Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Nat Commun ; 13(1): 874, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169117

ABSTRACT

IL-18 is emerging as an IL-22-induced and epithelium-derived cytokine which contributes to host defence against intestinal infection and inflammation. In contrast to its known role in Goblet cells, regulation of barrier function at the molecular level by IL-18 is much less explored. Here we show that IL-18 is a bona fide IL-22-regulated gate keeper for intestinal epithelial barrier. IL-22 promotes crypt immunity both via induction of phospho-Stat3 binding to the Il-18 gene promoter and via Il-18 independent mechanisms. In organoid culture, while IL-22 primarily increases organoid size and inhibits expression of stem cell genes, IL-18 preferentially promotes organoid budding and induces signature genes of Lgr5+ stem cells via Akt-Tcf4 signalling. During adherent-invasive E. coli (AIEC) infection, systemic administration of IL-18 corrects compromised T-cell IFNγ production and restores Lysozyme+ Paneth cells in Il-22-/- mice, but IL-22 administration fails to restore these parameters in Il-18-/- mice, thereby placing IL-22-Stat3 signalling upstream of the IL-18-mediated barrier defence function. IL-18 in return regulates Stat3-mediated anti-microbial response in Paneth cells, Akt-Tcf4-triggered expansion of Lgr5+ stem cells to facilitate tissue repair, and AIEC clearance by promoting IFNγ+ T cells.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Mucosal/immunology , Interleukin-18/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Animals , Crohn Disease/microbiology , Crohn Disease/pathology , Dysbiosis/microbiology , Escherichia coli/immunology , Interferon-gamma/immunology , Interleukin-18/genetics , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muramidase/metabolism , Organoids , Paneth Cells/immunology , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/metabolism , Tight Junctions/immunology , Interleukin-22
2.
Front Immunol ; 12: 761949, 2021.
Article in English | MEDLINE | ID: mdl-34938288

ABSTRACT

The decline in mucosal immunity during aging increases susceptibility, morbidity and mortality to infections acquired via the gastrointestinal and respiratory tracts in the elderly. We previously showed that this immunosenescence includes a reduction in the functional maturation of M cells in the follicle-associated epithelia (FAE) covering the Peyer's patches, diminishing the ability to sample of antigens and pathogens from the gut lumen. Here, co-expression analysis of mRNA-seq data sets revealed a general down-regulation of most FAE- and M cell-related genes in Peyer's patches from aged mice, including key transcription factors known to be essential for M cell differentiation. Conversely, expression of ACE2, the cellular receptor for SARS-Cov-2 virus, was increased in the aged FAE. This raises the possibility that the susceptibility of aged Peyer's patches to infection with the SARS-Cov-2 virus is increased. Expression of key Paneth cell-related genes was also reduced in the ileum of aged mice, consistent with the adverse effects of aging on their function. However, the increased expression of these genes in the villous epithelium of aged mice suggested a disturbed distribution of Paneth cells in the aged intestine. Aging effects on Paneth cells negatively impact on the regenerative ability of the gut epithelium and could indirectly impede M cell differentiation. Thus, restoring Paneth cell function may represent a novel means to improve M cell differentiation in the aging intestine and increase mucosal vaccination efficacy in the elderly.


Subject(s)
COVID-19 , Immunity, Mucosal/immunology , Immunosenescence/immunology , Paneth Cells/immunology , Peyer's Patches/immunology , Animals , Cell Differentiation/immunology , Mice , Mice, Inbred C57BL , SARS-CoV-2
3.
Sci Immunol ; 6(65): eabf7235, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34739338

ABSTRACT

Deficiency in X-linked inhibitor of apoptosis protein (XIAP) is the cause for X-linked lymphoproliferative syndrome 2 (XLP2). About one-third of these patients suffer from severe and therapy-refractory inflammatory bowel disease (IBD), but the exact cause of this pathogenesis remains undefined. Here, we used XIAP-deficient mice to characterize the mechanisms underlying intestinal inflammation. In Xiap−/− mice, we observed spontaneous terminal ileitis and microbial dysbiosis characterized by a reduction of Clostridia species. We showed that in inflamed mice, both TNF receptor 1 and 2 (TNFR1/2) cooperated in promoting ileitis by targeting TLR5-expressing Paneth cells (PCs) or dendritic cells (DCs). Using intestinal organoids and in vivo modeling, we demonstrated that TLR5 signaling triggered TNF production, which induced PC dysfunction mediated by TNFR1. TNFR2 acted upon lamina propria immune cells. scRNA-seq identified a DC population expressing TLR5, in which Tnfr2 expression was also elevated. Thus, the combined activity of TLR5 and TNFR2 signaling may be responsible for DC loss in lamina propria of Xiap−/− mice. Consequently, both Tnfr1−/−Xiap−/− and Tnfr2−/−Xiap−/− mice were rescued from dysbiosis and intestinal inflammation. Furthermore, RNA-seq of ileal crypts revealed that in inflamed Xiap−/− mice, TLR5 signaling was abrogated, linking aberrant TNF responses with the development of a dysbiosis. Evidence for TNFR2 signaling driving intestinal inflammation was detected in XLP2 patient samples. Together, these data point toward a key role of XIAP in mediating resilience of TLR5-expressing PCs and intestinal DCs, allowing them to maintain tissue integrity and microbiota homeostasis.


