28 results on '"GASTROINTESTINAL IMMUNE RESPONSE"'
Search Results
2. TiO₂ nanoparticles abrogate the protective effect of the Crohn’s disease-associated variation within the PTPN22 gene locus
- Author
-
Schwarzfischer, Marlene, Niechcia, Anna, Handler, Kristina, Morsy, Yasser, Wawrzyniak, Marcin, Laimbacher, Andrea S, Atrott, Kirstin, Manzini, Roberto, Baebler, Katharina, Hering, Larissa, Katkeviciutė, Egle, Häfliger, Janine, Lang, Silvia, Keller, Maja E, Woodtli, Jérôme, Eisenbeiss, Lisa, Kraemer, Thomas, Schraner, Elisabeth M, Wiesendanger, Mahesa, Zeissig, Sebastian, Rogler, Gerhard, Moor, Andreas E, Scharl, Michael, Spalinger, Marianne R, and University of Zurich
- Subjects
gut immunology ,10219 Clinic for Gastroenterology and Hepatology ,10077 Institute of Veterinary Anatomy ,gastrointestinal immune response ,IBD basic research ,inflammatory bowel disease ,Gastroenterology ,340 Law ,610 Medicine & health ,10218 Institute of Legal Medicine ,10244 Institute of Virology - Published
- 2023
3. When Eating Becomes a Pain in the Gut.
- Author
-
Adair, Taylor and Belz, Gabrielle T.
- Subjects
- *
VISCERAL pain , *IRRITABLE colon , *INTESTINES , *INGESTION , *INFLAMMATION , *IMMUNOLOGICAL tolerance - Abstract
Irritable bowel syndrome often appears following gastrointestinal infection and is marked by diarrhea, dysbiosis, fever, and intestinal pain following eating. A recent study by Aguilera-Lizarraga et al. now demonstrates that a breakdown in intestinal immunotolerance sparks an inflammatory response to typically tolerated food antigens and causes visceral pain. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
4. CARD9 in neutrophils protects from colitis and controls mitochondrial metabolism and cell survival
- Author
-
Camille Danne, Chloé Michaudel, Jurate Skerniskyte, Julien Planchais, Aurélie Magniez, Allison Agus, Marie-Laure Michel, Bruno Lamas, Gregory Da-Costa, Madeleine Spatz, Cyriane Oeuvray, Chloé Galbert, Maxime Poirier, Yazhou Wang, Alexia Lapiere, Nathalie Rolhion, Tatiana Ledent, Cédric Pionneau, Solenne Chardonnet, Floriant Bellvert, Edern Cahoreau, Amandine Rocher, Rafael Jose Argüello, Carole Peyssonnaux, Sabine Louis, Mathias L. Richard, Philippe Langella, Jamel El-Benna, Benoit Marteyn, Harry Sokol, MICrobiologie de l'ALImentation au Service de la Santé (MICALIS), AgroParisTech-Université Paris-Saclay-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre de Recherche Saint-Antoine (CRSA), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Paris Center for Microbiome Medicine (FHU PaCeMM), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Architecture et Réactivité de l'ARN (ARN), Institut de biologie moléculaire et cellulaire (IBMC), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS), Plateforme Post-génomique de la Pitié-Salpêtrière (PASS-P3S), Unité Mixte de Service Production et Analyse de données en Sciences de la vie et en Santé (PASS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), MetaboHUB-MetaToul, Génopole Toulouse Midi-Pyrénées [Auzeville] (GENOTOUL), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Ecole Nationale Vétérinaire de Toulouse (ENVT), Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), MetaToul FluxoMet (TBI-MetaToul), Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Génopole Toulouse Midi-Pyrénées [Auzeville] (GENOTOUL), Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Toulouse Biotechnology Institute (TBI), Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université Fédérale Toulouse Midi-Pyrénées-Institut National des Sciences Appliquées (INSA)-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université Fédérale Toulouse Midi-Pyrénées-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire d'Excellence : Biogenèse et pathologies du globule rouge (Labex Gr-Ex), Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Centre de recherche sur l'Inflammation (CRI (UMR_S_1149 / ERL_8252 / U1149)), Institut d’Etudes Avancées de l’Université de Strasbourg - Institute for Advanced Study (USIAS), Université de Strasbourg (UNISTRA), Pathogénèse des Infections vasculaires / Pathogenesis of Vascular Infections, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Funding was provided by the ANR-17-CE15-0019-01 grant and a Marie Skłodowska-Curie Actions fellowship., ANR-17-CE15-0019,MARYLAND,Modulation par Card9 du microbiote intestinal et de sa production d'agonistes Aryl hydrocarbon receptor: mécanisme, potentiel thérapeutique et comme biomarqueur dans les maladies inflammatoires chroniques de l'intestin(2017), LAMAS, Bruno, Modulation par Card9 du microbiote intestinal et de sa production d'agonistes Aryl hydrocarbon receptor: mécanisme, potentiel thérapeutique et comme biomarqueur dans les maladies inflammatoires chroniques de l'intestin - - MARYLAND2017 - ANR-17-CE15-0019 - AAPG2017 - VALID, MetaboHUB-Génopole Toulouse Midi-Pyrénées [Auzeville] (GENOTOUL), Université de Toulouse (UT)-Université de