Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 55(2): 237-253.e8, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35081371

ABSTRACT

The Th17 cell-lineage-defining cytokine IL-17A contributes to host defense and inflammatory disease by coordinating multicellular immune responses. The IL-17 receptor (IL-17RA) is expressed by diverse intestinal cell types, and therapies targeting IL-17A induce adverse intestinal events, suggesting additional tissue-specific functions. Here, we used multiple conditional deletion models to identify a role for IL-17A in secretory epithelial cell differentiation in the gut. Paneth, tuft, goblet, and enteroendocrine cell numbers were dependent on IL-17A-mediated induction of the transcription factor ATOH1 in Lgr5+ intestinal epithelial stem cells. Although dispensable at steady state, IL-17RA signaling in ATOH1+ cells was required to regenerate secretory cells following injury. Finally, IL-17A stimulation of human-derived intestinal organoids that were locked into a cystic immature state induced ATOH1 expression and rescued secretory cell differentiation. Our data suggest that the cross talk between immune cells and stem cells regulates secretory cell lineage commitment and the integrity of the mucosa.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestinal Mucosa/cytology , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin-17/metabolism , Stem Cells/metabolism , Animals , Cell Communication , Cell Differentiation/drug effects , Cell Lineage/drug effects , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/adverse effects , Humans , Interleukin-17/metabolism , Interleukin-17/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/metabolism , Intestines/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Receptors, Interleukin-17/deficiency , SOX9 Transcription Factor/metabolism , Signal Transduction , Stem Cells/cytology
2.
Immunity ; 44(3): 659-671, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26982366

ABSTRACT

Interleukin-17 (IL-17) and IL-17 receptor (IL-17R) signaling are essential for regulating mucosal host defense against many invading pathogens. Commensal bacteria, especially segmented filamentous bacteria (SFB), are a crucial factor that drives T helper 17 (Th17) cell development in the gastrointestinal tract. In this study, we demonstrate that Th17 cells controlled SFB burden. Disruption of IL-17R signaling in the enteric epithelium resulted in SFB dysbiosis due to reduced expression of α-defensins, Pigr, and Nox1. When subjected to experimental autoimmune encephalomyelitis, IL-17R-signaling-deficient mice demonstrated earlier disease onset and worsened severity that was associated with increased intestinal Csf2 expression and elevated systemic GM-CSF cytokine concentrations. Conditional deletion of IL-17R in the enteric epithelium demonstrated that there was a reciprocal relationship between the gut microbiota and enteric IL-17R signaling that controlled dysbiosis, constrained Th17 cell development, and regulated the susceptibility to autoimmune inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Gram-Positive Bacterial Infections/immunology , Gram-Positive Endospore-Forming Bacteria/immunology , Intestines/physiology , Receptors, Interleukin-17/metabolism , Th17 Cells/immunology , Animals , Dysbiosis/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Host-Pathogen Interactions , Immunity, Mucosal/genetics , Interleukin-17/metabolism , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Interleukin-17/genetics , Signal Transduction/genetics , Th17 Cells/microbiology , alpha-Defensins/genetics , alpha-Defensins/metabolism
3.
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
4.
J Immunol ; 206(4): 766-775, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33431657

ABSTRACT

Type 17 cytokines have been strongly implicated in mucosal immunity, in part by regulating the production of antimicrobial peptides. Using a mouse model of Citrobacter rodentium infection, which causes colitis, we found that intestinal IL-17RA and IL-17RC were partially required for control of infection in the colon and IL-17 regulates the production of luminal hydrogen peroxide as well as expression of Tnsf13 Reduced Tnfsf13 expression was associated with a profound defect in generating C. rodentium-specific IgA+ Ab-secreting cells. Taken together, intestinal IL-17R signaling plays key roles in controlling invading pathogens, in part by regulating luminal hydrogen peroxide as well as regulating the generation of pathogen-specific IgA+ Ab-secreting cells.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Oxidoreductases/immunology , Receptors, Interleukin-17/immunology , Signal Transduction/immunology , Animals , Disease Models, Animal , Enterobacteriaceae Infections/genetics , Humans , Hydrogen Peroxide/immunology , Immunoglobulin A, Secretory/genetics , Mice , Mice, Knockout , Oxidoreductases/genetics , Receptors, Interleukin-17/genetics , Signal Transduction/genetics
5.
J Immunol ; 206(7): 1540-1548, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33648937