Subject(s)
Inflammation/immunology , Intestines/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Toll-Like Receptor 5/immunology , X-Linked Inhibitor of Apoptosis Protein/immunology , Animals , Dendritic Cells/immunology , Dysbiosis/immunology , Humans , Immunity, Innate/immunology , Mice , Mice, Knockout , Paneth Cells/immunology , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type II/deficiency , X-Linked Inhibitor of Apoptosis Protein/deficiency
4.
Sci Immunol ; 6(65): eabf7473, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34739342

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by inappropriate immune responses to the microbiota in genetically susceptible hosts, but little is known about the pathways that link individual genetic alterations to microbiota-dependent inflammation. Here, we demonstrated that the loss of X-linked inhibitor of apoptosis protein (XIAP), a gene associated with Mendelian IBD, rendered Paneth cells sensitive to microbiota-, tumor necrosis factor (TNF)­, receptor-interacting protein kinase 1 (RIPK1)­, and RIPK3-dependent cell death. This was associated with deficiency in Paneth cell­derived antimicrobial peptides and alterations in the stratification and composition of the microbiota. Loss of XIAP was not sufficient to elicit intestinal inflammation but provided susceptibility to pathobionts able to promote granulomatous ileitis, which could be prevented by administration of a Paneth cell­derived antimicrobial peptide. These data reveal a pathway critical for host-microbial cross-talk, which is required for intestinal homeostasis and the prevention of inflammation and which is amenable to therapeutic targeting.


Subject(s)
Inflammation/immunology , Inhibitor of Apoptosis Proteins/immunology , Intestines/immunology , Microbiota/immunology , X-Linked Inhibitor of Apoptosis Protein/immunology , Animals , Antimicrobial Peptides/administration & dosage , Antimicrobial Peptides/biosynthesis , Antimicrobial Peptides/pharmacology , Female , Humans , Inflammation/drug therapy , Inflammation/pathology , Inhibitor of Apoptosis Proteins/deficiency , Inhibitor of Apoptosis Proteins/genetics , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Paneth Cells/chemistry , Paneth Cells/immunology , X-Linked Inhibitor of Apoptosis Protein/deficiency , X-Linked Inhibitor of Apoptosis Protein/genetics
5.
Sci Rep ; 11(1): 9915, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972646

ABSTRACT

Psychological stress has been reported to relate to dysbiosis, imbalance of the intestinal microbiota composition, and contribute to the onset and exacerbation of depression, though, underlying mechanisms of psychological stress-related dysbiosis have been unknown. It has been previously established that α-defensins, which are effector peptides of innate enteric immunity produced by Paneth cells in the small intestine, play an important role in regulation of the intestinal microbiota. However, the relationship between disruption of intestinal ecosystem and α-defensin under psychological stress is yet to be determined. Here we show using chronic social defeat stress (CSDS), a mouse depression model that (1) the exposure to CSDS significantly reduces α-defensin secretion by Paneth cells and (2) induces dysbiosis and significant composition changes in the intestinal metabolites. Furthermore, (3) they are recovered by administration of α-defensin. These results indicate that α-defensin plays an important role in maintaining homeostasis of the intestinal ecosystem under psychological stress, providing novel insights into the onset mechanism of stress-induced depression, and may further contribute to discovery of treatment targets for depression.


Subject(s)
Depression/immunology , Dysbiosis/immunology , Stress, Psychological/complications , alpha-Defensins/metabolism , Administration, Oral , Animals , Depression/drug therapy , Depression/microbiology , Depression/psychology , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/microbiology , Dysbiosis/psychology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Male , Mice , Paneth Cells/immunology , Paneth Cells/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Social Defeat , Stress, Psychological/drug therapy , Stress, Psychological/immunology , Stress, Psychological/psychology , alpha-Defensins/administration & dosage , alpha-Defensins/isolation & purification
6.
J Mol Med (Berl) ; 99(4): 517-530, 2021 04.
Article in English | MEDLINE | ID: mdl-33538854

ABSTRACT

The human gastrointestinal tract is in constant contact with microbial stimuli. Its barriers have to ensure co-existence with the commensal bacteria, while enabling surveillance of intruding pathogens. At the centre of the interaction lies the epithelial layer, which marks the boundaries of the body. It is equipped with a multitude of different innate immune sensors, such as Toll-like receptors, to mount inflammatory responses to microbes. Dysfunction of this intricate system results in inflammation-associated pathologies, such as inflammatory bowel disease. However, the complexity of the cellular interactions, their molecular basis and their development remains poorly understood. In recent years, stem cell-derived organoids have gained increasing attention as promising models for both development and a broad range of pathologies, including infectious diseases. In addition, organoids enable the study of epithelial innate immunity in vitro. In this review, we focus on the gastrointestinal epithelial barrier and its regional organization to discuss innate immune sensing and development.


