Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
J Int Med Res ; 52(6): 3000605241254788, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867509

ABSTRACT

OBJECTIVE: Neonatal necrotizing enterocolitis (NEC) is a serious intestinal inflammatory disease. We investigated intestinal fatty acid binding protein (I-FABP), I-FABP mRNA, and interleukin-6 (IL-6) as potential diagnostic biomarkers in NEC. METHODS: Forty mice were subjected to hypoxic-ischemic intestinal injury, and then serum I-FABP protein and mRNA levels were quantified. Ileal tissue pathological scores were determined by hematoxylin and eosin staining. I-FABP expression levels and translocation in these tissues were detected using western blotting and immunofluorescence, respectively. Samples from 30 human neonates with NEC and 30 healthy neonates had serum I-FABP protein/mRNA and IL-6 levels measured. RESULTS: The mouse ileal tissue pathological score and I-FABP levels, as well as serum I-FABP and I-FABP mRNA levels, were significantly higher in the model group than in the control group. Serum I-FABP, I-FABP mRNA, and IL-6 levels were significantly higher in human neonates with NEC than in the healthy group. Logistic regression and receiver operating curve analyses revealed that I-FABP protein/mRNA and IL-6 levels could be diagnostic biomarkers for NEC. CONCLUSIONS: I-FABP protein/mRNA and IL-6 levels are useful biomarkers of intestinal ischemic injury in neonates with NEC. The combined detection of I-FABP protein/mRNA and IL-6 is recommended rather than using a single biomarker.


Subject(s)
Biomarkers , Disease Models, Animal , Enterocolitis, Necrotizing , Fatty Acid-Binding Proteins , Interleukin-6 , Mice, Inbred BALB C , RNA, Messenger , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/diagnosis , Animals , Fatty Acid-Binding Proteins/blood , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Interleukin-6/blood , Interleukin-6/genetics , Infant, Newborn , Humans , Biomarkers/blood , Biomarkers/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/blood , Mice , Male , Female , Animals, Newborn , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ileum/metabolism , Ileum/pathology , Case-Control Studies , ROC Curve
2.
Sci Rep ; 14(1): 10783, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734725

ABSTRACT

Necrotising enterocolitis (NEC) has a complex pathophysiology but the common end-point is ischaemia reperfusion injury (IRI) and intestinal necrosis. We have previously reported that RIC significantly reduces the intestinal injury in a rat model of NEC. Here we describe the changes in intestinal mRNA occurring in the intestine of animals exposed to IRI, both with and without RIC. Related rat-pups were randomly assigned to four groups: SHAM, IRI only, RIC only and RIC + IRI. IRI animals, underwent 40 min of intestinal ischaemia, and 90 min of reperfusion. Animals that underwent RIC had three cycles of 5 min of alternating ischaemia/reperfusion by means of a ligature applied to the hind limb. Samples from the terminal ileum were immediately stored in RNA-preserving media for later next generation sequencing and transciptome analysis using R v 3.6.1. Differential expression testing showed that 868 genes differentially expressed in animals exposed to RIC alone compared to SHAM and 135 in the IRI and RIC group compared to IRI alone. Comparison between these two sets showed that 25 genes were differentially expressed in both groups. Pro-inflammatory molecules: NF-ĸß2, Cxcl1, SOD2 and Map3k8 all show reduced expression in response to RIC. Targeted gene analysis revealed increased expression in PI3K which is part of the so-called RISK-pathway which is a key part of the protective mechanisms of RIC in the heart. Overall, this transcriptomic analysis shows that RIC provides a protective effect to the intestine via anti-inflammatory pathways. This could be particularly relevant to treating and preventing NEC.


Subject(s)
Disease Models, Animal , Enterocolitis, Necrotizing , Gene Expression Profiling , Reperfusion Injury , Animals , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/metabolism , Rats , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Ischemic Preconditioning/methods , Transcriptome
3.
Pediatr Surg Int ; 40(1): 126, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717494

