New Frontiers of Necrotizing Enterocolitis: From Pathophysiology to Treatment

Abstract

Necrotizing enterocolitis [NEC] is an inflammatory disease of intestine largely occuring in preterm infants with a wide range of damage from minimal injury limited to mucosa to extensive necrosis of bowel wall and perforation. Despite advancements in neonatal care, mortality remains high [30% - 50%] and controversy still persists with regards to the most appropriate management of neonates with necrotizing enterocolitis. The main factors thought to be involved in the pathogenesis of NEC are: relatively hyper-reactive state of premature intestine, enteral feeding and bacterial colonization. In this review, we discuss current knowledge about the epidemiology, pathophysiology, imaging, medical and surgical management of necrotizing enterocolitis and describe novel strategies for prevention and treatment.

Share and Cite:

Zubarioglu, U. , Uslu, S. and Bulbul, A. (2017) New Frontiers of Necrotizing Enterocolitis: From Pathophysiology to Treatment. Health, 9, 106-123. doi: 10.4236/health.2017.91008.

1. Introduction

Necrotizing enterocolitis [NEC] is an inflammatory disease of intestine largely occur in preterm infants with a wide range of damage from minimal injury limited to mucosa to extensive necrosis of bowel wall and perforation. Necrotizing enterocolitis is the most common reason of death originated from intestinal tract in preterm babies especially for very low birth weight [VLBW] infants [1] . Although early recognition and aggressive treatment of this disorder has im- proved clinical outcomes, NEC accounts for substantial long-term morbidity in survivors of neonatal intensive care, particularly in VLBW infants.

Here, we will discuss current knowledge about epidemiology, pathophysiology, management strategies and describe new strategies about prevention and treat- ment.

2. Epidemiology

Worldwide, large multicentre studies coordinated by neonatal research networks have determined the incidence of NEC to be up to 13% among infants born ≤33 weeks of gestation or whose birth weight is ≤2500 g [1] - [9] . Although the ma- jority of cases of NEC occur among premature infants, a small subset of babies born at term or ≥35 weeks of gestation develop NEC-like gastrointestinal signs and symptoms, frequently in association with other conditions such as congeni- tal heart disease, perinatal asphyxia, polycythemia, sepsis, and respiratory disease [10] .

3. Pathogenesis

The main factors thought to be involved in the pathogenesis of NEC are: rela- tively hyper-reactive state of premature intestine, enteral feeding and intestinal microflora.

3.1. Hyper-Reactive State of Premature Intestine

To understand the reasons for why preterm babies are at a particularly high risk of developing NEC compared with term babies, researches have focused on identifying the differences between the premature and the full-term intestinal tract. These studies have shown significant differences in bacterial colonization, microcirculatory perfusion and the maturity of the innate immune system of gut [11] [12] . These differences can provide and explain us the multi-factorial pathogenesis of NEC. Major difference found in studies was expression of Toll- like receptor 4 [TLR4] at higher levels in the premature than the full-term intestine in mice, humans and other species [13] . The elevated expression of TLR4 in the premature gut is reflective to function of TLR4 exhibits in the regulation of normal gut development [13] . In the premature birth situation, intestinal TLR4 levels remain elevated and activation of TLR4 on the lining of the premature intestine by the Gram negative bacteria that colonize the pre- mature gut leads to a number of deleterious effects, including increased entero- cyte apoptosis, impaired mucosal healing and enhanced proinflammatory cyto- kine release, which in aggregate lead to the development of NEC [14] [15] . Also, the translocation of Gram-negative bacteria through the gut mucosa leads to activation of TLR4 on the lining of the endothelium of the premature bowel mesentery, resulting in a reduction in blood flow and the development of intes- tinal ischaemia and necrosis [16] . This explanation for the pathogenesis of NEC termed as “the cross-switching hypothesis” partially explains the reasons for which the premature infant is at risk of NEC development and why the disease occurs upon bacterial colonization.

Also additional factors are known to differ between the premature and full- term host that might contribute to this disease.

・ high baseline level of cellular endoplasmic reticulum stress within the prema- ture intestine which increases the likelihood of apoptosis in the epithelial lining [17] .

・ the decreased number of mucus-producing goblet cells in the premature intestine results in deficient mechanical protection [13] [18] .

・ the impaired clearance of luminal contents, owing to decreased motility [19] [20] [21] [22] .

・ decreased digestion and absorption as a result of enterocyte immaturity [23] [24] .

・ increased microvascular tone within the preterm intestinal mesentery [16] [25] .

・ presence of immature tight junctions [26] [27] .

All of these factors can render the bowel at risk of proinflammatory signalling, bacterial translocation and NEC development [19] [25] [28] . Notably, some of these important factors are linked to TLR4 signalling. Furthermore, T lym- phocytes have been shown to participate in the adaptation of the premature intestinal mucosa to bacterial colonization and contribute to NEC development [29] [30] . NEC is associated with lymphocyte imbalance within the intestinal mucosa, as TLR4 signalling in the intestinal epithelium leads to an upregulation of proinflammatory T helper 17 cells and a reduction in protective T regulatory cells [30] . Specifically, various investigators have identified roles for the in- creased expression and function of platelet-activating factor in the mucosal injury and barrier dysfunction associated with NEC [28] [31] [32] . Infants with NEC have high circulating levels of platelet-activating factor associated with the increased expression of this protein as well as with deficient activity of platelet- activating factor acetylhydrolase, the enzyme involved in its degradation [31] [33] [34] . Additionally, platelet-activating factor has been demonstrated to induce TLR4 expression and signalling [28] [35] .

3.2. Enteral Feeding

There have been 2 conditions known about relationship between enteral feeding and NEC; which are predominance of enterally fed premature babies in NEC epidemiology and protective effects of human milk against NEC [36] . A wide range of protective ingredients present in breast milk has been appointed and some part of them have been found as promising agents in NEC prevention and treatment [37] . Recent researches suggested the epigenetic effects of diet type on intestinal genomic structure. Human milk and other enterally given products change gene expression especially with methylation [38] . In studies performed in preterm infants and pig models, it was shown that proinflamatory genes became upregulated by enteral feeding [39] [40] . Also important risk factors in NEC pathogenesis such as intestinal microflora and intestinal splanhcnic perfusion has mutual interaction with enteral feeding type [41] [42] .

3.3. Intestinal Microflora

How bacterial pathogens involve in NEC pathophysiology still unclear but studies showed that they got interaction with several ways [36] . The most important evidence about bacterial involvement is occurence of NEC as out- breaks occasionally with the grown of same organisms from babies’ cultures and such clusters of cases got controlled with the start of infection control measures [43] [44] . However, differents centers reported separate microorganisms are grown in their outbreaks so someone can not claimed NEC development de- dicated to a distinct bacterial agent. Also researchers found that endotoxinemia, blood culture growns by bacteria and 30% hydrogen content of pneumatosis [an unique gas produced only by bacteria] in their studies which were demonstrate association of bacteria with NEC pathogenesis [45] [46] [47] .