Toulouse (UT)-Ecole Nationale Vétérinaire de Toulouse (ENVT), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-MetaboHUB-Génopole Toulouse Midi-Pyrénées [Auzeville] (GENOTOUL), Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Toulouse Biotechnology Institute (TBI), Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Institut National de la Recherche Agronomique (INRA)-AgroParisTech, MetaToul-MetaboHUB, Institut Cochin, INSERM, CNRS, Université de Paris, Laboratoire d'excellence GR-Ex, Institut Cochin, INSERM, CNRS, UNiversité de Paris, Laboratoire d'excellence GR-Ex, Institut Pasteur, Université de Paris, Inserm 1225 Unité de Pathogenèse des infections Vasculaires, Métabolomique et Fluxomique (MetaToul) (TBI-MetaToul), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Faculte de Medecine Xavier Bichat, Université Paris Diderot - Paris 7 (UPD7), and Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)
- Subjects
[SDV.IMM] Life Sciences [q-bio]/Immunology ,IBD ,Gastroenterology ,Apoptosis ,[SDV.TOX.TCA]Life Sciences [q-bio]/Toxicology/Toxicology and food chain ,mitochondrial activity ,Gastrointestinal immune response ,[SDV.TOX] Life Sciences [q-bio]/Toxicology ,[SDV.AEN] Life Sciences [q-bio]/Food and Nutrition ,neutrophils ,[SDV.TOX.TCA] Life Sciences [q-bio]/Toxicology/Toxicology and food chain ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,IBD-Genetics ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,gut inflammation ,CARD9 ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition - Abstract
ObjectivesInflammatory bowel disease (IBD) results from a combination of genetic predisposition, dysbiosis of the gut microbiota and environmental factors, leading to alterations in the gastrointestinal immune response and chronic inflammation. Caspase recruitment domain 9 (Card9), one of the IBD susceptibility genes, has been shown to protect against intestinal inflammation and fungal infection. However, the cell types and mechanisms involved in the CARD9 protective role against inflammation remain unknown.DesignWe used dextran sulfate sodium (DSS)-induced and adoptive transfer colitis models in total and conditional CARD9 knock-out mice to uncover which cell types play a role in the CARD9 protective phenotype. The impact of Card9 deletion on neutrophil function was assessed by an in vivo model of fungal infection and various functional assays, including endpoint dilution assay, apoptosis assay by flow cytometry, proteomics and real time bioenergetic profile analysis (Seahorse).ResultsLymphocytes are not intrinsically involved in the CARD9 protective role against colitis. CARD9 expression in neutrophils, but not in epithelial or CD11c+ cells, protects against DSS-induced colitis. In the absence of CARD9, mitochondrial dysfunction in neutrophils leads to their premature death through apoptosis, especially in oxidative environment. The decrease of fonctional neutrophils in tissues could explain the impaired containment of fungi and increased susceptibility to intestinal inflammation.ConclusionThese results provide new insight into the role of CARD9 in neutrophil mitochondrial function and its involvement in intestinal inflammation, paving the way for new therapeutic strategies targeting neutrophils.Summary boxWhat is already known about this subject?Inflammatory bowel disease (IBD) results from genetic predisposition, microbiota dysbiosis and environmental factors, but the alterations of the immune response leading to chronic intestinal inflammation are still not fully understood.Caspase recruitment domain 9 (Card9), one of the IBD susceptibility genes, has been shown to protect against intestinal inflammation and fungal infection.However, the cell types and cellular mechanisms involved in the CARD9 protective role against inflammation remain unknown.What are the new findings?CARD9 expression in neutrophils, but not in lymphocytes, epithelial cells or CD11c+ cells, protects against DSS-induced colitis.In the absence of CARD9, mitochondrial dysfunction in neutrophils leads to their premature death through apoptosis, especially in oxidative environment.The decrease of fonctional neutrophils in tissues could explain the impaired containment of fungi and increased susceptibility to intestinal inflammation.How might it impact on clinical practice in the foreseeable future?These results provide new insight into the role of CARD9 in neutrophil mitochondrial function and its involvement in intestinal inflammation.Understanding the role of neutrophils in chronic inflammation could lead to innovative therapeutic strategies targeting these key immune cells for various complex diseases.
- Published
- 2022
- Full Text
- View/download PDF
5. Disentangling tryptophan metabolism in inflammatory bowel diseases.
- Author
-
Becker C and Adolph TE
- Subjects
- Humans, Colon metabolism, Intestinal Mucosa metabolism, Tryptophan metabolism, Inflammatory Bowel Diseases metabolism