ABSTRACT

IL-17A and IL-22 derived from Th17 cells play a significant role in mucosal immunity and inflammation. TGF-ß and IL-6 promote Th17 differentiation; however, these cytokines have multiple targets. The identification and screening of additional molecules that regulate IL-17A and IL-22 responses in certain inflammatory conditions is of great clinical significance. In this study, we show that CDDO-Im, a specific Nrf2 activator, promotes IL-17A and IL-22 responses in murine Th17 cells. In contrast, CDDO-Im inhibits IL-17A response in multiple sclerosis patient-derived PBMCs. However, Nrf2 specifically regulates IL-22 response in vivo. Nrf2 acts through the regulation of antioxidant response element (ARE) binding motifs in target genes to induce or repress transcription. Promoter analysis revealed that Il17a, Rorc, and Ahr genes have several ARE motifs. We showed that Nrf2 bound to ARE repressor (ARE-R2) of Rorc and inhibited Rorc-dependent IL-17A transactivation. The luciferase reporter assay data showed that CDDO-Im regulated Ahr promoter activity. Chromatin immunoprecipitation quantitative PCR data showed that Nrf2 bound to ARE of AhR. Finally, we confirmed that the CDDO-Im-mediated induction of IL-22 production in CD4+ T cells was abrogated in CD4-specific Ahr knockout mice (AhrCD4 ). CH-223191, a specific AhR antagonist, inhibits CDDO-Im-induced IL-22 production in CD4+ T cells, which further confirmed the AhR-dependent regulation. Collectively, our data showed that Nrf2 via AhR pathways regulated IL-22 response in CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukins/metabolism , Multiple Sclerosis/immunology , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/immunology , Animals , Azo Compounds/metabolism , Gene Expression Regulation , Humans , Imidazoles/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/metabolism , Promoter Regions, Genetic/genetics , Pyrazoles/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Interleukin-22
6.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32179676

ABSTRACT

Late-onset sepsis (LOS) is a highly consequential complication of preterm birth and is defined by a positive blood culture obtained after 72 h of age. The causative bacteria can be found in patients' intestinal tracts days before dissemination, and cohort studies suggest reduced LOS risk in breastfed preterm infants through unknown mechanisms. Reduced concentrations of epidermal growth factor (EGF) of maternal origin within the intestinal tract of mice correlated to the translocation of a gut-resident human pathogen Escherichia coli, which spreads systemically and caused a rapid, fatal disease in pups. Translocation of Escherichia coli was associated with the formation of colonic goblet cell-associated antigen passages (GAPs), which translocate enteric bacteria across the intestinal epithelium. Thus, maternally derived EGF, and potentially other EGFR ligands, prevents dissemination of a gut-resident pathogen by inhibiting goblet cell-mediated bacterial translocation. Through manipulation of maternally derived EGF and alteration of the earliest gut defenses, we have developed an animal model of pathogen dissemination which recapitulates gut-origin neonatal LOS.


Subject(s)
Bacterial Translocation/immunology , ErbB Receptors/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Gastrointestinal Microbiome/immunology , Milk, Human/immunology , Neonatal Sepsis/immunology , Animals , Animals, Newborn , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Breast Feeding , Colon/metabolism , Colon/microbiology , Disease Models, Animal , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Transgenic , Milk, Human/metabolism , Neonatal Sepsis/metabolism , Neonatal Sepsis/microbiology , Signal Transduction/immunology , Time Factors
7.
J Biomed Inform ; 128: 104031, 2022 04.
Article in English | MEDLINE | ID: mdl-35183765