ABSTRACT

BACKGROUND: Neonatal necrotizing enterocolitis (NEC) is a common gastrointestinal emergency in neonates. MiRNA-192-5p was found associated with ulcerative colitis (UC) progression, also with aberrant expression in intestinal cancer tissue. However, the effects of miRNA-192-5p on NEC have not been reported. METHODS: Based on the bioinformatics analysis of the GEO dataset, miR-192-5p was identified as the differentially expressed miRNA in NEC, and activated leukocyte cell adhesion molecule (ALCAM) was predicted as its target. After that, in vitro, rat intestinal epithelial cell-6 (IEC-6) were stimulated with LPS to construct a cell model of NEC. IEC-6 cells were transfected with miRNA-192-5p mimics, miRNA-192-5p inhibitors, or miRNA-192-5p inhibitors + sh-ALCAM, and relevant negative control. In vivo, SD rats were treated with artificial feeding, hypoxic reoxygenation, cold stimulation, and LPS gavage to induce NEC, followed by injection of agomiR-NC or agomiRNA-192-5p. Then effects of miRNA-192-5p on NEC model IEC-6 cell viability, apoptosis, ALCAM expression, Interleukin (IL)-1ß and IL-6 levels, intestinal injury, intestinal permeability were detected. RESULTS: MiRNA-192-5p expression was downregulated in NEC IEC-6 cells, whose overexpression increased IEC-6 cell viability. MiRNA-192-5p inhibitors increased IL-1ß, IL-6 levels and promoted IEC-6 cell apoptosis. MiRNA-192-5p targeting of ALCAM decreased ALCAM expression, IL-1ß, and IL-6 levels. AgomiRNA-192-5p decreased ALCAM, IL-1ß, and IL-6 levels in intestinal tissue and pathological damage and increased miRNA-192-5p levels. CONCLUSION: MiR-192-5p protects against intestinal injury by inhibiting ALCAM-mediated inflammation and intestinal epithelial cells, which would provide a new idea for NEC treatment.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule , Disease Models, Animal , Enterocolitis, Necrotizing , MicroRNAs , Rats, Sprague-Dawley , Animals , Humans , Infant, Newborn , Rats , Animals, Newborn , Apoptosis/genetics , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Inflammation , MicroRNAs/genetics , Activated-Leukocyte Cell Adhesion Molecule/genetics , Activated-Leukocyte Cell Adhesion Molecule/metabolism
4.
Expert Rev Mol Med ; 26: e16, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557638

ABSTRACT

Epigenetic modifications, such as DNA methylation, are enzymatically regulated processes that directly impact gene expression patterns. In early life, they are central to developmental programming and have also been implicated in regulating inflammatory responses. Research into the role of epigenetics in neonatal health is limited, but there is a growing body of literature related to the role of DNA methylation patterns and diseases of prematurity, such as the intestinal disease necrotizing enterocolitis (NEC). NEC is a severe intestinal inflammatory disease, but the key factors that precede disease development remain to be determined. This knowledge gap has led to a failure to design effective targeted therapies and identify specific biomarkers of disease. Recent literature has identified altered DNA methylation patterns in the stool and intestinal tissue of neonates with NEC. These findings provide the foundation for a new avenue in NEC research. In this review, we will provide a general overview of DNA methylation and then specifically discuss the recent literature related to methylation patterns in neonates with NEC. We will also discuss how DNA methylation is used as a biomarker for other disease states and how, with further research, methylation patterns may serve as potential biomarkers for NEC.


Subject(s)
DNA Methylation , Enterocolitis, Necrotizing , Animals , Humans , Biomarkers , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Epigenesis, Genetic
5.
Microbiol Spectr ; 11(6): e0276623, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37909758

ABSTRACT

IMPORTANCE: Clostridium neonatale has been isolated from the fecal samples of asymptomatic neonates and cases of necrotizing enterocolitis (NEC). Taking advantage of a large collection of independent strains isolated from different spatio-temporal settings, we developed and established a cgMLST scheme for the molecular typing of C. neonatale. Both the cgMLST and cgSNP methods demonstrate comparable discrimination power. Results indicate geographic- and temporal- independent clustering of C. neonatale NEC-associated strains. No specific cgMLST clade of C. neonatale was genetically associated with NEC.


Subject(s)
Clostridium , Enterocolitis, Necrotizing , Infant, Newborn , Humans , Multilocus Sequence Typing/methods , Enterocolitis, Necrotizing/genetics , Genome, Bacterial
6.
Acta Biochim Pol ; 70(4): 905-910, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38015074

ABSTRACT

OBJECTIVE: Necrotizing enterocolitis (NEC) is a devastating inflammatory disease with high morbidity and mortality, mainly affecting premature infants. This study aimed to explore the role of miRNA-301a in the pathogenesis of NEC. METHODS: The differentially expressed miRNAs and mRNAs were screened by collating RNA-Seq data from the GEO database of intestinal tissue samples. The differential miRNA-mRNAs regulatory network was constructed based on functional enrichment analysis. Newborn BALB/c mice were used to establish the NEC model. Haematoxylin and eosin staining was used to assess intestinal damage. The levels of IL-8 and TNF-α in mouse serum were evaluated by ELISA. qRT-PCR was used to detect the expression of miRNA-301a in intestinal tissues. RESULTS: Bioinformatics analysis showed that miRNA-301a was involved in intestinal lesions. Intestinal tissue damage was reduced and serum levels of the inflammatory cytokines IL-8 and TNF-α were lower in NEC model mice treated with miRNA-301a antagonists. The level of miRNA-301a in intestinal tissues of NEC model mice was significantly higher than in the control group and miRNA-301a antagonists treated group. CONCLUSION: miRNA-301a plays an important role in the pathogenesis of NEC by promoting inflammation, and is a potential therapeutic target of NEC.