Earlier studies about NEC pathogenesis suggested the “dysbiosis” hypothesis [48] . Even so, recent studies claimed that bacterial diversity in microflora of gut disappeared just before beginning of NEC with following domination of path- ogenic bacteria [49] [50] .

4. Diagnosis

The cornerstone of effective NEC treatment relies on accurately diagnosing the disease, which can usually be established on the basis of readily available clinical, radiographic and laboratory data. The typical neonate with NEC is a premature infant who is thriving, yet suddenly presents with feeding intolerance, abdo- minal distension, bloody stools and signs of sepsis [11] [51] . For descriptive purposes and for disease stratification, the Bell scoring system has been widely utilized, which assesses the degree of NEC severity as mild [Bell stage I], mode- rate [Bell stage II] or severe [Bell stage III], as shown in Table 1.

Biomarkers and Noninvasive Testing for the Diagnosis of NEC

The relative nonspecificity of the readily available clinical and radiographic tests suggest the need for additional molecular markers to improve early diagnosis of NEC in premature infants. In this regard, the presence of several molecules that are detected in the blood have been assessed for their value in establishing the diagnosis of NEC and a number of them have shown considerable promise, including acute-phase reactants [such as C-reactive protein] and proinflammatory cytokines [for example, TNFα, IL-6 and IL-8] which were found as nonspecific [55] [56] [57] .

In addition, organ-specific biomarkers, such as those that would indicate enterocyte injury or intestinal barrier impairment, include intestinal fatty acid- binding protein, liver fatty acid-binding protein, faecal calprotectin studied for early identicifation of NEC [58] [59] . Among these circulating molecules, one of the most promising might be intestinal fatty acid-binding protein, a cytoplasmic protein involved in enterocyte lipid metabolism that is released into circulation and secreted into the urine after enterocyte damage, which has been suggested to be useful in the prediction of NEC development and to correlate with the extent of intestinal necrosis [53] [60] . Neverthless, I-FABP has handicaps in detection of NEC in early stages. Plasma half life is short and normal values in healthy preterm babies are variable that limits the use of I-FABP [36] . Also some NEC cases with extensive necrosis with ongoing damage can come against us with low

Table 1. Modified Bell staging criteria for necrotizing enterocolitis [52] [53] [54] .

I-FABP levels paradoxically because of this short half-life [53] [61] . In recent researches about biomarkers for NEC discovered novel urinary peptides and proteins which are linked to poor prognosis [62] [63] .

Although plain radiology still accepted as the primary imaging procedure for diagnosis and staging the infants with NEC some new modalities especially ultrasound [US] has attracted research interest [64] [65] . Abdominal US can detect both pneumatosis intestinalis and portal venous gas [PVG] which is earlier than plain radiography especially for PVG [66] . Also US gives more details about perfusion and thickness of gut wall and motility from plain radio- graphs which may detect babies with more advanced disease and those who may benefit from surgery [67] [68] .

Further information that can be obtained from ultrasound also includes the presence of free intraabdominal gas and the presence and nature of any free intraabdominal fluid that may be indicative of intestinal perforation [36] .

Doppler ultrasonography can be used for evaluating coeliac trunk and superior mesenteric artery blood flow velocity, which can demonstrate cases at risk of necrotizing enterocolitis with poor perfusion, also for assessment of the in- testinal wall viability in patients with NEC [54] [67] [68] [69] [70] .

Near infra-red spectroscopy [NIRS] has got research interest recently which may be useful for risk prediction of NEC, separation of cases with NEC from those without and detection of advanced NEC [71] [72] . Important limitation for NIRS is placement of probes on the skin and tissue penetration has short depth. So, measures of NIRS may give only oxygenation of underlying intestine of probes rather than entire intestine. On the other hand, NIRS measures can be use to detect differences in splanchnic tissue oxygenation in preterm babies who subsequently developed NEC and those who did not [71] [72] [73] .

5. Prevention

Given that NEC occurs in a well-defined population of patients who are pre- mature, there might be benefit in identifying specific preventive strategies that, if administered successfully to the appropriate patients, could reduce the inci- dence of NEC. In this regard, there has been tremendous interest in developing specific nutritional and pharmacological strategies to reduce the incidence of NEC.

5.1. The Use of Breast Milk

Multiple randomized clinical trials have now validated the empirical observation that breast milk statistically significantly reduces the incidence of NEC [74] [75] . Human milk contains a variety of beneficial bioactive factors, among which several have been shown to reduce NEC incidence and progression [74] [76] .

In Table 2, a list of human milk components which had protective effects against NEC presented.

Considerable research efforts have been deployed to identify these critical factors in the hope that new preventive strategies can be developed [75] . Although the precise mechanisms by which breast milk protects against NEC are not yet fully understood, emerging experimental evidence suggests that breast milk inhibits TLR4 signalling by preventing glycogen synthase kinase 3β activity [77] . Consequently, breast milk-mediated downregulation of TLR4 signalling can reverse the inhibition of intestinal stem cell proliferation and mucosal healing, which are themselves inhibited by TLR4 [77] [78] . Moreover, these effects were shown to be partially dependent upon activation of epidermal growth factor

Table 2. Protective factors of human milk against NEC.

receptor signalling [77] .

5.2. Donor Milk

The lack of availability of human breast milk [which can arise for a number of reasons, such as insufficient production by the mother of an infant remains a major challenge in neonatal care and has led to the use of donor breast milk as a potential substitute or supplement to formula-feeding [74] [79] . Multiple reports support the use of donor human milk as a potentially effective strategy for re- ducing the incidence of NEC [80] [81] .

5.3. Probiotics in the Prevention of NEC

Probiotics are defined as live microorganisms that provide a health benefit to the host. These agents have been shown to protect against NEC and reduce disease severity and overall mortality in premature infants [82] [83] . The finding that a degree of perturbation in the normal gut microbial flora exists in patients with NEC supports a rationale of using probiotics to treat and prevent this disease [84] [85] [86] . Considering the vulnerability of premature infants, routine admi- nistration of probiotic agents has elicited substantial controversy regarding the type of agent to be used, dosing and timing [83] [87] . A systematic review eva- luated the efficacy and safety of probiotics for preventing NEC and suggested that oral administration of probiotics decreases all-cause mortality and incidence of severe NEC in preterm infants; however, the precise probiotic agent, timing and length of therapy still remains to be established [83] [88] . Emerging con- sensus is that the use of probiotics in NEC could be effective in reducing the incidence of the disease without increasing rates of sepsis or other adverse events [88] [89] [90] [91] .