Subject(s)
Epithelial Cells/immunology , Gastrointestinal Tract/immunology , Immunity, Innate , Organoids , Adult , Animals , Biological Specimen Banks , Cell Polarity , Forecasting , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Goblet Cells/immunology , Humans , Immune Tolerance , Infant, Newborn , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Mice , Models, Immunological , NF-kappa B/physiology , Organ Specificity , Organoids/cytology , Organoids/immunology , Paneth Cells/immunology , Peyer's Patches/immunology , Stem Cells/immunology , Toll-Like Receptors/immunology
7.
Trends Parasitol ; 37(2): 165-175, 2021 02.
Article in English | MEDLINE | ID: mdl-33502317

ABSTRACT

The protozoan parasite Entamoeba histolytica is the causative agent of amebiasis, an infection that manifests as colitis and, in some cases, liver abscess. A better understanding of host protective factors is key to developing an effective remedy. Recently, significant advances have been made in understanding the mechanisms of MUC2 production by goblet cells upon amebic infection, regulation of antimicrobial peptide production by Paneth cells, the interaction of commensal microbiota with immune stimulation, and host genetics in conferring protection from amebiasis. In addition to host pathways that may serve as potential therapeutic targets, significant progress has also been made with respect to development of a vaccine against amebiasis. Here, we aim to highlight the current understanding and knowledge gaps critically.


Subject(s)
Entamoebiasis/immunology , Host-Parasite Interactions/immunology , Entamoeba histolytica , Entamoebiasis/genetics , Entamoebiasis/parasitology , Entamoebiasis/prevention & control , Goblet Cells/immunology , Goblet Cells/parasitology , Humans , Mucin-2/immunology , Paneth Cells/immunology , Pore Forming Cytotoxic Proteins/immunology , Protozoan Vaccines
8.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33483420

ABSTRACT

RNA helicases play roles in various essential biological processes such as RNA splicing and editing. Recent in vitro studies show that RNA helicases are involved in immune responses toward viruses, serving as viral RNA sensors or immune signaling adaptors. However, there is still a lack of in vivo data to support the tissue- or cell-specific function of RNA helicases owing to the lethality of mice with complete knockout of RNA helicases; further, there is a lack of evidence about the antibacterial role of helicases. Here, we investigated the in vivo role of Dhx15 in intestinal antibacterial responses by generating mice that were intestinal epithelial cell (IEC)-specific deficient for Dhx15 (Dhx15 f/f Villin1-cre, Dhx15ΔIEC). These mice are susceptible to infection with enteric bacteria Citrobacter rodentium (C. rod), owing to impaired α-defensin production by Paneth cells. Moreover, mice with Paneth cell-specific depletion of Dhx15 (Dhx15 f/f Defensinα6-cre, Dhx15ΔPaneth) are more susceptible to DSS (dextran sodium sulfate)-induced colitis, which phenocopy Dhx15ΔIEC mice, due to the dysbiosis of the intestinal microbiota. In humans, reduced protein levels of Dhx15 are found in ulcerative colitis (UC) patients. Taken together, our findings identify a key regulator of Wnt-induced α-defensins in Paneth cells and offer insights into its role in the antimicrobial response as well as intestinal inflammation.


Subject(s)
Colitis/immunology , Defensins/genetics , Enterobacteriaceae Infections/immunology , Paneth Cells/immunology , RNA Helicases/genetics , Wnt Signaling Pathway , Animals , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Defensins/immunology , Dextran Sulfate/administration & dosage , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Paneth Cells/microbiology , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA Helicases/immunology
9.
Gastroenterology ; 160(4): 1269-1283, 2021 03.
Article in English | MEDLINE | ID: mdl-33217447

ABSTRACT

BACKGROUND AND AIMS: Vitamin D exerts a regulatory role over mucosal immunity via the vitamin D receptor (VDR). Although Paneth cells and their products are known to regulate the commensal and pathogenic microbiota, the role that VDRs in Paneth cells play in these responses is unknown. METHODS: We identified the decreased intestinal VDR significantly correlated with reduction of an inflammatory bowel disease risk gene ATG16L1 and Paneth cell lysozymes in patients with Crohn's disease. We generated Paneth cell-specific VDR knockout (VDRΔPC) mice to investigate the molecular mechanisms. RESULTS: Lysozymes in the Paneth cells were significantly decreased in the VDRΔPC mice. Isolated VDRΔPC Paneth cells exhibited weakened inhibition of pathogenic bacterial growth and displayed reduced autophagic responses. VDRΔPC mice had significantly higher inflammation after Salmonella infections. VDRΔPC mice also showed high susceptibility to small intestinal injury induced by indomethacin, a nonsteroidal anti-inflammatory drug. Co-housing of VDRΔPC and VDRlox mice made the VDRΔPC less vulnerable to dextran sulfate sodium colitis, suggesting the transmission of protective bacterial from the VDRlox mice. Thus, a lack of VDR in Paneth cells leads to impaired antibacterial activities and consequently increased inflammatory responses. Genetically and environmentally regulated VDRs in the Paneth cells may set the threshold for the development of chronic inflammation, as observed in inflammatory bowel diseases. CONCLUSIONS: We provide new insights into the tissue-specific functions of VDRs in maintaining Paneth cell alertness to pathogens in intestinal disorders. Targeting the VDR affects multiple downstream events within Paneth cells that inhibit intestinal inflammation and establish host defense against enteropathogens.