Subject(s)
Enterocolitis, Necrotizing , MicroRNAs , Animals , Humans , Infant , Infant, Newborn , Mice , Disease Models, Animal , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Inflammation/pathology , Interleukin-8/genetics , MicroRNAs/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Epigenomics ; 15(8): 479-486, 2023 04.
Article in English | MEDLINE | ID: mdl-37309586

ABSTRACT

Background: Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants. Epigenetic changes in DNA methylation may be present prior to NEC onset. Methods: 24 preterm infants with NEC and 45 matched controls were included. Human DNA was isolated from stool samples and methylation of CTDSPL2, HERC1, NXPE3 and PTGDR was measured using pyrosequencing. Results: CTDSPL2 displayed a higher DNA methylation of 51% compared with 17% in controls, prior to NEC onset (p = 0.047). Discussion: Noninvasive measurement of methylation in stool allows for comparison with healthy preterm controls. This potentially allows future biomarker or risk predictor use. The effect of CTDSPL2 hypermethylation on gene expression remains unclear.


What is this article about? Necrotizing enterocolitis (NEC) is a common emergency condition affecting the gastrointestinal system of preterm infants. Epigenetic changes in DNA methylation may be present in infants before the onset of NEC. DNA methylation is a natural process that can help turn genes on or off, thereby affecting their function. This study focused on measuring the amount of DNA methylation in certain genes in preterm infants who developed NEC. What were the results? This study included 24 preterm infants with NEC and 45 matched healthy controls. The researchers isolated human DNA from stool samples, and the amount of DNA methylation of four specific genes was measured. They found that one of the genes, CTDSPL2, had significantly higher DNA methylation in infants who later developed NEC than in healthy infants. What do the results of the study mean? In this study, researchers found that CTDSPL2 showed a higher level of DNA methylation in stool samples of infants who later developed NEC. The effect of this change remains unclear, but may affect the way cells grow and respond to injury or infection, which could contribute to the development of NEC. Measuring DNA methylation in stool samples provides a noninvasive method for identifying DNA methylation changes in preterm infants. Comparing the amount of DNA methylation in healthy infants with that in preterm infants at risk of NEC may help predict the risk of developing NEC.


Subject(s)
Enterocolitis, Necrotizing , Infant, Premature , Humans , Infant , Infant, Newborn , DNA Methylation , Enterocolitis, Necrotizing/genetics , Feces
8.
PLoS Biol ; 21(5): e3002124, 2023 05.
Article in English | MEDLINE | ID: mdl-37205711

ABSTRACT

Necrotizing enterocolitis (NEC) is a gastrointestinal complication of premature infants with high rates of morbidity and mortality. A comprehensive view of the cellular changes and aberrant interactions that underlie NEC is lacking. This study aimed at filling in this gap. We combine single-cell RNA sequencing (scRNAseq), T-cell receptor beta (TCRß) analysis, bulk transcriptomics, and imaging to characterize cell identities, interactions, and zonal changes in NEC. We find an abundance of proinflammatory macrophages, fibroblasts, endothelial cells as well as T cells that exhibit increased TCRß clonal expansion. Villus tip epithelial cells are reduced in NEC and the remaining epithelial cells up-regulate proinflammatory genes. We establish a detailed map of aberrant epithelial-mesenchymal-immune interactions that are associated with inflammation in NEC mucosa. Our analyses highlight the cellular dysregulations of NEC-associated intestinal tissue and identify potential targets for biomarker discovery and therapeutics.