Administration of the probiotic bacteria Lactobacillus rhamnosus was shown to increase enterocyte proliferation and differentiation of Paneth cells in enteroids grown in a 3D bioscaffold [92] . Furthermore, treatment with CpG-containing bacterial DNA, which bypasses the potential adverse effects of live bacteria, is effective against experimental NEC in mice and piglets, and acts by activating Toll-like receptor 9 and inhibiting TLR4, providing a potential alternative to the use of live probiotics [93] .

6. Treatment

6.1. Medical Supportive Management

Most infants with Bell’s stage I or stage II NEC are managed with appropriate supportive therapies which includes cessation of enteral nutrition, support of ventilation, stabilization of fluid-electrolyte and acid-base balances, correction of ongoing coagulopathy and/or thrombocytopenia, bowel rest, and antibiotics [54] . About which antibiotics should be used for NEC, there is still no sufficient evidence and concencus in literature and this confusion was demonstrated by both an international survey and a Cochrane review [94] [95] . So, antibiotics may be ordered according to protocol of center and regulated according to growns in cultures and sensitivity results of the case. Similarly, the optimal duration time for withholding enteral feds and bowel rest is not on evidence- based treatment and based on institutional approach [94] .

6.2. Surgical Management

Surgical intervention is required in up to 50% of the NEC cases in large, po- pulation-based and hospital-based multicentre studies coordinated by neonatal research networks and typically includes the removal of necrotic intestine [5] [8] [9] [96] [97] . In rare cases, the placement of a peritoneal drain and abdominal irrigation might be sufficient. Although several studies have reported that pa- tients undergoing peritoneal drainage and laparotomy could have similar out- comes [87] [94] [95] . Several surgical guidelines have been published [89] [98] [99] [100] . Given that up to 74% of infants initially managed with peritoneal drainage will require a subsequent laparotomy [96] [99] a commonly accepted approach has been to reserve primary peritoneal drainage for those patients with substantially elevated intra-abdominal pressure that impairs ventilation, or for extremely small infants under 750 g.

6.3. New Medical Treatments in Research

In consequence of the role of hyper-reactive and immature immune system in NEC etiopathogenesis; research studies focused on new treatment agents that regulate the immune response. Pentoxifylline is one of these agents studied but a very weak evidence found for usage as combination therapy to antbiotics in neonatal sepsis in a Cochrane review with a relative risk of NEC of 0.62 [101] [102] . In another study which intra-peritoneal pentoxifylline used in neonatal rat model, it was found that NEC incidence and severity reduced [103] .

6.4. Promising New Agents

・ Stem cells; Recent evidences came from research groups suggested that am- niotic fluid stem cells [AFS], mesenchymal stem cells and enteric neural stem system cells have potential to change the trend of the NEC in experimental models [104] [105] [106] [107] [108] .

・ Amniotic fluid; In experimental animal models, in vitro proliferation and migration of gut epithelial cells was provided by both porcine and human amniotic fluid [109] . In another study which amniotic fluid used in postnatal minimal enteral feeding of preterm piglets; NEC incidence, severity and inflammation decreased [110] . In a mouse model, it was shown that NEC severity also decreased with amniotic fluid given enterally which contains epidermal growth factor [EGF] and its receptor as major factor in this effect [111] .

・ Growth Factors; Heparin-binding EGF-like growth factor has been identified as a biologic agent capable of preventing NEC in various animal models, and of reversing the effects of established NEC, via positive effects on mu- cosal healing, intestinal stem cell function and vascular perfusion [112] [113] [114] .

・ TLR4 inhibitor; nontoxic oligosaccharide that inhibits TLR4 was shown to prevent NEC in mice and piglets and to reduce intestinal inflammation in ex vivo human intestine obtained during the treatment of NEC [32] .

・ Human milk oligosaccharides; established an important role for in NEC prevention and treatment [115] [116] .

・ Lactoferrin; Emerging evidence also suggests a prophylactic benefit against the development of NEC by oral administration of lactoferrin with or with- out probiotics to preterm infants at risk of NEC [gestational age <32 weeks or birth weight <1500 g] [75] .

7. Outcome

Despite advancement in medical and surgical treatment over the last decades, the average mortality from NEC is 20% - 30%, with mortality as high as 50% in those infants requiring surgical management [117] . Necrotizing enterocolitis has also severe morbidities affects survived infants which are originated from intestinal or systemic insults [54] . These complications include;

・ Relapsing of NEC; About 10% of babies who had undergone surgery for NEC develop a relapsing episode and this causes long-term parenteral nutrition dependency [54] .

・ Intestinal strictures; About 25% of patients who had NEC and especially treated surgically, can develop one or more intestinal strictures [54] .

・ Gut failure; İnfants who had undergone surgery for NEC will develop in- testinal failure with high proportion and it depends on many factors (such as low birth weight, antibiotic use, ventilator use, and greater extent of bowel resection) also associated with NEC development [54] .

・ Parenteral nutrition associated complications

・ Neurodevelopmental disturbances; seen in the nearly half of neonates, but by which mechanisms this develops is stil not identified clearly [118] [119] . It is found that infants with NEC had white matter anomalies on magnetic resonance imaging at term which increases the risk for motor impairment [120] .

8. Conclusion

NEC is the most common and lethal gastrointestinal pathology that affects pre- mature infants. Characterized by high morbidity and mortality, complex patho- genesis and devastating short-term and long-term sequelae. Only within the past decade have substantial strides been made in the understanding of the molecular mechanisms that determine NEC pathogenesis. These advances undoubtedly hold the promise to improve the development of effective preventive and diag- nostic strategies to curtail the devastating consequences of the disease.