Subject(s)
Crohn Disease/immunology , Microbiota/immunology , Paneth Cells/immunology , Receptors, Calcitriol/metabolism , Animals , Autophagy , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Biopsy , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Crohn Disease/chemically induced , Crohn Disease/genetics , Crohn Disease/microbiology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Ileum/immunology , Ileum/microbiology , Ileum/pathology , Immunity, Mucosal , Male , Mice , Mice, Knockout , Muramidase/metabolism , Paneth Cells/metabolism , Receptors, Calcitriol/genetics , Vitamin D/metabolism
10.
Commun Biol ; 3(1): 252, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444775

ABSTRACT

Tumors have evolved mechanisms to escape anti-tumor immunosurveillance. They limit humoral and cellular immune activities in the stroma and render tumors resistant to immunotherapy. Sensitizing tumor cells to immune attack is an important strategy to revert immunosuppression. However, the underlying mechanisms of immune escape are still poorly understood. Here we discover Indoleamine-2,3-dioxygenase-1 (IDO1)+ Paneth cells in the stem cell niche of intestinal crypts and tumors, which promoted immune escape of colorectal cancer (CRC). Ido1 expression in Paneth cells was strictly Stat1 dependent. Loss of IDO1+ Paneth cells in murine intestinal adenomas with tumor cell-specific Stat1 deletion had profound effects on the intratumoral immune cell composition. Patient samples and TCGA expression data suggested corresponding cells in human colorectal tumors. Thus, our data uncovered an immune escape mechanism of CRC and identify IDO1+ Paneth cells as a target for immunotherapy.


Subject(s)
Colorectal Neoplasms/pathology , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Intestinal Neoplasms/pathology , Paneth Cells/immunology , STAT1 Transcription Factor/physiology , Animals , Colorectal Neoplasms/etiology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Intestinal Neoplasms/immunology , Intestinal Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Clin Transl Gastroenterol ; 12(1): e00296, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33464728

ABSTRACT

INTRODUCTION: Histopathological alterations in the ileum and colon in irritable bowel syndrome (IBS) are controversial, and normal values are poorly established. We hypothesized that changes in mucosal immune cells characterize IBS and key changes in immune composition are associated with the mucosa-associated microbiota (MaM). METHODS: A nested case-control study (48 IBS and 106 controls included) from 745 colonoscopy participants in a random population sample. Intraepithelial lymphocytes (IELs)/100 enterocytes and eosinophils/5 nonoverlapping high-power fields counted; mast cells identified by immunocytochemistry (CD117)/5 high-power fields. Paneth cells quantified per 5 crypts. 16S rRNA gene amplicon sequencing performed on available sigmoid MaM, n = 55 and fecal microbiota, n = 20. Microbiota profiles compared between samples with high and low IEL counts. RESULTS: IBS had increased IELs in the terminal ileum (relative risk ratio = 1.70, 95% confidence interval 1.08-2.76, P = 0.022 adjusted for age, sex, and smoking). Cecal IELs were increased in IBS-diarrhea (relative risk ratio = 2.03, 95% confidence interval 1.13-3.63, P = 0.017). No difference was observed in alpha diversity of MaM or fecal microbiota based on IEL count. There was no difference in beta diversity of the MaM according to IEL count in the terminal ileal (TI) (P = 0.079). High TI IEL counts associated with a significant expansion of the genus Blautia (P = 0.024) and unclassified Clostridiales (P = 0.036) in colon MaM. DISCUSSION: A modest but significant increase in IELs was observed in IBS vs. controls in a population-based setting. Subtle TI and cecal inflammation may play a pathogenic role in IBS but needs confirmation. Modest but discernible differences in the colonic MaM were seen according to TI IEL count but not IBS status.


Subject(s)
Colon/pathology , Gastrointestinal Microbiome/immunology , Ileum/pathology , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/immunology , Adult , Biopsy , Case-Control Studies , Clostridiales/genetics , Clostridiales/immunology , Clostridiales/isolation & purification , Colon/diagnostic imaging , Colon/immunology , Colon/microbiology , DNA, Bacterial/isolation & purification , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Humans , Ileum/diagnostic imaging , Ileum/immunology , Ileum/microbiology , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Male , Mast Cells/immunology , Middle Aged , Paneth Cells/immunology , RNA, Ribosomal, 16S/genetics
12.
Parasitol Int ; 74: 101923, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31054916

ABSTRACT

There is limited information regarding responses by slow cycling stem cells during T. spiralis-induced T-cell mediated intestinal inflammation and how such responses may relate to those of Paneth cells. Transgenic mice, in which doxycycline induces expression of histone 2B (H2B)-green fluorescent protein (GFP), were used. Following discontinuation of doxycycline ("chase" period), retention of H2B-GFP enabled the identification of slow cycling stem cells and long-lived Paneth cells. Inflammation in the small intestine (SI) was induced by oral administration of T. spiralis muscle larvae. Epithelial retention of H2B-GFP per crypt cell position (cp) was studied following immunohistochemistry and using the Score and Wincrypts program. Compared to non-infected controls, there was significant reduction in the number of H2B-GFP-retaining stem cells in T. spiralis-infected small intestines. H2B-GFP-retaining stem cells peaked at around cp 4 in control sections, but smaller peaks at higher cell positions (>10) were seen in sections of inflamed small intestines. In the latter, there was a significant increase in the total number of Paneth cells, with significant reduction in H2B-GFP-retaining Paneth cells, but a marked increase in unlabelled (H2B-GFP-negative) Paneth cells. In conclusion, following T. spiralis-infection, putative slow cycling stem cell numbers were reduced. A marked increase in newly generated Paneth cells at the crypt base led to higher cell positions of the remaining slow cycling stem cells.


Subject(s)
Intestine, Small/cytology , Paneth Cells/parasitology , Stem Cells/parasitology , Trichinellosis/immunology , Animals , Cell Cycle , Female , Green Fluorescent Proteins/genetics , Histones/genetics , Immunohistochemistry , Intestine, Small/immunology , Intestine, Small/parasitology , Male , Mice , Mice, Transgenic , Paneth Cells/immunology , Stem Cells/immunology , Trichinella spiralis
13.
Cell Mol Gastroenterol Hepatol ; 9(4): 611-625, 2020.
Article in English | MEDLINE | ID: mdl-31862317

ABSTRACT

BACKGROUND & AIMS: The protective intestinal mucosal barrier consists of multiple elements including mucus and epithelial layers and immune defense; nonetheless, barrier dysfunction is common in various disorders. The imprinted and developmentally regulated long noncoding RNA H19 is involved in many cell processes and diseases. Here, we investigated the role of H19 in regulating Paneth and goblet cells and autophagy, and its impact on intestinal barrier dysfunction induced by septic stress. METHODS: Studies were conducted in H19-deficient (H19-/-) mice, mucosal tissues from patients with sepsis, primary enterocytes, and Caco-2 cells. Septic stress was induced by cecal ligation and puncture (CLP), and gut permeability was detected by tracer fluorescein isothiocyanate-dextran assays. The function of Paneth and goblet cells was examined by immunostaining for lysozyme and mucin 2, respectively, and autophagy was examined by microtubule-associated proteins 1A/1B light chain 3 II immunostaining and Western blot analysis. Intestinal organoids were isolated from H19-/- and control littermate mice and treated with lipopolysaccharide (LPS). RESULTS: Intestinal mucosal tissues in mice 24 hours after exposure to CLP and in patients with sepsis showed high H19 levels, associated with intestinal barrier dysfunction. Targeted deletion of the H19 gene in mice enhanced the function of Paneth and goblet cells and promoted autophagy in the small intestinal mucosa. Knockout of H19 protected Paneth and goblet cells against septic stress, preserved autophagy activation, and promoted gut barrier function after exposure to CLP. Compared with organoids from control littermate mice, intestinal organoids isolated from H19-/- mice had increased numbers of lysozyme- and mucin 2-positive cells and showed increased tolerance to LPS. Conversely, ectopic overexpression of H19 in cultured intestinal epithelial cells prevented rapamycin-induced autophagy and abolished the rapamycin-induced protection of the epithelial barrier against LPS. CONCLUSIONS: In investigations of mice, human tissues, primary organoids, and intestinal epithelial cells, we found that increased H19 inhibited the function of Paneth and goblet cells and suppressed autophagy, thus potentially contributing to barrier dysfunction in intestinal pathologies.


Subject(s)
Autophagy/genetics , Goblet Cells/pathology , Paneth Cells/pathology , RNA, Long Noncoding/metabolism , Sepsis/pathology , Animals , Autophagy/immunology , Caco-2 Cells , Disease Models, Animal , Female , Goblet Cells/immunology , Humans , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/pathology , Male , Mice , Mice, Knockout , Organoids , Paneth Cells/immunology , Permeability , RNA, Long Noncoding/genetics , Sepsis/immunology
14.
Front Immunol ; 10: 2289, 2019.
Article in English | MEDLINE | ID: mdl-31608070

ABSTRACT

The main environmental risk factor associated with the development of Crohn's disease (CD) is cigarette smoking. Although the mechanism is still unknown, some studies have shown that cigarette exposure affects the intestinal barrier of the small bowel. Among the factors that may be involved in this process are Paneth cells. These specialized epithelial cells are located into the small intestine, and they are able to secrete antimicrobial peptides, having an essential role in the control of the growth of microorganisms. Alterations in its function are associated with inflammatory processes, such as CD. To study how cigarette components impact ileum homeostasis and Paneth cells integrity, we used intragastric administration of cigarette smoke condensate (CSC) in mice. Our results showed that inflammation was triggered after mucosal exposure of CSC, which induced particular alterations in Paneth cells granules, antimicrobial peptide production, and a reduction of bactericidal capacity. In fact, exposure to CSC generated an imbalance in the fecal bacterial population and increased the susceptibility of mice to develop ileal damage in response to bacterial infection. Moreover, our results obtained in mice unable to produce interleukin 10 (IL-10-/- mice) suggest that CSC treatment can induce a symptomatic enterocolitis with a pathological inflammation in genetically susceptible individuals.


Subject(s)
Ileum/immunology , Inflammation/immunology , Intestinal Mucosa/immunology , Tobacco Products/adverse effects , Animals , Crohn Disease/immunology , Crohn Disease/microbiology , Ileum/microbiology , Inflammation/microbiology , Interleukin-10/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/immunology , Paneth Cells/microbiology
15.
PLoS One ; 14(9): e0221618, 2019.
Article in English | MEDLINE | ID: mdl-31509557

ABSTRACT

Paneth cells (PCs) are specialized epithelial cells of the small bowel that contain multiple secretory granules filled with antimicrobial peptides and trophic factors, which are essential for the control of the microorganisms growth and maintaining intestinal integrity. Alterations in their function are associated with an imbalance of the normal microbiota, gastrointestinal infections and inflammatory processes, such as Crohn's disease (CD). One of the most common murine models for studying CD is IL-10-/- mouse. IL-10-/- mice when housed in conventional conditions and take contact with commensal microorganisms develop an acute enterocolitis mediated by a Th1 immune response. Even though, alterations in PCs function are related to CD, they had not been characterized yet in this mouse model. Here we show that in specific pathogen free conditions IL-10-/- mice have aberrant granules and a large number of immature PCs at the bottom of the crypt in the ileum of IL-10-/- mice before developing intestinal inflammation, along with a reduced expression of Indian Hedgehog. In addition, IL-10-/- Paneth cells presented a reduced expression of cryptidin-4, and a heterogeneous distribution of lysozyme+ granules. The alterations in the maturation of the PCs at the bottom of the crypt were not modified after the colonization by the conventional microbiota. On the other hand, depletion of microbiota altered the phenotype, but did not normalize PCs. Our results suggest that IL-10 could be necessary for the integrity of PCs. Moreover, our results help to explain why IL-10-/- mice develop enterocolitis in response to microorganisms.


Subject(s)
Interleukin-10/genetics , Paneth Cells/cytology , Secretory Vesicles/metabolism , alpha-Defensins/genetics , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Male , Mice , Microbiota , Paneth Cells/immunology , Paneth Cells/metabolism , Phenotype , Specific Pathogen-Free Organisms , Th1 Cells/immunology
16.
Gastroenterology ; 157(5): 1310-1322.e13, 2019 11.
Article in English | MEDLINE | ID: mdl-31352002

ABSTRACT

BACKGROUND & AIMS: Interferon lambda (IFNL) is expressed at high levels by intestinal epithelial cells (IECs) and mucosal immune cells in response to infection and inflammation. We investigated whether IFNL might contribute to pathogenesis of Crohn's disease (CD). METHODS: We obtained serum samples and terminal ileum biopsies from 47 patients with CD and 16 healthy individuals (controls). We measured levels of IFNL by enzyme-linked immunosorbent assay and immunohistochemistry and location of expression by confocal microscopy. Activation of IFNL signaling via STAT1 was measured in areas of no, mild, moderate, and severe inflammation and correlated with Paneth cell homeostasis and inflammation. IFNL expression and function were studied in wild-type mice and mice with intestinal epithelial cell-specific (ΔIEC) disruption or full-body disruption of specific genes (Mlkl-/-, Stat1ΔIEC, Casp8ΔIEC, Casp8ΔIECRipk3-/-, Casp8ΔIECTnfr-/-, Casp8ΔIECMlkl-/-, and Nod2-/- mice). Some mice were given tail vein injections of a vector encoding a secreted form of IFNL. Intestinal tissues were collected from mice and analyzed by immunohistochemistry and immunoblots. We generated 3-dimensional small intestinal organoids from mice and studied the effects of IFNL and inhibitors of STAT-signaling pathway. RESULTS: Patients with CD had significant increases in serum and ileal levels of IFNL compared with controls. Levels of IFNL were highest in ileum tissues with severe inflammation. High levels of IFNL associated with a reduced number of Paneth cells and increased cell death at the crypt bottom in inflamed ileum samples. Intestinal tissues from the ileum of wild-type mice injected with a vector expressing IFNL had reduced numbers of Paneth cells. IFNL-induced death of Paneth cells in mice did not occur via apoptosis, but required Mixed Lineage Kinase Domain Like (MLKL) and activation of Signal transducer and activator of transcription 1 (STAT1). In organoids, inhibitors of Janus kinase (JAK) signaling via STAT1 (glucocorticoids, tofacitinib, or filgotinib) reduced expression of proteins that mediate cell death and prevented Paneth cell death. CONCLUSIONS: Levels of IFNL are increased in serum and inflamed ileal tissues from patients with CD and associated with a loss of Paneth cells. Expression of a secreted form of IFNL in mice results in loss of Paneth cells from intestinal tissues, via STAT1 and MLKL, controlled by caspase 8. Strategies to reduce IFNL or block its effects might be developed for treatment of patients with CD affecting the terminal ileum.


Subject(s)
Crohn Disease/metabolism , Ileum/metabolism , Interferons/metabolism , Interleukins/metabolism , Paneth Cells/metabolism , STAT1 Transcription Factor/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Death , Crohn Disease/immunology , Crohn Disease/pathology , Disease Models, Animal , Humans , Ileum/immunology , Ileum/pathology , Interferons/genetics , Interleukins/genetics , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/immunology , Paneth Cells/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Signal Transduction , Tissue Culture Techniques , Up-Regulation
17.
J Crohns Colitis ; 13(8): 1081-1091, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31094420

ABSTRACT

AIMS: NOD2 and CD1d play a key role in innate immunity by recognizing conserved molecular patterns of pathogens. While NOD2-/- and CD1d-/- mice display structural and functional alterations in Paneth cells, animal studies have reported no impact of NOD2 or CD1d deficiency on experimental colitis. NOD2 mutations increase the susceptibility to inflammatory bowel diseases and the CD1d bound to α-galactosylceramide [α-GalCer] alleviates intestinal inflammation. We evaluated the effect of CD1d modulation on experimental colitis in NOD2-/- mice. METHODS: The effect of CD1d augmentation and depletion in NOD2-/- mice was assessed in a dextran sodium sulphate [DSS]-induced colitis model via administration of α-GalCer and construction of NOD2-/-CD1d-/- mice. The structural and functional changes in Paneth cells were evaluated using transmission electron microscopy and pilocarpine administration. Colitogenic taxa were analysed in the faeces of NOD2-/-CD1d-/- mice using 16S rRNA gene sequencing. RESULTS: In NOD2-/- mice, α-GalCer alleviated and CD1d depletion [NOD2-/-CD1d-/- mice] aggravated colitis activity and histology compared with co-housed littermates NOD2-/-, CD1d-/- and wild-type mice after administration of 3% DSS. In NOD2-/-CD1d-/- mice, the ultrastructure and degranulation ability of secretary granules in Paneth cells were altered and the intestinal microbial composition differed from that of their littermates. Faecal microbiota transplantation [FMT] with NOD2-/-CD1d-/- mice faeces into wild-type mice aggravated DSS-induced colitis, while FMT with wild-type mice faeces into NOD2-/-CD1d-/- mice alleviated DSS-induced colitis. Acetatifactor muris was identified only in NOD2-/-CD1d-/- mice faeces and the oral gavage of A. muris in wild-type mice aggravated DSS-induced colitis. CONCLUSION: CD1d modulates colonic inflammation in NOD2-/- mice by altering the intestinal microbial composition comprising A. muris.


Subject(s)
Antigens, CD1d/immunology , Clostridiales , Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases/immunology , Nod2 Signaling Adaptor Protein/immunology , Paneth Cells/immunology , Animals , Clostridiales/immunology , Clostridiales/isolation & purification , Disease Models, Animal , Inflammation/immunology , Inflammatory Bowel Diseases/pathology , Mice
18.
Kidney Int ; 95(4): 859-879, 2019 04.
Article in English | MEDLINE | ID: mdl-30777286

ABSTRACT

Intestinal Paneth cells play a critical role in ischemic acute kidney injury (AKI) by releasing interleukin 17A (IL-17A). Because Toll-like receptor 9 (TLR9) activation degranulates Paneth cells and necrotic tubular epithelial cells release several damage associated molecular patterns that target TLR9, we tested the hypothesis that intestinal TLR9 deficiency would protect against ischemic AKI and associated remote intestinal and hepatic dysfunction by decreasing Paneth cell degranulation. We generated mice lacking TLR9 in intestinal epithelia (TLR9fl/fl Villin Cre mice) and compared them to wild type (TLR9fl/fl) mice following right nephrectomy and left ischemia/reperfusion. To our surprise, mice lacking intestinal TLR9 had exacerbated kidney, liver, and small intestine injury after ischemia/reperfusion compared to wild type mice, characterized by increased kidney and intestinal inflammation, apoptosis, and necrosis as well as increased hepatic inflammation and apoptosis. Mice lacking intestinal TLR9 had larger Paneth cell granule size, pronounced intestinal macrophage infiltration, and higher intestinal crypt IL-17A expression. Administration of IL-17A neutralizing antibody prevented the exacerbation of ischemic AKI in mice lacking intestinal TLR9. These studies suggest that intestinal TLR9 activation protects against ischemic AKI and associated remote multi-organ dysfunction syndrome by regulating Paneth cell IL-17A synthesis.


Subject(s)
Acute Kidney Injury/immunology , Interleukin-17/metabolism , Multiple Organ Failure/immunology , Paneth Cells/pathology , Toll-Like Receptor 9/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis , Disease Models, Animal , Disease Progression , Humans , Hyperplasia/immunology , Hyperplasia/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestines/immunology , Intestines/pathology , Kidney/immunology , Kidney/pathology , Liver/immunology , Liver/pathology , Macrophages/immunology , Male , Mice , Mice, Transgenic , Multiple Organ Failure/pathology , Paneth Cells/immunology , Paneth Cells/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Toll-Like Receptor 9/genetics
19.
Semin Cell Dev Biol ; 88: 138-146, 2019 04.
Article in English | MEDLINE | ID: mdl-29355606

ABSTRACT

The gut is the biggest immune organ in the body that encloses commensal microbiota which aids in food digestion. Paneth cells, positioned at the frontline of host-microbiota interphase, can modulate the composition of microbiota. Paneth cells achieve this via the delivery of microbicidal substances, among which enteric α-defensins play the primary role. If microbiota is dysregulated, it can impact the function of the local mucosal immune system. Importantly, this system is also exposed to an enormous number of antigens which are derived from the gut-resident microbiota and processed food, and may potentially trigger undesirable local inflammatory responses. To understand the intricate regulations and liaisons between Paneth cells, microbiota and the immune system in this intestinal-specific setting, one must consider their mode of interaction in a wider context of regulatory processes which impose immune tolerance not only to self, but also to microbiota and food-derived antigens. These include, but are not limited to, tolerogenic mechanisms of central tolerance in the thymus and peripheral tolerance in the secondary lymphoid organs, and the intestine itself. Defects in these processes can compromise homeostasis in the intestinal mucosal immunity. In this review, which is focused on tolerance to intestinal antigens and its relevance for the pathogenesis of gut immune diseases, we provide an outline of such multilayered immune control mechanisms and highlight functional links that underpin their cooperative nature.


Subject(s)
Dysbiosis/prevention & control , Gastrointestinal Tract/immunology , Paneth Cells/immunology , Peripheral Tolerance , alpha-Defensins/immunology , Animals , Central Tolerance , Dysbiosis/immunology , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Gene Expression/immunology , Homeostasis/immunology , Humans , Immunity, Mucosal/drug effects , Inflammation , Paneth Cells/drug effects , Paneth Cells/microbiology , Symbiosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , alpha-Defensins/biosynthesis , alpha-Defensins/pharmacology
20.
Front Immunol ; 9: 2522, 2018.
Article in English | MEDLINE | ID: mdl-30455690

ABSTRACT

In gut epithelium, IL-22 transmits signals through STAT3 phosphorylation (pSTAT3) which provides intestinal immunity. Many components in the IL-22-pSTAT3 pathway have been identified as risk factors for inflammatory bowel disease (IBD) and some of them are considered as promising therapeutic targets. However, new perspectives are still needed to understand IL-22-pSTAT3 signaling for effective clinical interventions in IBD patients. Here, we revealed activating transcription factor 3 (ATF3), recently identified to be upregulated in patients with active IBD, as a crucial player in the epithelial IL-22-pSTAT3 signaling cascade. We found ATF3 is central to intestinal homeostasis and provides protection during colitis. Loss of ATF3 led to decreased crypt numbers, more shortened colon length, impaired ileal fucosylation at the steady state, and lethal disease activity during DSS-induced colitis which can be effectively ameliorated by rectal transplantation of wild-type colonic organoids. Epithelial stem cells and Paneth cells form a niche to orchestrate epithelial regeneration and host-microbe interactions, and IL-22-pSTAT3 signaling is a key guardian for this niche. We found ATF3 is critical for niche maintenance as ATF3 deficiency caused compromised stem cell growth and regeneration, as well as Paneth cell degeneration and loss of anti-microbial peptide (AMP)-producing granules, indicative of malfunction of Paneth/stem cell network. Mechanistically, we found IL-22 upregulates ATF3, which is required to relay IL-22 signaling leading to STAT3 phosphorylation and subsequent AMP induction. Intriguingly, ATF3 itself does not act on STAT3 directly, instead ATF3 regulates pSTAT3 by negatively targeting protein tyrosine phosphatases (PTPs) including SHP2 and PTP-Meg2. Furthermore, we identified ATF3 is also involved in IL-6-mediated STAT3 activation in T cells and loss of ATF3 leads to reduced capacity of Th17 cells to produce their signature cytokine IL-22 and IL-17A. Collectively, our results suggest that via IL-22-pSTAT3 signaling in the epithelium and IL-6-pSTAT3 signaling in Th17 cells, ATF3 mediates a cross-regulation in the barrier to maintain mucosal homeostasis and immunity.


Subject(s)
Activating Transcription Factor 3/metabolism , Colitis/pathology , Interleukin-6/metabolism , Interleukins/metabolism , STAT3 Transcription Factor/metabolism , Activating Transcription Factor 3/genetics , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/immunology , Colon/pathology , Epithelial Cells/metabolism , Homeostasis/immunology , Ileum/pathology , Immunity, Mucosal/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/immunology , Paneth Cells/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/immunology , Stem Cells/metabolism , Th17 Cells/immunology , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...