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2023
- Full Text
- View/download PDF
6. CARD9 in neutrophils protects from colitis and controls mitochondrial metabolism and cell survival.
- Author
-
Danne C, Michaudel C, Skerniskyte J, Planchais J, Magniez A, Agus A, Michel ML, Lamas B, Da Costa G, Spatz M, Oeuvray C, Galbert C, Poirier M, Wang Y, Lapière A, Rolhion N, Ledent T, Pionneau C, Chardonnet S, Bellvert F, Cahoreau E, Rocher A, Arguello RR, Peyssonnaux C, Louis S, Richard ML, Langella P, El-Benna J, Marteyn B, and Sokol H
- Subjects
- Mice, Animals, Neutrophils metabolism, Cell Survival, Inflammation metabolism, Mice, Knockout, Mitochondria metabolism, Dextran Sulfate toxicity, Disease Models, Animal, Mice, Inbred C57BL, CARD Signaling Adaptor Proteins metabolism, Colitis chemically induced, Colitis prevention & control, Inflammatory Bowel Diseases
- Abstract
Objectives: Inflammatory bowel disease (IBD) results from a combination of genetic predisposition, dysbiosis of the gut microbiota and environmental factors, leading to alterations in the gastrointestinal immune response and chronic inflammation. Caspase recruitment domain 9 ( Card9 ), one of the IBD susceptibility genes, has been shown to protect against intestinal inflammation and fungal infection. However, the cell types and mechanisms involved in the CARD9 protective role against inflammation remain unknown., Design: We used dextran sulfate sodium (DSS)-induced and adoptive transfer colitis models in total and conditional CARD9 knock-out mice to uncover which cell types play a role in the CARD9 protective phenotype. The impact of Card9 deletion on neutrophil function was assessed by an in vivo model of fungal infection and various functional assays, including endpoint dilution assay, apoptosis assay by flow cytometry, proteomics and real-time bioenergetic profile analysis (Seahorse)., Results: Lymphocytes are not intrinsically involved in the CARD9 protective role against colitis. CARD9 expression in neutrophils, but not in epithelial or CD11c+cells, protects against DSS-induced colitis. In the absence of CARD9, mitochondrial dysfunction increases mitochondrial reactive oxygen species production leading to the premature death of neutrophilsthrough apoptosis, especially in oxidative environment. The decreased functional neutrophils in tissues might explain the impaired containment of fungi and increased susceptibility to intestinal inflammation., Conclusion: These results provide new insight into the role of CARD9 in neutrophil mitochondrial function and its involvement in intestinal inflammation, paving the way for new therapeutic strategies targeting neutrophils., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
7. TiO 2 nanoparticles abrogate the protective effect of the Crohn's disease-associated variation within the PTPN22 gene locus.
- Author
-
Schwarzfischer M, Niechcial A, Handler K, Morsy Y, Wawrzyniak M, Laimbacher AS, Atrott K, Manzini R, Baebler K, Hering L, Katkeviciutė E, Häfliger J, Lang S, Keller ME, Woodtli J, Eisenbeiss L, Kraemer T, Schraner EM, Wiesendanger M, Zeissig S, Rogler G, Moor AE, Scharl M, and Spalinger MR
- Subjects
- Mice, Animals, CD8-Positive T-Lymphocytes metabolism, Inflammation complications, Dextran Sulfate, Disease Models, Animal, Mice, Inbred C57BL, Protein Tyrosine Phosphatase, Non-Receptor Type 22 genetics, Crohn Disease genetics, Crohn Disease complications, Colitis chemically induced, Colitis genetics, Colitis prevention & control, Inflammatory Bowel Diseases, Nanoparticles
- Abstract
Objective: Inflammatory bowel disease (IBD) is a multifactorial condition driven by genetic and environmental risk factors. A genetic variation in the protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene has been associated with autoimmune disorders while protecting from the IBD subtype Crohn's disease. Mice expressing the murine orthologous PTPN22-R619W variant are protected from intestinal inflammation in the model of acute dextran sodium sulfate (DSS)-induced colitis. We previously identified food-grade titanium dioxide (TiO
2 , E171) as a neglected IBD risk factor. Here, we investigate the interplay of the PTPN22 variant and TiO2 -mediated effects during IBD pathogenesis., Design: Acute DSS colitis was induced in wild-type and PTPN22 variant mice (PTPN22-R619W) and animals were treated with TiO2 nanoparticles during colitis induction. Disease-triggering mechanisms were investigated using bulk and single-cell RNA sequencing., Results: In mice, administration of TiO2 nanoparticles abrogated the protective effect of the variant, rendering PTPN22-R619W mice susceptible to DSS colitis. In early disease, cytotoxic CD8+ T-cells were found to be reduced in the lamina propria of PTPN22-R619W mice, an effect reversed by TiO2 administration. Normalisation of T-cell populations correlated with increased Ifng expression and, at a later stage of disease, the promoted prevalence of proinflammatory macrophages that triggered severe intestinal inflammation., Conclusion: Our findings indicate that the consumption of TiO2 nanoparticles might have adverse effects on the gastrointestinal health of individuals carrying the PTPN22 variant. This demonstrates that environmental factors interact with genetic risk variants and can reverse a protective mechanism into a disease-promoting effect., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