Conflict of Interests

The authors have indicated they have no financial relationships and conflict of interest relevant to this article to disclose.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Stoll, B.J., Hansen, N.I., Bell, E.F., et al. (2015) Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Trends in Care Practices, Morbidity, and Mortality of Extremely Preterm Neonates, 1993-2012. JAMA, 314, 1039-1051.
https://doi.org/10.1001/jama.2015.10244
[2] Holman, R.C., Stoll, B.J., Curns, A.T., et al. (2006) Necrotising Enterocolitis Hospitalisations among Neonates in the United States. Paediatric and Perinatal Epidemiology, 20, 498-506.
https://doi.org/10.1111/j.1365-3016.2006.00756.x
[3] Guillet, R., Stoll, B.J., Cotten, C.M., Gantz, M., McDonald, S., Poole, W.K. and Phelps, D.L. (2006) National Institute of Child Health and Human Development Neonatal Research Network. Association of H2-Blocker Therapy and Higher Incidence of Necrotizing Enterocolitis in Very Low Birth Weight Infants. Pediatrics, 117, 137-142.
https://doi.org/10.1542/peds.2005-1543
[4] Horbar, J.D., Badger, G.J., Carpenter, J.H., Fanaroff, A.A., Kilpatrick, S., LaCorte, M., Phibbs, R. and Soll, R.F. (2002) Members of the Vermont Oxford Network. Trends in Mortality and Morbidity for Very Low Birth Weight Infants, 1991-1999. Pediatrics, 110, 143-151.
https://doi.org/10.1542/peds.110.1.143
[5] Yee, W.H., Soraisham, A.S., Shah, V.S., et al. (2012) Incidence and Timing of Presentation of Necrotizing Enterocolitis in Preterm Infants. Pediatrics, 129, 298-304.
https://doi.org/10.1542/peds.2011-2022
[6] Luig, M., Lui, K., NSW & ACT NICUS Group (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.
https://doi.org/10.1111/j.1440-1754.2005.00583.x
[7] Luig, M., Lui, K., NSW & ACT NICUS Group (2005) Epidemiology of Necrotizing Enterocolitis—Part I: Changing Regional Trends in Extremely Preterm Infants over 14 Years. Journal of Paediatrics and Child Health, 41, 169-173.
https://doi.org/10.1111/j.1440-1754.2005.00582.x
[8] Sankaran, K., Puckett, B., Lee, D.S., et al. (2004) Variations in Incidence of Necrotizing Enterocolitis in Canadian Neonatal Intensive Care Units. Journal of Pediatric Gastroenterology and Nutrition, 39, 366-372.
https://doi.org/10.1097/00005176-200410000-00012
[9] Stoll, B.J., Hansen, N.I., Bell, E.F., et al. (2010) Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Neonatal Outcomes of Extremely Preterm Infants from the NICHD Neonatal Research Network. Pediatrics, 126, 443-456.
https://doi.org/10.1542/peds.2009-2959
[10] Christensen, R.D., Lambert, D.K., Baer, V.L. and Gordon, P.V. (2013) Necrotizing Enterocolitis in Term Infants. Clinics in Perinatology, 40, 69-78.
https://doi.org/10.1016/j.clp.2012.12.007
[11] Neu, J. and Walker, W.A. (2011) Necrotizing Enterocolitis. The New England Journal of Medicine, 364, 255-264.
https://doi.org/10.1056/NEJMra1005408
[12] Tanner, S.M., et al. (2015) Pathogenesis of Necrotizing Enterocolitis: Modeling the Innate Immune Response. American Journal of Pathology, 185, 4-16.
[13] Sodhi, C.P., et al. (2010) Toll-Like Receptor—4 Inhibits Enterocyte Proliferation via Impaired β-Catenin Signaling in Necrotizing Enterocolitis. Gastroenterology, 138, 185-196.
https://doi.org/10.1053/j.gastro.2009.09.045
[14] Gribar, S.C., et al. (2009) Reciprocal Expression and Signaling of TLR4 and TLR9 in the Pathogenesis and Treatment of Necrotizing Enterocolitis. The Journal of Immunology, 182, 636-646.
https://doi.org/10.4049/jimmunol.182.1.636
[15] Lu, P., Sodhi, C.P. and Hackam, D.J. (2014) Toll-Like Receptor Regulation of Intestinal Development and Inflammation in the Pathogenesis of Necrotizing Enterocolitis. Pathophysiology, 21, 81-93.
https://doi.org/10.1016/j.pathophys.2013.11.007
[16] Yazji, I., et al. (2013) Endothelial TLR4 Activation Impairs Intestinal Microcirculatory Perfusion in Necrotizing Enterocolitis via eNOS-NO-Nitrite Signaling. Proceedings of the National Academy of Sciences, 110, 9451-9456.
https://doi.org/10.1073/pnas.1219997110
[17] Afrazi, A., et al. (2014) Toll-Like Receptor 4-Mediated Endoplasmic Reticulum Stress in Intestinal Crypts Induces Necrotizing Enterocolitis. The Journal of Biological Chemistry, 289, 9584-9599.
https://doi.org/10.1074/jbc.M113.526517
[18] Deplancke, B. and Gaskins, H.R. (2001) Microbial Modulation of Innate Defense: Goblet Cells and the Intestinal Mucus Layer. The American Journal of Clinical Nutrition, 73, 1131-1141.
[19] Berseth, C.L. (1996) GastroIntestinal Motility in the Neonate. Clinics in Perinatology, 23, 179-190.
[20] Berseth, C.L. (1989) Gestational Evolution of Small Intestine Motility in Preterm and Term Infants. The Journal of Pediatrics, 115, 646-651.
https://doi.org/10.1016/S0022-3476(89)80302-6
[21] Hyman, P. and Thapar, N. (2013) Pediatric Neurogastroenterology: GastroIntestinal Motility and Functional Disorders in Children, Clinical Gastroenterology. In: Di Faure, L.T., Ed., Springer Inc., 257-270.
[22] Hackam, D.J., Upperman, J.S., Grishin, A. and Ford, H.R. (2005) Disordered Enterocyte Signaling and Intestinal Barrier Dysfunction in the Pathogenesis of Necrotizing Enterocolitis. Semin. Journal of Pediatric Surgery, 14, 49-57.
https://doi.org/10.1053/j.sempedsurg.2004.10.025
[23] Reisinger, K.W., et al. (2014) Breast-Feeding Improves Gut Maturation Compared with Formula Feeding in Preterm Babies. Journal of Pediatric Gastroenterology and Nutrition, 59, 720-724.
https://doi.org/10.1097/MPG.0000000000000523
[24] Lebenthal, A. and Lebenthal, E. (1999) The Ontogeny of the Small Intestinal Epithelium. Journal of Parenteral and Enteral Nutrition, 23, 3-6.
https://doi.org/10.1177/014860719902300502
[25] Watkins, D.J. and Besner, G.E. (2013) The Role of the Intestinal Microcirculation in Necrotizing Enterocolitis. Seminars in Pediatric Surgery, 22, 83-87.
https://doi.org/10.1053/j.sempedsurg.2013.01.004
[26] Anand, R.J., Leaphart, C.L., Mollen, K.P. and Hackam, D.J. (2007) The Role of the Intestinal Barrier in the Pathogenesis of Necrotizing Enterocolitis. Shock, 27, 124-133.
https://doi.org/10.1097/01.shk.0000239774.02904.65
[27] Liu, Z., Li, N. and Neu, J. (2005) Tight Junctions, Leaky Intestines, and Pediatric Diseases. Acta Paediatrica, 94, 386-393.
https://doi.org/10.1111/j.1651-2227.2005.tb01904.x
[28] Caplan, M.S., Simon, D. and Jilling, T. (2005) The Role of PAF, TLR, and the Inflammatory Response in Neonatal Necrotizing Enterocolitis. Seminars in Pediatric Surgery, 14, 145-151.
https://doi.org/10.1053/j.sempedsurg.2005.05.002
[29] Dingle, B.M., et al. (2013) FoxP3+ Regulatory T Cells Attenuate Experimental Necrotizing Enterocolitis. PLoS ONE, 8, e82963.
https://doi.org/10.1371/journal.pone.0082963
[30] Weitkamp, J.H., et al. (2014) Small Intestinal Intraepithelial TCRγδ+ T Lymphocytes Are Present in the Premature Intestine but Selectively Reduced in Surgical Necrotizing Enterocolitis. PLoS ONE, 9, e99042.
https://doi.org/10.1371/journal.pone.0099042
[31] Frost, B.L. and Caplan, M.S. (2013) Necrotizing Enterocolitis: Pathophysiology, Platelet-Activating Factor, and Probiotics. Seminars in Pediatric Surgery, 22, 88-93.
https://doi.org/10.1053/j.sempedsurg.2013.01.005
[32] Maheshwari, A. (2015) Immunologic and Hematological Abnormalities in Necrotizing Enterocolitis. Clinics in Perinatology, 42, 567-585.
https://doi.org/10.1016/j.clp.2015.04.014
[33] Rabinowitz, S.S., et al. (2001) Platelet-Activating Factor in Infants at Risk for Necrotizing Enterocolitis. The Journal of Pediatrics, 138, 81-86.
https://doi.org/10.1067/mpd.2001.110132
[34] Chatterton, D.E., Nguyen, D.N., Bering, S.B. and Sangild, P.T. (2013) Anti-Inflammatory Mechanisms of Bioactive Milk Proteins in the Intestine of Newborns. The International Journal of Biochemistry & Cell Biology, 45, 1730-1747.
https://doi.org/10.1016/j.biocel.2013.04.028
[35] Soliman, A., et al. (2010) Platelet-Activating Factor Induces TLR4 Expression in Intestinal Epithelial Cells: Implication for the Pathogenesis of Necrotizing Entero-colitis. PLoS ONE, 5, e15044.
https://doi.org/10.1371/journal.pone.0015044
[36] Eaton, S., Rees, C.M. and Hall, N.J. (2016) Current Research in Necrotizing Entero-colitis. Early Human Development, 97, 33-39.
https://doi.org/10.1016/j.earlhumdev.2016.01.013
[37] Caplan, M.S. (2010) Neonatal Necrotizing Enterocolitis: Clinical Observations, Pathophysiology, and Prevention. In: Martin, R.J. and Fanarof, A.A., Eds., Fanaroff and Martin’s Neonatal-Perinatal Medicine: Diseases of the Fetus and Infant, 9th Edition, Mosby, Missouri, 1431-1440.
[38] Verduci, E., Banderali, G., Barberi, S., Radaelli, G., Lops, A., Betti, F., et al. (2014) Epigenetic Effects of Human Breast Milk. Nutrients, 6, 1711-1724.
https://doi.org/10.3390/nu6041711
[39] Gao, F., Zhang, J., Jiang, P., Gong, D., Wang, J.W., Xia, Y., et al. (2014) Marked Methylation Changes in Intestinal Genes during the Perinatal Period of Preterm-neonates. BMC Genomics, 15, 716.
https://doi.org/10.1186/1471-2164-15-716
[40] Willems, R., Krych, L., Rybicki, V., Jiang, P., Sangild, P.T., Shen, R.L., et al. (2015) Introducing Enteral Feeding Induces Intestinal Subclinical Inflammation and Respective Chromatin Changes in Preterm Pigs. Epigenomics, 7, 553-565.
https://doi.org/10.2217/epi.15.13
[41] Trinchese, G., Cavaliere, G., Canani, R.B., Matamoros, S., Bergamo, P., De Filippo, C., et al. (2015) Human, Donkey and Cowmilk Differently Affects Energy Efficiency and Inflammatory State by Modulating Mitochondrial Function and Gut Microbiota. The Journal of Nutritional Biochemistry, 26, 1136-1146.
https://doi.org/10.1016/j.jnutbio.2015.05.003
[42] Schroeder, V.A., Mattioli, L.F., Kilkenny, T.A. and Belmont, J.M. (2014) Effects of Lactose-Containing vs Lactose-Free Infant Formula on Postprandial Superior-mesenteric Artery Flow in Term Infants. Journal of Parenteral and Enteral Nutrition, 38, 236-242.
https://doi.org/10.1177/0148607113478442
[43] Van Acker, J., de Smet, F., Muyldermans, G., Bougatef, A., Naessens, A. and Lauwers, S. (2001) Outbreak of Necrotizing Enterocolitis Associated with Enterobacter sakazakii in Powdered Milk Formula. Journal of Clinical Microbiology, 39, 293-297.
https://doi.org/10.1128/JCM.39.1.293-297.2001
[44] Rotbart, H.A. and Levin, M.J. (1983) How Contagious Is Necrotizing Enterocolitis? The Pediatric Infectious Disease, 2, 406-413.
https://doi.org/10.1097/00006454-198309000-00019
[45] Scheifele, D.W. (1990) Role of Bacterial Toxins in Neonatal Necrotizing Entero-colitis. The Journal of Pediatrics, 117, S44-S46.
https://doi.org/10.1016/S0022-3476(05)81129-1
[46] Scheifele, D.W., Olsen, E.M. and Pendray, M.R. (1985) Endotoxinemia and Throm-bocytopenia during Neonatal Necrotizing Enterocolitis. American Journal of Clinical Pathology, 83, 227-229.
https://doi.org/10.1093/ajcp/83.2.227
[47] Cheu, H.W., Brown, D.R. and Rowe, M.I. (1989) Breath Hydrogen Excretion as a Screening Test for the Early Diagnosis of Necrotizing Enterocolitis. The American Journal of Diseases of Children, 143, 156-159.
https://doi.org/10.1001/archpedi.1989.02150140042017
[48] Santulli, T.V., Schullinger, J.N., Heird, W.C., Gongaware, R.D., Wigger, J., Barlow, B., et al. (1975) Acute Necrotizing Enterocolitis in Infancy: A Review of 64 Cases. Pediatrics, 55, 376-387.
