schulzke, Deshpande and Patole14, Reference Hintz15). Half of all surgically managed NEC infants develop some degree of short-bowel syndrome/intestinal failure (Ref. Reference Murthy16), and poor growth is common, particularly in extremely low birth weight (ELBW, <1000 g) NEC infants (Ref. Reference Hintz15).

NEC pathogenesis and risk factors

Prematurity

NEC incidence and severity are most strongly associated with prematurity, quantified either as low gestational age (GA) or low weight at birth (Refs Reference Luig and Lui17, Reference Buch18, Reference Guthrie19). Briefly, NEC may arise on the basis of the interactions between two poorly developed systems, namely the intestine and the immune system (Refs Reference McElroy and Weitkamp20, Reference Battersby and Gibbons21, Reference Neu22) (Fig. 2). Immaturity of intestinal motility and mucosal/barrier functions facilitates a potentially harmful composition of the microbiome and bacterial translocation (Fig. 2a). Thus confronted with bacteria, the premature immune system responds by unleashing a violent inflammatory storm (Fig. 2e) that overwhelms the extant endogenous counter-regulatory mechanisms (Fig. 2f), leading to cell death and subsequent release of intracellular components such as stored cytokines termed alarmins (Fig. 2i) (Ref. Reference Bianchi23), thus perpetuating the inflammatory storm (Fig. 2g). As described below in detail, a poorly controlled, excessive inflammatory response is one of the major factors that not only triggers the cascade that ultimately leads to NEC, but also maintains disease activity as part of a vicious cycle (Fig. 2g).

Figure 2. Model of NEC pathogenesis in the preterm intestine. (a) Multiple factors are involved in the precipitation of NEC, including dysbiosis, formula feeding, and ischaemic/hypoxic assaults. (b) Inappropriate increases in abundance of, and signalling by, pro-inflammatory pattern recognition receptors (PRRs) such as TLR4 contribute to the initiation of a cascade that involves (c) antigen processing by antigen-presenting cells such as dendritic cells (DCs) and (d) activation of other immune cells such as T cells, monocytes, macrophages and regulatory T cells (Tregs), leading to (e) an inappropriate and excessive increase of pro-inflammatory cytokines, chemokines and transcription factors. (f) A deficiency in counter-regulatory mediators contributes to this pro-inflammatory milieu to self-perpetuate and spiral out of control – (g) a vicious cycle is formed. (h) Inflammation-, ischaemia/reperfusion- and hypoxia-associated injury compromises the endothelial integrity of the local blood vessels, which also feeds the vicious cycle. (i) Necrotic cell death of the intestinal epithelium ensues, further exacerbating tissue injury and inflammation. (j) In line with the clinical stages (see Fig. 1), NEC severity can range from mild intestinal injury to segmental or even complete destruction of the intestinal epithelium. (k) Disintegration of the intestinal epithelium compromises its barrier functions, ultimately leading to rampant bacterial translocation into the lamina propria and the systemic circulation. Sepsis, multi-organ failure and death ensue. *, systemic data. #, strong evidence to be harmful only from one paper.

Formula feeding

Formula feeding is a well-established risk factor for NEC (Fig. 2a), and the incidence of NEC in infants fed their own mother's milk is reduced compared with formula-fed infants (Ref. Reference Schanler, Shulman and Lau24). Exclusive feeding with their own mother's milk was also associated with fewer episodes of late-onset sepsis and/or NEC (OR 0.18; 95% CI 0.04–0.79, P = 0.02) and shorter duration of hospital stay compared with formula- or donor breast milk-fed infants (Ref. Reference Schanler25). A meta-analysis of studies comparing formula with donor breast milk in preterm or LBW infants revealed that formula triples the risk of NEC (Ref. Reference Quigley and McGuire26). Infant formula contains components such as unbound free fatty acids (Ref. Reference Penn27) that may facilitate NEC, and is deficient in potentially protective factors such as anti-inflammatory cytokines, immunoglobulins, growth factors, and microbiota, which are present in breast milk (Refs Reference Chatterton28, Reference Castanys-Munoz, Martin and Vazquez29). Further details are discussed in the relevant sections below.

Hypoxia–ischaemia

Historically, intestinal hypoxic–ischaemic injury was considered the single most important factor initiating and perpetuating NEC, a view consistent with the predominant pathologic finding being coagulative necrosis, a common sequela of prior ischaemia (Ref. Reference Ballance30). In addition, term neonates with NEC often have conditions such as chronic heart disease that favour hypoxic or ischaemic states (Fig. 2a) (Refs Reference Lambert31, Reference Fisher32). However, no primary hypoxic–ischaemic event can be identified in most preterm infants presenting with NEC. The appearance of NEC at 2–3 weeks of age (Ref. Reference Uauy33) (when pronounced or prolonged hypoxia/ischaemia is uncommon) rather points to a role of intestinal bacterial colonisation, which is usually nearly complete by this time.

Microbial colonisation

The gut microflora plays an important role in regulating gut immune homeostasis, e.g. by dampening excessive inflammatory responses and establishing an environment ‘tolerogenic’ for commensal bacteria (Ref. Reference Maynard34). This dampening process may be disrupted in NEC because of lower microflora diversity compared with preterm controls (Ref. Reference Wang35). It is currently unclear whether the dysbiosis (Fig. 2a) that often accompanies NEC is a consequence or one of the causes of abnormal immune interactions between gut bacteria and the preterm intestine. Nevertheless, the role of the initial microbial colonisation in NEC is probably important as experimental NEC does not develop in the absence of bacteria, i.e. in germ-free piglets (Ref. Reference Sangild36) or mice treated with antibiotics (Ref. Reference Sodhi37).

Of note, animal studies have implicated Clostridium butyricum in NEC (Refs Reference Bousseboua38, Reference Waligora-Dupriet39, Reference Azcarate-Peril40), and a recent study in human infants found this bacterium in the stool of 80% of NEC infants compared with 12% of controls (Ref. Reference Cassir41). Although these findings are promising, it is too early to conclude that C. butyricum is a bacterial cause of NEC.

Animal models of NEC

Much of our understanding of NEC pathogenesis stems from animal models of the disease, with the majority using rats, mice or piglets [reviewed in (Ref. Reference Lu42)]. Most published models employ one or several of the known risk factors that induce NEC-like intestinal injury. The earliest NEC model, dating to 1974, subjected newborn rats to formula feeding and hypoxic stress (Ref. Reference Barlow43). This model is still used today, the most common variant being to subject caesarean-born preterm rats to formula feeding, hypoxia and hypothermia. Other variants of the hypoxia–hypothermia model include using caesarean-born E18.5 mice (Ref. Reference Premkumar44), naturally delivered newborn mice (Ref. Reference Tian45) and 7–10-day-old mice (Ref. Reference Good46). As newborn mice are more difficult to feed and handle than rats, the variant using 10-day-old mice is widely used today. Less commonly, murine NEC is induced using 2,4,6-trinitrobenzene sulphonic acid by gavage or enema in 10-day-old mice (Ref. Reference MohanKumar47), ablation of Paneth cells in combination with gavage feeding of Klebsiella in 14–16-day-old mice (Ref. Reference Zhang48), and by oral administration of Cronobacter sakazakii in 3-day-old mice (Ref. Reference Emami49). Rabbit and hamster NEC models are occasionally employed, but most large animal-work on NEC is conducted in piglets. The gastrointestinal tract of newborn piglets closely resembles that of human babies in terms of anatomy, physiology, development and function. Piglet NEC models commonly comprise preterm birth, parenteral nutrition and formula feeding, but no exposure to hypoxia or hypothermia. NEC can also be modelled in primates, but such research is rarely undertaken as it requires preterm delivery and care for weeks in a NICU-like setting (Ref. Reference Namachivayam50). Another rare model is the gnotobiotic quail, primarily used for investigation of the role of clostridia in NEC (Refs Reference Waligora-Dupriet39, Reference Butel51).

NEC and Immunity

The relationship between the immature preterm immune system and NEC is complex. A number of innate and adaptive immune mediators have been implicated in NEC, as summarised in Figure 3; note the distinction between local and systemic events. It is also important to keep in mind that evidence from human resection specimens is virtually always obtained from advanced NEC stages; therefore, knowledge on the intestinal events occurring in early human NEC is all but non-existent.

Figure 3. Summary of the regulation and role of immune mediators in NEC. Green, protective; Grey, inconclusive; and Red, harmful. White text, animal data; purple text, human data; yellow text, animal and human data; black text outline, functional and/or genetic data. Ig, immunoglobulin; IFNγ, interferon gamma; IL, interleukin; IL-1Ra, interleukin-1 receptor antagonist; IL-1R8, IL-1 receptor 8; IL-17R, IL-17 receptor; MyD88, myeloid differentiation factor 88; NF-κB, nuclear factor-κB; NOD-2, nucleotide-binding oligomerisation domain-containing protein 2; PAF, platelet-activating factor; PAF-AH, PAF-acetylhydrolase; RORC, RAR-related orphan receptor C; TNF, tumour necrosis factor; TRIF, toll/IL-1R domain containing adaptor inducing IFNβ; TLR, toll-like receptor; TGF-β, transforming growth factor beta; *, may be protective in NEC, but Ig supplementation has not proven effective; #, strong evidence to be harmful only from one paper.

The immune system in preterm neonates

Detailed discussion of this topic is beyond the scope of this review, but briefly: The immune system is divided into two arms, innate and adaptive immunity. The newborn relies predominantly on innate immunity during early life as maturation of adaptive immunity lags behind that of innate immunity (Ref. Reference Dowling and Levy52). Within the adaptive arm, type 2 T-cell polarisation predominates in mother and foetus, thus protecting both from graft-versus-host-type rejections, which are mediated by type 1-polarised responses (Ref. Reference Levy53). Compared with term infants, other differences include lower immune cell counts (Ref. Reference Sharma54), lower expression of major histocompatibility class II molecules (Ref. Reference Jones, Holloway and Warner55), and reduced phagocytic ability of monocytes and neutrophils (Ref. Reference Strunk56).

Innate immunity

The innate arm of immunity is phylogenetically older than adaptive immunity and functions as the first line of defence against potential pathogens. Innate immunity has two key components; a static component that consists of epithelial surfaces such as the skin and the gastrointestinal epithelium, which serve as physical barriers against microbial entry, and a reactive component, which involves tissue-resident and patrolling immune cells that are poised to respond rapidly to potential threats.

Pattern recognition receptors (PRRs)

PRRs play a central role in innate immunity, as they recognise pathogen-associated molecular patterns of invading pathogens and initiate signalling cascades that lead to target-independent inflammatory responses. As they are expressed by most cell types, PRRs perform a key function in frontline surveillance (Ref. Reference Kumar, Kawai and Akira57). Two families of PRRs, Toll-like receptors (TLRs) and Nod-like receptors (NLRs), have been implicated in NEC.

Toll-like receptors

In the intestine, TLRs are expressed by immune cells and intestinal epithelial cells (IECs) (Ref. Reference Abreu58). A fine balance is required between preventing tissue invasion by gut bacteria on the one hand and establishing tolerance of a luminal commensal, symbiotic gut flora on the other. Therefore, the function of TLRs must be tightly controlled, particularly during the transition of the newborn gut from a germ-free intrauterine environment to postnatal exposure to colonising bacteria. Of note, much of our knowledge on TLRs in NEC stems from animal experiments, and it should be kept in mind that animal and human data are not always congruent.

TLR4

Among the TLRs, TLR4 has received by far the most attention in the context of NEC. TLR4 is activated by the Gram-negative bacterial cell wall component lipopolysaccharide (LPS), a prototypical trigger of inflammation. Abundance and function of TLR4 is tightly regulated: Late in murine pregnancy (up to day 18; normal duration 21 days), Tlr4 mRNA expression increases, but rapidly decreases immediately following birth, thus adapting innate responses to the new environment (Ref. Reference Gribar59). Functionally, murine foetal IECs are significantly more responsive to LPS than IECs isolated on postnatal days 1 and 6 (Ref. Reference Lotz60). Xenografts from more immature human foetal ileum also express 3-fold more TLR4 than more mature grafts when transplanted into SCID (severe combined immunodeficiency) mice (Ref. Reference Nanthakumar61).