Subject(s)
Enterocolitis, Necrotizing , Infant , Infant, Newborn , Humans , Enterocolitis, Necrotizing/genetics , Endothelial Cells , Intestine, Small , Infant, Premature , Intestines , Intestinal Mucosa
9.
Georgian Med News ; (336): 79-84, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37166885

ABSTRACT

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in preterm infants. Despite significant advances made in the prevention and treatment of disease so far, there has not been much change in the rate of mortality and morbidity associated with NEC. Although the factors affecting the development of necrotizing enterocolitis are not yet known precisely, prematurity is thought to be the most important risk factor for the development of NEC. This study aims to determine toll-like receptor (TLR) 2 and TLR4 expression levels in preterm neonates. A total of 50 preterm infants (patient: 42, control: 8) were included in the study. TLR2 and TLR4 expression levels were analysed by the RT-qPCR method. While there was no difference in infants' birth weight (g), gestational age (months), mother's age (years), father's age (years), and WBC (109/L); HGB (g/dL) and RBC (1012/L) were found to be significantly higher in the group with NEC (p<0.05). When TLR2 and TLR4 relative gene expression levels of neonates were evaluated (log2), it was determined that there was a significant difference between the two groups (below 1500 g) (p<0.001). TLR4 relative expression (2^-ddCt, above 1500 g) was higher in the NEC group than in the healthy group, while TLR2 relative expression (2^-ddCt, above 1500 g) was higher in the healthy group. TLR2 and TLR4 have been shown to have prominent roles in the development of NEC in experimental animal models and it would be significant to support this with human studies/animal models for a better understanding of the disease. Thus, it is recommended that future studies be carried out on experimental models that better replicate the human body, and dietary factors should be examined in detail.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Humans , Infant , Infant, Newborn , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/prevention & control , Gestational Age , Infant, Premature , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
10.
Int J Mol Sci ; 24(3)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36768695

ABSTRACT

Epigenetic immune cell counting is a DNA (de)methylation-based technique which can be used to quantify lymphocyte subsets on dried blood spots (DBS). The foregoing techniques allow for a retrospective investigation of immune cell profiles in newborns. In this study, we used this technique for determining lymphocyte subcounts as a potential biomarker for necrotizing enterocolitis (NEC). We investigated whether this technique can be implemented in the field of neonatology, by testing whether regulatory T cell (Treg) levels are pre-existently low in preterms with NEC. Newborn screening (NBS) cards from 32 preterms with NEC and 32 age- and weight-matched preterm controls, and 60 healthy term newborns, were analyzed. Relative and absolute cell counts were determined for CD3+, CD4+, CD8+, Th17, and Treg T cells. For both relative and absolute cell counts of CD3+, CD4+, CD8+, and Th17 T cells, significant differences were found between healthy term controls and both preterm groups, but not between preterm groups. For Tregs, no significant differences were found in either relative or absolute counts between any of the newborn groups. This study demonstrates the principle of epigenetic immune cell counting to analyze lymphocyte subsets in preterm neonates.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Infant , Female , Infant, Newborn , Humans , Infant, Premature , Retrospective Studies , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/genetics , Biomarkers , Lymphocyte Count , Epigenesis, Genetic
11.
Arq Bras Cir Dig ; 35: e1717, 2023.
Article in English | MEDLINE | ID: mdl-36629694

ABSTRACT

BACKGROUND: Abnormalities in the different stages of the intestinal maturation process cause metabolic and molecular changes. Among the genetic alterations associated with necrotizing enterocolitis, the -94ins/delATTG polymorphism in NFKB1 gene leads to unregulated activation of the NFKB protein due to an increase in the inherent pro-inflammatory state of the premature intestine. AIMS: To determine the prevalence of the -94ins/delATTG polymorphism in NFKB1 gene in neonates with and without necrotizing enterocolitis. METHODS: This is a case-control study, in which 25 neonates were evaluated as the case group and 50 neonates as the control group, of both genders. DNA was extracted from peripheral blood leukocytes, and the site encompassing the polymorphism was amplified by molecular techniques (polymerase chain reaction/polymorphism in restriction fragment length). RESULTS: Necrotizing enterocolitis was diagnosed in 25 (33%) neonates and, of these, 3 (12%) died. Male gender was more prevalent in both groups (p=0.1613): cases (52%) and controls (62%). Moderate and extreme preterm newborns were predominant in both groups: cases (80%) and controls (88%) (p=0.3036). Low birth weight and extremely low birth weight newborns were the most prevalent in cases (78%), and very low birth weight and extremely low birth weight were the most prevalent in controls (81%) (p=0.1073). Clinical treatment was successful in 72%, and hospital discharge was achieved in 88% of newborns with NEC. The -94ins/delATTG polymorphism in NFKB1 gene was not identified in all the 150 alleles analyzed (100%). CONCLUSIONS: The absence of the -94ins/delATTG polymorphism in NFKB1 gene in newborns with and without necrotizing enterocolitis does not rule out the possibility of alterations in this and/or in other genes in newborns with this condition, which reinforces the need for further research.