[49] Stewart, C.J., Marrs, E.C., Nelson, A., Lanyon, C., Perry, J.D., Embleton, N.D., et al. (2013) Development of the Preterm Gut Microbiome in Twins at Risk of Necro-tising Enterocolitis and Sepsis. PLoS One, 8, e73465.
https://doi.org/10.1371/journal.pone.0073465
[50] Brower-Sinning, R., Zhong, D., Good, M., Firek, B., Baker, R., Sodhi, C.P., et al. (2014) Mucosa Associated Bacterial Diversity in Necrotizing Enterocolitis. PLoS One, 9, e105046.
https://doi.org/10.1371/journal.pone.0105046
[51] Sharma, R. and Hudak, M.L. (2013) A Clinical Perspective of Necrotizing Entero-colitis: Past, Present, and Future. Clinics in Perinatology, 40, 27-51.
https://doi.org/10.1016/j.clp.2012.12.012
[52] Salhab, W.A., Perlman, J.M., Silver, L. and Sue Broyles, R. (2004) Necrotizing Enterocolitis and Neurodevelopmental Outcome in Extremely Low Birth Weight Infants <1000 g. Journal of Perinatology, 24, 534-540.
https://doi.org/10.1038/sj.jp.7211165
[53] Heida, F.H., Hulscher, J.B., Schurink, M., Timmer, A., Kooi, E.M., Bos, A.F., et al. (2015) Intestinal Fatty Acid-Binding Protein Levels in Necrotizing Enterocolitis Correlate with Extent of Necrotic Bowel: Results from a Multicenter Study. Journal of Pediatric Surgery, 50, 1115-1118.
https://doi.org/10.1016/j.jpedsurg.2014.11.037
[54] Zani, A. and Pierro, A. (2015) Necrotizing Enterocolitis: Controversies and Challenges. F1000Research, 4, pii: F1000 Faculty Rev-1373.
https://doi.org/10.12688/f1000research.6888.1
[55] Nantais-Smith, L. and Kadrofske, M. (2015) Noninvasive Biomarkers of Necrotizing Enterocolitis. The Journal of Perinatal & Neonatal Nursing, 29, 69-80.
https://doi.org/10.1097/JPN.0000000000000082
[56] 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.
https://doi.org/10.1136/archdischild-2014-307656
[57] Niemarkt, H.J., et al. (2015) Necrotizing Enterocolitis: A Clinical Review on Diagnostic Biomarkers and the Role of the Intestinal Microbiota. Inflammatory Bowel Diseases, 21, 436-444.
https://doi.org/10.1097/MIB.0000000000000184
[58] Ng, P.C. (2014) Biomarkers of Necrotising Enterocolitis. Seminars in Fetal and Neonatal Medicine, 19, 33-38.
https://doi.org/10.1016/j.siny.2013.09.002
[59] Thuijls, G., et al. (2010) Non-Invasive Markers for Early Diagnosis and Determination of the Severity of Necrotizing Enterocolitis. Annals of Surgery, 251, 1174-1180.
https://doi.org/10.1097/SLA.0b013e3181d778c4
[60] 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.
https://doi.org/10.1371/journal.pone.0121336
[61] Evennett, N., Hall, N., Pierro, A. and Eaton, S. (2010) Urinary Intestinal Fatty Acid-Binding Protein Concentration Predicts Extent of Disease in Necrotizing Enterocolitis. Journal of Pediatric Surgery, 45, 735-740.
https://doi.org/10.1016/j.jpedsurg.2009.09.024
[62] Sylvester, K.G., Ling, X.B., Liu, G.Y., Kastenberg, Z.J., Ji, J., Hu, Z., et al. (2014) Urine Protein Biomarkers for the Diagnosis and Prognosis of Necrotizing Entero-colitis in Infants. Journal of Pediatrics, 164, 607-612 e7.
https://doi.org/10.1016/j.jpeds.2013.10.091
[63] Sylvester, K.G., Ling, X.B., Liu, G.Y., Kastenberg, Z.J., Ji, J., Hu, Z., et al. (2014) A Novel Urine Peptide Biomarker-Based Algorithm for the Prognosis of Necrotising Enterocolitis in Human Infants. Gut, 63, 1284-1292.
https://doi.org/10.1136/gutjnl-2013-305130
[64] Bohnhorst, B. (2013) Usefulness of Abdominal Ultrasound in Diagnosing Necrotising Enterocolitis. Archives of Disease in Childhood—Fetal and Neonatal Edition, 98, 445-450.
https://doi.org/10.1136/archdischild-2012-302848
[65] Epelman, M., Daneman, A., Navarro, O.M., Morag, I., Moore, A.M., Kim, J.H., et al. (2007) Necrotizing Enterocolitis: Review of State-of-the-Art Imaging Findings with Pathologic Correlation. Radiographics, 27, 285-305.
https://doi.org/10.1148/rg.272055098
[66] Dordelmann, M., Rau, G.A., Bartels, D., Linke, M., Derichs, N., Behrens, C., et al. (2009) Evaluation of Portal Venous Gas Detected by Ultrasound Examination for Diagnosis of Necrotising Enterocolitis. Archives of Disease in Childhood—Fetal and Neonatal Edition, 94, 183-187.
https://doi.org/10.1136/adc.2007.132019
[67] Faingold, R., Daneman, A., Tomlinson, G., Babyn, P.S., Manson, D.E., Mohanta, A., et al. (2005) Necrotizing Enterocolitis: Assessment of Bowel Viability with Color Doppler US. Radiology, 235, 587-594.
https://doi.org/10.1148/radiol.2352031718
[68] Silva, C.T., Daneman, A., Navarro, O.M., et al. (2007) Correlation of Sonographic Findings and Outcome in Necrotizing Enterocolitis. Pediatric Radiology, 37, 274-282.
https://doi.org/10.1007/s00247-006-0393-x
[69] Murdoch, E.M., Sinha, A.K., Shanmugalingam, S.T., et al. (2006) Doppler Flow Velocimetry in the Superior Mesenteric Artery on the First Day of Life in Preterm Infants and the Risk of Neonatal Necrotizing Enterocolitis. Pediatrics, 118, 1999-2003.
https://doi.org/10.1542/peds.2006-0272
[70] Yikilmaz, A., Hall, N.J., Daneman, A., et al. (2014) Prospective Evaluation of the Impact of Sonography on the Management and Surgical Intervention of Neonates with Necrotizing Enterocolitis. Pediatric Surgery International, 30, 1231-1240.
https://doi.org/10.1007/s00383-014-3613-8
[71] DeWitt, A.G., Charpie, J.R., Donohue, J.E., Yu, S. and Owens, G.E. (2014) Splanchnic Near-Infrared Spectroscopy and Risk of Necrotizing Enterocolitis after Neonatal Heart Surgery. Pediatric Cardiology, 35, 1286-1294.
https://doi.org/10.1007/s00246-014-0931-5
[72] Patel, A.K., Lazar, D.A., Burrin, D.G., Smith, E.O., Magliaro, T.J., Stark, A.R., et al. (2014) Abdominal Near-Infrared Spectroscopy Measurements Are Lower in Preterm Infants at Risk for Necrotizing Enterocolitis. Pediatric Critical Care Medicine, 15, 735-741.