ABSTRACT

Preterm birth affects more than 10% of all births worldwide. Such infants are much more prone to Growth Faltering (GF), an issue that has been unsolved despite the implementation of numerous interventions aimed at optimizing preterm infant nutrition. To improve the ability for early prediction of GF risk for preterm infants we collected a comprehensive, large, and unique clinical and microbiome dataset from 3 different sites in the US and the UK. We use and extend machine learning methods for GF prediction from clinical data. We next extend graphical models to integrate time series clinical and microbiome data. A model that integrates clinical and microbiome data improves on the ability to predict GF when compared to models using clinical data only. Information on a small subset of the taxa is enough to help improve model accuracy and to predict interventions that can improve outcome. We show that a hierarchical classifier that only uses a subset of the taxa for a subset of the infants is both the most accurate and cost-effective method for GF prediction. Further analysis of the best classifiers enables the prediction of interventions that can improve outcome.


Subject(s)
Microbiota , Premature Birth , Humans , Infant , Infant, Newborn , Infant, Premature , Machine Learning
8.
Gastroenterology ; 158(5): 1402-1416.e2, 2020 04.
Article in English | MEDLINE | ID: mdl-31838076

ABSTRACT

BACKGROUND & AIMS: Trehalose is a disaccharide that might be used in the treatment of cardiometabolic diseases. However, trehalose consumption promotes the expansion of Clostridioides difficile ribotypes that metabolize trehalose via trehalose-6-phosphate hydrolase. Furthermore, brush border and renal trehalases can reduce the efficacy of trehalose by cleaving it into monosaccharides. We investigated whether a trehalase-resistant analogue of trehalose (lactotrehalose) has the same metabolic effects of trehalose without expanding C difficile. METHODS: We performed studies with HEK293 and Caco2 cells, primary hepatocytes from mice, and human intestinal organoids. Glucose transporters were overexpressed in HEK293 cells, and glucose tra2nsport was quantified. Primary hepatocytes were cultured with or without trehalose or lactotrehalose, and gene expression patterns were analyzed. C57B6/J mice were given oral antibiotics and trehalose or lactotrehalose in drinking water, or only water (control), followed by gavage with the virulent C difficile ribotype 027 (CD027); fecal samples were analyzed for toxins A (ToxA) or B (ToxB) by enzyme-linked immunosorbent assay. Other mice were given trehalose or lactotrehalose in drinking water for 2 days before placement on a chow or 60% fructose diet for 10 days. Liver tissues were collected and analyzed by histologic, serum biochemical, RNA sequencing, autophagic flux, and thermogenesis analyses. We quantified portal trehalose and lactotrehalose bioavailability by gas chromatography mass spectrometry. Fecal microbiomes were analyzed by 16S ribosomal RNA sequencing and principal component analyses. RESULTS: Lactotrehalose and trehalose each blocked glucose transport in HEK293 cells and induced a gene expression pattern associated with fasting in primary hepatocytes. Compared with mice on the chow diet, mice on the high-fructose diet had increased circulating cholesterol, higher ratios of liver weight-to-body weight, hepatic lipid accumulation (steatosis), and liver gene expression patterns of carbohydrate-responsive de novo lipogenesis. Mice given lactotrehalose while on the high-fructose diet did not develop any of these features and had increased whole-body caloric expenditure compared with mice given trehalose or water and fed a high-fructose diet. Livers from mice given lactotrehalose had increased transcription of genes that regulate mitochondrial energy metabolism compared with liver from mice given trehalose or controls. Lactotrehalose was bioavailable in venous and portal circulation and fecal samples. Lactotrehalose reduced fecal markers of microbial branched-chain amino acid biosynthesis and increased expression of microbial genes that regulate insulin signaling. In mice given antibiotics followed by CD027, neither lactotrehalose nor trehalose increased levels of the bacteria or its toxin in stool-in fact, trehalose reduced the abundance of CD027 in stool. Lactotrehalose and trehalose reduced markers of inflammation in rectal tissue after CD027 infection. CONCLUSIONS: Lactotrehalose is a trehalase-resistant analogue that increases metabolic parameters, compared with trehalose, without increasing the abundance or virulence of C difficile strain CD027. Trehalase-resistant trehalose analogues might be developed as next-generation fasting-mimetics for the treatment of diabetes and nonalcoholic fatty liver disease.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/prevention & control , Energy Metabolism/drug effects , Trehalose/pharmacology , Animals , Bacterial Proteins/metabolism , Caco-2 Cells , Clostridioides difficile/enzymology , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Disaccharidases/metabolism , Disease Models, Animal , Fasting/metabolism , Feces/microbiology , Glucose/metabolism , HEK293 Cells , Hepatocytes , Humans , Intestinal Mucosa/cytology , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Primary Cell Culture , Trehalose/analogs & derivatives , Trehalose/therapeutic use
9.
Pediatr Res ; 88(Suppl 1): 56-59, 2020 08.
Article in English | MEDLINE | ID: mdl-32855514

