Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
1.
Int Immunopharmacol ; 133: 112058, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38613883

ABSTRACT

Fetal growth restriction (FGR) is a major cause of premature and low-weight births, which increases the risk of necrotizing enterocolitis (NEC); however, the association remains unclear. We report a close correlation between placental polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and NEC. Newborns with previous FGR exhibited intestinal inflammation and more severe NEC symptoms than healthy newborns. Placental PMN-MDSCs are vital regulators of fetal development and neonatal gut inflammation. Placental single-cell transcriptomics revealed that PMN-MDSCs populations and olfactomedin-4 gene (Olfm4) expression levels were significantly increased in PMN-MDSCs in later pregnancy compared to those in early pregnancy and non-pregnant females. Female mice lacking Olfm4 in myeloid cells mated with wild-type males showed FGR during pregnancy, with a decreased placental PMN-MDSCs population and expression of growth-promoting factors (GPFs) from placental PMN-MDSCs. Galectin-3 (Gal-3) stimulated the OLFM4-mediated secretion of GPFs by placental PMN-MDSCs. Moreover, GPF regulation via OLFM4 in placental PMN-MDSCs was mediated via hypoxia inducible factor-1α (HIF-1α). Notably, the offspring of mothers lacking Olfm4 exhibited intestinal inflammation and were susceptible to NEC. Additionally, OLFM4 expression decreased in placental PMN-MDSCs from pregnancies with FGR and was negatively correlated with neonatal morbidity. These results revealed that placental PMN-MDSCs contributed to fetal development and ameliorate newborn intestinal inflammation.


Subject(s)
Fetal Growth Retardation , Myeloid-Derived Suppressor Cells , Placenta , Animals , Female , Pregnancy , Humans , Placenta/immunology , Placenta/metabolism , Infant, Newborn , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Fetal Growth Retardation/immunology , Mice , Mice, Knockout , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Mice, Inbred C57BL , Male , Galectins/metabolism , Galectins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestines/immunology , Intestines/pathology
2.
Cell Mol Immunol ; 21(6): 575-588, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38632385

ABSTRACT

Neonates are susceptible to inflammatory disorders such as necrotizing enterocolitis (NEC) due to their immature immune system. The timely appearance of regulatory immune cells in early life contributes to the control of inflammation in neonates, yet the underlying mechanisms of which remain poorly understood. In this study, we identified a subset of neonatal monocytes characterized by high levels of neuropilin-1 (Nrp1), termed Nrp1high monocytes. Compared with their Nrp1low counterparts, Nrp1high monocytes displayed potent immunosuppressive activity. Nrp1 deficiency in myeloid cells aggravated the severity of NEC, whereas adoptive transfer of Nrp1high monocytes led to remission of NEC. Mechanistic studies showed that Nrp1, by binding to its ligand Sema4a, induced intracellular p38-MAPK/mTOR signaling and activated the transcription factor KLF4. KLF4 transactivated Nos2 and enhanced the production of nitric oxide (NO), a key mediator of immunosuppression in monocytes. These findings reveal an important immunosuppressive axis in neonatal monocytes and provide a potential therapeutic strategy for treating inflammatory disorders in neonates.


Subject(s)
Animals, Newborn , Enterocolitis, Necrotizing , Inflammation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Monocytes , Neuropilin-1 , Monocytes/metabolism , Monocytes/immunology , Animals , Neuropilin-1/metabolism , Neuropilin-1/genetics , Inflammation/pathology , Inflammation/immunology , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/prevention & control , Mice , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Infant, Newborn , p38 Mitogen-Activated Protein Kinases/metabolism , Mice, Knockout
3.
J Pediatr Gastroenterol Nutr ; 75(1): 97-103, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35442233