8. Bioinformatic and experimental analysis reveals lack of support for measles mimicry in Crohn's disease.
- Author
-
Polymeros, Dimitrios, Tsiamoulos, Zacharias, Koutsoumpas, Andreas, Smyk, Daniel, Mytilinaiou, Maria, Triantafyllou, Konstantinos, Bogdanos, Dimitrios, and Ladas, Spiros
- Subjects
- *
CROHN'S disease , *MEASLES , *MOLECULAR mimicry , *AMINO acid sequence , *PEPTIDES , *AUTOIMMUNITY , *IMMUNE response - Abstract
Background A link between measles virus and Crohn's disease (CD) has been postulated. We assessed through bioinformatic and immunological approaches whether measles is implicated in CD induction, through molecular mimicry. Methods The BLAST2p program was used to identify amino acid sequence similarities between five measles virus and 56 intestinal proteins. Antibody responses to measles/human mimics were tested by an in-house ELISA using serum samples from 50 patients with CD, 50 with ulcerative colitis (UC), and 38 matched healthy controls (HCs). Results We identified 15 sets of significant (>70%) local amino acid homologies from two measles antigens, hemagglutinin-neuraminidase and fusion-glycoprotein, and ten human intestinal proteins. Reactivity to at least one measles 15-meric mimicking peptide was present in 27 out of 50 (54%) of patients with CD, 24 out of 50 (48%) with UC (CD versus UC, p = 0.68), and 13 out of 38 (34.2%) HCs (CD versus HC, p = 0.08). Double reactivity to at least one measles/human pair was present in four out of 50 (8%) patients with CD, three out of 50 (6%) with UC (p = 0.99), and in three out of 38 (7.9%) HCs (p >0.05 for all). Titration experiments yielded different extinction curves for anti-measles and anti-human intestinal double-reactive antibodies. Epitope prediction algorithms and three-dimensional modeling provided bioinformatic confirmation for the observed antigenicity of the main measles virus epitopic regions. Conclusions Measles sequences mimicking intestinal proteins are frequent targets of antibody responses in patients with CD, but this reactivity lacks disease specificity and does not initiate cross- reactive responses to intestinal mimics. We conclude that there is no involvement of measles/human molecular mimicry in the etiopathogenesis of CD. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
9. Location-specific cell identity rather than exposure to GI microbiota defines many innate immune signalling cascades in the gut epithelium
- Author
-
Nina Wallaschek, Malvika Pompaiah, Sina Bartfeld, Carolin Niklas, Isabella Pierotti, Özge Kayisoglu, Franziska Weiss, Armin Wiegering, and Christoph-Thomas Germer
- Subjects
Lipopolysaccharides ,Lipopolysaccharide ,Gut flora ,chemistry.chemical_compound ,Organoid ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Gut Microbiota ,Cells, Cultured ,Innate immune system ,biology ,Gastroenterology ,Epithelial Cells ,biology.organism_classification ,Epithelium ,Immunity, Innate ,Cell biology ,Gut Epithelium ,Gastrointestinal Microbiome ,Organoids ,medicine.anatomical_structure ,chemistry ,gastrointestinal immune response ,TLR4 ,mucosal immunity ,epithelial barrier ,Stem cell ,Signal Transduction - Abstract
ObjectiveThe epithelial layer of the GI tract is equipped with innate immune receptors to sense invading pathogens. Dysregulation in innate immune signalling pathways is associated with severe inflammatory diseases, but the responsiveness of GI epithelial cells to bacterial stimulation remains unclear.DesignWe generated 42 lines of human and murine organoids from gastric and intestinal segments of both adult and fetal tissues. Genome-wide RNA-seq of the organoids provides an expression atlas of the GI epithelium. The innate immune response in epithelial cells was assessed using several functional assays in organoids and two-dimensional monolayers of cells from organoids.ResultsResults demonstrate extensive spatial organisation of innate immune signalling components along the cephalocaudal axis. A large part of this organisation is determined before birth and independent of exposure to commensal gut microbiota. Spatially restricted expression of Toll-like receptor 4 (Tlr4) in stomach and colon, but not in small intestine, is matched by nuclear factor kappa B (NF-κB) responses to lipopolysaccharide (LPS) exposure. Gastric epithelial organoids can sense LPS from the basal as well as from the apical side.ConclusionWe conclude that the epithelial innate immune barrier follows a specific pattern per GI segment. The majority of the expression patterns and the function of TLR4 is encoded in the tissue-resident stem cells and determined primarily during development.
- Published
- 2019
10. Host transcriptome signatures in human faecal-washes predict histological remission in patients with IBD.
- Author
-
Ungar B, Yavzori M, Fudim E, Picard O, Kopylov U, Eliakim R, Shouval D, Levin Y, Savidor A, Ben-Moshe S, Manco R, Dan S, Egozi A, Bahar Halpern K, Mayer C, Barshack I, Ben-Horin S, and Itzkovitz S
- Abstract
Background: Colonoscopy is the gold standard for evaluation of inflammation in inflammatory bowel diseases (IBDs), yet entails cumbersome preparations and risks of injury. Existing non-invasive prognostic tools are limited in their diagnostic power. Moreover, transcriptomics of colonic biopsies have been inconclusive in their association with clinical features., Aims: To assess the utility of host transcriptomics of faecal wash samples of patients with IBD compared with controls., Methods: In this prospective cohort study, we obtained biopsies and faecal-wash samples from patients with IBD and controls undergoing lower endoscopy. We performed RNAseq of biopsies and matching faecal-washes, and associated them with endoscopic and histological inflammation status. We also performed faecal mass-spectrometry proteomics on a subset of samples. We inferred cell compositions using computational deconvolution and used classification algorithms to identify informative genes., Results: We analysed biopsies and faecal washes from 39 patients (20 IBD, 19 controls). Host faecal-transcriptome carried information that was distinct from biopsy RNAseq and faecal proteomics. Transcriptomics of faecal washes, yet not of biopsies, from patients with histological inflammation were significantly correlated to one another (p=5.3×10
-12 ). Faecal-transcriptome had significantly higher statistical power in identifying histological inflammation compared with transctiptome of intestinal biopsies (150 genes with area under the curve >0.9 in faecal samples vs 10 genes in biopsy RNAseq). These results were replicated in a validation cohort of 22 patients (10 IBD, 12 controls). Faecal samples were enriched in inflammatory monocytes, regulatory T cells, natural killer-cells and innate lymphoid cells., Conclusions: Faecal wash host transcriptome is a statistically powerful biomarker reflecting histological inflammation. Furthermore, it opens the way to identifying important correlates and therapeutic targets that may be obscured using biopsy transcriptomics., Competing Interests: Competing interests: SB-H received consulting and advisoryboard fees and/or research support from AbbVie, MSD, Janssen, Takeda andCellTrion. UK received speaker and advisory fees from: Abbvie, Jannsen, Gilead, MSD, Medtronic, Takeda and research support from: Jannsen, Medtronic, Takeda. RE received consultant and speaker fees from Janssen, Abbvie, Takeda and Medtronic. BU received consultation fees from Neopharm, Takeda, Janssen and Abbvie. DS received research support from Takeda and consultation and lecturing fees from AbbVie., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2022
- Full Text
- View/download PDF
11. NTPDase8 protects mice from intestinal inflammation by limiting P2Y 6 receptor activation: identification of a new pathway of inflammation for the potential treatment of IBD.