Subject(s)
Enterocolitis, Necrotizing , Rectal Neoplasms , Male , Humans , Infant, Newborn , Female , Case-Control Studies , Enterocolitis, Necrotizing/genetics , NF-kappa B p50 Subunit/genetics , Polymorphism, Genetic/genetics , Mutation
12.
Pediatr Res ; 94(1): 275-279, 2023 07.
Article in English | MEDLINE | ID: mdl-36596941

ABSTRACT

BACKGROUND: Vitamin D and its receptor (VDR) effects on the gastrointestinal system are among its most critical multisystemic effects. METHODS: This study aimed to reveal that VDR gene polymorphisms may constitute a risk factor for necrotizing enterocolitis (NEC). VDR Fok1-Bsm1-Apa single-nucleotide polymorphisms were analyzed in the NEC group (n = 74) and the control group (n = 147). Among 1112 babies at and below 36 weeks of gestational age who were hospitalized between January 2013 and December 2016 with a diagnosis of prematurity, 74 of a total of 148 patients who developed NEC during follow-up (NEC group) were included in the study. When NEC was diagnosed according to clinical and radiological findings and staged using Modified Bell criteria, 9 (12.1%) of 74 babies were stage 1A, 13 (17.5%) stage 1B, and 5 (6.7%) stage 2A, 33 (44.5%) stage 2B, 7 (9.4%) stage 3A, 7 (9.4%) stage 3B. Of 964 babies who did not develop NEC during follow-up, 147 were included as the control group in the study. Genotyping of VDR polymorphisms was assayed by real-time PCR. From 221 premature babies in the NEC and control groups, 2 ml peripheral blood was taken appropriately and meticulously into an EDTA tube. DNA was isolated from these blood samples. DNA amplification was performed using a thermal cycler (Applied Biosystems GeneAmp PCR System 9600). RESULTS: When the two groups were compared in terms of the prevalence of VDR Fok1 C/T genotype, it was found that TT genotype increased the risk of NEC by 2.697 times, and there was a significant relationship between TT genotype and the risk of NEC (p = 0.041). Multivariable logistic regression analysis was performed in terms of gestational age, birth weight, VDR gene polymorphism data between NEC and the control group. According to the analysis results, TT polymorphism, increased the risk of disease 4.5 times (p = 0.033). CONCLUSION: Fok 1 C > T polymorphism in the VDR gene plays a role in the development of NEC. Identifying the risk groups by detecting gene polymorphisms that cause increased susceptibility to NEC may assist in the follow-up of these patients and in making early treatment decisions for them. IMPACT: In this study examining the non-bone effects of the genetic differences in vitamin D metabolism in premature babies, Fok 1 polymorphism has been observed to be an essential risk factor for NEC. This is the first study in our country that has investigated the relationship between VDR gene polymorphism and necrotizing enterocolitis among the Turkish population. Identifying the risk groups by detecting gene polymorphisms that cause increased susceptibility to NEC may assist in the monitoring of these patients and in making early treatment decisions for them.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Female , Infant, Newborn , Humans , Receptors, Calcitriol/genetics , Enterocolitis, Necrotizing/genetics , Polymorphism, Single Nucleotide , Genotype , Real-Time Polymerase Chain Reaction , Vitamin D , Genetic Predisposition to Disease
13.
Immunol Invest ; 52(3): 257-269, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36576246

ABSTRACT

OBJECTIVE: Pleckstrin homology-like domain family A member 1 (PHLDA1) is involved in the progression of intestine-related diseases, but its role and related mechanisms in Necrotizing enterocolitis (NEC) are unclear. The aim of this study was to better understand the function of PHLDA1 in NEC and the underlying mechanisms. METHODS: A neonatal mouse model of NEC was established by hypoxic hypothermia, and sh-PHLDA1 was transfected into mice to observe the mortality of each group within 4 days. The levels of IL-1ß, IL-6, IL-18 and TNF-α were measured by PCR and ELISA. ROS, MDA, SOD, and GSH-Px levels were detected by Dihydroethidium (DHE) method and kit; expression of pyroptosis-related factors including NLRP3, ASC, cleaved-caspase1, GSDMD-N, IL-1ß, IL-18, and Nrf2 were detected by western-blot; mechanistically, the effects of transfection of sh-PHLDA1 and ML385 (Nrf2 inhibitor) were investigated, and the expression of pyroptosis-related factors was detected again. RESULTS: PHLDA1 was highly expressed in the intestinal tissues of NEC mice, and transfection of sh-PHLDA1 improved the survival rate, alleviated intestinal lesions, improved intestinal inflammation, oxidative stress and cellular scorching in NEC. In addition, sh-PHLDA1 was able to inhibit NLRP3 activation and pyroptosis by activating Nrf2. CONCLUSION: Knockdown of PHLDA1 attenuated necrotizing small intestinal colitis by enhancing Nrf2 expression to inhibit NLRP3 inflammasome activation and pyroptosis.