TLR4 gene and protein expression are elevated in the small intestinal mucosa of both human and mouse NEC compared with healthy controls (Fig. 2b) (Refs Reference Gribar59, Reference Chan62, Reference Leaphart63). This important signalling node is also target of mediators in breast milk such as soluble CD14, lactadherin, lactoferrin and 2′-fucosyllactose (Ref. Reference He, Lawlor and Newburg64). In a study in which lactating mice were milked under anaesthesia, mouse breast milk attenuated murine NEC by reducing TLR4 signalling, and overexpression of TLR4 in the intestinal epithelium reverses these protective effects (Ref. Reference Good65). In mice, excessive TLR4 expression was moreover linked to inhibition of intestinal repair, via activation of the p53-up-regulated modulator of apoptosis (Ref. Reference Neal66) as well as induction of endoplasmic reticulum (ER) stress in intestinal stem cells (Ref. Reference Afrazi67). Increased ER stress and apoptosis have been observed in the intestinal crypts of human NEC patients (Ref. Reference Afrazi67).

A pathogenic role of TLR4 in NEC appears likely, as TLR4-deficient mice (Ref. Reference Sodhi37) and mice with non-functional TLR4 (Ref. Reference Leaphart63) were protected against NEC-associated tissue damage, and a small molecule TLR4 inhibitor (C34) administered by oral gavage reduced ileal NEC injury (Ref. Reference Neal68). Interestingly, enterocyte-specific deletion of TLR4 also efficiently protected from NEC, suggesting that the epithelium participates in this aspect of the disease (Ref. Reference Sodhi37). Indeed, there is evidence that TLR4 expression in the intestinal epithelium may influence the recruitment and polarisation of T cells in the intestinal mucosa (Ref. Reference Egan69).

TLR9

Interestingly, TLR9, which recognises the characteristically CpG-rich bacterial DNA, acts as a counter-regulator of the disease-promoting effects of TLR4 in NEC (Ref. Reference Gribar59). Regulation of Tlr9 gene expression in the murine ileum is opposite to that of TLR4, so that Tlr9 decreases during late pregnancy, but increases at birth (Ref. Reference Gribar59). Mouse pups receiving two injections of 1 mg/kg CpG-DNA per day (Ref. Reference Gribar59) or once-daily oral CpG-DNA (Ref. Reference Good46) exhibited reduced NEC severity compared to vehicle-treated pups, demonstrating a functional relevance for TLR9 in NEC. Conversely, a mutation rendering TLR9 unresponsive to CpG-DNA causes increased NEC severity in mice (Ref. Reference Gribar59). Similarly, Lactobacillus rhamnosus-mediated protection in murine NEC is also dependent on TLR9 activation, as protection was abolished upon selective lentiviral knockdown of intestinal epithelial TLR9 (Ref. Reference Good46). A small human study showed that TLR9 protein abundance was reduced in NEC patients compared with controls (Fig. 2b) (Ref. Reference Gribar59); however, a protective function of TLR9 has not been confirmed in humans.

TLR5 and other TLRs

Gene expression of Tlr1, -2, -3, -6 and -7 was increased in ileal tissue of NEC rats compared with dam-fed controls, with only Tlr5 decreased (Refs Reference Liu70, Reference Le Mandat Schultz71). A NEC-associated decrease in Tlr5 is consistent with TLR5 knockout mice developing spontaneous colitis (Refs Reference Vijay-Kumar72, Reference Carvalho73). The underlying mechanism between decreased TLR5 and chronic intestinal inflammation remains unknown, but it was speculated that absence of epithelial TLR5 may reduce epithelial barrier functions and thus increase bacterial translocation (Ref. Reference Vijay-Kumar72). Alternatively, the decrease in Tlr5 mRNA may be secondary to increased TLR2 and -4 activation (Ref. Reference van Aubel74); notably, Tlr2 and -4 are elevated in resected intestinal tissue from infants with stage III NEC (Ref. Reference Chan62).

In summary, aberrantly elevated TLR4 signalling has a pathogenic role in NEC, whereas TLR9 and possibly TLR5 act as counter-regulators of TLR4. The functional relevance of other TLRs in the disease remains poorly defined.

Nucleotide-binding oligomerisation domain (NOD)-like receptors

NLRs are intracellular PRRs and are critical mediators of the assembly of the inflammasome, which converts the pro-forms of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 into their mature, active forms. Data on NLRs in NEC are scant.

NOD-2

NOD-2 is a sensor of bacterial cell-wall fragments, specifically muramyl dipeptide (MDP). NOD-2 mediates production of anti-bacterial defensins in epithelial Paneth cells (Ref. Reference Voss75) and elicits immune responses through the nuclear factor (NF)-κB pathway (Ref. Reference Philpott76). NOD-2 activity may exert protective effects in NEC as daily injections of MDP almost completely abolished NEC-associated intestinal tissue damage in mice (Ref. Reference Richardson77). Similarly, in humans, NOD-2 loss-of-function mutations has been associated with Crohn's disease (CD) (Refs Reference Hugot78, Reference Ogura79) and VLBW infant carriers of two or more NOD-2 loss-of-function alleles had an increased risk for NEC requiring surgery (OR 3.57; 95% CI 1.3–10.0, P = 0.03) (Ref. Reference Hartel80).

Mediators of innate immunity

IL-1

IL-1 is the prototypical pro-inflammatory cytokine, and is induced in numerous cell types by a wide variety of triggers. Active at picogram concentrations, IL-1 induces a plethora of inflammatory effects, including the production of other pro-inflammatory mediators, tissue damage and fever (Ref. Reference Dinarello, Simon and van der Meer81). The two isoforms, IL-1α and IL-1β, bind to the same heterodimeric cell surface receptor (Ref. Reference Dinarello, Simon and van der Meer81). Activation and release of IL-1β are tightly controlled by post-translational mechanisms such as processing by caspase-1, which in turn is regulated by the inflammasome. Therefore, data on IL1B mRNA not accompanied by protein measurements may not be indicative of biological activity and should be interpreted with great caution. IL-1 binding to its receptor triggers a signalling cascade that results in activation of pro-inflammatory transcription factors such as NF-κB and AP-1, which in turn induce pro-inflammatory cytokines such as IL-6, tumour necrosis factor (TNF) and IL-1 itself (Ref. Reference Dinarello, Simon and van der Meer81).

Studies on IL-1α in NEC are rare. In caesarean-delivered preterm piglets with NEC, lysates of the small intestine exhibited increased IL1A mRNA abundance compared to colostrum-fed controls (Refs Reference Siggers82, Reference Siggers83). This increase in IL1A expression was rapid, occurring at 8 h and persisting for up to 34 h post-NEC induction (Ref. Reference Siggers82).

IL-1β protein was elevated systemically (Ref. Reference Guven84) and in intestinal tissue in animal models of NEC (Fig. 2e) (Refs Reference Liu70, Reference Yurttutan85). In newborn rats, 48 h of formula feeding alone increased IL-1β protein in the terminal ileum 3-fold compared with dam-fed controls (Ref. Reference Liu70). Induction of NEC increased IL-1β up to 6-fold compared with dam-fed controls (Refs Reference Liu70, Reference Yurttutan85). Importantly, the authors highlighted that increases in IL-1β preceded tissue injury, which did not occur before 72 h (Ref. Reference Liu70).

In one of the few human studies on IL-1β in NEC, ileal IL1B mRNA in surgical NEC infants was more than 10-fold higher compared with GA-matched non-NEC controls (Ref. Reference Weitkamp86). Similarly, in situ hybridisation experiments showed a more than 2-fold increase in IL1B mRNA in full-thickness sections of stage III NEC infants compared with surgical controls (Ref. Reference Viscardi87). Systemically, there was no difference between the pre-operative serum IL-1β abundance in NEC babies and non-NEC controls (Ref. Reference Benkoe88). Similarly, limited time course experiments in human NEC infants beginning at NEC onset (defined by a combination of clinical and laboratory findings) and covering 8, 24, 48 and 72 h showed no significant change in serum IL-1β (Ref. Reference Edelson, Bagwell and Rozycki89). However, there was a trend towards higher IL-1β abundance in stage III infants compared with stage I and II infants (Ref. Reference Edelson, Bagwell and Rozycki89).

Overall, the available data indicate that increased IL-1 precedes NEC injury, suggesting that IL-1 aggravates tissue damage and contributes to NEC initiation and perpetuation of the vicious cycle (Fig. 2g).

IL-1 receptor antagonist (IL-1Ra)

IL-1Ra is an anti-inflammatory cytokine that functions by competitively inhibiting the binding of the two pro-inflammatory ligands IL-1α and IL-1β to their receptor. IL-1Ra is in clinical use as reviewed in (Ref. Reference Dinarello, Simon and van der Meer81), though at present not in NEC.

As IL-1Ra is one of the endogenous counter-regulatory mechanisms induced by inflammation, its abundance is often associated with disease severity in inflammatory diseases. However, the considerable increases in IL-1Ra observed in NEC (Ref. Reference Edelson, Bagwell and Rozycki89) clearly do not curtail the overwhelming inflammation that underpins NEC; perhaps IL-1Ra concentrations are insufficiently elevated in the gut where the inflammatory damage is occurring. Interestingly, IL-1Ra was decreased 2–3 weeks prior to NEC onset in buccal swabs from at-risk infants (Ref. Reference Murgas Torrazza90), suggesting a causative connection between NEC and IL-1Ra deficiency (Fig. 2f). Indeed, IL-1Ra shows promise as a NEC biomarker as described below.

Tumour necrosis factor

TNF, like IL-1, is a key pro-inflammatory cytokine that activates inflammatory mediators such as NF-κB in virtually any cell type.

TNF was increased systemically (Ref. Reference Caplan91) and in intestinal tissue (Ref. Reference Baregamian92) of NEC patients compared with non-NEC controls (Fig. 2e), but was not indicative of disease severity (Refs Reference Benkoe88, Reference Harris93, Reference Morecroft94, Reference Morecroft95). Ileal and systemic TNF were also increased in rat models of NEC (Refs Reference Sheng96, Reference Cetinkaya97, Reference Seitz98), with the mRNA rising as early as 1.5 h after the first feed (Ref. Reference Rentea99). Although others did not observe such increases in TNF (Ref. Reference Nadler100), functional data indicate a disease-promoting role for TNF. Inhibition of TNF via administration of a monoclonal anti-TNF antibody (Refs Reference Seitz98, Reference Halpern101), pentoxiphylline (Ref. Reference Travadi102), etanercept (Ref. Reference Yurttutan103) or infliximab (Ref. Reference Tayman104) significantly reduced intestinal inflammation and tissue injury in neonatal NEC rats. However, others have reported no significant improvement with pentoxiphylline in hypoxia/reperfusion-induced rabbit NEC (Ref. Reference Erdener105).

These observations suggest that TNF contributes to NEC progression, likely with a major role in the early stages of the disease. The usefulness of TNF as a biomarker in NEC appears limited.

IL-6

IL-6 is an important acute phase immune mediator; for example, it stimulates hepatocytes to produce acute-phase proteins such as C-reactive protein (CRP). In fact, both CRP and IL-6 are in clinical use as biomarkers of acute inflammation (Ref. Reference Tanaka and Kishimoto106).

It is likely that excessive IL-6 plays a pathogenic role in NEC. Genetic analysis of IL-6 single nucleotide polymorphisms (SNPs) in neonates of 32 weeks gestation or less revealed that Caucasians with IL-6 rs1800795, an SNP that is associated with increased plasma IL-6 in neonates (Ref. Reference Kilpinen107), were six times more likely to develop NEC and seven times more likely to progress to stage III disease (Ref. Reference Franklin108). These observations agree with studies that demonstrated elevated IL-6 protein (Ref. Reference Ren109) and mRNA expression (Refs Reference Chan62, Reference Chan110) in resected intestinal tissue of stage III NEC patients compared with controls (Fig. 2e). IL-6 may thus be useful as a biomarker in NEC; see the Biomarkers section.

IL-10

IL-10 is an important dampener of immune responses in the intestine, and loss of IL-10 or its receptor (IL-10R) results in early-onset inflammatory bowel disease in humans (Ref. Reference Kotlarz111) and mice (Ref. Reference Kuhn112). Although the interaction between the intestinal microbiome and immunity is not part of this review, it is interesting to note that the intestinal inflammation of IL-10-deficient mice does not develop in a pathogen-free environment (Ref. Reference Kuhn112).