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in the neonatal ICU with minimal progress in the research. METHODS: Federal webpages were queried to look for funding opportunity announcements (FOAs) and to develop lists of funded projects on NEC to identify gaps in NEC-related research topics. RESULTS: Over the past 30 years, the National Institutes of Health (NIH) issued two FOAs to stimulate research on NEC with $4.1 million set aside for the first year of respective funding. We identified 23 recently funded studies of which 18 were research projects, 4 training grants, and 1 conference grant support. Only one grant focused on parent and family engagement in the NICU. CONCLUSION: There are significant research gaps that can be addressed with adequate funding from the federal government on the prevention and treatment of NEC.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/therapy , Financing, Government , Intensive Care, Neonatal/organization & administration , Neonatology/organization & administration , Clinical Trials as Topic , Family Health , Federal Government , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases , Intensive Care Units, Neonatal , National Institutes of Health (U.S.) , Research Design , Research Support as Topic , Treatment Outcome , United States
10.
Pediatr Res ; 88(Suppl 1): 35-40, 2020 08.
Article in English | MEDLINE | ID: mdl-32855511

ABSTRACT

The 2019 Necrotizing Enterocolitis (NEC) Symposium expanded upon the NEC Society's goals of bringing stakeholders together to discuss cutting-edge science, potential therapeutics and preventative measures, as well as the patient-family perspectives of NEC. The Symposium facilitated discussions and shared knowledge with the overarching goal of creating "A World Without NEC." To accomplish this goal, new research to advance the state of the science is necessary. Over the last decade, several established investigators have significantly improved our understanding of the pathophysiology of NEC and they have paved the way for the next generation of clinician-scientists funded to perform NEC research. This article will serve to highlight the contributions of these young clinician-scientists that seek to elucidate how immune, microbial and nervous system dysregulation contributes to the pathophysiology of NEC.


Subject(s)
Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/therapy , Neonatology/trends , Anemia/complications , Blood Transfusion , Cell Transplantation/methods , Congresses as Topic , Enteric Nervous System , Enterocolitis, Necrotizing/immunology , Family Health , Gastrointestinal Microbiome , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases , Infant, Premature , Mesenchymal Stem Cells/cytology , Parents , Translational Research, Biomedical
11.
Pediatr Res ; 88(1): 66-76, 2020 07.
Article in English | MEDLINE | ID: mdl-31242501

ABSTRACT

BACKGROUND: Preterm infants are susceptible to unique pathology due to their immaturity. Mouse models are commonly used to study immature intestinal disease, including necrotizing enterocolitis (NEC). Current NEC models are performed at a variety of ages, but data directly comparing intestinal developmental stage equivalency between mice and humans are lacking. METHODS: Small intestines were harvested from C57BL/6 mice at 3-4 days intervals from birth to P28 (n = 8 at each age). Preterm human small intestine samples representing 17-23 weeks of completed gestation were obtained from the University of Pittsburgh Health Sciences Tissue Bank, and at term gestation during reanastamoses after resection for NEC (n = 4-7 at each age). Quantification of intestinal epithelial cell types and messenger RNA for marker genes were evaluated on both species. RESULTS: Overall, murine and human developmental trends over time are markedly similar. Murine intestine prior to P10 is most similar to human fetal intestine prior to viability. Murine intestine at P14 is most similar to human intestine at 22-23 weeks completed gestation, and P28 murine intestine is most similar to human term intestine. CONCLUSION: Use of C57BL/6J mice to model the human immature intestine is reasonable, but the age of mouse chosen is a critical factor in model development.