Subject(s)
Enterocolitis, Necrotizing , Inflammasomes , Transcription Factors , Animals , Mice , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Inflammasomes/metabolism , Interleukin-18 , NF-E2-Related Factor 2/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis
14.
Am J Physiol Gastrointest Liver Physiol ; 324(1): G24-G37, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36410023

ABSTRACT

Single immunoglobulin interleukin-1-related receptor (SIGIRR), toll-interacting protein (TOLLIP), and A20 are major inhibitors of toll-like receptor (TLR) signaling induced postnatally in the neonatal intestine. Short-chain fatty acids (SCFAs), fermentation products of indigestible carbohydrates produced by symbiotic bacteria, inhibit intestinal inflammation. Herein, we investigated the mechanisms by which SCFAs regulate SIGIRR, A20, and TOLLIP expression and mitigate experimental necrotizing enterocolitis (NEC). Butyrate induced NOTCH activation by repressing sirtuin 1 (SIRT1)-mediated deacetylation of the Notch intracellular domain (NICD) in human intestinal epithelial cells (HIECs). Overexpression of NICD induced SIGIRR, A20, and TOLLIP expression. Chromatin immunoprecipitation revealed that butyrate-induced NICD binds to the SIGIRR, A20, and TOLLIP gene promoters. Notch1-shRNA suppressed butyrate-induced SIGIRR/A20 upregulation in mouse enteroids and HIEC. Flagellin (TLR5 agonist)-induced inflammation in HIEC was inhibited by butyrate in a SIGIRR-dependent manner. Neonatal mice fed butyrate had increased NICD, A20, SIGIRR, and TOLLIP expression in the ileal epithelium. Butyrate inhibited experimental NEC-induced intestinal apoptosis, cytokine expression, and histological injury. Our data suggest that SCFAs can regulate the expression of the major negative regulators of TLR signaling in the neonatal intestine through Notch1 and ameliorate experimental NEC. Enteral SCFAs supplementation in preterm infants provides a promising bacteria-free, therapeutic option for NEC.NEW & NOTEWORTHY Short-chain fatty acids (SCFAs), such as propionate and butyrate, metabolites produced by symbiotic gut bacteria are known to be anti-inflammatory, but the mechanisms by which they protect against NEC are not fully understood. In this study, we reveal that SCFAs regulate intestinal inflammation by inducing the key TLR and IL1R inhibitors, SIGIRR and A20, through activation of the pluripotent transcriptional factor NOTCH1. Butyrate-mediated SIGIRR and A20 induction represses experimental NEC in the neonatal intestine.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn , Animals , Mice , Humans , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/genetics , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Infant, Premature , Inflammation/metabolism , Intestinal Mucosa/metabolism , Fatty Acids, Volatile/pharmacology , Fatty Acids, Volatile/metabolism , Butyrates/metabolism , Immunoglobulins/metabolism , Interleukin-1/metabolism , Receptor, Notch1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
15.
BMC Pediatr ; 22(1): 658, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371157

ABSTRACT

OBJECTIVE: To detect differentially expressed genes in patients with neonatal necrotizing enterocolitis (NEC) by bioinformatics methods and to provide new ideas and research directions for the prevention, early diagnosis and treatment of NEC. METHODS: Gene chip data were downloaded from the Gene Expression Omnibus database. The genes that were differentially expressed in NEC compared with normal intestinal tissues were screened with GEO2R. The functions, pathway enrichment and protein interactions of these genes were analyzed with DAVID and STRING. Then, the core network genes and significant protein interaction modules were detected using Cytoscape software. RESULTS: Overall, a total of 236 differentially expressed genes were detected, including 225 upregulated genes and 11 downregulated genes, and GO and KEGG enrichment analyses were performed. The results indicated that the upregulated differentially expressed genes were related to the dimerization activity of proteins, while the downregulated differentially expressed genes were related to the activity of cholesterol transporters. KEGG enrichment analysis revealed that the differentially expressed genes were significantly concentrated in metabolism, fat digestion and absorption pathways. Through STRING analysis, 9 key genes in the protein network interaction map were identified: EPCAM, CDH1, CFTR, IL-6, APOB, APOC3, APOA4, SLC2A and NR1H4. CONCLUSION: Metabolic pathways and biological processes may play important roles in the development of NEC. The screening of possible core targets by bioinformatics is helpful in clarifying the pathogenesis of NEC at the gene level and in providing references for further research.


