Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Hum Genet ; 108(6): 1126-1137, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34010604

ABSTRACT

Dysregulated transforming growth factor TGF-ß signaling underlies the pathogenesis of genetic disorders affecting the connective tissue such as Loeys-Dietz syndrome. Here, we report 12 individuals with bi-allelic loss-of-function variants in IPO8 who presented with a syndromic association characterized by cardio-vascular anomalies, joint hyperlaxity, and various degree of dysmorphic features and developmental delay as well as immune dysregulation; the individuals were from nine unrelated families. Importin 8 belongs to the karyopherin family of nuclear transport receptors and was previously shown to mediate TGF-ß-dependent SMADs trafficking to the nucleus in vitro. The important in vivo role of IPO8 in pSMAD nuclear translocation was demonstrated by CRISPR/Cas9-mediated inactivation in zebrafish. Consistent with IPO8's role in BMP/TGF-ß signaling, ipo8-/- zebrafish presented mild to severe dorso-ventral patterning defects during early embryonic development. Moreover, ipo8-/- zebrafish displayed severe cardiovascular and skeletal defects that mirrored the human phenotype. Our work thus provides evidence that IPO8 plays a critical and non-redundant role in TGF-ß signaling during development and reinforces the existing link between TGF-ß signaling and connective tissue defects.


Subject(s)
Bone Diseases/etiology , Cardiovascular Diseases/etiology , Connective Tissue Diseases/etiology , Immunity, Cellular/immunology , Loss of Function Mutation , Loss of Heterozygosity , beta Karyopherins/genetics , Adolescent , Adult , Animals , Bone Diseases/pathology , Cardiovascular Diseases/pathology , Child , Connective Tissue Diseases/pathology , Female , Humans , Infant , Male , Middle Aged , Pedigree , Phenotype , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Young Adult , Zebrafish , beta Karyopherins/metabolism
4.
PLoS One ; 13(10): e0205826, 2018.
Article in English | MEDLINE | ID: mdl-30365510

ABSTRACT

Mutations in interleukin-10 receptor (IL-10R) genes are one cause of very early-onset inflammatory bowel disease with perianal lesions, which can be cured by hematopoietic stem cell transplantation. Using a functional test, which assesses responsiveness of peripheral monocytes to IL-10, we identified three unrelated Portuguese patients carrying two novel IL-10RB mutations. In the three patients, sequencing of genomic DNA identified the same large deletion of exon 3 which precluded protein expression. This mutation was homozygous in two patients born from consanguineous families and heterozygous in the third patient born from unrelated parents. Microsatellite analysis of the IL10RB genomic region revealed a common haplotype in the three Portuguese families pointing to a founder deletion inherited from a common ancestor 400 years ago. In the third patient, surface expression of IL-10R was normal but signaling in response to IL-10 was impaired. Complementary DNA sequencing and next-generation sequencing of IL10RB locus with custom-made probes revealed a ≈ 6 Kb duplication encompassing the exon 6 which leads to a frameshift mutation and a loss of the TYK2-interacting Box 2 motif. Altogether, we describe two novel copy number variations in IL10RB, one with founder effect and one preserving cell surface expression but abolishing signaling.


Subject(s)
DNA Copy Number Variations , Interleukin-10 Receptor beta Subunit/deficiency , Interleukin-10 Receptor beta Subunit/genetics , Alleles , Amino Acid Motifs , DNA, Complementary/genetics , Exons , Family Health , Female , Founder Effect , Genome, Human , Haplotypes , Heterozygote , Homozygote , Humans , Infant , Leukocytes, Mononuclear/cytology , Male , Microsatellite Repeats , Mutation , Portugal , Signal Transduction
5.
J Crohns Colitis ; 12(9): 1104-1112, 2018 08 29.
Article in English | MEDLINE | ID: mdl-29788237

ABSTRACT

BACKGROUND AND AIMS: An expanding number of monogenic defects have been identified as causative of severe forms of very early-onset inflammatory bowel diseases [VEO-IBD]. The present study aimed at defining how next-generation sequencing [NGS] methods can be used to improve identification of known molecular diagnosis and to adapt treatment. METHODS: A total of 207 children were recruited in 45 paediatric centres through an international collaborative network [ESPGHAN GENIUS working group] with a clinical presentation of severe VEO-IBD [n = 185] or an anamnesis suggestive of a monogenic disorder [n = 22]. Patients were divided at inclusion into three phenotypic subsets: predominantly small bowel inflammation, colitis with perianal lesions, and colitis only. Methods to obtain molecular diagnosis included functional tests followed by specific Sanger sequencing, custom-made targeted NGS, and in selected cases whole exome sequencing [WES] of parents-child trios. Genetic findings were validated clinically and/or functionally. RESULTS: Molecular diagnosis was achieved in 66/207 children [32%]: 61% with small bowel inflammation, 39% with colitis and perianal lesions, and 18% with colitis only. Targeted NGS pinpointed gene mutations causative of atypical presentations, and identified large exonic copy number variations previously missed by WES. CONCLUSIONS: Our results lead us to propose an optimised diagnostic strategy to identify known monogenic causes of severe IBD.