- Author
-
Salem M, Lecka J, Pelletier J, Gomes Marconato D, Dumas A, Vallières L, Brochu G, Robaye B, Jobin C, and Sévigny J
- Subjects
- Adenosine Triphosphatases genetics, Animals, Apoptosis, Bone Marrow Transplantation, Colon metabolism, Cytokines metabolism, Dextran Sulfate pharmacology, Disease Models, Animal, Epithelial Cells metabolism, Humans, Immunohistochemistry, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Mice, Inbred C57BL, Mice, Knockout, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Thiourea pharmacology, Mice, Adenosine Triphosphatases metabolism, Colitis metabolism, Isothiocyanates pharmacology, Receptors, Purinergic P2 genetics, Receptors, Purinergic P2 metabolism, Thiourea analogs & derivatives
- Abstract
Objective: Nucleotides are danger signals that activate inflammatory responses via binding P2 receptors. The nucleoside triphosphate diphosphohydrolase-8 (NTPDase8) is an ectonucleotidase that hydrolyses P2 receptor ligands. We investigated the role of NTPDase8 in intestinal inflammation., Design: We generated NTPDase8-deficient ( Entpd8
-/- ) mice to define the role of NTPDase8 in the dextran sodium sulfate (DSS) colitis model. To assess inflammation, colons were collected and analysed by histopathology, reverse transcriptase-quantitative real-time PCR (RT-qPCR) and immunohistochemistry. P2 receptor expression was analysed by RT-qPCR on primary intestinal epithelium and NTPDase8 activity by histochemistry. The role of intestinal P2Y6 receptors was assessed by bone marrow transplantation experiments and with a P2Y6 receptor antagonist., Results: NTPDase8 is the dominant enzyme responsible for the hydrolysis of nucleotides in the lumen of the colon. Compared with wild-type (WT) control mice, the colon of Entpd8-/- mice treated with DSS displayed significantly more histological damage, immune cell infiltration, apoptosis and increased expression of several proinflammatory cytokines. P2Y6 was the dominant P2Y receptor expressed at the mRNA level by the colonic epithelia. Irradiated P2ry6-/- mice transplanted with WT bone marrow were fully protected from DSS-induced intestinal inflammation. In agreement, the daily intrarectal injection of a P2Y6 antagonist protected mice from DSS-induced intestinal inflammation in a dose-dependent manner. Finally, human intestinal epithelial cells express NTPDase8 and P2Y6 similarly as in mice., Conclusion: NTPDase8 protects the intestine from inflammation most probably by limiting the activation of P2Y6 receptors in colonic epithelial cells. This may provide a novel therapeutic strategy for the treatment of inflammatory bowel disease., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2022
- Full Text
- View/download PDF
12. Compartment-specific immunity in the human gut: properties and functions of dendritic cells in the colon versus the ileum
- Author
-
J. Landy, Alyssa Parian, Stella C. Knight, Nicholas R. English, Xuhang Li, Simon T. Peake, Gui Han Lee, Ailsa Hart, T Elliott, Henning Spranger, Elizabeth R. Mann, Mark Lazarev, Steven R. Brant, Hafid O. Al-Hassi, David Bernardo, and Ripple Man
- Subjects
0301 basic medicine ,Receptors, CCR7 ,Receptors, CCR4 ,Colon ,T-Lymphocytes ,GUT IMMUNOLOGY ,T cell ,Receptors, Cell Surface ,chemical and pharmacologic phenomena ,Ileum ,C-C chemokine receptor type 7 ,Biology ,T-Lymphocytes, Regulatory ,Immune tolerance ,Flow cytometry ,Molecular Imprinting ,Receptors, CCR ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, CD ,medicine ,Humans ,Receptors, Immunologic ,Membrane Glycoproteins ,medicine.diagnostic_test ,Gastroenterology ,Interleukin ,FOXP3 ,Dendritic Cells ,Flow Cytometry ,Molecular biology ,MUCOSAL IMMUNOLOGY ,Microscopy, Electron ,GASTROINTESTINAL IMMUNE RESPONSE ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Cytokines ,Lymphocyte Culture Test, Mixed ,Gut Immunity ,Integrin alpha Chains ,030215 immunology - Abstract
Objective Dendritic cells (DC) mediate intestinal immune tolerance. Despite striking differences between the colon and the ileum both in function and bacterial load, few studies distinguish between properties of immune cells in these compartments. Furthermore, information of gut DC in humans is scarce. We aimed to characterise human colonic versus ileal DC. Design Human DC from paired colonic and ileal samples were characterised by flow cytometry, electron microscopy or used to stimulate T cell responses in a mixed leucocyte reaction. Results A lower proportion of colonic DC produced pro-inflammatory cytokines (tumour necrosis factor-α and interleukin (IL)-1β) compared with their ileal counterparts and exhibited an enhanced ability to generate CD4 + FoxP3 + IL-10 + (regulatory) T cells . There were enhanced proportions of CD103 + Sirpα − DC in the colon, with increased proportions of CD103 + Sirpα + DC in the ileum. A greater proportion of colonic DC subsets analysed expressed the lymph-node-homing marker CCR7, alongside enhanced endocytic capacity, which was most striking in CD103 + Sirpα + DC. Expression of the inhibitory receptor ILT3 was enhanced on colonic DC. Interestingly, endocytic capacity was associated with CD103 + DC, in particular CD103 + Sirpα + DC. However, expression of ILT3 was associated with CD103 − DC. Colonic and ileal DC differentially expressed skin-homing marker CCR4 and small-bowel-homing marker CCR9, respectively, and this corresponded to their ability to imprint these homing markers on T cells. Conclusions The regulatory properties of colonic DC may represent an evolutionary adaptation to the greater bacterial load in the colon. The colon and the ileum should be regarded as separate entities, each comprising DC with distinct roles in mucosal immunity and imprinting.