IL-10 functionality in macrophages curtails intestinal inflammation, as specific knockout of IL-10R signalling in intestinal lamina propria-resident macrophages results in severe spontaneous colitis in mice (Ref. Reference Zigmond113). The number of regulatory T cells (Treg), an important source of intestinal IL-10 (Ref. Reference Uhlig114), was reduced in the ileum of NEC rats compared to dam-fed controls (Fig. 2f) (Ref. Reference Liu115). Similarly, in humans, the total number of CD4+Foxp3+ Treg and the Treg/T effector ratio was reduced in the lamina propria of surgical NEC infants compared to surgical controls (Ref. Reference Weitkamp86). Mice deficient in IL-10 exhibited more severe epithelial damage and overall NEC injury than wild-type controls (Fig. 2f) (Ref. Reference Emami116). Moreover, administration of exogenous IL-10 to IL-10-deficient mice prior to NEC induction prevented mucosal injury (Ref. Reference Emami116). IL-10 as a protective factor in NEC is supported by the observation that human breast milk contains high concentrations of bioactive IL-10 (Ref. Reference Garofalo117) and lower IL-10 abundance in breast milk correlates with increased human NEC incidence (Ref. Reference Fituch118).

However, a deficiency in IL-10 is not observed in human NEC; indeed, both serum and ileal IL-10 were markedly increased in infants diagnosed with NEC, particularly in those with advanced NEC (Refs Reference Weitkamp86, Reference Benkoe88, Reference Edelson, Bagwell and Rozycki89), which, as with IL-1Ra, is likely part of the immune system's inadequate attempt at countering the excessive inflammation. As NEC predominantly affects preterm infants, it should also be noted that prematurity does not predispose to IL-10 deficiency (Refs Reference Dembinski119, Reference Blanco-Quiros120) or inducibility by TLR agonists (Refs Reference Dembinski119, Reference Lavoie121, Reference Dirix, Vermeulen and Mascart122).

It thus appears likely that IL-10 contributes to dampening inflammation in NEC, but its precise role in NEC pathogenesis remains unclear.

Mediators of innate immune signalling

Nuclear factor-κB

NF-κB is the prototypical pro-inflammatory transcription factor, with many pathways converging at this central node of inflammatory signalling. TLR-, IL-1 receptor (IL-1R)-, and TNFR-activation trigger a cascade that leads to release of cytoplasmic NF-κB from its inhibitory protein, the inhibitor of κB (IκB), allowing NF-κB to translocate to the nucleus and to actuate the transcription of pro-inflammatory mediators, including cytokines, chemokines and leukocyte adhesion molecules (Ref. Reference Li and Verma123). Developmental regulation of NF-κB pathway components may favour NEC, e.g. a reduced abundance of IκB in foetal primary IEC compared with mature adult enterocytes (called T84 cells) (Ref. Reference Claud124).

In animals, vaginal birth may trigger a transient, low-grade increase in NF-κB activation in the small intestine, possibly allowing a tolerogenic immune surveillance of the early stages of bacterial colonisation (Ref. Reference Lotz60): NF-κB was activated in murine IECs as early as 60 min after natural birth in the absence of inflammatory stimuli (Ref. Reference Lotz60) before its activation returned to baseline by 24 h (Ref. Reference Rentea99). Conversely, NF-κB activity was nearly undetectable in the small intestine of newborn rats delivered by caesarean section (Ref. Reference De Plaen125). These findings may contribute to the unexpected observation that vaginal birth is a risk factor for early onset NEC (defined as <14 days, stage II or higher) in human preterm infants of <33 weeks GA (Ref. Reference Yee126). However, the association between vaginal birth and intestinal NF-κB activation has not been demonstrated in human infants.

On the other hand, there is clear evidence for an involvement of NF-κB in NEC. First, NEC severity was correlated with increased NF-κB activity in the epithelial cells of caesarean-born pups (Fig. 2e) (Refs Reference Le Mandat Schultz71, Reference Rentea99, Reference De Plaen125), and second, specific inhibition of NF-κB (using a NEMO-binding domain peptide) in NEC rats markedly reduces disease incidence and severity (Ref. Reference De Plaen125). Furthermore, in a human study, 100% of NEC infants were carriers of the NFKB1 variant –94delATTG, which leads to more pronounced inflammatory responses to LPS (Ref. Reference Adamzik127), compared to 65% of the non-NEC infants (Ref. Reference Sampath128).

MyD88 (myeloid differentiation factor 88), TRIF [Toll/IL-1R domain containing adaptor inducing interferon (IFN)β] and IL-1R8 (IL-1 receptor 8, previously called SIGIRR)

The first step in the TLR- and IL-1R signalling cascades is recruitment of adapter molecules to the intracellular domains of the receptors. For example, TLR4 activates two signalling pathways, one via the adapter MyD88 and one via TRIF (Ref. Reference O'Neill and Bowie129).

In concordance with the finding that TLR4-deficient mice were protected from NEC injury (Ref. Reference Sodhi37), deficiency in MyD88 (Ref. Reference Yang130) and TRIF (Ref. Reference Sodhi37) also attenuated the disease (Fig. 2b). Unexpectedly, the protection conferred by the absence of MyD88 was not as complete as that observed in mice deficient in TLR4 and TRIF, indicating an important role for TRIF-dependent signalling in NEC (Ref. Reference Sodhi37). Similarly, a deficiency in IL-1R8, which is a negative regulator of TLR- and IL-1R signalling (Refs Reference Garlanda131, Reference Nold-Petry132), may also be important as a small study associated NEC infants with stop-, missense- or splice region-IL-1R8 variants (Fig. 2b) (Ref. Reference Sampath133).

Adaptive immunity

The immune system's adaptive arm responds to highly specific antigens, which must be processed and presented, again in a highly specific fashion, by antigen-presenting cells (APC). The prototypic APC are dendritic cells (DC), which present antigens to T and B cells, the major effector cells of adaptive immunity. Such presentation results in the polarisation of naïve CD4+ T helper (Th) cells into different subsets, including Th1, Th2, Th17 and Treg, with the subset determination depending on the state of the APC, the antigen, its presentation, and the local cytokine milieu. Each subset is characterised by predominance of a transcription factor (T-bet, GATA-3, Ror-γt and Foxp3, respectively) and signature cytokines (IFNγ, IL-4, IL-17A and IL-10, respectively). Generally, the subsets antagonise each other, e.g. Th1 cytokines inhibit Th2 polarisation.

There are conflicting data on the lymphocyte fraction of the inflammatory tissue infiltrate in NEC: Whereas a lamina propria CD4+ T cell component of 30–40% in NEC mouse pups and human infants was reported (Ref. Reference Egan69), others observed a paucity of lymphocytes in the inflammatory infiltrate in human NEC infants (Refs Reference MohanKumar47, Reference Remon134). Thus, the data discussed below need to be interpreted with caution. Nevertheless, some animal studies provide evidence to support a role for CD4+ T cell influx as an important pathogenic event in NEC. For example, recombination activating gene-deficient (Rag1−/−) mice, which are deficient in functional T and B cells, exhibit significantly reduced NEC-associated intestinal injury and Il1b expression compared with wild-type controls (Ref. Reference Egan69). In addition, adoptive transfer of naïve CD4+ T cells to Rag1−/− mice prior to NEC induction restored susceptibility to severe NEC (Ref. Reference Egan69). Furthermore, transfer and repopulation of Rag1−/− mice with CD4+ T cells from wild-type mice with NEC led to intestinal damage and increased Il1b expression after 48 h (Ref. Reference Egan69). RNA sequencing of ileal samples from surgical NEC infants also revealed strongly altered T and B cell signalling in NEC compared with non-NEC preterm controls (Ref. Reference Tremblay135). Although surprisingly little information is available on the role of Th subsets in initiation and/or perpetuation of NEC, some of the signature cytokines have been investigated.

Th1 Cytokines

IFNγ

IFNγ is the signature cytokine of Th1 immune responses. It contributes to the differentiation of Th1 cells and exerts pro-inflammatory actions by inducing Th1 chemokines, activating macrophages and facilitating phagocytosis (Ref. Reference Schoenborn and Wilson136). The combined effects of IFNγ are critical to clearance of intracellular pathogens. Of note, prematurity is associated with a reduced capacity to mount Th1 responses and produce IFNγ (Ref. Reference Basha, Surendran and Pichichero137).

Whereas one human study reported no difference between peri-operative serum IFNγ in NEC infants and non-NEC controls (Ref. Reference Benkoe88), others found a 4-fold higher frequency of cells spontaneously secreting IFNγ in peripheral blood mononuclear cells (PBMCs) isolated from stage II and III NEC infants at diagnosis compared with age-matched healthy controls (Ref. Reference Abdelhamid138). Similarly, contradictory observations were made on IFNG mRNA in intestinal resection specimens (Refs Reference Ford139, Reference Pender140).

In rats and mice, the data more clearly point to a disease-promoting role for IFNγ, as ileal IFNγ protein abundance dramatically increased after induction of experimental NEC compared with dam-fed controls (Fig. 2e) (Refs Reference Liu70, Reference Leaphart141). Mechanistically, excessive IFNγ interferes with epithelial barrier integrity and regeneration, including function of intercellular gap junctions and IEC migration (two processes impaired in wild-type NEC mice but unaffected in IFNγ-deficient NEC mice) (Ref. Reference Leaphart141). Abrogation of these detrimental effects of IFNγ is likely to contribute to the observation that 10-day-old IFNγ-deficient mice are completely protected from NEC-associated ileal tissue damage (Ref. Reference Leaphart141).

IL-12

The principal function of IL-12 is to promote and maintain Th1 polarisation, for example by induction of IFNγ. Animal studies of NEC are inconclusive about IL-12, one reporting lower (Ref. Reference Nadler100), others higher (Refs Reference Coursodon-Boyiddle142, Reference Halpern143), expression. Interestingly, in human infants, reduced IL-12 abundance might be a risk factor for NEC: Preterm infants with a low bioactivity IL-12p40 promoter polymorphism exhibited a higher risk of NEC (CTCTAA allele, OR 2.9, 95% CI 1.4–6.0, P = 0.004) compared with infants with homozygous IL-12 CTCTGC alleles (Ref. Reference Bokodi144).

IL-18

IL-18 is a pleiotropic cytokine with functions in innate and adaptive immunity. In concert with IL-12, IL-18 enhances IFNγ production and promotes Th1 differentiation (Ref. Reference Akdis145).

In experimental NEC, IL-18 appears to aggravate the disease process. Ileal IL-18 protein abundance increased progressively with severity of NEC injury in rats (Fig. 2e) (Refs Reference Halpern143, Reference Halpern146). Furthermore, IL-18-deficient mice were partially protected from NEC injury (Ref. Reference Halpern147), and the protection of anti-TNF treatment was associated with reduced intestinal IL-18 protein (Ref. Reference Halpern101).

However, the available human evidence disagrees with the animal findings. Ileal IL18 mRNA was decreased in NEC infants compared with controls (Ref. Reference Weitkamp86). Similarly, a low-expression polymorphism (IL-18 A-607) was more frequent in infants with stage III NEC than in those with stage I/II (Ref. Reference Heninger148), and plasma IL-18 was moderately reduced in ELBW infants who subsequently developed NEC compared with infants that did not (Ref. Reference Maheshwari149).

Th2 Cytokines

Th2 cytokines studied in NEC include IL-4, IL-5, and IL-13. IL-4 is the signature cytokine of the Th2 subset as it promotes Th2 polarisation, suppresses Th1 responses, and induces B cell immunoglobulin class switching to IgE. The functions of IL-13 are similar to those of IL-4, including IgE class switching and activation of mast cells and eosinophils. IL-5 acts on eosinophils, promoting their activation, survival, and adhesion (Ref. Reference Akdis145). The intrauterine environment favours Th2 polarisation (Ref. Reference Levy53).

Increased ileal IL-4 and IL-5 accompanies NEC progression in rats (Ref. Reference Liu70). Similarly, in a small human study, PBMCs isolated from stage II and III NEC infants at diagnosis exhibited 3-fold more cells spontaneously secreting IL-4 than GA-matched healthy controls (Ref. Reference Abdelhamid138). However, comparing pre-operative NEC infants and GA-matched controls, serum IL-4 was not different, while IL-5 was 50% lower (Ref. Reference Benkoe88), a surprising finding as onset of NEC coincides with eosinophilia (Ref. Reference Calhoun150). Moreover, infants affected by NEC less frequently carried a high-bioactivity variant of the IL-4Rα chain (Ref. Reference Treszl151).