https://doi.org/10.1097/PCC.0000000000000211
[73] Cortez, J., Gupta, M., Amaram, A., Pizzino, J., Sawhney, M. and Sood, B.G. (2011) Noninvasive Evaluation of Splanchnic Tissue Oxygenation Using Near-Infrared Spectroscopy in Preterm Neonates. The Journal of Maternal-Fetal & Neonatal Medicine, 24, 574-582.
https://doi.org/10.3109/14767058.2010.511335
[74] Section on, Breastfeeding (2012) Breastfeeding and the Use of Human Milk. Pediatrics, 129, 827-841.
https://doi.org/10.1542/peds.2011-3552
[75] Pammi, M. and Abrams, S.A. (2015) Oral Lactoferrin for the Prevention of Sepsis and Necrotizing Enterocolitis in Preterm Infants. The Cochrane Database of Systematic Reviews, 2, CD007137.
https://doi.org/10.1002/14651858.cd007137.pub4
[76] Lonnerdal, B. (2010) Bioactive Proteins in Human Milk: Mechanisms of Action. Journal of Pediatrics, 156, 26-30.
https://doi.org/10.1016/j.jpeds.2009.11.017
[77] 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.
https://doi.org/10.1038/mi.2015.30
[78] 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. The Journal of Biological Chemistry, 287, 37296-37308.
https://doi.org/10.1074/jbc.M112.375881
[79] Good, M., Sodhi, C.P. and Hackam, D.J. (2014) Evidence-Based Feeding Strategies before and after the Development of Necrotizing Enterocolitis. Expert Review of Clinical Immunology, 10, 875-884.
https://doi.org/10.1586/1744666X.2014.913481
[80] Quigley, M. and McGuire, W. (2014) Formula versus Donor Breast Milk for Feeding Preterm or Low Birth Weight Infants. The Cochrane Database of Systematic Reviews, 4, CD002971.
https://doi.org/10.1002/14651858.cd002971.pub3
[81] Sullivan, S., et al. (2010) An Exclusively Human Milk-Based Diet Is Associated with a Lower Rate of Necrotizing Enterocolitis than a Diet of Human Milk and Bovine Milk-Based Products. Journal of Pediatrics, 156, 562-567 e1.
https://doi.org/10.1016/j.jpeds.2009.10.040
[82] Neu, J. (2014) Probiotics and Necrotizing Enterocolitis. Clinics in Perinatology, 41, 967-978.
https://doi.org/10.1016/j.clp.2014.08.014
[83] Robinson, J. (2014) Cochrane in Context: Probiotics for Prevention of Necrotizing Enterocolitis in Preterm Infants. Evidence-Based Child Health, 9, 672-674.
https://doi.org/10.1002/ebch.1977
[84] Vongbhavit, K. and Underwood, M.A. (2016) Prevention of Necrotizing Entero-colitis through Manipulation of the Intestinal Microbiota of the Premature Infant. Clinical Therapeutics, 38, 716-732.
https://doi.org/10.1016/j.clinthera.2016.01.006
[85] Coggins, S.A., Wynn, J.L. and Weitkamp, J.H. (2015) Infectious Causes of Necro-tizing Enterocolitis. Clinics in Perinatology, 42, 133-154.
https://doi.org/10.1016/j.clp.2014.10.012
[86] Neu, J. (2015) Preterm Infant Nutrition, Gut Bacteria, and Necrotizing Entero-colitis. Current Opinion in Clinical Nutrition & Metabolic Care, 18, 285-288.
https://doi.org/10.1097/MCO.0000000000000169
[87] Fleming, P., Hall, N.J. and Eaton, S. (2015) Probiotics and Necrotizing Enterocolitis. Pediatric Surgery International, 31, 1111-1118.
https://doi.org/10.1007/s00383-015-3790-0
[88] AlFaleh, K. and Anabrees, J. (2014) Probiotics for Prevention of Necrotizing Enterocolitis in Preterm Infants. Evidence-Based Child Health, 9, 584-671.
https://doi.org/10.1002/ebch.1976
[89] Downard, C.D., et al. (2012) Treatment of Necrotizing Enterocolitis: An American Pediatric Surgical Association Outcomes and Clinical Trials Committee Systematic Review. Journal of Pediatric Surgery, 47, 2111-2122.
https://doi.org/10.1016/j.jpedsurg.2012.08.011
[90] Floch, M.H., et al. (2015) Recommendations for Probiotic Use—2015 Update: Proceedings and Consensus Opinion. Journal of Clinical Gastroenterology, 49, 69-73.
https://doi.org/10.1097/mcg.0000000000000420
[91] Houghteling, P.D. and Walker, W.A. (2015) From Birth to “Immunohealth”, Allergies and Enterocolitis. Journal of Clinical Gastroenterology, 49, 7-12.
https://doi.org/10.1097/MCG.0000000000000355
[92] Shaffiey, S.A., et al. (2016) Intestinal Stem Cell Growth and Differentiation on a Tubular Scaffold with Evaluation in Small and Large Animals. Regenerative Medicine, 11, 45-61.
https://doi.org/10.2217/rme.15.70
[93] 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, 1021-1032.
https://doi.org/10.1152/ajpgi.00452.2013
[94] Zani, A., Eaton, S., Puri, P., et al. (2015) International Survey on the Management of Necrotizing Enterocolitis. European Journal of Pediatric Surgery, 25, 27-33.
https://doi.org/10.1055/s-0034-1387942
[95] Shah, D. and Sinn, J.K. (2012) Antibiotic Regimens for the Empirical Treatment of Newborn Infants with Necrotising Enterocolitis. The Cochrane Database of Systematic Reviews, 8, CD007448.
https://doi.org/10.1002/14651858.cd007448.pub2
[96] Stey, A., et al. (2015) Outcomes and Costs of Surgical Treatments of Necrotizing Enterocolitis. Pediatrics, 135, 1190-1197.
https://doi.org/10.1542/peds.2014-1058
[97] 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.
https://doi.org/10.1016/j.jpedsurg.2013.08.006
[98] Rao, S.C., Basani, L., Simmer, K., Samnakay, N. and Deshpande, G. (2011) Peritoneal Drainage versus Laparotomy as Initial Surgical Treatment for Perforated Necrotizing Enterocolitis or Spontaneous Intestinal Perforation in Preterm Low Birth Weight Infants. The Cochrane Database of Systematic Reviews, CD006182.
https://doi.org/10.1002/14651858.cd006182.pub2
[99] Raval, M.V., Hall, N.J., Pierro, A. and Moss, R.L. (2013) Evidence-Based Prevention and Surgical Treatment of Necrotizing Enterocolitis—A Review of Randomized Controlled Trials. Seminars in Pediatric Surgery, 22, 117-121.