- Published
- 2015
- Full Text
- View/download PDF
13. Ceramide-CD300f binding suppresses experimental colitis by inhibiting ATP-mediated mast cell activation
- Author
-
Jiro Kitaura, Akie Maehara, Mariko Takahashi, Ayako Kaitani, Yoshinori Yamanishi, Takanori Teshima, Masamichi Isobe, Ko Okumura, Kumi Izawa, Toshio Kitamura, and Toshihiro Matsukawa
- Subjects
0301 basic medicine ,Ceramide ,Ceramides ,Inflammatory bowel disease ,Flow cytometry ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Adenosine Triphosphate ,Extracellular ,medicine ,Animals ,Mast Cells ,Colitis ,Receptors, Immunologic ,Receptor ,Lamina propria ,biology ,medicine.diagnostic_test ,EXPERIMENTAL COLITIS ,Inflammatory Bowel Disease ,Gastroenterology ,medicine.disease ,Molecular biology ,IBD MODELS ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,GASTROINTESTINAL IMMUNE RESPONSE ,chemistry ,Immunology ,biology.protein ,Antibody - Abstract
Objective Extracellular ATP mediates mast cell-dependent intestinal inflammation via P2X7 purinoceptors. We have previously shown that CD300f (also called the leucocyte mono-immunoglobulin-like receptor 3 (LMIR3)) suppresses immunoglobulin E-dependent and mast cell-dependent allergic responses by binding to ceramide. The aim of the present study was to clarify the role of ceramide–LMIR3 interaction in the development of IBD. Design The dextran sodium sulfate (DSS)-induced colitis model was used in wild-type (WT), LMIR3 −/− , mast cell-deficient Kit W-sh/W-sh , Kit W-sh/W-sh LMIR3 −/− or Kit W-sh/W-sh mice engrafted with WT or LMIR3 −/− bone marrow-derived mast cells (BMMCs). The severity of colitis was determined by clinical and histological criteria. Lamina propria cell populations were assessed by flow cytometry. Production of chemical mediators from lamina propria cells was measured by real-time reverse transcription PCR. Production of chemical mediators from ATP-stimulated BMMCs in the presence or absence of ceramide was measured by ELISA. The severity of DSS-induced colitis was assessed in mice given either an Fc fusion protein containing an extracellular domain of LMIR3, and anticeramide antibody, or ceramide liposomes. Results LMIR3 deficiency exacerbated DSS-induced colitis in mice. Kit W-sh/W-sh mice harbouring LMIR3 −/− mast cells exhibited more severe colitis than those harbouring WT mast cells. Ceramide–LMIR3 interaction inhibited ATP-stimulated activation of BMMCs. DSS-induced colitis was aggravated by disrupting the ceramide–LMIR3 interaction, whereas it was suppressed by treating with ceramide liposomes. Conclusions LMIR3-deficient colonic mast cells were pivotal in the exacerbation of DSS-induced colitis in LMIR3 −/− mice. Ceramide liposomes attenuated DSS-induced colitis by inhibiting ATP-mediated activation of colonic mast cells through ceraimide–LMIR3 binding.
- Published
- 2015
14. Location-specific cell identity rather than exposure to GI microbiota defines many innate immune signalling cascades in the gut epithelium.
- Author
-
Kayisoglu O, Weiss F, Niklas C, Pierotti I, Pompaiah M, Wallaschek N, Germer CT, Wiegering A, and Bartfeld S
- Subjects
- Animals, Cells, Cultured, Humans, Lipopolysaccharides immunology, Signal Transduction, Epithelial Cells immunology, Gastrointestinal Microbiome immunology, Immunity, Innate physiology, Intestinal Mucosa cytology, Intestinal Mucosa immunology, Organoids immunology
- Abstract
Objective: The epithelial layer of the GI tract is equipped with innate immune receptors to sense invading pathogens. Dysregulation in innate immune signalling pathways is associated with severe inflammatory diseases, but the responsiveness of GI epithelial cells to bacterial stimulation remains unclear., Design: We generated 42 lines of human and murine organoids from gastric and intestinal segments of both adult and fetal tissues. Genome-wide RNA-seq of the organoids provides an expression atlas of the GI epithelium. The innate immune response in epithelial cells was assessed using several functional assays in organoids and two-dimensional monolayers of cells from organoids., Results: Results demonstrate extensive spatial organisation of innate immune signalling components along the cephalocaudal axis. A large part of this organisation is determined before birth and independent of exposure to commensal gut microbiota. Spatially restricted expression of Toll-like receptor 4 ( Tlr4 ) in stomach and colon, but not in small intestine, is matched by nuclear factor kappa B (NF-κB) responses to lipopolysaccharide (LPS) exposure. Gastric epithelial organoids can sense LPS from the basal as well as from the apical side., Conclusion: We conclude that the epithelial innate immune barrier follows a specific pattern per GI segment. The majority of the expression patterns and the function of TLR4 is encoded in the tissue-resident stem cells and determined primarily during development., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
15. The vagal innervation of the gut and immune homeostasis
- Author
-
Gianluca Matteoli and Guy E. Boeckxstaens
- Subjects
Neuroimmunomodulation ,Neuroimmunology ,Inflammation ,Biology ,Enteric Nervous System ,Immune system ,Ileus ,medicine ,Gut Immunology ,Homeostasis ,Humans ,Immunity, Mucosal ,Gastroenterology ,Recent Advances in Basic Science ,Vagus Nerve ,Inflammatory Bowel Diseases ,Acetylcholine ,Vagus nerve ,Intestines ,Nicotinic agonist ,Immunology ,Cholinergic ,Enteric nervous system ,medicine.symptom ,Neuroscience ,Gastrointestinal Immune Response ,medicine.drug ,Signal Transduction - Abstract