Subject(s)
Epithelium/growth & development , Gene Expression Regulation, Developmental , Intestines/growth & development , Animals , Enterocolitis, Necrotizing/metabolism , Epithelium/pathology , ErbB Receptors/metabolism , Gene Expression Profiling , Homeostasis , Humans , Intestinal Diseases/metabolism , Intestines/pathology , Mice , Mice, Inbred C57BL
12.
Proc Natl Acad Sci U S A ; 114(7): 1672-1677, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137842

ABSTRACT

Enteroviruses are among the most common viral infectious agents of humans and are primarily transmitted by the fecal-oral route. However, the events associated with enterovirus infections of the human gastrointestinal tract remain largely unknown. Here, we used stem cell-derived enteroids from human small intestines to study enterovirus infections of the intestinal epithelium. We found that enteroids were susceptible to infection by diverse enteroviruses, including echovirus 11 (E11), coxsackievirus B (CVB), and enterovirus 71 (EV71), and that contrary to an immortalized intestinal cell line, enteroids induced antiviral and inflammatory signaling pathways in response to infection in a virus-specific manner. Furthermore, using the Notch inhibitor dibenzazepine (DBZ) to drive cellular differentiation into secretory cell lineages, we show that although goblet cells resist E11 infection, enteroendocrine cells are permissive, suggesting that enteroviruses infect specific cell populations in the human intestine. Taken together, our studies provide insights into enterovirus infections of the human intestine, which could lead to the identification of novel therapeutic targets and/or strategies to prevent or treat infections by these highly clinically relevant viruses.


Subject(s)
Enterovirus Infections/virology , Enterovirus/physiology , Intestine, Small/virology , Organoids/virology , Caco-2 Cells , Cell Lineage/drug effects , Cell Lineage/genetics , Cells, Cultured , Dibenzazepines/pharmacology , Disease Resistance/genetics , Enterovirus Infections/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Profiling/methods , Host-Pathogen Interactions , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Intestine, Small/cytology , Intestine, Small/metabolism , Organoids/cytology , Organoids/metabolism , Signal Transduction/genetics
13.
Br J Nutr ; 120(6): 665-680, 2018 09.
Article in English | MEDLINE | ID: mdl-30176959

ABSTRACT

Necrotising enterocolitis (NEC) is a devastating disease that typically affects formula-fed premature infants, suggesting that dietary components may influence disease pathogenesis. TAG are the major fat components of infant formula, and their digestion requires pancreatic lipases, which may be naturally deficient in premature neonates. We hypothesise that NEC develops partly from the accumulation of incompletely digested long-chain TAG-containing unsaturated fatty acids within the intestinal epithelial cells, leading to oxidative stress and enterocyte damage. We further hypothesise that the administration of a formula that contains reduced TAG ('pre-digested fat') that do not require lipase action may reduce NEC severity. To test these hypotheses, we induced NEC in neonatal mice using three different fat formulations, namely 'standard fat', 'pre-digested fat' or 'very low fat', and determined that mice fed 'standard fat' developed severe NEC, which was significantly reduced in mice fed 'pre-digested fat' or 'very low fat'. The expression level of the critical fat-digesting enzyme carboxyl ester lipase was significantly lower in the newborn compared with older pups, leading to impaired fat digestion. The accumulation of mal-digested fat resulted in the significant accumulation of fat droplets within the intestinal epithelium of the distal ileum, resulting in the generation of reactive oxygen species and intestinal inflammation. Strikingly, these changes were prevented in pups fed 'pre-digested fat' or 'very low fat' formulas. These findings suggest that nutritional formula containing a pre-digested fat system may overcome the natural lipase deficiency of the premature gut, and serve as a novel approach to prevent NEC.