ABSTRACT

OBJECTIVES: Human milk reduces the incidence of necrotizing enterocolitis (NEC). Prior studies have demonstrated that exogenous surfactant protein-A (SP-A) modulates intestinal inflammation, reduces NEC-like pathology in SP-A-deficient (SPAKO) pups, and may contribute to breast milk's immunomodulatory potential. We hypothesize that SP-A is present in milk and impacts inflammatory responses in the terminal ileum of neonatal mice. METHODS: Human milk was collected at postpartum days 1-3 and 28. Mouse milk was collected at postpartum days 1-10. SP-A was detected in milk through immunoprecipitation and western blot analysis. The impact of murine wild-type (WT) milk on SPAKO pup ileum was evaluated in a model of intestinal inflammation via cross-rearing experiments. Terminal ileum was evaluated for inflammatory cytokine and toll-like receptor 4 (TLR4) mRNA expression via quantitative real-time RT-PCR. RESULTS: SP-A was detected in human milk and wild type (WT) mouse milk, but not in SPAKO mouse milk. Expression of TLR4, interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α was decreased in SPAKO pups reared with WT dams compared to SPAKO pups reared with SPAKO dams, with a peak effect at day of life 14. When inflammation was induced using a lipopolysaccharide-induced model of inflammation, expression of TLR4, IL-1ß, IL-6, CXCL-1, and TNF-α was significantly lower in SPAKO pups reared with WT dams compared to SPAKO pups reared with SPAKO dams. CONCLUSIONS: SP-A is present in human and murine milk and plays a role in lowering inflammation in murine pup terminal ileum. Both baseline inflammation and induced inflammatory responses are reduced via exposure to SP-A in milk with the effect amplified in inflammatory conditions.


Subject(s)
Enterocolitis, Necrotizing , Milk, Human , Pulmonary Surfactant-Associated Protein A , Toll-Like Receptor 4 , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/immunology , Female , Humans , Immunomodulating Agents/pharmacology , Infant, Newborn , Inflammation/immunology , Inflammation/metabolism , Interleukin-6 , Mice , Milk, Human/drug effects , Milk, Human/immunology , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Surface-Active Agents , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Mediators Inflamm ; 2021: 6259381, 2021.
Article in English | MEDLINE | ID: mdl-34675753

ABSTRACT

OBJECTIVE: To determine the role of sodium butyrate in intestinal inflammation via regulation of high-mobility group box-1 (HMGB1), we analyzed the potential mechanism in necrotizing enterocolitis (NEC) in a neonatal mouse model. METHODS: A NEC model was created with hypoxia and cold exposure and artificial overfeeding. C57BL/6 neonatal mice were randomized into three groups: the control, untreated NEC, and sodium butyrate (150 mM)-pretreated NEC groups. Pathological variations in ileocecal intestinal tissue were observed by HE staining and scored in a double-blind manner. The mRNA expression levels of HMGB1, Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), and inflammatory cytokines in intestinal tissues were determined by quantitative real-time PCR. The protein levels of HMGB1 and associated cytokines in intestinal tissues were evaluated using ELISA. The relative protein expression levels of TLR4 and NF-κB in intestinal tissues were quantified by western blot. RESULTS: Sodium butyrate administration improved the body weight and survival rate of NEC mice; relieved intestinal pathological injury; reduced the intestinal expression of HMGB1, TLR4, NF-κB, interleukin- (IL-) 1ß, IL-6, IL-8, and TNF-α; and increased the intestinal expression of IL-10 (P < 0.05). Treatment with butyrate decreased the proportion of opportunistic Clostridium_sensu_stricto_1 and Enterococcus and increased the proportion of beneficial Firmicutes and Lactobacillus in the NEC model. CONCLUSIONS: Sodium butyrate intervention relieves intestinal inflammation and partially corrects the disrupted intestinal flora in mice with NEC.


Subject(s)
Butyric Acid/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Animals , Butyric Acid/pharmacology , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/pathology , Female , HMGB1 Protein/genetics , HMGB1 Protein/physiology , Intestines/microbiology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/physiology , Random Allocation , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
5.
Biomed Res Int ; 2021: 1877075, 2021.
Article in English | MEDLINE | ID: mdl-34712727

ABSTRACT

The objective of this study was to explore the therapeutic effects of berberine on necrotic enteritis (NE) in broilers caused by Clostridium perfringens. A total of 240 1-day-old Arbor Acres chicks were divided into four groups, as negative controls (NC), positive controls (PC), berberine- (BER-) treated, or lincomycin- (LMY-) treated groups. Broilers were challenged with C. perfringens at 15-21 days of age, followed by BER or LMY supplied in drinking water for 7 days. Experimental results showed that C. perfringens infection significantly decreased growth performance and increased intestinal necrosis index and the number of C. perfringens present to 6.45 Log10CFU/g (P < 0.001). Proinflammatory cytokines in the ileum were significantly increased, but the expression of ileal tight junction proteins occludin and claudin-1 was significantly reduced. Both BER and LMY ameliorated some of these observations. Compared with the PC group, the number of C. perfringens in the cecum was significantly decreased following treatment (P < 0.001), and growth performance and small intestine morphology were similar to those of the NC group (P > 0.05). IL-1ß, IL-6, and TNF-α levels as well as occludin and claudin-1 expression were also significantly improved (P < 0.05). BER has the potential to replace antibiotics for NE caused by C. perfringens.


Subject(s)
Berberine/pharmacology , Chickens/immunology , Chickens/microbiology , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/microbiology , Intestines/microbiology , Intestines/pathology , Animals , Cecum/drug effects , Cecum/microbiology , Clostridium perfringens/drug effects , Cytokines/metabolism , Diet , Enterocolitis, Necrotizing/genetics , Female , Gene Expression Regulation/drug effects , Ileum/metabolism , Intestines/immunology , Lincomycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
7.
BMC Microbiol ; 21(1): 225, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362295

ABSTRACT

BACKGROUND: Necrotising enterocolitis (NEC) is a devastating bowel disease, primarily affecting premature infants, with a poorly understood aetiology. Prior studies have found associations in different cases with an overabundance of particular elements of the faecal microbiota (in particular Enterobacteriaceae or Clostridium perfringens), but there has been no explanation for the different results found in different cohorts. Immunological studies have indicated that stimulation of the TLR4 receptor is involved in development of NEC, with TLR4 signalling being antagonised by the activated TLR9 receptor. We speculated that differential stimulation of these two components of the signalling pathway by different microbiota might explain the dichotomous findings of microbiota-centered NEC studies. Here we used shotgun metagenomic sequencing and qPCR to characterise the faecal microbiota community of infants prior to NEC onset and in a set of matched controls. Bayesian regression was used to segregate cases from control samples using both microbial and clinical data. RESULTS: We found that the infants suffering from NEC fell into two groups based on their microbiota; one with low levels of CpG DNA in bacterial genomes and the other with high abundances of organisms expressing LPS. The identification of these characteristic communities was reproduced using an external metagenomic validation dataset. We propose that these two patterns represent the stimulation of a common pathway at extremes; the LPS-enriched microbiome suggesting overstimulation of TLR4, whilst a microbial community with low levels of CpG DNA suggests reduction of the counterbalance to TLR4 overstimulation. CONCLUSIONS: The identified microbial community patterns support the concept of NEC resulting from TLR-mediated pathways. Identification of these signals suggests characteristics of the gastrointestinal microbial community to be avoided to prevent NEC. Potential pre- or pro-biotic treatments may be designed to optimise TLR signalling.


Subject(s)
Enterocolitis, Necrotizing/microbiology , Epithelial Cells/immunology , Gastrointestinal Microbiome/genetics , Infant, Premature, Diseases/microbiology , Toll-Like Receptor 4/immunology , Bayes Theorem , DNA, Bacterial/genetics , Enterocolitis, Necrotizing/immunology , Epithelial Cells/microbiology , Feces/microbiology , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/immunology , Metagenomics , RNA, Ribosomal, 16S/genetics , Toll-Like Receptor 4/genetics
8.
Gut Microbes ; 13(1): 1951113, 2021.
Article in English | MEDLINE | ID: mdl-34264803

ABSTRACT

Premature birth, especially if born before week 32 of gestation, is associated with increased risk of neonatal morbidity and mortality. Prophylactic use of probiotics has been suggested to protect preterm infants via supporting a healthy gut microbiota (GM) development, but the suggested strains and doses vary between studies. In this study, we profiled the GM of 5, 10 and 30-day fecal samples from two cohorts of preterm neonates (born <30 weeks of gestation) recruited in the same neonatal intensive care unit. One cohort (n = 165) was recruited from September 2006 to January 2009 before probiotics were introduced in the clinic. The second cohort (n = 87) was recruited from May 2010 to October 2011 after introducing Lacticaseibacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis BB-12 supplementation policy. Through V3-V4 region 16S rRNA gene amplicon sequencing, a distinct increase of L. rhamnosus and B. animalis was found in the fecal samples of neonates supplemented with probiotics. During the first 30 days of life, the preterm GM went through similarly patterned progression of bacterial populations. Staphylococcus and Weissella dominated in early samples, but was gradually overtaken by Veillonella, Enterococcus and Enterobacteriaceae. Probiotic supplementation was associated with pronounced reduction of Weissella, Veillonella spp. and the opportunistic pathogen Klebsiella. Potential nosocomial pathogens Citrobacter and Chryseobacterium species also gradually phased out. In conclusion, probiotic supplementation to preterm neonates affected gut colonization by certain bacteria, but did not change the overall longitudinal bacterial progression in the neonatal period.Abbreviations: GM: Gut microbiota; ASV: Amplicon sequence variant; NEC: Necrotizing enterocolitis; DOL: Days of life; NICU: Neonatal intensive care unit; ESPGHAN: European Society for Pediatric Gastroenterology, Hepatology and Nutrition; Db-RDA: Distance-based redundancy analysis; PERMANOVA: Permutational multivariate analysis of variance; ANCOM: Analysis of compositions of microbiomes; LGG: Lacticaseibacillus (former Lactobacillus) rhamnosus GG; BB-12: Bifidobacterium animalis ssp. lactis BB-12; DGGE: Denaturing Gradient Gel Electrophoresis.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/immunology , Gastrointestinal Microbiome/drug effects , Infant, Premature/growth & development , Probiotics/pharmacology , Probiotics/therapeutic use , Bifidobacterium animalis/isolation & purification , Cohort Studies , Denmark , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Humans , Infant, Newborn , Infant, Newborn, Diseases/drug therapy , Infant, Newborn, Diseases/immunology , Infant, Premature/immunology , Lactobacillus/isolation & purification , Male
9.
Cell Rep Med ; 2(6): 100320, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34195684

ABSTRACT

Necrotizing enterocolitis (NEC) is a deadly intestinal inflammatory disorder that primarily affects premature infants and lacks adequate therapeutics. Interleukin (IL)-22 plays a critical role in gut barrier maintenance, promoting epithelial regeneration, and controlling intestinal inflammation in adult animal models. However, the importance of IL-22 signaling in neonates during NEC remains unknown. We investigated the role of IL-22 in the neonatal intestine under homeostatic and inflammatory conditions by using a mouse model of NEC. Our data reveal that Il22 expression in neonatal murine intestine is negligible until weaning, and both human and murine neonates lack IL-22 production during NEC. Mice deficient in IL-22 or lacking the IL-22 receptor in the intestine display a similar susceptibility to NEC, consistent with the lack of endogenous IL-22 during development. Strikingly, treatment with recombinant IL-22 during NEC substantially reduces inflammation and enhances epithelial regeneration. These findings may provide a new therapeutic strategy to attenuate NEC.


Subject(s)
Enterocolitis, Necrotizing/immunology , Interleukins/genetics , Intestinal Mucosa/immunology , Recombinant Proteins/pharmacology , Regeneration/immunology , Animals , Animals, Newborn , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Disease Models, Animal , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/pathology , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/microbiology , Infant, Newborn, Diseases/pathology , Infant, Premature , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukins/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Regeneration/genetics , Signal Transduction , Weaning , Interleukin-22
10.
Int Immunopharmacol ; 96: 107787, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34162151

ABSTRACT

Despite the fact that melatonin regulates the expression of long noncoding RNAs (lncRNAs) under different physiological and pathological conditions, it has not been confirmed whether melatonin-induced lncRNAs regulate the differentiation of Treg and Th17 cells. Herein, we show that the expression of LINC01512 is significantly down-regulated and correlates with imbalanced Treg/Th17 ratios in necrotising enterocolitis (NEC) tissues. Through gain- and loss-of-function approaches, we found that LINC01512 promotes the differentiation of Treg cells but interferes with that of Th17 cells. Mechanistically, LINC01512 promotes SIRT1 in Treg and Th17 cells, and subsequently enhances the differentiation of Treg cells and inhibits that of Th17 cells. Furthermore, we demonstrate that melatonin up-regulates the transcription of LINC01512 via the AMPK signalling pathway and that the blockade of AMPK represses LINC01512 expression in Treg and Th17 cells. Overall, our results confirm that SIRT1-regulated differentiation of Treg/Th17 cells is actually modulated by melatonin-induced LINC0512. Moreover, manipulation of the AMPK/LINC01512/SIRT1 axis via melatonin may be a novel therapeutic approach to reduce inflammation.


Subject(s)
Enterocolitis, Necrotizing/immunology , Melatonin/pharmacology , RNA, Long Noncoding , Sirtuin 1/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Cell Differentiation , Humans , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
11.
Front Immunol ; 12: 650709, 2021.
Article in English | MEDLINE | ID: mdl-33968047

ABSTRACT

Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.


Subject(s)
Enterocolitis, Necrotizing/immunology , Gastrointestinal Microbiome/immunology , Infant, Newborn, Diseases/immunology , Prenatal Exposure Delayed Effects/immunology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Infant, Very Low Birth Weight/immunology , Pregnancy
12.
Biomed Res Int ; 2021: 5568724, 2021.
Article in English | MEDLINE | ID: mdl-33880370

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is one of the most serious gastrointestinal disease-causing high morbidity and mortality in premature infants. However, the underlying mechanism of the pathogenesis of NEC is still not fully understood. METHODS: RNA sequencing of intestinal specimens from 9 NEC and 5 controls was employed to quantify the gene expression levels. RNA sequencing was employed to quantify the gene expression levels. DESeq2 tool was used to identify the differentially expressed genes. The biological function, pathways, transcription factors, and immune cells dysregulated in NEC were characterized by gene set enrichment analysis. RESULTS: In the present study, we analyzed RNA sequencing data of NECs and controls and revealed that immune-related pathways were highly activated, while some cellular responses to external stimuli-related pathways were inactivated in NEC. Moreover, B cells, macrophages M1, and plasma cells were identified as the major cell types involved in NEC. Furthermore, we also found that inflammation-related transcription factor genes, such as STAT1, STAT2, and IRF2, were significantly activated in NEC, further suggesting that these TFs might play critical roles in NEC pathogenesis. In addition, NEC samples exhibited heterogeneity to some extent. Interestingly, two subgroups in the NEC samples were identified by hierarchical clustering analysis. Notably, B cells, T cells, Th1, and Tregs involved in adaptive immune were predicted to highly infiltrate into subgroup I, while subgroup II was significantly infiltrated by neutrophils. The heterogeneity of immune cells in NEC indicated that both innate and adaptive immunes might induce NEC-related inflammatory response. CONCLUSIONS: In summary, we systematically analyzed inflammation-related genes, signaling pathways, and immune cells to characterize the NEC pathogenesis and samples, which greatly improved our understanding of the roles of inflammatory responses in NEC.


Subject(s)
Computational Biology , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/immunology , Infant, Premature/immunology , Inflammation/genetics , Signal Transduction/genetics , Case-Control Studies , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Infant, Newborn , Transcription Factors/metabolism
13.
J Neuroinflammation ; 18(1): 66, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33676524

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal disease in premature neonates with high mortality and morbidity, while the underlining mechanism of intestinal injury and profound neurological dysfunction remains unclear. Here, we aimed to investigate the involvement of NLPR3 inflammasome activation in NEC-related enterocolitis and neuroinflammation, especially long-term cognitive impairment, meanwhile, explore the protective effect of NLRP3 inhibitor MCC950 on NEC in mice. METHODS: NLRP3 inflammasome activation in the intestine and brain was assessed in the NEC mouse model, and NLRP3 inhibitor MCC950 was administrated during the development of NEC. Survival rate, histopathological injury of the intestine and brain, and expression of mature IL-1ß and other pro-inflammatory cytokines were analyzed. Long-term cognitive impairment was evaluated by behavioral test. RESULTS: The expression of NLRP3 and mature IL-1ß in the intestine and brain was greatly upregulated in NEC mice compared to the controls. MCC950 treatment efficiently improved NEC survival rate, reduced intestinal and brain inflammation, and ameliorated the severity of pathological damage in both organs. Additionally, in vivo blockage of NLRP3 inflammasome with MCC950 in early life of NEC pups potently protected against NEC-associated long-term cognitive impairment. CONCLUSIONS: Our findings suggest that NLRP3 inflammasome activation participates in NEC-induced intestinal and brain injury, and early intervention with NLRP3 inhibitor may provide beneficial therapeutic effect on NEC infants.


Subject(s)
Cognitive Dysfunction/immunology , Enterocolitis, Necrotizing/immunology , Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Furans/pharmacology , Indenes/pharmacology , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sulfonamides/pharmacology
14.
Inflamm Res ; 70(3): 343-358, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33564933

ABSTRACT

BACKGROUND: Activation of intestinal macrophages is implicated in the pathogenesis of neonatal necrotizing enterocolitis (NEC), yet its precise mechanisms remain unclear. OBJECTIVE: The purpose of this study is to investigate the role of macrophages and TNF-α via an inflammatory MicroRNA in NEC. MATERIALS AND METHODS: Immunofluorescence (IF) staining of CD68, iNOS, and Arg-1 was employed to identify phenotypes of macrophage in the intestines of NEC infants and NEC mice. Expression of TNF-α, c-kit, and miR-222 was evaluated by qRT-PCR, Western blot, and immunochemical staining from the tissue samples. RESULTS: Large number of M1 macrophage infiltration was found in the NEC intestines. Expression of CD68, iNOS, and TNF-α were significantly increased, while c-kit was decreased distinctly in the NEC group. In the early phase of NEC mouse model, inhibition of M1 macrophages reduced the incidence of NEC and intestinal inflammation. We found that TNF-α upregulated the expression of miRNA-222 and inhibited the expression of c-kit. Conversely, such decrease of c-kit expression could be reversed by miR-222 antagonists. Furtherly, dual-luciferase assay confirmed that c-kit can be inhibited by miR-222 directly. CONCLUSION: Macrophages activation in NEC intestine results in an increased inflammatory response and TNF-α production, accompanied with miR-222 upregulation and c-kit suppression. Modulations of M1 macrophages, TNF-α or miR-222 may be potential therapeutic targets for NEC treatment.


Subject(s)
Enterocolitis, Necrotizing/immunology , Macrophages/immunology , MicroRNAs , Proto-Oncogene Proteins c-kit/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Enterocolitis, Necrotizing/pathology , Female , Humans , Infant, Newborn , Intestine, Small/immunology , Intestine, Small/pathology , Macrophage Activation , Male , Mice, Inbred C57BL , Up-Regulation
15.
Nat Commun ; 12(1): 1042, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589625

ABSTRACT

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Enterocolitis, Necrotizing/genetics , Indoles/administration & dosage , Milk, Human/physiology , Receptors, Aryl Hydrocarbon/genetics , Toll-Like Receptor 4/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/immunology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/immunology , Diet/methods , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/prevention & control , Female , Gene Expression Regulation , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Ligands , Maternal Exposure , Mice , Pregnancy , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , Swine , Toll-Like Receptor 4/immunology
16.
Gut Microbes ; 13(1): 1-16, 2021.
Article in English | MEDLINE | ID: mdl-33382952

ABSTRACT

Preterm infants are at risk of multiple morbidities including necrotizing enterocolitis (NEC). Suspected NEC patients receive intravenous antibiotics (AB) to prevent sepsis, although enteral AB is arguably more effective at reducing NEC but is rarely used due to the risk of AB resistance. Fecal microbiota transplantation (FMT) has shown protective effects against NEC in animal experiments, but the interaction between AB and FMT has not been investigated in neonates. We hypothesized that administration of enteral AB followed by rectal FMT would effectively prevent NEC with negligible changes in AB resistance and systemic immunity. Using preterm piglets, we examined host and gut microbiota responses to AB, FMT, or a sequential combination thereof, with emphasis on NEC development. In a saline-controlled experiment, preterm piglets (n = 67) received oro-gastric neomycin (50 mg/kg/d) and amoxicillin-clavulanate (50/12.5 mg/kg/d) (hereafter AB) for four days after cesarean delivery, and were subsequently given rectal FMT from healthy suckling piglet donors. Whereas AB protected the stomach and small intestine, and FMT primarily protected the colon, the sequential combination treatment surprisingly provided no NEC protection. Furthermore, minor changes in the gut microbiota composition were observed in response to either treatment, although AB treatment decreased species diversity and increased AB resistance among coliform bacteria and Enterococci, which were both partly reversed by FMT. Besides, enteral AB treatment suppressed cellular and functional systemic immune development, which was not prevented by subsequent FMT. We discovered an antagonistic relationship between enteral AB and FMT in terms of NEC development. The outcome may depend on choice of AB compounds, FMT composition, doses, treatment duration, and administration routes, but these results challenge the applicability of enteral AB and FMT in preterm infants.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Enterocolitis, Necrotizing/therapy , Fecal Microbiota Transplantation , Animals , Animals, Newborn , Bacterial Adhesion/drug effects , Combined Modality Therapy , Disease Models, Animal , Drug Resistance, Microbial/drug effects , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/microbiology , Gastrointestinal Microbiome/drug effects , Immunity, Mucosal/drug effects , Intubation, Gastrointestinal , Lymphocytes/drug effects , Myeloid Cells/drug effects , Premature Birth , Swine , Treatment Outcome
17.
Am J Perinatol ; 38(11): 1150-1157, 2021 09.
Article in English | MEDLINE | ID: mdl-32446253

ABSTRACT

OBJECTIVE: Feeding intolerance (FI) is a common presentation of necrotizing enterocolitis (NEC) and sepsis. NEC and sepsis are associated with hematological changes, but these changes alone are not reliable biomarkers for early diagnosis. This study examined whether the combination of hematological indices and FI can be used as an early diagnostic tool for NEC or sepsis. STUDY DESIGN: This retrospective cohort study included infants born at <1,500 g or <30 weeks who had symptoms of FI. The exclusion criteria were congenital or chromosomal disorders, thrombocytopenia or platelet transfusion before the onset of FI, and history of bowel resection. We compared the hematological indices from infants with pathologic FI (due to NEC or sepsis) to infants with benign FI. RESULTS: During the study period, 211 infants developed FI; 185 met the inclusion criteria. Infants with pathologic FI (n = 90, 37 cases with NEC and 53 with sepsis) had lower birth gestational age and weight compared with 95 infants with benign FI (n = 95). Pathologic FI was associated with lower platelet count (median 152 × 103/µL vs. 285 × 103/µL, p < 0.001) and higher immature-to-total neutrophil (I/T) ratio (median 0.23 vs. 0.04, p < 0.001) at the onset of FI. Pathologic FI was also associated with a decrease in baseline platelets compared with an increase in benign FI. For diagnosis of pathologic FI, a decrease ≥10% in platelets from baseline had a sensitivity and specificity of 0.64 and 0.73, respectively, I/T ratio ≥0.1 had a sensitivity and specificity of 0.71 and 0.78, respectively, and the combination of both parameters had a sensitivity and specificity of 0.50 and 0.97, respectively. CONCLUSION: FI caused by NEC or sepsis was associated with a decrease in platelets from baseline, and a lower platelet level and higher I/T ratio at the onset of FI. These findings can help clinicians in the management of preterm infants with FI. KEY POINTS: · FI is a common presentation of NEC and sepsis in preterm infants.. · FI due to NEC or sepsis is associated with changes in platelets and I/T ratio.. · These changes could be useful as early markers for diagnosis..


Subject(s)
Enterocolitis, Necrotizing/diagnosis , Food Intolerance/etiology , Neutrophils/immunology , Sepsis/diagnosis , Biomarkers/blood , Blood Platelets , Early Diagnosis , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/immunology , Female , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Leukocyte Count , Logistic Models , Male , Neutrophils/metabolism , Retrospective Studies , Sensitivity and Specificity , Sepsis/blood , Sepsis/immunology
18.
Pediatr Res ; 89(1): 91-101, 2021 01.
Article in English | MEDLINE | ID: mdl-32221473

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Ileum/drug effects , Intestinal Mucosa/drug effects , Lactose/analogs & derivatives , Milk, Human/chemistry , Toll-Like Receptor 4/antagonists & inhibitors , Trisaccharides/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Humans , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lactose/isolation & purification , Lactose/pharmacology , Mice , Molecular Docking Simulation , Signal Transduction , Sus scrofa , Toll-Like Receptor 4/metabolism , Trisaccharides/isolation & purification , Weight Loss/drug effects
19.
Nutrients ; 14(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35011027

ABSTRACT

Necrotizing enterocolitis (NEC) is a significant cause of mortality and morbidity in preterm infants. The pathogenesis of NEC is not completely understood; however, intestinal immaturity and excessive immunoreactivity of intestinal mucosa to intraluminal microbes and nutrients appear to have critical roles. Dietary fats are not only the main source of energy for preterm infants, but also exert potent effects on intestinal development, intestinal microbial colonization, immune function, and inflammatory response. Preterm infants have a relatively low capacity to digest and absorb triglyceride fat. Fat may thereby accumulate in the ileum and contribute to the development of NEC by inducing oxidative stress and inflammation. Some fat components, such as long-chain polyunsaturated fatty acids (LC-PUFAs), also exert immunomodulatory roles during the early postnatal period when the immune system is rapidly developing. LC-PUFAs may have the ability to modulate the inflammatory process of NEC, particularly when the balance between n3 and n6 LC-PUFAs derivatives is maintained. Supplementation with n3 LC-PUFAs alone may have limited effect on NEC prevention. In this review, we describe how various fatty acids play different roles in the pathogenesis of NEC in preterm infants.


Subject(s)
Dietary Fats , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/prevention & control , Infant Nutritional Physiological Phenomena/physiology , Infant, Premature , Dietary Fats/administration & dosage , Dietary Fats/analysis , Dietary Fats/metabolism , Digestion/physiology , Enterocolitis, Necrotizing/immunology , Fatty Acids/analysis , Fatty Acids/chemistry , Humans , Infant Formula/analysis , Infant, Newborn , Inflammation , Milk, Human/chemistry , Oxidative Stress
20.
Nat Commun ; 11(1): 5794, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188181

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe, currently untreatable intestinal disease that predominantly affects preterm infants and is driven by poorly characterized inflammatory pathways. Here, human and murine NEC intestines exhibit an unexpected predominance of type 3/TH17 polarization. In murine NEC, pro-inflammatory type 3 NKp46-RORγt+Tbet+ innate lymphoid cells (ILC3) are 5-fold increased, whereas ILC1 and protective NKp46+RORγt+ ILC3 are obliterated. Both species exhibit dysregulation of intestinal TLR repertoires, with TLR4 and TLR8 increased, but TLR5-7 and TLR9-12 reduced. Transgenic IL-37 effectively protects mice from intestinal injury and mortality, whilst exogenous IL-37 is only modestly efficacious. Mechanistically, IL-37 favorably modulates immune homeostasis, TLR repertoires and microbial diversity. Moreover, IL-37 and its receptor IL-1R8 are reduced in human NEC epithelia, and IL-37 is lower in blood monocytes from infants with NEC and/or lower birthweight. Our results on NEC pathomechanisms thus implicate type 3 cytokines, TLRs and IL-37 as potential targets for novel NEC therapies.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/immunology , Adaptive Immunity , Animals , Animals, Newborn , Biomarkers/metabolism , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/pathology , Homeostasis , Humans , Immunity, Innate , Infant, Newborn , Inflammation Mediators/metabolism , Interleukin-1 , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...