Subject(s)
High-Throughput Nucleotide Sequencing , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Adolescent , Age of Onset , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Inflammatory Bowel Diseases/therapy , Male , Predictive Value of Tests
6.
EMBO Mol Med ; 10(4)2018 04.
Article in English | MEDLINE | ID: mdl-29567797

ABSTRACT

Herein, we report the first identification of biallelic-inherited mutations in ALPI as a Mendelian cause of inflammatory bowel disease in two unrelated patients. ALPI encodes for intestinal phosphatase alkaline, a brush border metalloenzyme that hydrolyses phosphate from the lipid A moiety of lipopolysaccharides and thereby drastically reduces Toll-like receptor 4 agonist activity. Prediction tools and structural modelling indicate that all mutations affect critical residues or inter-subunit interactions, and heterologous expression in HEK293T cells demonstrated that all ALPI mutations were loss of function. ALPI mutations impaired either stability or catalytic activity of ALPI and rendered it unable to detoxify lipopolysaccharide-dependent signalling. Furthermore, ALPI expression was reduced in patients' biopsies, and ALPI activity was undetectable in ALPI-deficient patient's stool. Our findings highlight the crucial role of ALPI in regulating host-microbiota interactions and restraining host inflammatory responses. These results indicate that ALPI mutations should be included in screening for monogenic causes of inflammatory bowel diseases and lay the groundwork for ALPI-based treatments in intestinal inflammatory disorders.


Subject(s)
Alkaline Phosphatase/metabolism , Inflammatory Bowel Diseases/metabolism , Alkaline Phosphatase/deficiency , Alkaline Phosphatase/genetics , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Homeostasis , Humans , Inflammatory Bowel Diseases/genetics , Intestines , Mutation/genetics , Signal Transduction/physiology
8.
J Pediatr Gastroenterol Nutr ; 64(3): 378-384, 2017 03.
Article in English | MEDLINE | ID: mdl-27253662

ABSTRACT

OBJECTIVE: Early-onset inflammatory bowel diseases can result from a wide spectrum of rare mendelian disorders. Early molecular diagnosis is crucial in defining treatment and in improving life expectancy. Herein we aimed at defining the mechanism of an immunodeficiency-polyendrocrinopathy and enteropathy-X-linked (IPEX)-like disease combined with a severe immunodeficiency in 2 siblings born from distantly related parents. METHODS: Whole exome sequencing was performed on blood-extracted genomic DNA from the 2 affected children and their parents on the genomic platform of Institut IMAGINE. Candidate gene mutation was identified using the in-house software PolyWeb and confirmed by Sanger sequencing. Protein expression was determined by western blot. Flow cytometry was used to assess consequences of the mutation on lymphocyte phenotype and nuclear factor-kappa B (NF-κB) activation at diagnosis and after treatment by hematopoietic stem cell transplantation. RESULTS: We identified a homozygous missense mutation in mucosa-associated lymphoid tissue lymphoma translocation 1 gene (MALT1), which precluded protein expression. In keeping with the known function of MALT1, NF-κB-dependent lymphocyte activation was severely impaired. Moreover, there was a drastic reduction in Forkhead box P3 (FOXP3) regulatory T cells accounting for the IPEX-like phenotype. Following identification of the mutation, both children received hematopoietic stem cell transplantation, which permitted full clinical recovery. Immunological workup at 6 and 12 months after transplantation showed normal NF-κB activation and correction of regulatory T cells frequency. CONCLUSIONS: Along with FOXP3, interleukin 2 receptor alpha chain (IL2RA), and cytotoxic T-lymphocyte protein 4 precursor (CTLA-4) mutations, MALT1 deficiency should now be considered as a possible cause of IPEX-like syndrome associated with immunodeficiency that can be cured by hematopoietic stem cell transplantation.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/genetics , Genetic Diseases, X-Linked/genetics , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/deficiency , Mutation, Missense , Child , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diarrhea/diagnosis , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Markers , Homozygote , Humans , Immune System Diseases/diagnosis , Immune System Diseases/genetics , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Siblings
9.
J Allergy Clin Immunol ; 134(5): 1131-41.e9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24942515