The central nervous system interacts dynamically with the immune system to modulate inflammation through humoral and neural pathways. Recently, in animal models of sepsis, the vagus nerve (VN) has been proposed to play a crucial role in the regulation of the immune response, also referred to as the cholinergic anti-inflammatory pathway. The VN, through release of acetylcholine, dampens immune cell activation by interacting with α-7 nicotinic acetylcholine receptors. Recent evidence suggests that the vagal innervation of the gastrointestinal tract also plays a major role controlling intestinal immune activation. Indeed, VN electrical stimulation potently reduces intestinal inflammation restoring intestinal homeostasis, whereas vagotomy has the reverse effect. In this review, we will discuss the current understanding concerning the mechanisms and effects involved in the cholinergic anti-inflammatory pathway in the gastrointestinal tract. Deeper investigation on this counter-regulatory neuroimmune mechanism will provide new insights in the cross-talk between the nervous and immune system leading to the identification of new therapeutic targets to treat intestinal immune disease.
- Published
- 2013
- Full Text
- View/download PDF
16. Gut symbiotic microbes imprint intestinal immune cells with the innate receptor SLAMF4 which contributes to gut immune protection against enteric pathogens
- Author
-
Amale Laouar, Young-Soon Jang, Allison Cabinian, Yasmina Laouar, May Tang, Bongkum Choi, and Daniel Sinsimer
- Subjects
0301 basic medicine ,Cell ,Gut flora ,Real-Time Polymerase Chain Reaction ,digestive system ,Microbiology ,03 medical and health sciences ,Mice ,Immune system ,Intestinal mucosa ,Signaling Lymphocytic Activation Molecule Family ,medicine ,Animals ,Germ-Free Life ,Humans ,ENTERIC INFECTIONS ,Intestinal Mucosa ,Gut Microbiota ,Receptor ,Symbiosis ,Immunity, Mucosal ,biology ,INTESTINAL BACTERIA ,Gastroenterology ,biology.organism_classification ,Flow Cytometry ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,030104 developmental biology ,Lymphatic system ,medicine.anatomical_structure ,GASTROINTESTINAL IMMUNE RESPONSE ,Immunology ,MUCOSAL IMMUNITY ,Bone marrow ,Signal transduction ,ANTIBIOTICS ,Signal Transduction - Abstract
BackgroundInteractions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field.AimWe have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is.MethodsExpression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtgbone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection withListeria monocytogenesandCytobacter rodentium.ResultsSLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC)TNLG8Aare key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death.ConclusionsSLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.
- Published
- 2016
17. A basal gradient of Wnt and stem-cell number influences regional tumour distribution in human and mouse intestinal tracts
- Author
-
Pedro Rodenas-Cuadrado, Manuel Rodriguez-Justo, Kimberley Howarth, Ian Tomlinson, Stefania Segditsas, James E. East, Simon J. Leedham, Satish Keshav, Annabelle Lewis, Sreelakshmi Mallappa, Simon Travis, Susan K. Clark, Rosemary Jeffery, Trevor A. Graham, and Hayley Davis
- Subjects
Pathology ,Cell Count ,colon carcinogenesis ,Mice ,0302 clinical medicine ,Intestinal mucosa ,mucosal defense ,colorectal cancer genes ,Homeostasis ,colonic adenomas ,Intestinal Mucosa ,Wnt Signaling Pathway ,Tissue homeostasis ,In Situ Hybridization ,beta Catenin ,Wnt signalling ,colorectal function ,0303 health sciences ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,cancer syndromes ,Stem Cells ,Gastroenterology ,Wnt signaling pathway ,colonic neoplasms ,LRP5 ,just-right hypothesis ,Cell biology ,Barrett's oesophagus ,Intestines ,Crohn's disease ,colonic polyps ,030220 oncology & carcinogenesis ,histopathology ,adenoma ,necrotising enterocolitis ,epithelial barrier ,Stem cell ,carcinogenesis ,Genetic Markers ,medicine.medical_specialty ,Beta-catenin ,Genes, APC ,Colon ,Intestinal Neoplasm ,gastrointestinal cancer ,IBD ,cancer genetics ,Mice, Transgenic ,colorectal adenomas ,colorectal cancer ,In situ hybridization ,5-aminosalicylic acid (5-ASA) ,03 medical and health sciences ,colorectal carcinoma ,Intestinal Neoplasms ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,cancer ,paneth cells ,mathematical modelling ,030304 developmental biology ,clinical trials ,Barretts carcinoma ,Original Articles ,colorectal diseases ,epithelial cells ,Wnt Proteins ,colorectal neogenesis ,inflammation ,gastrointestinal immune response ,Mutation ,IBD basic research ,biology.protein ,mucosal immunity ,microsatellite instability ,infliximab ,celiac disease - Abstract
OBJECTIVE: Wnt signalling is critical for normal intestinal development and homeostasis. Wnt dysregulation occurs in almost all human and murine intestinal tumours and an optimal but not excessive level of Wnt activation is considered favourable for tumourigenesis. The authors assessed effects of pan-intestinal Wnt activation on tissue homeostasis, taking into account underlying physiological Wnt activity and stem-cell number in each region of the bowel. DESIGN: The authors generated mice that expressed temporally controlled, stabilised β-catenin along the crypt-villus axis throughout the intestines. Physiological Wnt target gene activity was assessed in different regions of normal mouse and human tissue. Human intestinal tumour mutation spectra were analysed. RESULTS: In the mouse, β-catenin stabilisation resulted in a graduated neoplastic response, ranging from dysplastic transformation of the entire epithelium in the proximal small bowel to slightly enlarged crypts of non-dysplastic morphology in the colorectum. In contrast, stem and proliferating cell numbers were increased in all intestinal regions. In the normal mouse and human intestines, stem-cell and Wnt gradients were non-identical, but higher in the small bowel than large bowel in both species. There was also variation in the expression of some Wnt modulators. Human tumour analysis confirmed that different APC mutation spectra are selected in different regions of the bowel. CONCLUSIONS: There are variable gradients in stem-cell number, physiological Wnt activity and response to pathologically increased Wnt signalling along the crypt-villus axis and throughout the length of the intestinal tract. The authors propose that this variation influences regional mutation spectra, tumour susceptibility and lesion distribution in mice and humans.