Subject(s)
Diet , Dietary Fats/pharmacology , Digestion , Enterocolitis, Necrotizing/metabolism , Infant Formula/chemistry , Intestinal Mucosa/drug effects , Triglycerides/pharmacology , Animals , Animals, Newborn , Dietary Fats/metabolism , Enterocolitis, Necrotizing/etiology , Enterocytes/drug effects , Enterocytes/metabolism , Enterocytes/pathology , Fatty Acids, Unsaturated/metabolism , Food, Formulated , Humans , Ileum/drug effects , Ileum/metabolism , Infant Nutritional Physiological Phenomena , Infant, Newborn , Inflammation/etiology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipase/metabolism , Mice , Oxidative Stress , Severity of Illness Index , Triglycerides/metabolism
14.
J Immunol ; 197(3): 859-71, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27307558

ABSTRACT

We seek to define the mechanisms leading to the development of lung disease in the setting of neonatal necrotizing enterocolitis (NEC), a life-threatening gastrointestinal disease of premature infants characterized by the sudden onset of intestinal necrosis. NEC development in mice requires activation of the LPS receptor TLR4 on the intestinal epithelium, through its effects on modulating epithelial injury and repair. Although NEC-associated lung injury is more severe than the lung injury that occurs in premature infants without NEC, the mechanisms leading to its development remain unknown. In this study, we now show that TLR4 expression in the lung gradually increases during postnatal development, and that mice and humans with NEC-associated lung inflammation express higher levels of pulmonary TLR4 than do age-matched controls. NEC in wild-type newborn mice resulted in significant pulmonary injury that was prevented by deletion of TLR4 from the pulmonary epithelium, indicating a role for pulmonary TLR4 in lung injury development. Mechanistically, intestinal epithelial TLR4 activation induced high-mobility group box 1 release from the intestine, which activated pulmonary epithelial TLR4, leading to the induction of the neutrophil recruiting CXCL5 and the influx of proinflammatory neutrophils to the lung. Strikingly, the aerosolized administration of a novel carbohydrate TLR4 inhibitor prevented CXCL5 upregulation and blocked NEC-induced lung injury in mice. These findings illustrate the critical role of pulmonary TLR4 in the development of NEC-associated lung injury, and they suggest that inhibition of this innate immune receptor in the neonatal lung may prevent this devastating complication of NEC.


Subject(s)
Enterocolitis, Necrotizing/complications , Lung Injury/etiology , Respiratory Mucosa/metabolism , Toll-Like Receptor 4/biosynthesis , Animals , Animals, Newborn , Chemokine CXCL5/metabolism , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Knockdown Techniques , HMGB1 Protein/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Lung Injury/immunology , Lung Injury/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/immunology
15.
Cytokine ; 100: 1-10, 2017 12.
Article in English | MEDLINE | ID: mdl-28687373

ABSTRACT

Interleukin-33 (IL-33) is a member of the IL-1 cytokine family that has been widely studied since its discovery in 2005 for its dichotomous functions in homeostasis and inflammation. IL-33, along with its receptor suppression of tumorigenicity 2 (ST2), has been shown to modulate both the innate and adaptive immune system. Originally, the IL-33/ST2 signaling axis was studied in the context of inducing type 2 immune responses with the expression of ST2 by T helper 2 (TH2) cells. However, the role of IL-33 is not limited to TH2 responses. Rather, IL-33 is a potent activator of TH1 cells, group 2 innate lymphoid cells (ILC2s), regulatory T (Treg) cells, and CD8+ T cells. The intestine is uniquely important in this discussion, as the intestinal epithelium is distinctively positioned to interact with both pathogens and the immune cells housed in the mucosa. In the intestine, IL-33 is expressed by the pericryptal fibroblasts and its expression is increased particularly in disease states. Moreover, IL-33/ST2 signaling aberrancy is implicated in the pathogenesis of inflammatory bowel disease (IBD). Accordingly, for this review, we will focus on the role of IL-33 in the regulation of intestinal immunity, involvement in intestinal disease, and implication in potential therapeutics.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunity, Innate , Interleukin-33/immunology , Interleukin-33/metabolism , Intestines/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Humans , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/physiopathology , Interleukin-1/immunology , Interleukin-33/therapeutic use , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th2 Cells/immunology
16.
J Surg Res ; 213: 158-165, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28601308