A marked increase in ileal IL-13 in NEC rats occurred after onset of tissue injury (Ref. Reference Liu70). Others have proposed that IL-13 protects the gut by curbing excessive IL-17 and limiting its colitogenic effects (Ref. Reference Mannon and Reinisch152). However, IL-13 also causes epithelial dysfunction such as goblet cell hyperplasia and mucus hypersecretion.

Th17 Cytokines

The Th17 signature cytokine, IL-17A, has several pro-inflammatory effects that are important for host protection against extracellular bacteria, including induction of chemokines (CXCL1, CXCL6 and CXCL10) and neutrophil recruitment and activation (Ref. Reference Akdis145). IL-23 induces Th17 polarisation, stimulates IL-17A in effector T cells, and is necessary for differentiation and effector functions of Th17 cells. Dysregulation of the Th17 pathway has been linked to inflammatory bowel diseases such as CD and ulcerative colitis (Ref. Reference Khor, Gardet and Xavier153).

Th17 responses likely also play a pathogenic role in NEC. For example, RNA sequencing has revealed remarkable similarities in the signalling pathways affected by NEC, CD and paediatric CD (Ref. Reference Tremblay135). Lamina propria CD4+ Th17 cells were more than 2-fold more abundant in NEC mice compared with controls (Ref. Reference Egan69), and intestinal IL-17A and IL-17 receptor A (IL-17RA) was increased in mouse and human NEC (Ref. Reference Egan69). These observations are in agreement with formula-fed preterm NEC baboons who exhibit a 5-fold increase in ileal IL17A gene expression compared with GA-matched non-NEC preterm controls (Ref. Reference Namachivayam50), and with ileal Il23 mRNA being 6-fold higher in NEC rats than in dam-fed controls (Refs Reference Coursodon-Boyiddle142, Reference Underwood154). Moreover, intraperitoneal injection of recombinant IL-17A in newborn mice led to loss of intercellular tight junctions in the villi, reduced enterocyte proliferation and increased crypt apoptosis (Ref. Reference Egan69). The detrimental effects of IL-17A in murine NEC were mediated by IL-17R, as these effects were abrogated by blockade of IL-17R with an antibody (Ref. Reference Egan69). Similarly, inhibition of STAT3, a critical mediator of T cell differentiation towards a Th17 phenotype, using the compound WP1066 was also protective against murine NEC; WP1066 reduced Th17 cells and increased Tregs (Ref. Reference Egan69). In fact, the balance between Tregs and Th17 cells may be critical in facilitating NEC, as one of the consequences of TLR4 deficiency was restoration of the Treg/Th17 ratio and near complete prevention of the NEC-associated intestinal infiltration of CD4+ T cells (Ref. Reference Egan69).

By contrast, systemic IL-17 was reduced in 21-day-old babies that subsequently developed NEC compared with infants that did not (Ref. Reference Maheshwari149). Likewise, there were 50% fewer of the Th17-associated intestinal intraepithelial γδ-T cells in the ileum of acute surgical NEC infants than in non-NEC controls (Ref. Reference Weitkamp155). Furthermore, expression of the Th17 transcription factor RAR-related orphan receptor C (RORC) was 10-fold less in the ileal mucosa of NEC infants compared to non-NEC controls (Ref. Reference Weitkamp155).

In summary, although a disease-promoting role for Th17 polarisation may be emerging (Ref. Reference Egan69), the data from humans frequently contradict those from animals in the field of adaptive immunity in NEC, and there are only few mechanistic studies. Moreover, the possibility that different Th subsets may dominate during different NEC stages remains poorly studied; thus, current evidence does not allow a conclusion on the relevance of Th polarisation in NEC.

Immunoglobulins

Immunoglobulins (Ig) are produced by B cells in five isotypes (IgA, IgD, IgE, IgG and IgM) and function as antibodies or receptors that target foreign invaders such as bacteria, viruses, fungi, parasites and toxins, assisting in their neutralisation in cooperation with other immune cells. Ig-mediated host defence in the gut is immature even in infants born at term and is thus temporarily dependent on Ig transfer from the mother (Ref. Reference Brandtzaeg156). Breast milk is a major source of Ig for the newborn infant and has been proposed as one of the major factors by which breast milk protects against NEC. The Ig content in infant formula is low or absent (Ref. Reference Chatterton28).

Ig supplementation was suggested as a prophylactic for NEC, with two small human studies reporting successful reduction of NEC incidence in infants orally administered either IgG alone (Ref. Reference Rubaltelli, Benini and Sala157) or a mixture of IgA and IgG (Ref. Reference Eibl158). However, in a larger study, oral supplementation of IgG alone did not reduce NEC incidence (Ref. Reference Lawrence159). A systematic review of these studies concluded that IgG or IgG+IgA demonstrated no significant reduction in incidence of definite NEC, suspected NEC, need for surgery or death from NEC in preterm and LBW infants (Ref. Reference Foster, Cole and Seth160). Similarly, a systematic review of intravenous immunoglobulin administration to preterm or LBW infants or both also did not find any statistically significant difference in the incidence of NEC (Ref. Reference Ohlsson and Lacy161). Thus, current evidence does not support the administration of oral or intravenous Ig for the prevention of NEC.

Chemokines

IL-8

IL-8 is a member of the CXC chemokine family that is produced by a variety of immune and non-immune cells (Ref. Reference Akdis145). Its main effector role is to recruit neutrophils to the site of inflammation.

The premature human gut readily produces IL-8 (Ref. Reference Nanthakumar162) and unlike in the adult immune system, IL-8 production is also a major T-cell effector function in preterm infants (Ref. Reference Gibbons163). IL8 mRNA expression was increased in intestinal resection specimens from NEC infants compared with non-NEC controls (Fig. 2e) (Refs Reference Weitkamp86, Reference Nadler164) and serum IL-8 holds substantial potential as a diagnostic marker for NEC (see the ‘Biomarkers’ section).

Other mediators

Transforming growth factor beta (TGF-β)

The biological activities of TGF-β are pleiotropic and strongly dependent on the target cell/organ and the local cytokine milieu. In the context of adaptive immunity, TGF-β may support anti- and pro-inflammatory responses, for example suppressing Th1- and Th2-polarisation and promoting Treg functions, but also inducing Th17 cell differentiation (Ref. Reference Travis and Sheppard165).

However, there is good evidence that TGF-β-deficiency promotes NEC. Disruption of TGF-β-signalling via depletion of TGF-βRII significantly increased the severity of platelet-activating factor (PAF) + LPS-induced NEC injury in 10–12-day-old mice compared with controls (Ref. Reference Maheshwari166). Moreover, tissue damage was ameliorated by enteral TGF-β2-supplementation in the PAF + LPS model and in formula-, hypoxia-, and cold stress-triggered mouse NEC (Ref. Reference Maheshwari166). Likewise, oral administration of TGF-β1 to NEC rats resulted in moderate suppression of NF-κB activation in ileal IECs and was associated with a 20% overall reduction in NEC incidence compared with vehicle-fed controls (Ref. Reference Shiou167).

Intestinal TGF-β2 bioactivity, protein abundance, and gene expression were markedly reduced in NEC patients compared to GA-matched non-NEC controls (Fig. 2f) (Ref. Reference Maheshwari166) and preterm versus term infants (Refs Reference Namachivayam50, Reference Maheshwari166). A similar TGF-β2 deficiency was observed in the intestine of formula-fed preterm baboons, and was even more pronounced in preterm baboons with NEC (Ref. Reference Namachivayam50). In fact, the protective properties of human breast milk may in part be mediated by TGF-β2, which it contains in high quantities (Ref. Reference Chatterton28).

Mechanistic studies showed that human adult PBMC-derived macrophages develop increasing LPS tolerance when exposed to media conditioned by increasingly mature intestines, an effect mediated primarily through TGF-β2 and to a lesser extent by TGF-β1 (Ref. Reference Maheshwari166). In the developing intestine, macrophage production of pro-inflammatory cytokines may thus be suppressed by TGF-β, promoting tolerogenicity to commensal bacteria (Ref. Reference Maheshwari166). This function combines with TGF-β2-mediated cytoprotection (Refs Reference Nguyen168, Reference Nguyen169), rendering TGF-β2 a key protective player in NEC.

Platelet-activating factor

PAF is a pro-inflammatory phospholipid mediator that activates pathways such as protein-kinase C (PKC), mitogen-activated protein kinases (MAPK), phosphatidylinositol 3-kinase (PI3K) and NF-κB (Refs Reference Izumi and Shimizu170, Reference De Plaen171).

Intravenous administration of PAF in adult rats causes NEC-like ischaemic necrosis in the small intestine (Ref. Reference Hsueh172). Pre-treatment with a low dose of LPS further aggravates these lesions, suggesting a synergistic effect between TLR signalling and PAF in intestinal disease (Ref. Reference Hsueh172).

Whereas a moderate elevation of circulating and stool PAF is physiological upon commencement of enteral feeding in newborn babies (Refs Reference MacKendrick173, Reference Amer174), this increase is more pronounced in formula-fed infants. Even higher PAF concentrations were observed in NEC patients compared with non-NEC controls (Fig. 2e) (Refs Reference Caplan91, Reference Amer174).

The human neonate has a reduced capability to control substantial increases in PAF as the activity of its degrading enzyme, PAF-acetylhydrolase (PAF-AH) remains low in the first few weeks of life (Fig. 2f) (Ref. Reference Caplan175). Unlike formula, breast milk contains PAF-AH, which likely contributes to breast milk-mediated protection from NEC (Ref. Reference Moya176). Although postnatally the circulating PAF-AH concentrations are similar in term and preterm infants (Ref. Reference Caplan175), in the setting of NEC, PAF-AH activity is reduced by more than 50% compared with non-NEC controls (Ref. Reference Caplan91). PAF-AH-deficient mice were more than twice as likely to develop experimental NEC than wild-type mice and exhibited a significantly higher abundance of inflammatory mediators such as CXCL1 and inducible nitric oxide synthase (Ref. Reference Lu177). A beneficial role of PAF-AH is supported by the demonstration that intravenously administered recombinant PAF-AH protected against experimental NEC injury (Ref. Reference Caplan178). Furthermore, blockade of the PAF receptor ameliorated NEC-associated tissue damage in rats (Ref. Reference Caplan179) and piglets (Ref. Reference Ewer180).

Biomarkers

Among the major challenges clinicians face in caring for infants who may have NEC, unequivocal identification of the disease in its early stages, differentiating it from sepsis or spontaneous intestinal perforation (SIP), and deciding if and when to proceed with surgery, stand out. Identifying and validating biomarkers to guide clinical decision making would represent a major advance in neonatal medicine. A recent review summarised potential biomarkers in NEC (Ref. Reference Ng, Ma and Lam181); here, we focus on promising candidates with a relevance to immunology.

The acute phase reactant CRP is widely used as a marker of inflammation in NEC and many other diseases. Whereas CRP is non-specific and cannot be used to differentiate NEC from sepsis, it may be useful to determine disease progression; for example, a persistently elevated CRP may be indicative of treatment failure, whereas normalisation may indicate success.

IL-6 may have greater sensitivity and specificity than CRP for charting NEC disease. In surgical NEC infants, serum IL-6 was up to 60-fold higher than in controls (Refs Reference Benkoe88, Reference Ren109, Reference Chan110), and was correlated with disease severity. In small studies on blood samples obtained within 48 h of NEC diagnosis (Refs Reference Morecroft94, Reference Morecroft95), IL-6 was undetectable in stage I, whereas the mean concentrations were 127 pg/ml in stage II and 3127 pg/ml in stage III patients. Post-operatively, stage III patients exhibited a decline in serum IL-6 to stage II levels, and importantly, mean IL-6 was 3-fold higher in infants that subsequently died than in survivors (Ref. Reference Morecroft95). Furthermore, pre-operative IL-6 concentrations were markedly lower in SIP, a condition that is sometimes difficult to differentiate from NEC (Refs Reference Chan62, Reference Bhatia182). Similarly, a mathematical model employing the sequential use of IL-10, IL-6 and RANTES plasma measurements predicted the development of disseminated intravascular coagulation in VLBW infants with severe sepsis and NEC (Ref. Reference Ng183). Thus, IL-6 may assist clinicians in assessing NEC disease severity and progression, and in distinguishing between NEC and SIP, but not sepsis.