Subject(s)
Computational Biology , Enterocolitis, Necrotizing , Humans , Infant, Newborn , Computational Biology/methods , Gene Regulatory Networks , Gene Expression Profiling/methods , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , Protein Interaction Maps
16.
Pediatr Surg Int ; 38(12): 1971-1979, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36208323

ABSTRACT

PURPOSE: Preterm infants are more susceptible to necrotizing enterocolitis (NEC) than term Queryinfants. This may be due to a relative paucity of Lgr5+ or Bmi1+-expressing intestinal stem cells (ISCs) which are responsible for promoting intestinal recovery after injury. We hypothesized that the cellular markers of Lgr5+ and Bmi1+, which represent the two distinct ISC populations, would be lower in younger mice compared to older mice. In addition, we hypothesized that experimental NEC would result in a greater loss of Lgr5+ expression compared to Bmi1+ expression. METHODS: Transgenic mice with EGFP-labeled Lgr5 underwent euthanasia at 10 different time points from E15 to P56 (n = 8-11/group). Lgr5+-expressing ISCs were quantified by GFP ELISA and Bmi1+ was assessed by qPCR. In addition, Lgr5EGFP mice underwent experimental NEC via formula feeding and hypoxic and hypothermic stress. Additional portions of the intestine underwent immunostaining with anti-GFP or anti-Bmi1+ antibodies to confirm ELISA and PCR results. For statistical analysis, p < 0.05 was significant. RESULTS: Lgr5+ and Bmi1+expression was lowest in embryonal and early postnatal mice and increased with age in all segments of the intestine. Experimental NEC was associated with loss of Lgr5+-expressing ISCs but no significant change in Bmi1+ expression. CONCLUSION: Lgr5+ and Bmi1+ expression increase with age. Lgr5+-expressing ISCs are lower following experimental necrotizing enterocolitis while Bmi1+ expression remains relatively unchanged. Developing a targeted medical therapy to protect the low population of ISCs in preterm infants may promote tissue recovery and regeneration after injury from NEC.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant, Newborn , Humans , Mice , Animals , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Intestinal Mucosa/metabolism , Infant, Premature , Stem Cells/metabolism , Intestines , Mice, Transgenic
17.
BMC Pediatr ; 22(1): 617, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289463

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a disastrous gastrointestinal disease of newborns, and the mortality rate of infants with NEC is approximately 20%-30%. The exploration of pathogenic targets of NEC will be conducive to timely diagnosis of NEC. METHODS: The whole transcriptome RNA sequencing was performed on NEC samples to reveal the expression of lncRNAs, circRNAs, miRNAs and mRNAs. Using differential expression analysis, cross analysis, target prediction, enrichment analysis, the pathogenic ceRNA network and target was found. RESULTS: Preliminarily, 281 DEmRNAs, 21 DEmiRNAs, 253 DElncRNAs and 207 DEcircRNAs were identified in NEC samples compared with controls. After target prediction and cross analyses, a key ceRNA regulatory network was built including 2 lncRNAs, 4 circRNAs, 2 miRNAs and 20 mRNAs. These 20 mRNAs were significantly enriched in many carbohydrate metabolism related pathways. After cross analysis of hypoxia-, carbohydrate metabolism-related genes, and 20 core genes, one gene HK2 was finally obtained. Dendritic cells activated were significantly differentially infiltrated and negatively correlated with HK2 expression in NEC samples. CONCLUSIONS: The promising pathogenic hypoxia-related gene HK2 has been firstly identified in NEC, which might also involve in the carbohydrate metabolism in NEC.


Subject(s)
Enterocolitis, Necrotizing , MicroRNAs , RNA, Long Noncoding , Humans , Infant , Infant, Newborn , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , Hypoxia/genetics , RNA, Circular , RNA, Long Noncoding/genetics , RNA, Messenger/genetics
18.
OMICS ; 26(8): 440-450, 2022 08.
Article in English | MEDLINE | ID: mdl-35900271

ABSTRACT

Breastfeeding reduces the risk of necrotizing enterocolitis (NEC), one of the most common causes of morbidity and mortality in preterm infants. However, the molecular substrates by which human milk (HM) offers protection against NEC are not well known. Using fetal intestinal epithelial cells treated with known NEC aggravators, namely lipopolysaccharide (LPS) and platelet-activating factor (PAF), we mapped the time-course of changes in targeted expression analysis of 35 NEC-associated genes, so-called the NEC signature. We found, first, that HM treatment fully rescued LPS/PAF-induced fetal intestinal cell death at 12 and 24 h (n = 5). Differential gene expression and bioinformatics revealed that HM did not mitigate inflammatory and cell death signals, but instead promoted cell proliferation and stress response pathways to mitigate LPS/PAF-induced inflammatory cell death. From this, epidermal growth factor (EGF) synthesis emerged as the central player in rescue of the fetal intestinal cell death. Functional validation was supported by reversal of the cellular rescue by HM following EGF knockdown by small interfering RNA. In conclusion, this study suggests that HM might offer protection against NEC through enhancing intestinal EGF production to rescue the inflammatory cell death. Future studies are warranted to verify these HM molecular protective effects in NEC models in vivo. The findings reported herein also support future research avenues to discover new therapeutics to boost intrinsic EGF production in the injured intestinal tissues in neonates with NEC, for example, by bioactive components in human milk, natural compounds, or small molecules.


Subject(s)
Enterocolitis, Necrotizing , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Epidermal Growth Factor/analysis , Epidermal Growth Factor/pharmacology , Epithelial Cells , Humans , Infant , Infant, Newborn , Infant, Premature , Lipopolysaccharides/adverse effects , Lipopolysaccharides/analysis , Milk, Human/chemistry
19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(3): 414-420, 2022 May.
Article in Chinese | MEDLINE | ID: mdl-35642148

ABSTRACT

Objective: To investigate the expression and role of programmed death ligand-1 (PD-L1) in a mouse model of necrotizing enterocolitis (NEC). Methods: A total of 20 wild-type C57 BL/6 J mice were randomly assigned to the control and the model groups. Mice in the control group were breastfed, while mice in the model group were given lipopolysaccharide, formula feeding, hypoxia, and cold stimulation for NEC induction. Then, the intestines of the mice were collected in order to assess the pathological changes through HE staining, to examine PD-L1 expression and localization with immunofluorescence co-localization, and to evaluate intestinal PD-L1 expression with Western blot. Peripheral blood was collected for flow cytometry to examine leukocyte subpopulations and their PD-L1 expression. On the other hand, 14 PD-L1 (+/+) mice and 14 PD-L1 (-/-) mice were randomly divided into their respective genotype control groups and model groups. The same induction method as was already mentioned was adopted for the model groups. The intestines of the mice were collected for HE staining to evaluate the pathological change and peripheral blood was collected to examine the expression of inflammatory factors. Results: The NEC mouse model was successfully constructed. PD-L1 was widely expressed in enterocytes and inflammatory cells in the mouse intestines and in T cells, monocytes, and neutrophils in peripheral blood. The expression of PD-L1 in NEC mouse intestines increased in comparison with that of the control group. In the peripheral blood of NEC mice, the proportion of T cells and monocytes and their PD-L1 expression showed no significant changes compared with those of the control group, while the proportion of neutrophils and their PD-L1 expression increased by about 140% and 150%, respectively, in comparison with those of the control group ( P<0.05). According to the results of the PD-L1 gene mouse experiment, the control groups of PD-L1 (+/+) mice and PD-L1 (-/-) mice showed no significant difference in their intestinal conditions and serum inflammatory factor levels, while the PD-L1 (-/-) NEC mouse had worse intestinal pathological changes and increased mean pathological scores compared with those of PD-L1 (+/+) NEC mouse ( P<0.05). In addition, serum interleukin (IL)-10 in PD-L1 (-/-) NEC mouse decreased by about 44% compared with that of PD-L1 (+/+) NEC mice, and chemokine (C-X-C motif) ligand 1/IL-6/IL-1ß all increased by more than 25% (all P<0.05). Conclusion: PD-L1 is widely expressed in inflammatory cells and enterocytes in mice. Knocking out PD-L1 aggravates the degree of NEC inflammation and intestinal pathological changes. PD-L1 plays a protective role by reducing inflammation in the pathogenesis of NEC, the mechanism of which may be related to the regulation of neutrophils/enterocytes.


Subject(s)
B7-H1 Antigen , Enterocolitis, Necrotizing , Animals , B7-H1 Antigen/genetics , Disease Models, Animal , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Inflammation , Interleukin-1beta , Mice , Mice, Inbred C57BL
20.
BMC Pediatr ; 22(1): 225, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35468817

ABSTRACT

Necrotizing enterocolitis (NEC) is a multifactorial and complex disease. Our knowledge of the cellular and genetic basis of NEC have expanded considerably as new molecular mechanisms have been identified. This article will focus on the current understanding of the molecular pathogenesis of NEC with a focus on the inflammatory, immune, infectious, and genetic mechanisms that drive disease development.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Infant, Premature, Diseases , Enterocolitis, Necrotizing/genetics , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...