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a severe intestinal disease of premature infants with high mortality. Studies suggest a causative relationship between red blood cell (RBC) transfusion and NEC; however, whether RBC transfusion leads to worse outcomes in NEC is unknown. We sought to determine whether RBC transfusion was associated with an increased risk of surgical NEC and mortality. METHODS: In this retrospective study, 115 patients were enrolled with NEC Bell's stage 2A or greater from 2010-2015. Patients were classified based on the timing of RBC transfusion before NEC: ≤72 h, >72 h, and no transfusion. Variables including gestational age (GA), birth weight (BW), feedings, and hematocrit levels were analyzed. Outcomes were surgical intervention for NEC following RBC transfusion and mortality. RESULTS: Twenty-three (20%) infants developed NEC ≤ 72 h after RBC transfusion, 16 (69.6%) required surgery with a mortality rate of 21.7% (n = 5). Seventeen (15%) infants developed NEC > 72 h after RBC transfusion, 12 (70.6%) required surgery with a mortality rate of 23.5% (n = 4). 75 (65%) patients developed NEC without RBC transfusion, 17 (22.7%) required surgery with a mortality rate of 4% (n = 3). Lower GA and BW were significantly associated with RBC transfusion and the need for surgical intervention. RBC transfusion ≤72 h before NEC was associated with surgical NEC (pairwise adjusted P < 0.001) and mortality (pairwise adjusted P = 0.048). However, multivariable logistic regression analysis revealed RBC transfusion is not an independent risk factor for surgical NEC. CONCLUSIONS: Infants of lower GA and BW were more likely to receive an RBC transfusion before NEC, which was significantly associated with surgical intervention and an increasing risk of mortality. Judicious use of transfusions in premature infants may improve NEC outcomes.


Subject(s)
Enterocolitis, Necrotizing , Erythrocyte Transfusion/adverse effects , Infant, Premature, Diseases , Enterocolitis, Necrotizing/mortality , Enterocolitis, Necrotizing/surgery , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/mortality , Infant, Premature, Diseases/surgery , Logistic Models , Male , Multivariate Analysis , Retrospective Studies , Risk Factors , Severity of Illness Index , Treatment Outcome
17.
J Immunol ; 194(10): 4931-9, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25862813

ABSTRACT

The mechanisms that lead to the development of remote lung injury after trauma remain unknown, although a central role for the gut in the induction of lung injury has been postulated. We hypothesized that the development of remote lung injury after trauma/hemorrhagic shock requires activation of TLR4 in the intestinal epithelium, and we sought to determine the mechanisms involved. We show that trauma/hemorrhagic shock caused lung injury in wild-type mice, but not in mice that lack TLR4 in the intestinal epithelium, confirming the importance of intestinal TLR4 activation in the process. Activation of intestinal TLR4 after trauma led to increased endoplasmic reticulum (ER) stress, enterocyte apoptosis, and the release of circulating HMGB1, whereas inhibition of ER stress attenuated apoptosis, reduced circulating HMGB1, and decreased lung injury severity. Neutralization of circulating HMGB1 led to reduced severity of lung injury after trauma, and mice that lack HMGB1 in the intestinal epithelium were protected from the development of lung injury, confirming the importance of the intestine as the source of HMGB1, whose release of HMGB1 induced a rapid protein kinase C ζ-mediated internalization of surface tight junctions in the pulmonary epithelium. Strikingly, the use of a novel small-molecule TLR4 inhibitor reduced intestinal ER stress, decreased circulating HMGB1, and preserved lung architecture after trauma. Thus, intestinal epithelial TLR4 activation leads to HMGB1 release from the gut and the development of lung injury, whereas strategies that block upstream TLR4 signaling may offer pulmonary protective strategies after trauma.