Several other biomarkers of immune function and intestinal injury have been suggested to predict the progress of NEC. Higher plasma and urinary abundance of I-FABP is correlated with more severe intestinal damage, and predicted the need for surgery (Refs Reference Gregory184, Reference Edelson185, Reference Schurink186). Similarly, increased serum IL-1Ra (>130 000 pg/ml) at NEC onset was 92% specific in identifying infants whose disease subsequently progressed to stage III (Ref. Reference Edelson, Bagwell and Rozycki89). Moreover, serum IL-1Ra (>60 000 pg/ml) at NEC onset was 100% specific and 68% sensitive in classifying patients as suspected (stage I) or definite (stages II and III) NEC (Ref. Reference Edelson, Bagwell and Rozycki89). NEC patients exhibited higher serum IL-8 than healthy infants and babies with sepsis and non-inflammatory intestinal conditions (Refs Reference Benkoe88, Reference Ren109, Reference Maheshwari149, Reference Harris187).

Increased serum IL-8 at diagnosis of NEC predicted the need for surgery and correlated with 60-day mortality (Ref. Reference Benkoe188). Pre-operative serum IL-8 moreover predicted subsequent NEC severity, with 20-fold more IL-8 in pan-intestinal than in focal NEC cases (2750 versus 171 pg/ml) (Ref. Reference Benkoe189). Furthermore, compared with pre-operative abundance, serum IL-8 dropped by 60% in focal NEC, by 92% in multifocal NEC, and by 96% in pan-intestinal NEC by post-operative day 3 in infants that survived the disease; note there were no postoperative data on non-survivors, as most of them died within 24 h (Ref. Reference Benkoe189). Like IL-6, IL-8 also differentiated NEC from SIP (Ref. Reference Bhatia182).

Deficiencies in anti-inflammatory mediators such as TGF-β1 and inter-alpha inhibitor protein (IaIp) may serve as predictive biomarkers for NEC onset. ELBW infants who subsequently developed NEC exhibited low circulating TGF-β1 from the first day of life, a concentration <1380 pg/ml predicted 64% of NEC cases (Ref. Reference Maheshwari149). Similarly, low plasma IaIp differentiated between NEC and non-specific abdominal disorders (Ref. Reference Chaaban190).

Stool measurement of calprotectin also has potential for predicting NEC onset and severity (Refs Reference Yoon191, Reference Albanna, Ahmed and Awad192, Reference Bin-Nun193). However, larger studies are required to resolve the wide variations in calprotectin concentrations in faecal matter and to establish universal thresholds for NEC diagnosis (Ref. Reference Ng, Ma and Lam181).

It is common practice to combine several biomarkers into scores to achieve maximal diagnostic and predictive power. An example in NEC is the combined use of serum amyloid A and apoplipoprotein CII (ApoSAA score) that can guide the decision to initiate antibiotic treatment, as the ApoSAA score stratifies infants into low- and high-risk groups for sepsis/NEC (Ref. Reference Ng194). The LIT [liver-fatty acid binding protein, intestinal-fatty acid binding protein (I-FABP) and trefoil factor-3] score can be used to determine NEC from sepsis and, more importantly, to differentiate the need for surgery and predict chance of survival (Ref. Reference Ng195).

Clinical trials

No immune mediator or inhibitor has been tested for efficacy in treatment of NEC. The clinical trials landscape is dominated by probiotics, which are thought to restore the gut flora to its healthy, diverse state, thus indirectly modulating the preterm gut immune system towards its tolerogenic poise.

Conclusion and Outlook

Although the current data paints a complex picture of the vicious disease cycle in NEC (Fig. 2), two features stand out. First, there is a clear link between marked increases in certain pro-inflammatory mediators, including TLR4, TNF, IL-18, IFNγ, PAF, IL-6, IL-8, IL-1β, NF-κB and possibly IL-17A, in intestinal tissue on the one hand, and increased NEC severity on the other. Thus, beyond confirming that NEC occurs in the setting of excessive inflammation, research should focus on the aforementioned mediators in order to identify potential therapeutic targets. Second, it is likely that deficiencies in protective mediators such as TLR9, IL-1R8, IL-1Ra, TGFβ2, PAF-AH, and IL-10, as well as in Tregs, permit development of excessive inflammation in NEC, and thereby predispose infants to the disease.

On the biomarker front, immune mediators such as IL-1Ra, IL-6, IL-8, and TGF-β1 have emerged as promising candidates. Measurement of gut-specific markers such as I-FABP also demonstrates potential for management of NEC. Before clinical implementation, these observations need to be confirmed in larger multicentre trials.

At present we have insufficient evidence to draw a conclusion on the involvement of adaptive immunity in NEC initiation and perpetuation; however, recent data on a pathogenic role of Th17 responses and a Th17/Treg imbalance invite further exploration. On a cautionary note, findings in animal models and in the human have frequently proven contradictory, pointing to the danger inherent to relying too heavily on animal work. Furthermore, increasing the use of ‘omics’-approaches in NEC research will identify yet unknown mediators that contribute to NEC pathogenesis, and studies addressing the contribution of different cell types to the disease (e.g. IEC versus macrophages versus lymphocytes) are needed. The large datasets that are becoming increasingly available should also be mined in search for abnormalities such as SNPs or other genetic variants relevant to NEC.

While we have in recent years made progress in understanding some aspects of NEC, it is clear that a major research effort is required if an immunology-based treatment for NEC is to emerge. Only such an effort can banish the spectre of NEC, which looms over NICUs and continues to kill preterm infants.

Acknowledgements and funding

We thank Sue Panckridge for both her artistic advice on the design of the figures as well as her contributions towards the graphic elements. This work was supported by two Project Grants by the National Health and Medical Research Council (M.F.N. and C.A.N., grant numbers 1012353 and 1043845); the Marian and E.H. Flack Trust (M.F.N. and C.A.N.); the Future Leader Fellowship from the National Heart Foundation of Australia (C.A.N., CF14/3517), the ANZ-Trustees (C.A.N., CT-20681); Monash University's Larkins Fellowship (M.F.N.); the Hudson Institute's Star Recruitment Fellowship (M.F.N.); the Monash University's Faculty Postgraduate Research Scholarship (S.X.C.) and the Victorian Government's Operational Infrastructure Support Program.

Conflict of interest

None.

Further reading, resources and contacts

http://www.morgansfund.org/

The Morgan Leary Vaughan Fund for Necrotizing Enterocolitis (NEC) is an all-volunteer, public charity dedicated to promoting public awareness about NEC and the potentially devastating effects it can have on preemies and their families, and to advancing research to prevent, diagnose, treat, and ultimately, cure NEC.

http://necsociety.org/

The NEC society is a non-profit organisation that seeks to raise awareness of the risk factors of NEC in the wider community, advocate for better policies and practices to best protect preterm infants from NEC and assist in encouraging future NEC research.

http://www.preemieparentalliance.wildapricot.org/

The Preemie Parent Alliance represents a diverse set of organisations that provide information, support and resources to parents of preterm infants.

References

1. Al Tawil, K. et al. (2013) Risk factors, characteristics and outcomes of necrotizing enterocolitis in late preterm and term infants. Journal of Neonatal-Perinatal Medicine 6, 125-130 Google Scholar
2. Patel, R.M. et al. (2015) Causes and timing of death in extremely premature infants from 2000 through 2011. New England Journal of Medicine 372, 331-340 CrossRefGoogle ScholarPubMed
3. Henry, M.C. and Moss, R.L. (2009) Necrotizing enterocolitis. Annual Review of Medicine 60, 111-124 Google Scholar
4. Stoll, B.J. et al. (2010) Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics 126, 443-456 Google Scholar
5. Fitzgibbons, S.C. et al. (2009) Mortality of necrotizing enterocolitis expressed by birth weight categories. Journal of Pediatric Surgery 44, 1072-1075; discussion 75-6Google Scholar
6. Blakely, M.L. et al. (2005) Postoperative outcomes of extremely low birth-weight infants with necrotizing enterocolitis or isolated intestinal perforation: a prospective cohort study by the NICHD Neonatal Research Network. Annals of Surgery 241, 984-989; discussion 89-94Google Scholar
7. ANZNN (Australian and New Zealand Neonatal Network). (2012) Report of the Australian and New Zealand Neonatal Network 2008 and 2009. Sydney: ANZNN.Google Scholar
8. Hall, N.J., Eaton, S. and Pierro, A. (2013) Royal Australasia of Surgeons Guest Lecture. Necrotizing enterocolitis: prevention, treatment, and outcome. Journal of Pediatric Surgery 48, 2359-2367 Google Scholar
9. Bell, M.J. et al. (1978) Neonatal necrotizing enterocolitis. Therapeutic decisions based upon clinical staging. Annals of Surgery 187, 1-7 Google Scholar
10. Walsh, M.C. and Kliegman, R.M. (1986) Necrotizing enterocolitis: treatment based on staging criteria. Pediatric Clinics of North America 33, 179-201 CrossRefGoogle ScholarPubMed
11. Bisquera, J.A., Cooper, T.R. and Berseth, C.L. (2002) Impact of necrotizing enterocolitis on length of stay and hospital charges in very low birth weight infants. Pediatrics 109, 423-428 CrossRefGoogle ScholarPubMed
12. Ganapathy, V., Hay, J.W. and Kim, J.H. (2012) Costs of necrotizing enterocolitis and cost-effectiveness of exclusively human milk-based products in feeding extremely premature infants. Breastfeeding Medicine: the Official Journal of the Academy of Breastfeeding Medicine 7, 29-37 CrossRefGoogle ScholarPubMed
13. Pike, K. et al. (2012) Outcomes at 7 years for babies who developed neonatal necrotising enterocolitis: the ORACLE Children Study. Archives of disease in childhood. Fetal and Neonatal Edition 97, F318-F322 Google Scholar
14. Schulzke, S.M., Deshpande, G.C. and Patole, S.K. (2007) Neurodevelopmental outcomes of very low-birth-weight infants with necrotizing enterocolitis: a systematic review of observational studies. Archives of Pediatrics & Adolescent Medicine 161, 583-590 CrossRefGoogle ScholarPubMed
15. Hintz, S.R. et al. (2005) Neurodevelopmental and growth outcomes of extremely low birth weight infants after necrotizing enterocolitis. Pediatrics 115, 696-703 Google Scholar
16. Murthy, K. et al. (2014) Short-term outcomes for preterm infants with surgical necrotizing enterocolitis. Journal of Perinatology: Official Journal of the California Perinatal Association 34, 736-740 Google Scholar
17. Luig, M. and Lui, K. (2005) Epidemiology of necrotizing enterocolitis. Part II. Risks and susceptibility of premature infants during the surfactant era: a regional study. Journal of Paediatrics and Child Health 41, 174-179 Google Scholar
18. Buch, N.A. et al. (2001) An epidemiological study of neonatal necrotizing enterocolitis. Saudi Medical Journal 22, 231-237 Google Scholar
19. Guthrie, S.O. et al. (2003) Necrotizing enterocolitis among neonates in the United States. Journal of Perinatology: Official Journal of the California Perinatal Association 23, 278-285 Google Scholar
20. McElroy, S.J. and Weitkamp, J.H. (2011) Innate immunity in the small intestine of the preterm infant. Neoreviews 12, e517-e526 CrossRefGoogle ScholarPubMed
21. Battersby, A.J. and Gibbons, D.L. (2013) The gut mucosal immune system in the neonatal period. Pediatric Allergy and Immunology 24, 414-421 Google Scholar
22. Neu, J. (2007) Gastrointestinal development and meeting the nutritional needs of premature infants. American Journal of Clinical Nutrition 85, 629S-634S Google Scholar
23. Bianchi, M.E. (2007) DAMPs, PAMPs and alarmins: all we need to know about danger. Journal of Leukocyte Biology 81, 1-5 Google Scholar
24. Schanler, R.J., Shulman, R.J. and Lau, C. (1999) Feeding strategies for premature infants: beneficial outcomes of feeding fortified human milk versus preterm formula. Pediatrics 103(6 Pt 1), 1150-1157 Google Scholar
25. Schanler, R.J. et al. (2005) Randomized trial of donor human milk versus preterm formula as substitutes for mothers’ own milk in the feeding of extremely premature infants. Pediatrics 116, 400-406 Google Scholar
26. Quigley, M. and McGuire, W. (2014) Formula versus donor breast milk for feeding preterm or low birth weight infants. Cochrane Database of Systematic Reviews 4, CD002971 Google Scholar
27. Penn, A.H. et al. (2012) Digested formula but not digested fresh human milk causes death of intestinal cells in vitro: implications for necrotizing enterocolitis. Pediatric Research 72, 560-567 CrossRefGoogle Scholar
28. Chatterton, D.E. et al. (2013) Anti-inflammatory mechanisms of bioactive milk proteins in the intestine of newborns. International Journal of Biochemistry & Cell Biology 45, 1730-1747 Google Scholar
29. Castanys-Munoz, E., Martin, M.J. and Vazquez, E. (2016) Building a beneficial microbiome from birth. Advances in Nutrition 7, 323-330 Google Scholar
30. Ballance, W.A. et al. (1990) Pathology of neonatal necrotizing enterocolitis: a ten-year experience. Journal of pediatrics 117(1 Pt 2), S6-S13 Google Scholar
31. Lambert, D.K. et al. (2007) Necrotizing enterocolitis in term neonates: data from a multihospital health-care system. Journal of Perinatology: Official Journal of the California Perinatal Association 27, 437-443 Google Scholar
32. Fisher, J.G. et al. (2014) Serious congenital heart disease and necrotizing enterocolitis in very low birth weight neonates. Journal of the American College of Surgeons 220, 1018-1026 Google Scholar
33. Uauy, R.D. et al. (1991) Necrotizing enterocolitis in very low birth weight infants: biodemographic and clinical correlates. National Institute of Child Health and Human Development Neonatal Research Network. The Journal of Pediatrics 119, 630-638 Google Scholar
34. Maynard, C.L. et al. (2012) Reciprocal interactions of the intestinal microbiota and immune system. Nature 489, 231-241 Google Scholar
35. Wang, Y. et al. (2009) 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. ISME Journal 3, 944-954 Google Scholar
36. Sangild, P.T. et al. (2006) Diet- and colonization-dependent intestinal dysfunction predisposes to necrotizing enterocolitis in preterm pigs. Gastroenterology 130, 1776-1792 Google Scholar
37. Sodhi, C.P. et al. (2012) Intestinal epithelial Toll-like receptor 4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology 143, 708-718 e1-5CrossRefGoogle ScholarPubMed
38. Bousseboua, H. et al. (1989) Experimental cecitis in gnotobiotic quails monoassociated with Clostridium butyricum strains isolated from patients with neonatal necrotizing enterocolitis and from healthy newborns. Infection and Immunity 57, 932-936 Google Scholar
39. Waligora-Dupriet, A.J. et al. (2005) Evidence for clostridial implication in necrotizing enterocolitis through bacterial fermentation in a gnotobiotic quail model. Pediatric Research 58, 629-635 Google Scholar
40. Azcarate-Peril, M.A. et al. (2011) Acute necrotizing enterocolitis of preterm piglets is characterized by dysbiosis of ileal mucosa-associated bacteria. Gut Microbes 2, 234-243 Google Scholar
41. Cassir, N. et al. (2015) Clostridium butyricum strains and Dysbiosis linked to Necrotizing Enterocolitis in preterm Neonates. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America 61, 1107-15Google Scholar
42. Lu, P. et al. (2014) Animal models of gastrointestinal and liver diseases. Animal Models of Necrotizing Enterocolitis: Pathophysiology, Translational Relevance, and Challenges. American Journal of Physiology. Gastrointestinal and Liver Physiology 306, G917-G928 Google Scholar
43. Barlow, B. et al. (1974) An experimental study of acute neonatal enterocolitis--the importance of breast milk. Journal of Pediatric Surgery 9, 587-595 Google Scholar
44. Premkumar, M.H. et al. (2014) Argininosuccinate lyase in enterocytes protects from development of necrotizing enterocolitis. American Journal of Physiology – Gastrointestinal and Liver Physiology 307, G347-G354 Google Scholar
45. Tian, R. et al. (2010) Characterization of a necrotizing enterocolitis model in newborn mice. International Journal of Clinical and Experimental Medicine 3, 293-302 Google Scholar
46. Good, M. et al. (2014) Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: evidence in mice for a role of TLR9. American Journal of Physiology – Gastrointestinal and Liver Physiology 306, G1021-G1032 CrossRefGoogle ScholarPubMed
47. MohanKumar, K. et al. (2012) Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model. American Journal of Physiology – Gastrointestinal and Liver Physiology 303, G93-102 Google Scholar
48. Zhang, C. et al. (2012) Paneth cell ablation in the presence of Klebsiella pneumoniae induces necrotizing enterocolitis (NEC)-like injury in the small intestine of immature mice. Disease Models & Mechanisms 5, 522-532 Google Scholar
49. Emami, C.N. et al. (2012) Role of neutrophils and macrophages in the pathogenesis of necrotizing enterocolitis caused by Cronobacter sakazakii . Journal of Surgical Research 172, 18-28 Google Scholar
50. Namachivayam, K. et al. (2013) Smad7 inhibits autocrine expression of TGF-beta2 in intestinal epithelial cells in baboon necrotizing enterocolitis. American Journal of Physiology – Gastrointestinal and Liver Physiology 304, G167-G180 Google Scholar
51. Butel, M.J. et al. (1998) Clostridial pathogenicity in experimental necrotising enterocolitis in gnotobiotic quails and protective role of bifidobacteria. Journal of Medical Microbiology 47, 391-399 Google Scholar
52. Dowling, D.J. and Levy, O. (2014) Ontogeny of early life immunity. Trends in Immunology 35, 299-310 Google Scholar
53. Levy, O. (2007) Innate immunity of the newborn: basic mechanisms and clinical correlates. Nature Reviews Immunology 7, 379-390 Google Scholar
54. Sharma, A.A. et al. (2012) The developing human preterm neonatal immune system: a case for more research in this area. Clinical Immunology 145, 61-68 Google Scholar
55. Jones, C.A., Holloway, J.A. and Warner, J.O. (2002) Phenotype of fetal monocytes and B lymphocytes during the third trimester of pregnancy. Journal of Reproductive Immunology 56, 45-60 Google Scholar
56. Strunk, T. et al. (2004) Differential maturation of the innate immune response in human fetuses. Pediatric Research 56, 219-226 Google Scholar
57. Kumar, H., Kawai, T. and Akira, S. (2011) Pathogen recognition by the innate immune system. International Review of Immunology 30, 16-34 Google Scholar
58. Abreu, M.T. (2010) Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nature Reviews Immunology 10, 131-144 Google Scholar
59. Gribar, S.C. et al. (2009) Reciprocal expression and signaling of TLR4 and TLR9 in the pathogenesis and treatment of necrotizing enterocolitis. Journal of Immunology 182, 636-646 Google Scholar
60. Lotz, M. et al. (2006) Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells. Journal of Experimental Medicine 203, 973-984 Google Scholar
61. Nanthakumar, N. et al. (2011) The mechanism of excessive intestinal inflammation in necrotizing enterocolitis: an immature innate immune response. PLoS ONE 6, e17776 Google Scholar
62. Chan, K.Y. et al. (2014) Genome-wide expression profiles of Necrotizing Enterocolitis versus spontaneous intestinal perforation in human intestinal tissues: dysregulation of functional pathways. Annals of Surgery 260, 1128-1137 Google Scholar
63. Leaphart, C.L. et al. (2007) A critical role for TLR4 in the pathogenesis of necrotizing enterocolitis by modulating intestinal injury and repair. Journal of Immunology 179, 4808-4820 Google Scholar
64. He, Y., Lawlor, N.T. and Newburg, D.S. (2016) Human milk components modulate toll-like receptor-mediated inflammation. Advances in Nutrition 7, 102-111 Google Scholar
65. Good, M. et al. (2015) 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 Immunology 8, 1166-1179 Google Scholar
66. Neal, M.D. et al. (2012) Toll-like receptor 4 is expressed on intestinal stem cells and regulates their proliferation and apoptosis via the p53 up-regulated modulator of apoptosis. Journal of Biological Chemistry 287, 37296-37308 Google Scholar
67. Afrazi, A. et al. (2014) Toll-like receptor 4-mediated endoplasmic reticulum stress in intestinal crypts induces necrotizing enterocolitis. Journal of Biological Chemistry 289, 9584-9599 Google Scholar
68. Neal, M.D. et al. (2013) Discovery and validation of a new class of small molecule Toll-like receptor 4 (TLR4) inhibitors. PLoS ONE 8, e65779 Google Scholar
69. Egan, C.E. et al. (2015) Toll-like receptor 4-mediated lymphocyte influx induces neonatal necrotizing enterocolitis. Journal of Clinical Investigation Google Scholar
70. Liu, Y. et al. (2009) Changes in intestinal Toll-like receptors and cytokines precede histological injury in a rat model of necrotizing enterocolitis. American Journal of Physiology – Gastrointestinal and Liver Physiology 297, G442-G450 Google Scholar
71. Le Mandat Schultz, A. et al. (2007) Expression of TLR-2, TLR-4, NOD2 and pNF-kappaB in a neonatal rat model of necrotizing enterocolitis. PLoS ONE 2, e1102 Google Scholar
72. Vijay-Kumar, M. et al. (2007) Deletion of TLR5 results in spontaneous colitis in mice. Journal of Clinical Investigation 117, 3909-3921 Google Scholar
73. Carvalho, F.A. et al. (2012) Interleukin-1beta (IL-1beta) promotes susceptibility of Toll-like receptor 5 (TLR5) deficient mice to colitis. Gut 61, 373-384 Google Scholar
74. van Aubel, R.A. et al. (2007) Ligand-induced differential cross-regulation of Toll-like receptors 2, 4 and 5 in intestinal epithelial cells. Molecular Immunology 44, 3702-3714 Google Scholar
75. Voss, E. et al. (2006) NOD2/CARD15 mediates induction of the antimicrobial peptide human beta-defensin-2. Journal of Biological Chemistry 281, 2005-2011 Google Scholar
76. Philpott, D.J. et al. (2014) NOD proteins: regulators of inflammation in health and disease. Nature Reviews Immunology 14, 9-23 Google Scholar
77. Richardson, W.M. et al. (2010) Nucleotide-binding oligomerization domain-2 inhibits toll-like receptor-4 signaling in the intestinal epithelium. Gastroenterology 139, 904-917, 17 e1-6Google Scholar
78. Hugot, J.P. et al. (2001) Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature 411, 599-603 Google Scholar
79. Ogura, Y. et al. (2001) A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature 411, 603-606 Google Scholar
80. Hartel, C. et al. (2016) NOD2 loss-of-function mutations and risks of necrotizing Enterocolitis or focal intestinal perforation in very low-birth-weight Infants. Inflammatory Bowel Diseases 22, 249-256 Google Scholar
81. Dinarello, C.A., Simon, A. and van der Meer, J.W. (2012) Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nature Reviews Drug Discovery 11, 633-652 Google Scholar
82. Siggers, J. et al. (2011) Transition from parenteral to enteral nutrition induces immediate diet-dependent gut histological and immunological responses in preterm neonates. American Journal of Physiology – Gastrointestinal and Liver Physiology 301, G435-G445 Google Scholar
83. Siggers, J. et al. (2013) Postnatal amniotic fluid intake reduces gut inflammatory responses and necrotizing enterocolitis in preterm neonates. American Journal of Physiology – Gastrointestinal and Liver Physiology 304, G864-G875 Google Scholar
84. Guven, A. et al. (2011) Melatonin ameliorates necrotizing enterocolitis in a neonatal rat model. Journal of Pediatric Surgery 46, 2101-2107 Google Scholar
85. Yurttutan, S. et al. (2013) Protective effects of colchicine in an experimental model of necrotizing enterocolitis in neonatal rats. Journal of Surgical Research 183, 156-162 Google Scholar
86. Weitkamp, J.H. et al. (2013) Necrotising enterocolitis is characterised by disrupted immune regulation and diminished mucosal regulatory (FOXP3)/effector (CD4, CD8) T cell ratios. Gut 62, 73-82 Google Scholar
87. Viscardi, R.M. et al. (1997) Inflammatory cytokine mRNAs in surgical specimens of necrotizing enterocolitis and normal newborn intestine. Pediatric Pathology & Laboratory Medicine: Journal of the Society for Pediatric Pathology, Affiliated with the International Paediatric Pathology Association 17, 547-559 Google Scholar
88. Benkoe, T. et al. (2013) Comprehensive evaluation of 11 cytokines in premature infants with surgical necrotizing enterocolitis. PLoS ONE 8, e58720 Google Scholar
89. Edelson, M.B., Bagwell, C.E. and Rozycki, H.J. (1999) Circulating pro- and counterinflammatory cytokine levels and severity in necrotizing enterocolitis. Pediatrics 103(4 Pt 1), 766-771 Google Scholar
90. Murgas Torrazza, R. et al. (2013) Pilot study using proteomics to identify predictive biomarkers of necrotizing enterocolitis from buccal swabs in very low birth weight infants. Neonatology 104, 234-242 Google Scholar
91. Caplan, M.S. et al. (1990) Role of platelet activating factor and tumor necrosis factor-alpha in neonatal necrotizing enterocolitis. Journal of Pediatrics 116, 960-964 Google Scholar
92. Baregamian, N. et al. (2009) Tumor necrosis factor-alpha and apoptosis signal-regulating kinase 1 control reactive oxygen species release, mitochondrial autophagy, and c-Jun N-terminal kinase/p38 phosphorylation during necrotizing enterocolitis. Oxidative Medicine and Cellular Longevity 2, 297-306 Google Scholar
93. Harris, M.C. et al. (1994) Cytokine elevations in critically ill infants with sepsis and necrotizing enterocolitis. Journal of Pediatrics 124, 105-111 Google Scholar
94. Morecroft, J.A. et al. (1994) Plasma cytokine levels in necrotizing enterocolitis. Acta Paediatrica 396, 18-20 Google Scholar
95. Morecroft, J.A. et al. (1994) Plasma interleukin-6 and tumour necrosis factor levels as predictors of disease severity and outcome in necrotizing enterocolitis. Journal of Pediatric Surgery 29, 798-800 Google Scholar
96. Sheng, Q. et al. (2014) Human beta-defensin-3 promotes intestinal epithelial cell migration and reduces the development of necrotizing enterocolitis in a neonatal rat model. Pediatric Research 76, 269-279 Google Scholar
97. Cetinkaya, M. et al. (2013) CDP-choline reduces severity of intestinal injury in a neonatal rat model of necrotizing enterocolitis. Journal of Surgical Research 183, 119-128 Google Scholar
98. Seitz, G. et al. (2005) Protective effect of tumor necrosis factor alpha antibody on experimental necrotizing enterocolitis in the rat. Journal of Pediatric Surgery 40, 1440-1445 Google Scholar
99. Rentea, R.M. et al. (2013) Early enteral stressors in newborns increase inflammatory cytokine expression in a neonatal necrotizing enterocolitis rat model. European Journal of Pediatric Surgery:Official Journal of Austrian Association of Pediatric Surgery = Zeitschrift fur Kinderchirurgie 23, 39-47 Google Scholar
100. Nadler, E.P. et al. (2000) Expression of inducible nitric oxide synthase and interleukin-12 in experimental necrotizing enterocolitis. Journal of Surgical Research 92, 71-77 Google Scholar
101. Halpern, M.D. et al. (2006) Reduction of experimental necrotizing enterocolitis with anti-TNF-alpha. American Journal of Physiology – Gastrointestinal and Liver Physiology 290, G757-G764 Google Scholar
102. Travadi, J. et al. (2006) Pentoxifylline reduces the incidence and severity of necrotizing enterocolitis in a neonatal rat model. Pediatric Research 60, 185-189 Google Scholar
103. Yurttutan, S. et al. (2014) Beneficial effects of Etanercept on experimental necrotizing enterocolitis. Pediatric Surgery International 30, 71-77 Google Scholar
104. Tayman, C. et al. (2016) TNF-alpha blockade efficiently reduced severe intestinal damage in necrotizing enterocolitis. Journal of Investigative Surgery 18, 1-9 Google Scholar
105. Erdener, D. et al. (2004) Pentoxifylline does not prevent hypoxia/reoxygenation-induced necrotizing enterocolitis. An experimental study. Biology of the Neonate 86, 29-33 Google Scholar
106. Tanaka, T. and Kishimoto, T. (2014) The biology and medical implications of interleukin-6. Cancer Immunology Research 2, 288-294 Google Scholar
107. Kilpinen, S. et al. (2001) The promoter polymorphism of the interleukin-6 gene regulates interleukin-6 production in neonates but not in adults. European Cytokine Network 12, 62-68 Google Scholar
108. Franklin, A.L. et al. (2015) Are immune modulating single nucleotide polymorphisms associated with necrotizing enterocolitis? Scientific Reports 5, 18369 Google Scholar
109. Ren, Y. et al. (2005) Up-regulation of macrophage migration inhibitory factor in infants with acute neonatal necrotizing enterocolitis. Histopathology 46, 659-667 Google Scholar
110. Chan, K.Y. et al. (2012) Immunoregulatory protein profiles of necrotizing enterocolitis versus spontaneous intestinal perforation in preterm infants. PLoS ONE 7, e36977 Google Scholar
111. Kotlarz, D. et al. (2012) Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology 143, 347-355 Google Scholar
112. Kuhn, R. et al. (1993) Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75, 263-274 Google Scholar
113. Zigmond, E. et al. (2014) Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 40, 720-733 Google Scholar
114. Uhlig, H.H. et al. (2006) Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of colitis. Journal of Immunology 177, 5852-5860 Google Scholar
115. Liu, Y. et al. (2013) Lactobacillus reuteri DSM 17938 changes the frequency of Foxp3+ regulatory T cells in the intestine and mesenteric lymph node in experimental necrotizing enterocolitis. PLoS ONE 8, e56547 Google Scholar
116. Emami, C.N. et al. (2012) Role of interleukin-10 in the pathogenesis of necrotizing enterocolitis. American Journal of Surgery 203, 428-435 Google Scholar
117. Garofalo, R. et al. (1995) Interleukin-10 in human milk. Pediatric Research 37(4 Pt 1), 444-449 Google Scholar
118. Fituch, C.C. et al. (2004) Concentrations of IL-10 in preterm human milk and in milk from mothers of infants with necrotizing enterocolitis. Acta Paediatrica 93, 1496-1500 Google Scholar
119. Dembinski, J. et al. (2003) Modulation of pro- and anti-inflammatory cytokine production in very preterm infants. Cytokine 21, 200-206 Google Scholar
120. Blanco-Quiros, A. et al. (2000) Cord blood interleukin-10 levels are increased in preterm newborns. European Journal of Pediatrics 159, 420-423 Google Scholar
121. Lavoie, P.M. et al. (2010) Profound lack of interleukin (IL)-12/IL-23p40 in neonates born early in gestation is associated with an increased risk of sepsis. Journal of Infectious Diseases 202, 1754-1763 Google Scholar
122. Dirix, V., Vermeulen, F. and Mascart, F. (2013) Maturation of CD4+ regulatory T lymphocytes and of cytokine secretions in infants born prematurely. Journal of Clinical Immunology 33, 1126-1133 Google Scholar
123. Li, Q. and Verma, I.M. (2002) NF-kappaB regulation in the immune system. Nature Reviews Immunology 2, 725-734 Google Scholar
124. Claud, E.C. et al. (2004) Developmentally regulated IkappaB expression in intestinal epithelium and susceptibility to flagellin-induced inflammation. Proceedings of the National Academy of Sciences of the United States of America 101, 7404-7408 Google Scholar
125. De Plaen, I.G. et al. (2007) Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis. Pediatric Research 61, 716-721 Google Scholar
126. Yee, W.H. et al. (2012) Incidence and timing of presentation of necrotizing enterocolitis in preterm infants. Pediatrics 129, e298-e304 Google Scholar
127. Adamzik, M. et al. (2013) The NFKB1 promoter polymorphism (-94ins/delATTG) alters nuclear translocation of NF-kappaB1 in monocytes after lipopolysaccharide stimulation and is associated with increased mortality in sepsis. Anesthesiology 118, 123-133 Google Scholar
128. Sampath, V. et al. (2011) The NFKB1 (g.-24519delATTG) variant is associated with necrotizing enterocolitis (NEC) in premature infants. Journal of Surgical Research 169, e51-e57 Google Scholar
129. O'Neill, L.A. and Bowie, A.G. (2007) The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nature Reviews Immunology 7, 353-364 Google Scholar
130. Yang, G. et al. (2014) Critical role of myeloid differentiation factor 88 in necrotizing enterocolitis. Pediatric Research 75, 707-715 Google Scholar
131. Garlanda, C. et al. (2013) Negative regulatory receptors of the IL-1 family. Seminars in Immunology 25, 408-415 Google Scholar
132. Nold-Petry, C.A. et al. (2015) IL-37 requires the receptors IL-18Ralpha and IL-1R8 (SIGIRR) to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nature Immunology 16, 354-365 Google Scholar
133. Sampath, V. et al. (2015) SIGIRR genetic variants in premature infants with necrotizing enterocolitis. Pediatrics 135, e1530-4 Google Scholar
134. Remon, J.I. et al. (2015) Depth of bacterial invasion in resected intestinal tissue predicts mortality in surgical necrotizing enterocolitis. Journal of Perinatology 35, 755-762 Google Scholar
135. Tremblay, E. et al. (2016) Gene expression profiling in necrotizing enterocolitis reveals pathways common to those reported in Crohn's disease. BMC Medical Genomics 9, 6 Google Scholar
136. Schoenborn, J.R. and Wilson, C.B. (2007) Regulation of interferon-gamma during innate and adaptive immune responses. Advances in Immunology 96, 41-101 Google Scholar
137. Basha, S., Surendran, N. and Pichichero, M. (2014) Immune responses in neonates. Expert Review of Clinical Immunology 10, 1171-1184 Google Scholar
138. Abdelhamid, A.E. et al. (2011) In vitro cow's milk protein-specific inflammatory and regulatory cytokine responses in preterm infants with necrotizing enterocolitis and sepsis. Pediatric Research 69, 165-169 Google Scholar
139. Ford, H. et al. (1997) The role of inflammatory cytokines and nitric oxide in the pathogenesis of necrotizing enterocolitis. Journal of Pediatric Surgery 32, 275-282 Google Scholar
140. Pender, S.L. et al. (2003) Matrix metalloproteinases in necrotising enterocolitis. Pediatric Research 54, 160-164 Google Scholar
141. Leaphart, C.L. et al. (2007) Interferon-gamma inhibits intestinal restitution by preventing gap junction communication between enterocytes. Gastroenterology 132, 2395-2411 Google Scholar
142. Coursodon-Boyiddle, C.F. et al. (2012) Pomegranate seed oil reduces intestinal damage in a rat model of necrotizing enterocolitis. American Journal of Physiology – Gastrointestinal and Liver Physiology 303, G744-G751 Google Scholar
143. Halpern, M.D. et al. (2002) Up-regulation of IL-18 and IL-12 in the ileum of neonatal rats with necrotizing enterocolitis. Pediatric Research 51, 733-739 Google Scholar
144. Bokodi, G. et al. (2007) Association of interferon gamma T+874A and interleukin 12 p40 promoter CTCTAA/GC polymorphism with the need for respiratory support and perinatal complications in low birthweight neonates. Archives of Disease in Childhood. Fetal and Neonatal Edition 92, F25-F29 Google Scholar
145. Akdis, M. et al. (2011) Interleukins, from 1 to 37, and interferon-gamma: receptors, functions, and roles in diseases. Journal of Allergy and Clinical Immunology 127, 701-721 e1-70Google Scholar
146. Halpern, M.D. et al. (2003) Ileal cytokine dysregulation in experimental necrotizing enterocolitis is reduced by epidermal growth factor. Journal of Pediatric Gastroenterology and Nutrition 36, 126-133 Google Scholar
147. Halpern, M.D. et al. (2008) Decreased development of necrotizing enterocolitis in IL-18-deficient mice. American Journal of Physiology – Gastrointestinal and Liver Physiology 294, G20-G26 Google Scholar
148. Heninger, E. et al. (2002) Genetic variants of the interleukin-18 promoter region (-607) influence the course of necrotising enterocolitis in very low birth weight neonates. European Journal of Pediatrics 161, 410-411 Google Scholar
149. Maheshwari, A. et al. (2014) Cytokines associated with necrotizing enterocolitis in extremely-low-birth-weight infants. Pediatric Research 76, 100-108 Google Scholar
150. Calhoun, D.A. et al. (2000) Granulocyte-macrophage colony-stimulating factor and interleukin-5 concentrations in premature neonates with eosinophilia. Journal of Perinatology: Official Journal of the California Perinatal Association 20, 166-171 Google Scholar
151. Treszl, A. et al. (2003) Lower prevalence of IL-4 receptor alpha-chain gene G variant in very-low-birth-weight infants with necrotizing enterocolitis. Journal of Pediatric Surgery 38, 1374-1378 Google Scholar
152. Mannon, P. and Reinisch, W. (2012) Interleukin 13 and its role in gut defence and inflammation. Gut 61, 1765-1773 Google Scholar
153. Khor, B., Gardet, A. and Xavier, R.J. (2011) Genetics and pathogenesis of inflammatory bowel disease. Nature 474, 307-317 Google Scholar
154. Underwood, M.A. et al. (2014) Bifidobacterium longum subsp. infantis in experimental necrotizing enterocolitis: alterations in inflammation, innate immune response, and the microbiota. Pediatric Research 76, 326-333 Google Scholar
155. Weitkamp, J.H. et al. (2014) Small intestinal intraepithelial TCRgammadelta+ T lymphocytes are present in the premature intestine but selectively reduced in surgical necrotizing enterocolitis. PLoS ONE 9, e99042 Google Scholar
156. Brandtzaeg, P. (2003) Mucosal immunity: integration between mother and the breast-fed infant. Vaccine 21, 3382-3388 Google Scholar
157. Rubaltelli, F.F., Benini, F. and Sala, M. (1991) Prevention of necrotizing enterocolitis in neonates at risk by oral administration of monomeric IgG. Developmental Pharmacology & Therapeutics 17, 138-143 Google Scholar
158. Eibl, M.M. et al. (1988) Prevention of necrotizing enterocolitis in low-birth-weight infants by IgA–IgG feeding. New England Journal of Medicine 319, 1-7 Google Scholar
159. Lawrence, G. et al. (2001) Enteral human IgG for prevention of necrotising enterocolitis: a placebo-controlled, randomised trial. Lancet 357, 2090-2094 Google Scholar
160. Foster, J.P., Cole, M.J. and Seth, R. (2016) Oral immunoglobulin for preventing necrotizing enterocolitis in preterm and low birth weight neonates. Cochrane Database of Systematic Reviews 4, CD001816 Google Scholar
161. Ohlsson, A. and Lacy, J.B. (2013) Intravenous immunoglobulin for preventing infection in preterm and/or low birth weight infants. Cochrane Database of Systematic Reviews 7, CD000361Google Scholar
162. Nanthakumar, N.N. et al. (2000) Inflammation in the developing human intestine: a possible pathophysiologic contribution to necrotizing enterocolitis. Proceedings of the National Academy of Sciences of the United States of America 97, 6043-6048 Google Scholar
163. Gibbons, D. et al. (2014) Interleukin-8 (CXCL8) production is a signatory T cell effector function of human newborn infants. Nature Medicine 20, 1206-1210 Google Scholar
164. Nadler, E.P. et al. (2001) Intestinal cytokine gene expression in infants with acute necrotizing enterocolitis: interleukin-11 mRNA expression inversely correlates with extent of disease. Journal of Pediatric Surgery 36, 1122-1129 Google Scholar
165. Travis, M.A. and Sheppard, D. (2014) TGF-beta activation and function in immunity. Annual Review of Immunology 32, 51-82 Google Scholar
166. Maheshwari, A. et al. (2011) TGF-beta2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology 140, 242-253 Google Scholar
167. Shiou, S.R. et al. (2013) Oral administration of transforming growth factor-beta1 (TGF-beta1) protects the immature gut from injury via Smad protein-dependent suppression of epithelial nuclear factor kappaB (NF-kappaB) signaling and proinflammatory cytokine production. Journal of Biological Chemistry 288, 34757-34766 Google Scholar
168. Nguyen, D.N. et al. (2014) Transforming growth factor-beta2 and endotoxin interact to regulate homeostasis via interleukin-8 levels in the immature intestine. American Journal of Physiology – Gastrointestinal and Liver Physiology 307, G689-G699 Google Scholar
169. Nguyen, D.N. et al. (2015) Protective effects of transforming growth factor beta2 in intestinal epithelial cells by regulation of proteins associated with stress and endotoxin responses. PLoS ONE 10, e0117608 Google Scholar
170. Izumi, T. and Shimizu, T. (1995) Platelet-activating factor receptor: gene expression and signal transduction. Biochimica et Biophysica Acta 1259, 317-333 Google Scholar
171. De Plaen, I.G. et al. (2000) Lipopolysaccharide activates nuclear factor kappaB in rat intestine: role of endogenous platelet-activating factor and tumour necrosis factor. British Journal of Pharmacology 129, 307-314 Google Scholar
172. Hsueh, W. et al. (2003) Neonatal necrotizing enterocolitis: clinical considerations and pathogenetic concepts. Pediatric and Developmental Pathology 6, 6-23 Google Scholar
173. MacKendrick, W. et al. (1993) Increase in plasma platelet-activating factor levels in enterally fed preterm infants. Biology of the Neonate 64, 89-95 Google Scholar
174. Amer, M.D. et al. (2004) Platelet-activating factor concentration in the stool of human newborns: effects of enteral feeding and neonatal necrotizing enterocolitis. Biology of the Neonate 85, 159-166 Google Scholar
175. Caplan, M. et al. (1990) Serum PAF acetylhydrolase increases during neonatal maturation. Prostaglandins 39, 705-714 Google Scholar
176. Moya, F.R. et al. (1994) Platelet-activating factor acetylhydrolase in term and preterm human milk: a preliminary report. Journal of Pediatric Gastroenterology and Nutrition 19, 236-239 Google Scholar
177. Lu, J. et al. (2010) Dual roles of endogenous platelet-activating factor acetylhydrolase in a murine model of necrotizing enterocolitis. Pediatric Research 68, 225-230 Google Scholar
178. Caplan, M.S. et al. (1997) The role of recombinant platelet-activating factor acetylhydrolase in a neonatal rat model of necrotizing enterocolitis. Pediatric Research 42, 779-783 Google Scholar
179. Caplan, M.S. et al. (1997) The platelet-activating factor receptor antagonist WEB 2170 prevents neonatal necrotizing enterocolitis in rats. Journal of Pediatric Gastroenterology and Nutrition 24, 296-301 Google Scholar
180. Ewer, A.K. et al. (2004) The role of platelet activating factor in a neonatal piglet model of necrotising enterocolitis. Gut 53, 207-213 Google Scholar
181. Ng, P.C., Ma, T.P. and Lam, H.S. (2015) The use of laboratory biomarkers for surveillance, diagnosis and prediction of clinical outcomes in neonatal sepsis and necrotising enterocolitis. Archives of Disease in Childhood. Fetal and Neonatal Edition 100, F448-F452 Google Scholar
182. Bhatia, A.M. et al. (2014) Cytokine levels in the preterm infant with neonatal intestinal injury. American Journal of Perinatology 31, 489-496 Google Scholar
183. Ng, P.C. et al. (2006) Early prediction of sepsis-induced disseminated intravascular coagulation with interleukin-10, interleukin-6, and RANTES in preterm infants. Clinical Chemistry 52, 1181-1189 Google Scholar
184. Gregory, K.E. et al. (2014) Urinary intestinal fatty acid binding protein predicts necrotizing enterocolitis. Journal of Pediatrics 164, 1486-1488 Google Scholar
185. Edelson, M.B. et al. (1999) Plasma intestinal fatty acid binding protein in neonates with necrotizing enterocolitis: a pilot study. Journal of Pediatric Surgery 34, 1453-1457 Google Scholar
186. Schurink, M. et al. (2015) Intestinal fatty acid-binding protein as a diagnostic marker for complicated and uncomplicated necrotizing enterocolitis: a prospective cohort study. PLoS ONE 10, e0121336 Google Scholar
187. Harris, M.C. et al. (2005) Cytokine elaboration in critically ill infants with bacterial sepsis, necrotizing entercolitis, or sepsis syndrome: correlation with clinical parameters of inflammation and mortality. Journal of Pediatrics 147, 462-468 Google Scholar
188. Benkoe, T. et al. (2014) Interleukin-8 predicts 60-day mortality in premature infants with necrotizing enterocolitis. Journal of Pediatric Surgery 49, 385-389 Google Scholar
189. Benkoe, T. et al. (2012) Interleukin 8 correlates with intestinal involvement in surgically treated infants with necrotizing enterocolitis. Journal of Pediatric Surgery 47, 1548-1554 Google Scholar
190. Chaaban, H. et al. (2010) Inter-alpha inhibitor protein level in neonates predicts necrotizing enterocolitis. Journal of Pediatrics 157, 757-761 Google Scholar
191. Yoon, J.M. et al. (2014) Fecal calprotectin concentration in neonatal necrotizing enterocolitis. Korean Journal of Pediatrics 57, 351-356 Google Scholar
192. Albanna, E.A., Ahmed, H.S. and Awad, H.A. (2014) Stool calprotectin in necrotizing enterocolitis. Journal of Clinical Neonatoloy 3, 16-19 Google Scholar
193. Bin-Nun, A. et al. (2015) Rapid fecal calprotectin (FC) analysis: point of care testing for diagnosing early necrotizing enterocolitis. American Journal of Perinatology 32, 337-342 Google Scholar
194. Ng, P.C. et al. (2010) Host-response biomarkers for diagnosis of late-onset septicemia and necrotizing enterocolitis in preterm infants. Journal of Clinical Investigation 120, 2989-3000 Google Scholar
195. Ng, E.W. et al. (2013) Gut-associated biomarkers L-FABP, I-FABP, and TFF3 and LIT score for diagnosis of surgical necrotizing enterocolitis in preterm infants. Annals of Surgery 258, 1111-1118 Google Scholar
Figure 0