ABSTRACT

BACKGROUND: Crohn disease is an inflammatory bowel disease (IBD) with a complex mode of inheritance. Although nucleotide binding and oligomerization domain containing 2 (NOD2) is the strongest risk factor, the cause of Crohn disease remains unknown in the majority of the cases. X-linked inhibitor of apoptosis (XIAP) deficiency causes X-linked lymphoproliferative syndrome type 2. IBD has been reported in some XIAP-deficient patients. OBJECTIVE: We characterize the IBD affecting a large cohort of patients with mutations in XIAP and examine the possible pathophysiologic mechanisms. METHODS: We performed a phenotypical and histologic analysis of the IBD affecting 17 patients with hemizygous mutations in XIAP, including 3 patients identified by screening 83 patients with pediatric-onset IBD. The X chromosome inactivation was analyzed in female carriers of heterozygous XIAP mutations, including 2 adults with IBD. The functional consequences of XIAP deficiency were analyzed. RESULTS: Clinical presentation and histology of IBD in patients with XIAP deficiency overlapped with those of patients with Crohn disease. The age at onset was variable (from 3 months to 41 years), and IBD was severe and difficult to treat. In 2 patients hematopoietic stem cell transplantation fully restored intestinal homeostasis. Monocytes of patients had impaired NOD2-mediated IL-8 and monocyte chemoattractant protein 1 (MCP-1) production, as well as IL-10, in response to NOD2 and Toll-like receptor 2/4 costimulation. Nucleotide binding and oligomerization domain containing 1 (NOD1)-mediated IL-6 and IL-8 production was defective in fibroblasts from XIAP-deficient patients. The 2 heterozygous female carriers of XIAP mutations with IBD displayed abnormal expression of the XIAP mutated allele, resulting in impaired activation of the NOD2 pathway. CONCLUSION: IBD in patients with XIAP deficiency is similar to Crohn disease and is associated with defective NOD2 function in monocytes. Importantly, we report that it is not restricted to male patients because we identified 2 symptomatic female heterozygous carriers of XIAP mutations.


Subject(s)
Chromosomes, Human, X , Crohn Disease , Genetic Diseases, X-Linked , Hemizygote , Heterozygote , Lymphoproliferative Disorders , X-Linked Inhibitor of Apoptosis Protein/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Chromosomes, Human, X/genetics , Chromosomes, Human, X/metabolism , Cohort Studies , Crohn Disease/blood , Crohn Disease/genetics , Crohn Disease/pathology , Cytokines/blood , Cytokines/genetics , Female , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Humans , Infant , Lymphoproliferative Disorders/blood , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Male , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism
10.
Inflamm Bowel Dis ; 19(13): 2820-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24216686

ABSTRACT

OBJECTIVE: Early-onset inflammatory bowel disease starting within the first months of life could be due to a particular genetic defect. We set up the GENetically determined ImmUne-mediated enteropathieS (GENIUS) network and collected infants with a proven defect of the IL10 axis for accurate phenotyping of disease presentation and evolution. DESIGN: Ten patients with early-onset inflammatory bowel disease with confirmed mutations in IL10, IL10RA, or IL10RB genes were characterized on clinical, endoscopic-histological, immunobiological, and radiological findings. Functional assays to confirm defective responses to IL10 were performed on peripheral blood mononuclear cells. RESULTS: A functional defect in IL10 signaling was confirmed in all IL10R patients tested. Disease started with severe diarrhea within the first 12 weeks in all patients. All infants showed Crohn's disease-like ulcerations limited to the colon with marked perianal inflammation (fissures, abscess, and fistula); disease progression to the small bowel occurred in only 1 patient. Four of the 10 patients had granulomata on histology, and all patients showed Crohn's disease-like mesenteric infiltration on imaging. Disease pattern was indistinguishable between IL10R alpha or beta chain or IL10 defects; autoimmunity was not observed. Mutations in IL10 were more frequently associated with bacterial and viral infections. Patients responded partially to treatment with steroids or anti-tumor necrosis factor drugs, whereas hematopoietic stem cell transplantation proved efficacious. CONCLUSION: The importance of the IL10 pathway within the colonic mucosa is highlighted by the development of severe colitis within a few weeks in infants with mutations in IL10, IL10RA, or IL10RB. Immunosuppression failed to correct the defect in this pathway, which seems to be a key to controlling inflammation in the colon.


Subject(s)
Colitis, Ulcerative/genetics , Crohn Disease/genetics , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10/genetics , Mutation/genetics , Receptors, Interleukin/genetics , Adolescent , Age of Onset , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Male , Phenotype , Prognosis
11.
Immunity ; 37(1): 108-21, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22705105

ABSTRACT

With the goal in mind to define how interleukin-15 (IL-15) contributes to acute intestinal inflammation, we have used a mouse model of ileitis induced by oral infection with Toxoplasma gondii. We observed that a crosstalk between IL-15 and interleukin-18 (IL-18) promoted intestinal recruitment of inflammatory monocytes, where these cells participated in parasite control but also in tissue damage. A stromal source of IL-15 controlled the development of lamina propria NKp46(+)NK1.1(+) cells, whereas IL-18 produced during T. gondii infection stimulated their production of the chemokine CCL3. In turn, CCL3 attracted inflammatory monocytes via their chemokine receptor CCR1, which was indispensable for their recruitment into the inflamed gut. Collectively, these results identify the IL-15-dependent subset of intestinal NKp46(+) cells as an important source of CCL3, which can amplify intestinal inflammation via the recruitment of CCR1(+) inflammatory monocytes. Preliminary evidence suggests that this pathway might operate in Crohn's disease.


Subject(s)
Antigens, Ly/metabolism , Enteritis/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Monocytes/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Adolescent , Animals , Chemokine CCL3/metabolism , Child , Crohn Disease/immunology , Crohn Disease/metabolism , Enteritis/metabolism , Enteritis/parasitology , Humans , Interleukin-15/genetics , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, CCR1/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Toxoplasma/immunology , Toxoplasma/metabolism
12.
Am J Gastroenterol ; 106(8): 1544-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21519361

ABSTRACT

OBJECTIVES: Early onset inflammatory bowel diseases (EO-IBD) developing during the first year of life are likely to reflect inherited defects in key mechanism(s) controlling intestinal homeostasis, as recently suggested for interleukin 10 (IL10). Thus, we aimed to further elaborate the hypothesis of defective anti-inflammatory responses in patients with IBD. METHODS: The capacities of transforming growth factor ß (TGFß) and IL10 to inhibit proinflammatory cytokine production by monocyte-derived dendritic cells (MoDC) or peripheral blood cells (PBMC) was analyzed in 75 children with IBD, including 13 infants with EO-IBD (in whom autoimmune diseases or classical immunodeficiencies were ruled out). IL10 receptor-A/-B expression, STAT3 activation in response to IL6, IL10, IL21, IL22 were analyzed by FACS and western blotting. IL10RA and B genes were sequenced. The response to IL22 was tested in ileal/colonic tissue cultures. Tissue gene expression was analyzed by Taqman real-time polymerase chain reaction. RESULTS: Production of IL10 in response to bacterial motifs was normal in all IBD patients. In contrast to our original hypothesis, no defect of the anti-inflammatory potential of TGFß and IL10 was observed in children with IBD or EO-IBD except two infants who presented with granuloma-positive colitis at 3 months of life: no response to IL10 was observed secondary to mutations in the α (p.R262C) or ß (p.E141X) chain of IL10R, respectively, although a fully functional Jak-STAT3 pathway was present in both patients. When analyzing the regulation of intestinal bacterial clearance, we detected a defect in the patient with absent IL10 RB to upregulate protective transcripts in response to IL22, whereas all other EO-IBD patients, including the patient with an abnormal α chain, responded normally. CONCLUSIONS: Impaired IL10 signaling characterizes a subgroup of IBD patients, whereas the majority of children with severe IBD including EO forms normally produces and responds to IL10. Defective IL22 signaling may additionally impair intestinal epithelial clearance. Our data point out the complexity of IBD, which represent a group of distinct diseases with several pathogenetic abnormalities.


Subject(s)
Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Blotting, Western , Crohn Disease/genetics , Crohn Disease/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Humans , Infant , Interleukin-10 Receptor beta Subunit/metabolism , Interleukins/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mucin-1/metabolism , Mucins/metabolism , Peptide Fragments/metabolism , Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , beta-Defensins/metabolism , Interleukin-22
13.
Gastroenterology ; 139(3): 770-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20537998

ABSTRACT

BACKGROUND & AIMS: Little is known about the pathophysiology of early onset forms of autoimmune enteropathy (AIE). AIE has been associated with mutations in FOXP3-a transcription factor that controls regulatory T-cell development and function. We analyzed the molecular basis of neonatal or early postnatal AIE using clinical, genetic, and functional immunological studies. METHODS: Gastroenterological and immunological features were analyzed in 9 boys and 2 girls with AIE that began within the first 5 months of life. FOXP3 and IL2RA were genotyped in peripheral blood monocytes. FOXP3 messenger RNA and protein expression were analyzed using reverse-transcription polymerase chain reaction, flow cytometry, and confocal immunofluorescence of CD4(+) T cells. Regulatory T-cell function (CD4(+)CD25(+)) was assayed in coculture systems. RESULTS: AIE associated with extraintestinal autoimmunity was severe and life-threatening; all patients required total parenteral nutrition. Regulatory T cells from 7 patients had altered function and FOXP3 mutations that resulted in lost or reduced FOXP3 protein expression; 2 infants had reduced regulatory T-cell activity and reduced levels of FOXP3 protein, although we did not detect mutations in FOXP3 coding region, poly-A site, or promoter region (called FOXP3-dependent AIE). Two patients had a normal number of regulatory T cells that expressed normal levels of FOXP3 protein and normal regulatory activity in in vitro coculture assays (called FOXP3-independent AIE). No mutations in IL2RA were found. CONCLUSIONS: Most cases of AIE are associated with alterations in regulatory T-cell function; some, but not all, cases have mutations that affect FOXP3 expression levels. Further studies are needed to identify mechanisms of AIE pathogenesis.


Subject(s)
Autoimmune Diseases/immunology , Forkhead Transcription Factors/blood , Intestinal Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Age of Onset , Autoimmune Diseases/genetics , Autoimmune Diseases/mortality , Autoimmune Diseases/therapy , CD4 Lymphocyte Count , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Coculture Techniques , Down-Regulation , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Humans , Immunosuppressive Agents/therapeutic use , Infant , Infant, Newborn , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2 Receptor alpha Subunit/genetics , Intestinal Diseases/genetics , Intestinal Diseases/mortality , Intestinal Diseases/therapy , Male , Microscopy, Confocal , Mutation , Open Reading Frames , Parenteral Nutrition, Total , Promoter Regions, Genetic , RNA, Messenger/blood , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Treatment Outcome
14.
Gastroenterology ; 130(7): 1962-74, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16762619

ABSTRACT

BACKGROUND & AIMS: Few data exist on the molecular events causing intestinal epithelial destruction during inflammatory processes, such as inflammatory bowel disease (IBD). In this work, we analyzed the potential implication of tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) in these inflammatory lesions. METHODS: TRAIL and TRAIL-receptor expression were analyzed in normal, inflammatory ileum/colon and human intestinal epithelial cell (IEC) lines (HIEC), Caco-2, and HT-29 using RNase protection assay, real-time and reverse-transcription polymerase chain reaction (RT-PCR), immunohistochemistry, and Western blot analysis. TRAIL-induced activation of NF-kappaB was determined by electrophoretic mobility shift assay. Caspase-recruitment domain (CARD)15 expression and interleukin-(IL)8 production were studied by RT-PCR and enzyme-linked immunosorbent assay. Apoptosis was monitored using Annexin-V/caspase-3 assays. RESULTS: Normal mature IEC expressed low TRAIL levels, whereas, in inflammatory lesions, TRAIL messenger RNA and protein were markedly up-regulated in IEC and lamina propria lymphocytes at levels comparable with trinitrobenzene sulfonic acid-induced colitis. Interferon-gamma and TNF-alpha potently induced TRAIL in IEC. In vitro analyses revealed a dual biologic effect of TRAIL on HIEC: Under noninflammatory conditions, TRAIL up-regulated via nuclear factor-kappaB CARD15 and IL-8, whereas, under inflammatory conditions, TRAIL became a potent inducer of apoptosis in HIEC, which was confirmed ex vivo using ileal organ cultures. TNF-alpha markedly increased the expression of the proapoptotic receptor TRAIL-R2. TRAIL-induced IEC apoptosis required a functional caspase cascade. CONCLUSIONS: TRAIL is a new inflammatory mediator implicated in the homeostasis of intestinal epithelial barrier functions. TRAIL is highly up-regulated in IEC in inflammatory ileum and colon. It may augment in an auto-/paracrine fashion the elimination of IEC via apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Epithelial Cells/cytology , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Blotting, Western , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Female , Flow Cytometry , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sampling Studies , Sensitivity and Specificity , Statistics, Nonparametric , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism
15.
J Biol Chem ; 279(41): 42984-92, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15302888

ABSTRACT

The capacity of non-pathogenic enteric bacteria to induce a pro-inflammatory response is under debate in terms of its effect on the symbiosis between the mammalian host and its commensal gut microflora. Activation of NF-kappaB and induction of interleukin-8 (IL-8) and CCL-20 by the commensal Escherichia coli strain MG1655 were first studied in vitro in the human intestinal epithelial cell (IECs) lines HT29-19A and Caco-2, transfected or not with plasmids encoding dominant negative Toll-like receptor (TLR) 5 and myeloid differentiation factor-88 (MyD88) adaptor protein. The response of enterocytes in situ was then assessed using murine ileal biopsies mounted in Ussing chambers. Commensal E. coli induced NF-kappaB DNA binding, NF-kappaB transcriptional activity, CCL-20 expression, and IL-8 secretion in the human IEC lines. E. coli MG1655 flagellin was necessary and sufficient to trigger this pro-inflammatory pathway via its interaction with TLR5 and the subsequent recruitment of the adaptor protein MyD88. Following epithelial cell polarization, signaling could be induced by live E. coli and flagellin on the apical side of HT29-19A. The in vivo relevance of our findings was confirmed, because immunohistochemical staining of murine ileum demonstrated expression of TLR5 in the apical part of enterocytes in situ. Furthermore, flagellin added on the mucosal side of murine ileal biopsies mounted in Ussing chambers induced a basolateral production of KC, a functional murine homolog of human IL-8. These findings provide strong evidence that flagellin released by flagellated commensal bacteria in the intestinal lumen can induce a pro-inflammatory response in enterocytes in vivo.


Subject(s)
Escherichia coli/metabolism , Intestinal Mucosa/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/metabolism , Biopsy , Caco-2 Cells , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Chemokine CCL20 , Chemokines, CC/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Enterocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/cytology , Flagellin/chemistry , Flagellin/metabolism , Humans , Ileum/pathology , Inflammation , Interleukin-8/metabolism , Lipopolysaccharides/metabolism , Macrophage Inflammatory Proteins/metabolism , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred C3H , Mutation , Myeloid Differentiation Factor 88 , NF-kappa B/metabolism , Phenotype , Plasmids/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/chemistry , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Toll-Like Receptor 5 , Toll-Like Receptors , Transcription, Genetic , Transfection
16.
Gastroenterology ; 125(3): 730-45, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949719

ABSTRACT

BACKGROUND & AIMS: The mechanism of intraepithelial lymphocyte hyperplasia, a hallmark of celiac disease, is unknown. We have investigated the role of epithelium-derived interleukin (IL)-15 in the alterations of epithelial homeostasis in refractory celiac sprue, a privileged situation to study the first step of lymphoid transformation and the contribution of intraepithelial lymphocytes to villous atrophy in celiac disease. METHODS: IL-15 expression was assessed in biopsy specimens and isolated enterocytes by combining immunohistochemistry, flow cytometry, and real-time quantitative polymerase chain reaction. The ability of IL-15 to induce growth and survival of clonal intraepithelial lymphocytes lacking surface CD3 and to induce their cytotoxicity and secretion of interferon gamma was tested using soluble IL-15 and coculture in the presence of epithelial cell lines expressing membrane IL-15. RESULTS: IL-15 was massively overexpressed not only in lamina propria but also in the intestinal epithelium of patients with active celiac disease and refractory celiac sprue. IL-15 was not secreted but delivered at the surface of enterocytes. IL-15 specifically induced the expansion and survival of the clonal abnormal intraepithelial lymphocytes that characterize refractory celiac sprue and triggered their secretion of interferon gamma and their cytotoxicity against intestinal epithelial cells. Comparable activating signals could be delivered by IL-15 expressed at the membrane of the T84 enterocyte cell line. CONCLUSIONS: These data provide strong evidence that uncontrolled overexpression of IL-15 in refractory celiac sprue perpetuates epithelial damage and promotes the emergence of T-cell clonal proliferations. Blocking IL-15 might prove useful to treat this severe complication of celiac disease.


Subject(s)
Celiac Disease/immunology , Interleukin-15/physiology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Lymphoma/etiology , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Granzymes , Homeostasis , Humans , Integrin alpha Chains/analysis , Interferon-gamma/genetics , Middle Aged , Serine Endopeptidases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...