Subject(s)
Acute Lung Injury/immunology , Intestinal Mucosa/immunology , Shock, Hemorrhagic/complications , Toll-Like Receptor 4/immunology , Animals , Cell Line , Disease Models, Animal , HMGB1 Protein/biosynthesis , HMGB1 Protein/immunology , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism
18.
J Biol Chem ; 290(18): 11309-20, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25802340

ABSTRACT

Nuclear factor κB (NF-κB) is an important signaling molecule that plays a critical role in the development of acute pancreatitis. Current methods for examining NF-κB activation involve infection of an adenoviral NF-κB-luciferase reporter into cell lines or electrophoretic mobility shift assay of lysate. The use of adeno-associated viruses (AAVs) has proven to be an effective method of transfecting whole organs in live animals. We examined whether intrapancreatic duct infusion of AAV containing an NF-κB-luciferase reporter (AAV-NF-κB-luciferase) can reliably measure pancreatic NF-κB activation. We confirmed the infectivity of the AAV-NF-κB-luciferase reporter in HEK293 cells using a traditional luciferase readout. Mice were infused with AAV-NF-κB-luciferase 5 weeks before induction of pancreatitis (caerulein, 50 µg/kg). Unlike transgenic mice that globally express NF-κB-luciferase, AAV-infused mice showed a 15-fold increase in pancreas-specific NF-κB bioluminescence following 12 h of caerulein compared with baseline luminescence (p < 0.05). The specificity of the NF-κB-luciferase signal to the pancreas was confirmed by isolating the pancreas and adjacent organs and observing a predominant bioluminescent signal in the pancreas compared with liver, spleen, and stomach. A complementary mouse model of post-ERCP-pancreatitis also induced pancreatic NF-κB signals. Taken together these data provide the first demonstration that NF-κB activation can be examined in a live, dynamic fashion during pancreatic inflammation. We believe this technique offers a valuable tool to study real-time activation of NF-κB in vivo.


Subject(s)
Dependovirus/genetics , Luminescent Measurements , Molecular Imaging , NF-kappa B/metabolism , Pancreas/metabolism , Pancreas/virology , Animals , Ceruletide/metabolism , Dependovirus/physiology , HEK293 Cells , Humans , Luciferases/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , Organ Specificity , Signal Transduction
19.
Infect Immun ; 84(3): 782-9, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26729763

ABSTRACT

Interleukin 22 (IL-22) is an IL-10-related cytokine produced by T helper 17 (Th17) cells and other immune cells that signals via IL-22 receptor alpha 1 (IL-22Ra1), which is expressed on epithelial tissues, as well as hepatocytes. IL-22 has been shown to have hepatoprotective effects that are mediated by signal transducer and activator of transcription 3 (STAT3) signaling. However, it is unclear whether IL-22 can directly regulate antimicrobial programs in the liver. To test this hypothesis, hepatocyte-specific IL-22Ra1 knockout (Il22Ra1(Hep-/-)) and Stat3 knockout (Stat3(Hep-/-)) mice were generated and subjected to intra-abdominal infection with Klebsiella pneumoniae, which results in liver injury and necrosis. We found that overexpression of IL-22 or therapeutic administration of recombinant IL-22 (rIL-22), given 2 h postinfection, significantly reduced the bacterial burden in both the liver and spleen. The antimicrobial activity of rIL-22 required hepatic Il22Ra1 and Stat3. Serum from rIL-22-treated mice showed potent bacteriostatic activity against K. pneumoniae, which was dependent on lipocalin 2 (LCN2). However, in vivo, rIL-22-induced antimicrobial activity was only partially reduced in LCN2-deficient mice. We found that rIL-22 also induced serum amyloid A2 (SAA2) and that SAA2 had anti-K. pneumoniae bactericidal activity in vitro. These results demonstrate that IL-22, through IL-22Ra1 and STAT3 singling, can induce intrinsic antimicrobial activity in the liver, which is due in part to LCN2 and SAA2. Therefore, IL-22 may be a useful adjunct in treating hepatic and intra-abdominal infections.


Subject(s)
Interleukins/metabolism , Intraabdominal Infections/metabolism , Klebsiella Infections/metabolism , Klebsiella pneumoniae/physiology , Animals , Female , Humans , Interleukins/administration & dosage , Interleukins/genetics , Intraabdominal Infections/drug therapy , Intraabdominal Infections/genetics , Intraabdominal Infections/microbiology , Klebsiella Infections/drug therapy , Klebsiella Infections/genetics , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL