Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 302
1.
BMJ Case Rep ; 17(6)2024 Jun 05.
Article En | MEDLINE | ID: mdl-38839401

Retinopathy of prematurity (ROP) and necrotising enterocolitis (NEC) are complications of prematurity. Despite being quite different in terms of incidence, pathogenesis and consequences, both share a pathogenic role of aberrant vascularisation: increased in ROP, deficient for NEC. Current therapy for ROP includes the use of anti-vascular endothelial growth factor (anti-VEGF) agents, which are able to interrupt retinal hypervascularity. Despite being delivered intravitreously, anti-VEGF used in ROP can be absorbed into circulation and exert systemic effects. We present here a case of an ex-27 weeks gestational age infant, presenting multiple NEC risk factors, treated at 2 months of age with low-dose ranibizumab, who developed a large bowel NEC episode in the first week after treatment. We believe that this further report of an association between anti-VEGF agents and NEC could be interesting for the identification of children at risk of severe adverse events and stimulating further research on the topic.


Angiogenesis Inhibitors , Enterocolitis, Necrotizing , Intravitreal Injections , Ranibizumab , Retinopathy of Prematurity , Humans , Retinopathy of Prematurity/drug therapy , Enterocolitis, Necrotizing/drug therapy , Ranibizumab/administration & dosage , Ranibizumab/therapeutic use , Infant, Newborn , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Male , Infant, Premature , Female , Vascular Endothelial Growth Factor A/antagonists & inhibitors
2.
Sci Rep ; 14(1): 7665, 2024 04 01.
Article En | MEDLINE | ID: mdl-38561398

The integrity of the intestinal mucus barrier is crucial for human health, as it serves as the body's first line of defense against pathogens. However, postnatal development of the mucus barrier and interactions between maturity and its ability to adapt to external challenges in neonatal infants remain unclear. In this study, we unveil a distinct developmental trajectory of the mucus barrier in preterm piglets, leading to enhanced mucus microstructure and reduced mucus diffusivity compared to term piglets. Notably, we found that necrotizing enterocolitis (NEC) is associated with increased mucus diffusivity of our large pathogen model compound, establishing a direct link between the NEC condition and the mucus barrier. Furthermore, we observed that addition of sodium decanoate had varying effects on mucus diffusivity depending on maturity and health state of the piglets. These findings demonstrate that regulatory mechanisms governing the neonatal mucosal barrier are highly complex and are influenced by age, maturity, and health conditions. Therefore, our results highlight the need for specific therapeutic strategies tailored to each neonatal period to ensure optimal gut health.


Decanoic Acids , Enterocolitis, Necrotizing , Mucus , Infant, Newborn , Animals , Humans , Swine , Inflammation , Dietary Supplements , Enterocolitis, Necrotizing/drug therapy , Intestinal Mucosa
3.
Life Sci ; 339: 122420, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-38218534

Human milk is the most valuable source of nutrition for infants. The structure and function of human milk oligosaccharides (HMOs), which are key components of human milk, have long been attracting particular research interest. Several recent studies have found HMOs to be efficacious in the prevention and treatment of necrotizing enterocolitis (NEC). Additionally, they could be developed in the future as non-invasive predictive markers for NEC. Based on previous findings and the well-defined functions of HMOs, we summarize potential protective mechanisms of HMOs against neonatal NEC, which include: modulating signal receptor function, promoting intestinal epithelial cell proliferation, reducing apoptosis, restoring intestinal blood perfusion, regulating microbial prosperity, and alleviating intestinal inflammation. HMOs supplementation has been demonstrated to be protective against NEC in both animal studies and clinical observations. This calls for mass production and use of HMOs in infant formula, necessitating more research into the safety of industrially produced HMOs and the appropriate dosage in infant formula.


Enterocolitis, Necrotizing , Milk, Human , Infant , Animals , Infant, Newborn , Humans , Milk, Human/chemistry , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/prevention & control , Intestines , Cell Proliferation , Oligosaccharides/pharmacology , Oligosaccharides/therapeutic use , Oligosaccharides/analysis
4.
Int Immunopharmacol ; 128: 111360, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-38176339

Bile acids (BAs) have increasingly been implicated in the onset and progression of necrotizing enterocolitis (NEC); multiple findings have demonstrated their ability to induce damage to the intestinal epithelium, thereby exacerbating disease severity. Although we previously showed that melatonin was able to treat NEC by correcting the Treg/Th17 imbalance, the modulatory effect of melatonin on BAs remains unclear. In this study, we conducted transcriptome analysis on intestinal tissues from patients with NEC and validated these findings. Subsequently, we treated mice with melatonin alone or in combination with an agonist/inhibitor of Sirtuin 1 (SIRT1) to assess faecal and serum BA levels, the expression levels of BA transporters and regulators, and the extent of intestinal injury. Our transcriptome results indicated dysregulation of BA metabolism and abnormal expression of BA transporters in patients with NEC, which were also observed in our NEC mouse model. Furthermore, exogenous BAs were found to aggravate NEC severity in mice. Notably, melatonin effectively restored the aberrant expression of BA transporters, such as apical membrane sodium-dependent bile acid transporters (ASBT), ileal bile acid-binding protein (IBABP), and organic solute transporter-alpha (OST-α), by upregulating SIRT1 expression while reducing farnesoid X receptor (FXR) acetylation, consequently leading to decreased serum and faecal BA levels and mitigated NEC severity. Thus, we propose a potential mechanism through which melatonin reduces BA levels via the SIRT1/FXR signalling axis in an NEC mouse model. Collectively, these results highlight that melatonin holds promise for reducing BA levels and represents a promising therapeutic strategy for treating NEC.


Enterocolitis, Necrotizing , Melatonin , Animals , Humans , Mice , Bile Acids and Salts/metabolism , Disease Models, Animal , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Intestines , Liver , Melatonin/pharmacology , Melatonin/therapeutic use , Sirtuin 1/metabolism
5.
J Am Coll Surg ; 238(1): 82-98, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-37870229

BACKGROUND: Necrotizing enterocolitis (NEC) is a devastating condition where inflammatory changes and necrosis in the gut results in activation of brain microglia and subsequent neurodevelopmental impairment. Chondroitin sulfate (CS) is a glycosaminoglycan in human breast milk that is absent in conventional formulas. We hypothesized that oral formula supplementation with CS during a murine model of experimental NEC would not only attenuate intestinal injury, but also brain injury. STUDY DESIGN: NEC was induced in mouse pups on postnatal days (PNDs) 5 to 8. Three conditions were studied: (1) breastfed controls, (2) NEC, and (3) NEC+enteral CS (formula+200 mg/kg/d of CS). Pups were euthanized on PND 9 or reunited with dams by the evening of PND 8. Intestinal segments were H&E stained, and immunohistochemistry was performed on brain tissue for Iba-1 to assess for microglial morphology and cortical changes. Neurodevelopmental assays were performed on mice reunited with foster dams on PND 9. Single-cell RNA-sequencing analysis was performed on human intestinal epithelial cells exposed to (1) nothing, (2) hydrogen peroxide (H 2 O 2 ) alone, or (3) H 2 O 2 + CS to look at the differential gene expression between groups. Groups were compared with ANOVA or Kruskal-Wallis tests as appropriate with p < 0.05 considered significant. RESULTS: Compared with NEC, mice treated with oral CS showed improved clinical outcomes, decreased intestinal injury, and attenuated microglial activation and deleterious cortical change. Mice with CS performed better on early neurodevelopmental assays when compared with NEC alone. Single-cell analysis of HIEC-6 cells demonstrated that CS treatment down regulated several inflammatory pathways including nuclear factor κB-suggesting an explanation for the improved Th17 intestinal cytokine profile. CONCLUSIONS: Oral CS supplementation improved both physiological, clinical, and developmental outcomes. These data suggest that CS is a safe compound for formula supplementation for the prevention of NEC.


Brain Injuries , Enterocolitis, Necrotizing , Female , Animals , Mice , Infant, Newborn , Humans , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Brain Injuries/metabolism , Dietary Supplements , Disease Models, Animal , Intestinal Mucosa
6.
J Perinatol ; 44(4): 587-593, 2024 Apr.
Article En | MEDLINE | ID: mdl-37863983

OBJECTIVE: Evaluate the impact of a multidisciplinary guideline standardizing antibiotic duration and enteral feeding practices following medical necrotizing enterocolitis (mNEC). STUDY DESIGN: For preterm infants with Bell Stage 2 A mNEC and negative blood culture, antibiotic treatment was standardized to 7 days. Trophic feeds of unfortified human milk began 72 h after resolution of pneumatosis. Feeds were advanced by 20 cc/kg/day starting on the last day of antibiotics. Primary outcomes were antibiotic days and days to full feeds, defined as 120 cc/kg/day of enteral nutrition. Secondary outcomes included central line days and length of stay (LOS). RESULTS: Antibiotic duration decreased 23%. Time to start trophic feeds and time to full feeds decreased 33 and 16% respectively. Central line use dropped (98 to 72% of infants) and central line days were reduced by 59%. CONCLUSION: Implementation of a mNEC QI package reduced antibiotic duration, time to full feeds, central line use and CL days.


Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant, Newborn , Humans , Infant, Premature , Enterocolitis, Necrotizing/drug therapy , Quality Improvement , Enteral Nutrition , Anti-Bacterial Agents/therapeutic use , Infant, Very Low Birth Weight
7.
Physiol Rep ; 11(17): e15819, 2023 09.
Article En | MEDLINE | ID: mdl-37697223

Necrotizing enterocolitis (NEC) continues to be a devastating disease in preterm neonates and has a paucity of medical management options. Chondroitin sulfate (CS) is a naturally occurring glycosaminoglycan (GAG) in human breast milk (HM) and has been shown to reduce inflammation. We hypothesized that supplementation with CS in an experimental NEC model would alter microbial diversity, favorably alter the cytokine profile, and (like other sulfur compounds) improve outcomes in experimental NEC via the eNOS pathway. NEC was induced in 5-day-old pups. Six groups were studied (n = 9-15/group): (1) WT breastfed and (2) Formula fed controls, (3) WT NEC, (4) WT NEC + CS, (5) eNOS KO (knockout) NEC, and (6) eNOS KO NEC + CS. Pups were monitored for clinical sickness score and weights. On postnatal day 9, the pups were killed. Stool was collected from rectum and microbiome analysis was done with 16 s rRNA sequencing. Intestinal segments were examined histologically using a well-established injury scoring system and segments were homogenized and analyzed for cytokine profile. Data were analyzed using GraphPad Prism with p < 0.05 considered significant. CS supplementation in formula improved experimental NEC outcomes when compared to NEC alone. CS supplementation resulted in similar improvement in NEC in both the WT and eNOS KO mice. CS supplementation did not result in microbial changes when compared to NEC alone. Our data suggest that although CS supplementation improved outcomes in NEC, this protection is not conferred via the eNOS pathway or alteration of microbial diversity. CS therapy in NEC does improve the intestinal cytokine profile and further experiments will explore the mechanistic role of CS in altering immune pathways in this disease.


Enterocolitis, Necrotizing , Fetal Diseases , Female , Infant, Newborn , Humans , Animals , Mice , Chondroitin Sulfates/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Disease Models, Animal , Dietary Supplements , Cytokines
8.
J Pediatr Surg ; 58(12): 2391-2398, 2023 Dec.
Article En | MEDLINE | ID: mdl-37684170

BACKGROUND: Hydrogen sulfide (H2S) has been shown to improve outcomes in a murine model of necrotizing enterocolitis (NEC). There is evidence in humans that H2S relies on endothelial nitric oxide synthase (eNOS) to exert its protective effects, potentially through the persulfidation of eNOS at the Cysteine 443 residue. We obtained a novel mouse strain with a mutation at this residue (eNOSC440G) and hypothesized that this locus would be critical for GYY4137 (an H2S donor) to exert its protective effects. METHODS: Necrotizing enterocolitis was induced in 5-day old wild type (WT) and eNOSC440G mice using intermittent exposure to hypoxia and hypothermia in addition to gavage formula feeds. On postnatal day 9, mice were humanely euthanized. Data collected included daily weights, clinical sickness scores, histologic lung injury, intestinal injury (macroscopically and histologically), and intestinal perfusion. During the NEC model, pups received daily intraperitoneal injections of either GYY4137 (50 mg/kg) or PBS (vehicle). Data were tested for normality and compared using t-test or Mann-Whitney, and a p-value <0.05 was considered significant. RESULTS: In WT mice, the administration of GYY4137 significantly improved clinical sickness scores, attenuated intestinal and lung injury, and improved mesenteric perfusion compared to vehicle (p < 0.05). In eNOSC440G mice, the treatment and vehicle groups had similar clinical sickness scores, intestinal and lung injury scores, and intestinal perfusion. CONCLUSIONS: GYY4137 administration improves clinical outcomes, attenuates intestinal and lung injury, and improves perfusion in a murine model of necrotizing enterocolitis. The beneficial effects of GYY4137 are dependent on the Cys440 residue of eNOS.


Enterocolitis, Necrotizing , Hydrogen Sulfide , Infant, Newborn, Diseases , Lung Injury , Humans , Infant, Newborn , Animals , Mice , Nitric Oxide Synthase Type III , Hydrogen Sulfide/pharmacology , Enterocolitis, Necrotizing/drug therapy , Disease Models, Animal , Nitric Oxide
9.
Sci Rep ; 13(1): 13395, 2023 08 17.
Article En | MEDLINE | ID: mdl-37591866

Prostaglandin E2 (PGE2) is implicated in intestinal inflammation and intestinal blood flow regulation with a paradoxical effect on the pathogenesis of necrotizing enterocolitis (NEC), which is not yet well understood. In the current study, we found that PGE2, EP4, and COX-2 varied at different distances from the most damaged area in the terminal ileum obtained from human infants with NEC. PGE2 administration alleviated the phenotype of experimental NEC and the intestinal microvascular features in experimental NEC, but this phenomenon was inhibited by eNOS depletion, suggesting that PGE2 promoted intestinal microcirculatory perfusion through eNOS. Furthermore, PGE2 administration increased the VEGF content in MIMECs under TNFα stress and promoted MIMEC proliferation. This response to PGE2 was involved in eNOS phosphorylation and nitric oxide (NO) production and was blocked by the EP4 antagonist in vitro, suggesting that targeting the PGE2-EP4-eNOS axis might be a potential clinical and therapeutic strategy for NEC treatment. The study is reported in accordance with ARRIVE guidelines ( https://arriveguidelines.org ).


Dinoprostone , Enterocolitis, Necrotizing , Infant , Humans , Infant, Newborn , Microcirculation , Intestines , Cyclooxygenase 2 , Enterocolitis, Necrotizing/drug therapy
10.
J Pediatr Surg ; 58(10): 1982-1989, 2023 Oct.
Article En | MEDLINE | ID: mdl-37479571

BACKGROUND: Antibiotic overutilization in the neonatal intensive care unit (NICU) has many adverse effects, and necrotizing enterocolitis (NEC) is one of the most common indications for antibiotics in premature infants. Evidence for a preferred antibiotic regimen for NEC is lacking. This project aims to reduce piperacillin-tazobactam use and overall antibiotic duration in neonates with NEC through the implementation of an antibiotic stewardship pathway based on the modified Bell stage classification system. METHODS: A multidisciplinary team consisting of neonatology, pharmacy, infectious disease, and surgery developed an antibiotic protocol for the management of NEC based on Bell stage. Recommendations included 48 h of ampicillin/gentamicin (AG) for stage I, 5-10 days of AG for stage II, the addition of metronidazole for stage IIIA, and 7-14 days of piperacillin-tazobactam (PT) for stage IIIB. We evaluated overall antibiotic and PT exposure, progression to surgical NEC, NEC recurrence, antibiotic resistance, bacteremia/fungemia, and mortality 1 year pre- and post-protocol implementation. RESULTS: 27 patients pre-intervention and 44 post-intervention were analyzed. Antibiotic exposure was reduced from a median 119.19 to 80.65 days of therapy (DOT) per 1000 patient days (p = 0.11). PT exposure decreased after protocol implementation (median 68.78 vs. 7.97 DOT per 1000 patient days, p = 0.002). There were no significant differences in morbidity or mortality outcomes. CONCLUSIONS: Antibiotic stewardship strategies can be implemented in the NICU without compromising outcomes in patients with NEC. Bell stage stratification appears to be an effective method for antibiotic selection. Further studies are needed in a larger population to optimize regimens and ensure safety. TYPE OF STUDY: Retrospective comparative study. LEVEL OF EVIDENCE: Level III.


Antimicrobial Stewardship , Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Infant , Female , Infant, Newborn , Humans , Enterocolitis, Necrotizing/drug therapy , Quality Improvement , Retrospective Studies , Infant, Premature , Anti-Bacterial Agents/therapeutic use , Ampicillin/therapeutic use , Piperacillin, Tazobactam Drug Combination/therapeutic use
11.
Tissue Cell ; 83: 102128, 2023 Aug.
Article En | MEDLINE | ID: mdl-37413858

AIM: This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway. METHODS: NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1ß and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL. RESULTS: NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway. CONCLUSION: AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.


Enterocolitis, Necrotizing , NF-kappa B , Rats , Animals , NF-kappa B/metabolism , Animals, Newborn , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Ligands , Rats, Sprague-Dawley , Signal Transduction , Inflammation/pathology , Disease Models, Animal
12.
Cochrane Database Syst Rev ; 6: CD004205, 2023 06 20.
Article En | MEDLINE | ID: mdl-37338074

BACKGROUND: Mortality and morbidity due to neonatal sepsis and necrotising enterocolitis (NEC) remain high despite the use of potent antimicrobial agents. Agents that modulate inflammation may improve outcomes. Pentoxifylline (PTX), a phosphodiesterase inhibitor, is one such agent. This is an update of a review first published in 2003 and updated in 2011 and 2015. OBJECTIVES: To assess the effectiveness and safety of intravenous PTX as an adjunct to antibiotic therapy on mortality and morbidity in neonates with suspected or confirmed sepsis and neonates with NEC. SEARCH METHODS: We searched CENTRAL, MEDLINE, Embase, CINAHL, and trial registries in July 2022. We also searched the reference lists of identified clinical trials and handsearched conference abstracts.   SELECTION CRITERIA: We included randomised controlled trials (RCTs) or quasi-RCTs assessing the efficacy of PTX with antibiotics (any dose or duration) for treatment of suspected or confirmed sepsis or NEC in neonates. We included three comparisons: (1) PTX with antibiotics compared to placebo or no intervention with antibiotics; (2) PTX with antibiotics compared to PTX with antibiotics and adjunct treatments such as immunoglobulin M-enriched intravenous immunoglobulin (IgM-enriched IVIG); (3) PTX with antibiotics compared to adjunct treatments such as IgM-enriched IVIG with antibiotics. DATA COLLECTION AND ANALYSIS: We reported typical risk ratio (RR) and risk difference (RD) with 95% confidence intervals (CI) for dichotomous outcomes, and mean difference (MD) for continuous outcomes derived from a fixed-effect model of meta-analysis. We calculated the number needed to treat for an additional beneficial outcome (NNTB) if there was a statistically significant reduction in RD. MAIN RESULTS: We identified no new studies for this update. We included six RCTs (416 neonates). All of the included studies examined neonates with sepsis; we identified no studies on neonates with NEC. Four of the six trials had high risk of bias for at least one risk of bias domain. Comparison 1: PTX with antibiotics compared to placebo with antibiotics, or antibiotics alone, in neonates with sepsis may reduce all-cause mortality during hospital stay (typical RR 0.57, 95% CI 0.35 to 0.93; typical RD -0.08, 95% CI -0.14 to -0.01; NNTB 13, 95% CI 7 to 100; 6 studies, 416 participants, low-certainty evidence) and may decrease length of hospital stay (LOS) (MD -7.74, 95% CI -11.72 to -3.76; 2 studies, 157 participants, low-certainty evidence). The evidence is very uncertain that PTX with antibiotics compared to placebo or no intervention results in any change in chronic lung disease (CLD) (RR 1.50, 95% CI 0.45 to 5.05; 1 study, 120 participants, very low-certainty evidence), severe intraventricular haemorrhage (sIVH) (RR 0.75, 95% CI 0.28 to 2.03; 1 study, 120 participants, very low-certainty evidence), periventricular leukomalacia (PVL) (RR 0.50, 95% CI 0.10 to 2.63; 1 study, 120 participants, very low-certainty evidence), NEC (RR 0.56, 95% CI 0.29 to 1.06; 6 studies, 405 participants, very low-certainty evidence), or retinopathy of prematurity (ROP) (RR 0.40, 95% CI 0.08 to 1.98; 1 study, 120 participants, very low-certainty evidence) in neonates with sepsis. Comparison 2: the evidence is very uncertain that PTX with antibiotics compared to PTX with antibiotics and IgM-enriched IVIG has any effect on mortality (RR 0.71, 95% CI 0.24 to 2.10; 102 participants, 1 study, very low-certainty evidence) or development of NEC in neonates with sepsis (RR 1.33, 95% CI 0.31 to 5.66; 1 study, 102 participants, very low-certainty evidence). The outcomes of CLD, sIVH, PVL, LOS, and ROP were not reported. Comparison 3: the evidence is very uncertain that PTX with antibiotics compared to IgM-enriched IVIG with antibiotics has any effect on mortality (RR 1.25, 95% CI 0.36 to 4.39; 102 participants, 1 study, very low-certainty evidence) or development of NEC (RR 1.33, 95% CI 0.31 to 5.66; 102 participants, 1 study, very low-certainty evidence) in neonates with sepsis. The outcomes of CLD, sIVH, PVL, LOS, and ROP were not reported. All of the included studies evaluated adverse effects due to PTX, but none were reported in the intervention group in any of the comparisons. AUTHORS' CONCLUSIONS: Low-certainty evidence suggests that adjunct PTX therapy in neonatal sepsis may decrease mortality and length of hospital stay without any adverse effects. The evidence is very uncertain if PTX with antibiotics compared to PTX with antibiotics and IgM-enriched IVIG, or PTX with antibiotics compared to IgM-enriched IVIG with antibiotics, has any effect on mortality or development of NEC. We encourage researchers to undertake well-designed multicentre trials to confirm or refute the effectiveness and safety of pentoxifylline in reducing mortality and morbidity in neonates with sepsis or NEC.


Enterocolitis, Necrotizing , Lung Diseases , Neonatal Sepsis , Pentoxifylline , Retinopathy of Prematurity , Sepsis , Humans , Infant, Newborn , Anti-Bacterial Agents/adverse effects , Enterocolitis, Necrotizing/drug therapy , Immunoglobulin M , Immunoglobulins, Intravenous/therapeutic use , Infant, Premature , Neonatal Sepsis/drug therapy , Pentoxifylline/adverse effects , Sepsis/drug therapy
13.
Early Hum Dev ; 183: 105797, 2023 08.
Article En | MEDLINE | ID: mdl-37300991

Necrotizing enterocolitis (NEC) is a devastating neonatal disease that affects neonates worldwide and often leads to high morbidity and mortality rates. Despite extensive research, the cause of NEC remains unclear, and current treatment options are limited. An important novel finding is the potential role of intestinal Alkaline Phosphatase (IAP) in both pathogenesis and treatment of NEC. IAP can play a vital role in detoxifying liposaccharides (LPS), a key mediator of many pathological processes, thereby reducing the inflammatory response associated with NEC. Furthermore, IAP can help prevent dysbiosis, improve intestinal perfusion, and promote autophagy. In this comprehensive review, we present evidence of the possible connection between IAP and the LPS/Toll-like receptor 4 (TLR4) pathway, impaired gut immunity, and dysbiosis in the preterm gut. Based on these findings, the administration of exogenous IAP might provide promising preventive and therapeutic avenues for the management of NEC.


Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant, Newborn , Humans , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Dysbiosis/drug therapy , Lipopolysaccharides/therapeutic use , Infant, Newborn, Diseases/drug therapy
14.
Semin Pediatr Surg ; 32(3): 151308, 2023 Jun.
Article En | MEDLINE | ID: mdl-37295297

Antibiotic therapy remains a cornerstone of treatment of both medical and surgical presentations of necrotizing enterocolitis (NEC). However, guidelines regarding the administration of antibiotics for the treatment of NEC are lacking and practices vary amongst clinicians. Although the pathogenesis of NEC is unknown, there is consensus that the infant gastrointestinal microbiome contributes to the disease. The presumed connection between dysbiosis and NEC has prompted some to study whether early prophylactic enteral antibiotics can prevent NEC. Yet others have taken an opposing approach, studying whether perinatal antibiotic exposure increases the risk of NEC by inducing a state of dysbiosis. This narrative review summarizes what is known about antibiotics and their association with the infant microbiome and NEC, current antibiotic prescribing practices for infants with medical and surgical NEC, as well as potential strategies to further optimize the use of antibiotics in this population of infants.


Enterocolitis, Necrotizing , Fetal Diseases , Gastrointestinal Microbiome , Infant, Newborn, Diseases , Infant , Pregnancy , Female , Infant, Newborn , Humans , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/etiology , Dysbiosis/complications , Dysbiosis/drug therapy , Anti-Bacterial Agents/therapeutic use
15.
Medicine (Baltimore) ; 102(24): e34011, 2023 Jun 16.
Article En | MEDLINE | ID: mdl-37327300

The objective of this study was to evaluate the effect of maturing fetal lung on clinical efficacy of acetaminophen in the treatment of premature infants with patent ductus arteriosus (PDA). A total of 441 premature infants admitted to our hospital from May 2020 to May 2021 were recruited, including 152 premature infants receiving fetal lung maturation (13 cases of PDA closure with drug use and 2 cases failed) and 289 cases without maturing fetal lung (17 cases of PDA closure and 8 cases failed). Finally, a total of 30 cases were enrolled in this clinical trial. All infants were divided into groups A and B according to whether fetal lung maturation was adopted before delivery. In group A, 13 infants received fetal lung maturation, and 17 in group B did not undergo fetal lung maturation. Infants in both groups were orally given with acetaminophen. After 3-day treatment, the second course of treatment was given immediately if PDA was not closed. The PDA closure rate and patency rate of PDA at the end of 2 treatment courses were statistically compared between 2 groups. The feeding intolerance, upper gastrointestinal bleeding, renal failure, necrotizing enterocolitis, bronchopulmonary dysplasia, periventricular-intraventricular hemorrhage, the age at total enteral nutrition and the length of hospital stay were also compared between 2 groups. After the 1st and 2nd treatment courses, the PDA closure rate in group A was 84.61%, significantly higher than 52.94% in group B (P < .05), whereas there was no significant difference in the PDA patency rate between 2 groups (P > .05). No significant differences were observed regarding the feeding intolerance, renal failure, necrotizing enterocolitis, periventricular-intraventricular hemorrhage, bronchopulmonary dysplasia, the length of hospital stay and the age at total enteral nutrition between 2 groups (all P > .05). In addition, the incidence of upper gastrointestinal bleeding in group A was 7.69%, slightly lower than 5.88% in group B (P > .05). Compared with premature infants untreated with fetal lung maturation interventions before delivery, premature infants who receive fetal lung maturation interventions combined with acetaminophen for PDA are likely to obtain a higher PDA closure rate and a lower incidence rate of the upper gastrointestinal bleeding.


Bronchopulmonary Dysplasia , Ductus Arteriosus, Patent , Enterocolitis, Necrotizing , Humans , Infant, Newborn , Acetaminophen/therapeutic use , Bronchopulmonary Dysplasia/drug therapy , Cerebral Hemorrhage/drug therapy , Ductus Arteriosus, Patent/drug therapy , Enterocolitis, Necrotizing/drug therapy , Gastrointestinal Hemorrhage/drug therapy , Infant, Premature , Lung
16.
Gut Microbes ; 15(1): 2221758, 2023.
Article En | MEDLINE | ID: mdl-37358104

Microbiome-targeting therapies have received great attention as approaches to prevent disease in infants born preterm, but their safety and efficacy remain uncertain. Here we summarize the existing literature, focusing on recent meta-analyses and systematic reviews that evaluate the performance of probiotics, prebiotics, and/or synbiotics in clinical trials and studies, emphasizing interventions for which the primary or secondary outcomes were prevention of necrotizing enterocolitis, late-onset sepsis, feeding intolerance, and/or reduction in hospitalization length or all-cause mortality. Current evidence suggests that probiotics and prebiotics are largely safe but conclusions regarding their effectiveness in the neonatal intensive care unit have been mixed. To address this ambiguity, we evaluated publications that collectively support benefits of probiotics with moderate to high certainty evidence in a recent comprehensive network meta-analysis, highlighting limitations in these trials that make it difficult to support with confidence the routine, universal administration of probiotics to preterm infants.


Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Microbiota , Probiotics , Humans , Infant, Newborn , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/prevention & control , Infant, Premature , Intensive Care Units, Neonatal , Prebiotics , Probiotics/adverse effects
17.
Sci Rep ; 13(1): 8241, 2023 05 22.
Article En | MEDLINE | ID: mdl-37217588

Necrotizing enterocolitis (NEC) brain injury is mediated through Toll-like receptor 4 (TLR4) on the intestinal epithelium and brain microglia. Our aim was to determine whether postnatal and/or prenatal NAC can modify NEC associated intestinal and brain TLR4 expression and brain glutathione levels in a rat model of NEC. Newborn Sprague-Dawley rats were randomized into three groups: Control (n = 33); NEC (n = 32)-hypoxia and formula feeding; and NEC-NAC (n = 34)-received NAC (300 mg/kg IP) in addition to NEC conditions. Two additional groups included pups of dams treated once daily with NAC (300 mg/kg IV) for the last 3 days of pregnancy: NAC-NEC (n = 33) or NAC-NEC-NAC (n = 36) with additional postnatal NAC. Pups were sacrificed on the fifth day, and ileum and brains harvested for TLR-4 and glutathione protein levels. Brain and ileum TLR-4 protein levels were significantly increased in NEC offspring as compared to control (brain 2.5 ± 0.6 vs. 0.88 ± 0.12 U and ileum 0.24 ± 0.04 vs. 0.09 ± 0.01, p < 0.05). When NAC was administered only to dams (NAC-NEC) a significant decrease in TLR-4 levels was demonstrated in both offspring brain (1.53 ± 0.41 vs. 2.5 ± 0.6 U, p < 0.05) and ileum (0.12 ± 0.03 vs. 0.24 ± 0.04 U, p < 0.05) as compared to NEC. The same pattern was demonstrated when NAC was administered only or postnatally. The decrease in brain and ileum glutathione levels observed in NEC offspring was reversed with all NAC treatment groups. NAC reverses the increase in ileum and brain TLR-4 levels and the decrease in brain and ileum glutathione levels associated with NEC in a rat model, and thus may protect from NEC associated brain injury.


Brain Injuries , Enterocolitis, Necrotizing , Animals , Rats , Acetylcysteine/pharmacology , Animals, Newborn , Brain/metabolism , Brain Injuries/metabolism , Disease Models, Animal , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Glutathione/metabolism , Ileum/metabolism , Intestines , Rats, Sprague-Dawley , Rodentia/metabolism , Toll-Like Receptor 4/metabolism
18.
Food Funct ; 14(8): 3769-3778, 2023 Apr 24.
Article En | MEDLINE | ID: mdl-36995017

Necrotizing enterocolitis (NEC) is a life-threatening risk to the health of neonates, but thus far, there is no very effective treatment. Although many studies have confirmed the therapeutic role of peptides in diseases, the effect of peptides in NEC remains poorly understood. This study investigated the role of casein-derived peptide YFYPEL in NEC cells and animal models. We synthesized YFYPEL and analysed its protective effects on NEC both in vitro and in vivo. YFYPEL integration in the intestine increased rat survival and clinical conditions, lowered the incidence of NEC, alleviated bowel inflammation, and enhanced intestinal cell migration. Furthermore, YFYPEL significantly decreased interleukin 6 expression and increased intestinal epithelial cell migration. Moreover, YFYPEL alleviated intestinal epithelial cell dysfunction through the PI3K/AKT pathway, as demonstrated by western blotting and bioinformatics analysis. A selective PI3K activator reversed the protective effect of YFYPEL on lipopolysaccharide-stimulated intestinal epithelial cells. Our study showed that YFYPEL reduced inflammatory cytokine expression and enhanced migration by regulating the PI3K/AKT pathway. The use of YFYPEL may thus develop into a novel modality in NEC treatment.


Enterocolitis, Necrotizing , Intestinal Mucosa , Animals , Rats , Intestinal Mucosa/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Caseins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Epithelial Cells/metabolism , Enterocolitis, Necrotizing/drug therapy , Cytokines/metabolism , Disease Models, Animal
19.
Pediatr Surg Int ; 39(1): 102, 2023 Feb 04.
Article En | MEDLINE | ID: mdl-36738350

PURPOSE: Our previous clinical pilot study reported that miconazole (MCZ) prevented morbidity from surgical necrotizing enterocolitis (NEC). The present study re-investigated this effect in a long-term cohort over 20 years. METHODS: We conducted a retrospective cohort study from April 1998 to March 2020. A total of 1169 extremely low-birth-weight infants (ELBWIs) admitted to our neonatal intensive care unit, including 45 with NEC (3.8%), underwent surgery. Since 2002, protocol MCZ administration for 3 weeks has been applied for neonates born before 26 weeks' gestation or weighing under 1000 g. We compared the background characteristics and clinical outcomes between patients with and without MCZ administration. RESULTS: The morbidity rate decreased after applying the MCZ protocol, but no improvement in mortality was seen. A propensity score-matched analysis indicated that treated patients by MCZ showed a delay in developing surgical NEC by 12 days. The MCZ protocol also helped increase body weight at surgery. Prophylactic MCZ administration did not improve the neurological development of the language-social and postural-motor domains in the surgical NEC patients. But cognitive-adaptive domain caught up by a chronological age of 3 years old. CONCLUSIONS: Revising the protocol to extend the dosing period may improve the outcomes of surgical NEC after the onset.


Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant , Infant, Newborn , Humans , Child, Preschool , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/surgery , Miconazole/therapeutic use , Retrospective Studies , Pilot Projects , Morbidity
20.
Tissue Cell ; 81: 102003, 2023 Apr.
Article En | MEDLINE | ID: mdl-36682224

OBJECTIVE: Necrotizing enterocolitis (NEC) is a severe disease in newborns, this study aimed to investigate the protective effect of dimethyl fumarate (DMF) on NEC and its possible mechanism. METHODS: In vivo, the mice were divided into the control, NEC, and NEC+DMF group. The NEC model was established by artificial feeding, hypoxic for 4 days, and lipopolysaccharide (LPS) stimulation on day 2 and day 3. DMF (25 mg/kg/d) was administered to NEC mice on day 1 and day 3. On the 11th day, the blood and intestinal tissues of mice were taken for enzyme-linked immunosorbent assay (ELISA), pathological examination, quantitative real-time PCR (RT-qPCR), Western blot, and immunohistochemical (IHC) detection. In vitro, human colorectal cells (FHC) were induced by LPS (100 ng/mL) and was divided into the control, LPS, and LPS+DMF group. The effect of DMF (20 µM) on cell viability and TLR4 signal transduction was detected by MTT and RT-qPCR, respectively. RESULTS: Compared to the NEC mice, DMF attenuated NEC-induced weight loss and abdominal distension diarrhea in mice, and alleviated NEC-induced intestinal pathological injuries. In addition, DMF reduced the expression of IL-6, IL-1ß, TNF-α, NF-κB, and TLR4 in NEC mice intestinal tissues. Furthermore, DMF inhibited NEC-induced intestinal cell apoptosis as well as the protein expression of BCL2-Associated X (BAX), caspase-3, caspase-9, and increased Bcl-2 (B-cell lymphoma-2) expression. In vitro, DMF improved cell viability, and restrained NF-κB and TLR4 expression in LPS-induced NEC cells. CONCLUSION: DMF has a protective effect against intestine damage of NEC, which is related to the inhibition of the TLR signaling pathway, alleviating the inflammatory response.


Enterocolitis, Necrotizing , NF-kappa B , Animals , Mice , Infant, Newborn , Humans , NF-kappa B/metabolism , Dimethyl Fumarate/pharmacology , Toll-Like Receptor 4/metabolism , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/metabolism , Lipopolysaccharides/toxicity , Signal Transduction , Intestines/pathology , Disease Models, Animal
...