Figure 1. Modified Bell's staging criteria for necrotising enterocolitis, adapted from (Ref. 10).

Figure 1

Figure 2. Model of NEC pathogenesis in the preterm intestine. (a) Multiple factors are involved in the precipitation of NEC, including dysbiosis, formula feeding, and ischaemic/hypoxic assaults. (b) Inappropriate increases in abundance of, and signalling by, pro-inflammatory pattern recognition receptors (PRRs) such as TLR4 contribute to the initiation of a cascade that involves (c) antigen processing by antigen-presenting cells such as dendritic cells (DCs) and (d) activation of other immune cells such as T cells, monocytes, macrophages and regulatory T cells (Tregs), leading to (e) an inappropriate and excessive increase of pro-inflammatory cytokines, chemokines and transcription factors. (f) A deficiency in counter-regulatory mediators contributes to this pro-inflammatory milieu to self-perpetuate and spiral out of control – (g) a vicious cycle is formed. (h) Inflammation-, ischaemia/reperfusion- and hypoxia-associated injury compromises the endothelial integrity of the local blood vessels, which also feeds the vicious cycle. (i) Necrotic cell death of the intestinal epithelium ensues, further exacerbating tissue injury and inflammation. (j) In line with the clinical stages (see Fig. 1), NEC severity can range from mild intestinal injury to segmental or even complete destruction of the intestinal epithelium. (k) Disintegration of the intestinal epithelium compromises its barrier functions, ultimately leading to rampant bacterial translocation into the lamina propria and the systemic circulation. Sepsis, multi-organ failure and death ensue. *, systemic data. #, strong evidence to be harmful only from one paper.

Figure 2

Figure 3. Summary of the regulation and role of immune mediators in NEC. Green, protective; Grey, inconclusive; and Red, harmful. White text, animal data; purple text, human data; yellow text, animal and human data; black text outline, functional and/or genetic data. Ig, immunoglobulin; IFNγ, interferon gamma; IL, interleukin; IL-1Ra, interleukin-1 receptor antagonist; IL-1R8, IL-1 receptor 8; IL-17R, IL-17 receptor; MyD88, myeloid differentiation factor 88; NF-κB, nuclear factor-κB; NOD-2, nucleotide-binding oligomerisation domain-containing protein 2; PAF, platelet-activating factor; PAF-AH, PAF-acetylhydrolase; RORC, RAR-related orphan receptor C; TNF, tumour necrosis factor; TRIF, toll/IL-1R domain containing adaptor inducing IFNβ; TLR, toll-like receptor; TGF-β, transforming growth factor beta; *, may be protective in NEC, but Ig supplementation has not proven effective; #, strong evidence to be harmful only from one paper.