https://doi.org/10.1053/j.sempedsurg.2013.01.009
[100] Rees, C.M., et al. (2008) Peritoneal Drainage or Laparotomy for Neonatal Bowel Perforation? A Randomized Controlled Trial. Annals of Surgery, 248, 44-51.
https://doi.org/10.1097/SLA.0b013e318176bf81
[101] Pammi, M. and Haque, K.N. (2015) Pentoxifylline for Treatment of Sepsis and Necrotizing Enterocolitis in Neonates. The Cochrane Database of Systematic Reviews, 3, Cd004205.
https://doi.org/10.1002/14651858.cd004205.pub3
[102] Akdag, A., Dilmen, U., Haque, K., Dilli, D., Erdeve, O. and Goekmen, T. (2014) Role of Pentoxifylline and/or IgM-Enriched Intravenous Immunoglobulin in the Management of Neonatal Sepsis. American Journal of Perinatology, 31, 905-912.
https://doi.org/10.1055/s-0033-1363771
[103] Travadi, J., Patole, S., Charles, A., Dvorak, B., Doherty, D. and Simmer, K. (2006) Pentoxifylline Reduces the Incidence and Severity of Necrotizing Enterocolitis in a Neonatal rat Model. Pediatric Research, 60, 185-189.
https://doi.org/10.1203/01.pdr.0000228325.24945.ac
[104] Eaton, S., Zani, A., Pierro, A. and De, C.P. (2013) Stemcells as a Potential Therapy for Necrotizing Enterocolitis. Expert Opinion on Biological Therapy, 13, 1683-1689.
https://doi.org/10.1517/14712598.2013.849690
[105] Zani, A., Cananzi, M., Fascetti-Leon, F., Lauriti, G., Smith, V.V., Bollini, S., et al. (2014) Amniotic Fluid Stem Cells Improve Survival and Enhance Repair of Damaged Intestine in Necrotising Enterocolitis via a COX-2 Dependent Mechanism. Gut, 63, 300-309.
https://doi.org/10.1136/gutjnl-2012-303735
[106] Tayman, C., Uckan, D., Kilic, E., Ulus, A.T., Tonbul, A., Murat, H.I., et al. (2011) Mesenchymal Stem Cell Therapy in Necrotizing Enterocolitis: A Rat Study. Pediatric Research, 70, 489-494.
https://doi.org/10.1203/PDR.0b013e31822d7ef2
[107] Yang, J.X., Watkins, D., Chen, C.L., Bhushan, B., Zhou, Y. and Besner, G.E. (2012) Heparin-Binding Epidermal Growth Factor-Like Growth Factor Andmesenchymal Stem Cells Act Synergistically to Prevent Experimental Necrotizing Enterocolitis. Journal of the American College of Surgeons, 215, 534-545.
https://doi.org/10.1016/j.jamcollsurg.2012.05.037
[108] Wei, J., Zhou, Y. and Besner, G.E. (2015) Heparin-Binding EGF-Like Growth Factor and Enteric Neural Stem Cell Transplantation in the Prevention of Experimental Necrotizing Enterocolitis in Mice. Pediatric Research, 78, 29-37.
https://doi.org/10.1038/pr.2015.63
[109] Ostergaard, M.V., Bering, S.B., Jensen, M.L., Thymann, T., Purup, S., Diness, M., et al. (2014) Modulation of Intestinal Inflammation by Minimal Enteral Nutrition with Amniotic Fluid in Preterm Pigs. Journal of Parenteral and Enteral Nutrition, 38, 576-586.
https://doi.org/10.1177/0148607113489313
[110] Siggers, J., Ostergaard, M.V., Siggers, R.H., Skovgaard, K., Molbak, L., Thymann, T., et al. (2013) Postnatal Amniotic Fluid Intake Reduces Gut Inflammatory Responses and Necrotizing Enterocolitis in Pretermneonates. American Journal of Physiology—GastroIntestinal and Liver Physiology, 304, 864-875.
https://doi.org/10.1152/ajpgi.00278.2012
[111] Good, M., Siggers, R.H., Sodhi, C.P., Afrazi, A., Alkhudari, F., Egan, C.E., et al. (2012) Amniotic Fluid Inhibits Toll-Like Receptor 4 Signaling in the Fetal and Neonatal Intestinal Epithelium. Proceedings of the National Academy of Sciences, 109, 11330-11335.
https://doi.org/10.1073/pnas.1200856109
[112] Su, Y., Yang, J. and Besner, G.E. (2013) HB-EGF Promotes Intestinal Restitution by Affecting Integrin-Extracellular Matrix Interactions and Intercellular Adhesions. Growth Factors, 31, 39-55.
https://doi.org/10.3109/08977194.2012.755966
[113] Chen, C.L., et al. (2012) Heparin-Binding EGF-Like Growth Factor Protects Intestinal Stem Cells from Injury in a Rat Model of Necrotizing Enterocolitis. Laboratory Investigation, 92, 331-344.
https://doi.org/10.1038/labinvest.2011.167
[114] Yang, J., Su, Y., Zhou, Y. and Besner, G.E. (2014) Heparin-Binding EGF-Like Growth Factor [HB-EGF] Therapy for Intestinal Injury: Application and Future Prospects. Pathophysiology, 21, 95-104.
https://doi.org/10.1016/j.pathophys.2013.11.008
[115] Newburg, D.S. (2009) Neonatal Protection by an Innate Immune System of Human Milk Consisting of Oligosaccharides and Glycans. Journal of Animal Science, 87, 26-34.
https://doi.org/10.2527/jas.2008-1347
[116] Jantscher-Krenn, E., et al. (2012) The Human Milk Oligosaccharide Disialyllacto-N-Tetraose Prevents Necrotising Enterocolitis in Neonatal Rats. Gut, 61, 1417-1425.
https://doi.org/10.1136/gutjnl-2011-301404
[117] Fitzgibbons, S.C., Ching, Y., Yu, D., et al. (2009) Mortality of Necrotizing Entero-colitis Expressed by Birth Weight Categories. Journal of Pediatric Surgery, 44, 1072-1076.
https://doi.org/10.1016/j.jpedsurg.2009.02.013
[118] Rees, C.M., Pierro, A. and Eaton, S. (2007) Neurodevelopmental Outcomes of Neonates with Medically and Surgically Treated Necrotizing Enterocolitis. Archives of Disease in Childhood Fetal and Neonatal Edition, 92, 193-198.
https://doi.org/10.1136/adc.2006.099929
[119] Hintz, S.R., Kendrick, D.E., Stoll, B.J., et al. (2005) Neurodevelopmental and Growth Outcomes of Extremely Low Birth Weight Infants after Necrotizing Ente-rocolitis. Pediatrics, 115, 696-703.
https://doi.org/10.1542/peds.2004-0569
[120] Shah, D.K., Doyle, L.W., Anderson, P.J., et al. (2008) Adverse Neurodevelopment in Preterm Infants with Postnatal Sepsis or Necrotizing Enterocolitis Is Mediated by White Matter Abnormalities on Magnetic Resonance Imaging at Term. Journal of Pediatrics, 153, 170-175.
https://doi.org/10.1016/j.jpeds.2008.02.033

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.