- Published
- 2013
18. Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2-dependent manner
- Author
-
Terrence E. Riehl, Monica R. Walker, Xueping Ee, William F. Stenson, Matthew A. Ciorba, Clara Moon, Jeffrey M. Marinshaw, Thaddeus S. Stappenbeck, Gerardo M. Nava, and M. Suprada Rao
- Subjects
bacterial interactions ,crohn's disease ,gastrointestinal pathology ,Apoptosis ,Intestinal radioprotection ,radiation therapy ,Mice ,radiation enteritis ,0302 clinical medicine ,Intestinal mucosa ,epithelial proliferation ,Lactobacillus ,Prostaglandin E2 ,Intestinal Mucosa ,Mice, Knockout ,0303 health sciences ,biology ,Lacticaseibacillus rhamnosus ,Reverse Transcriptase Polymerase Chain Reaction ,Gastroenterology ,Intestinal epithelium ,3. Good health ,cancer immunobiology ,macrophages ,inflammatory mechanisms ,Radiation Injuries, Experimental ,medicine.anatomical_structure ,cyclooxygenase-2 ,030220 oncology & carcinogenesis ,anaerobic bacterial fermentation ,Female ,Whole-Body Irradiation ,probiotic ,medicine.drug ,inflammatory mediators ,Crypt ,IBD ,antibacterial peptide ,digestive system ,03 medical and health sciences ,Lactobacillus rhamnosus ,inflammatory bowel disease ,stem cells ,medicine ,Animals ,030304 developmental biology ,ulcerative colitis ,Lamina propria ,mesenchymal stem cells ,Probiotics ,bacterial infection ,biology.organism_classification ,Molecular biology ,Toll-Like Receptor 2 ,Mice, Inbred C57BL ,Cyclooxygenase 2 ,gastrointestinal immune response ,Immunology ,Myeloid Differentiation Factor 88 ,colorectal neoplasia - Abstract
Background The small intestinal epithelium is highly sensitive to radiation and is a major site of injury during radiation therapy and environmental overexposure. Objective To examine probiotic bacteria as potential radioprotective agents in the intestine. Methods 8-week-old C57BL/6 wild-type or knockout mice were administered probiotic by gavage for 3 days before 12 Gy whole body radiation. The intestine was evaluated for cell-positional apoptosis (6 h) and crypt survival (84 h). Results Gavage of 5×107 Lactobacillus rhamnosus GG (LGG) improved crypt survival about twofold (p
- Published
- 2011
19. Rifaximin is associated with modest, transient decreases in multiple taxa in the gut microbiota of patients with diarrhoea-predominant irritable bowel syndrome.
- Author
-
Fodor AA, Pimentel M, Chey WD, Lembo A, Golden PL, Israel RJ, and Carroll IM
- Subjects
- Adult, Aged, Aged, 80 and over, Anti-Bacterial Agents administration & dosage, Anti-Bacterial Agents pharmacology, Diarrhea drug therapy, Double-Blind Method, Feces microbiology, Female, Gastrointestinal Agents administration & dosage, Gastrointestinal Agents pharmacology, Humans, Irritable Bowel Syndrome physiopathology, Male, Middle Aged, Prospective Studies, RNA, Ribosomal, 16S genetics, Rifaximin administration & dosage, Young Adult, Gastrointestinal Microbiome drug effects, Irritable Bowel Syndrome drug therapy, Irritable Bowel Syndrome microbiology, Rifaximin pharmacology
- Abstract
Rifaximin, a non-systemic antibiotic, is efficacious for the treatment of diarrhoea-predominant irritable bowel syndrome (IBS-D). Given the emerging association between the gut microbiota and IBS, this study examined potential effects of rifaximin on the gastrointestinal microbial community in patients with IBS-D. TARGET 3 was a randomised, double-blind, placebo-controlled, phase 3 study. Patients with IBS-D initially received open-label rifaximin 550 mg 3 times daily (TID) for 2 weeks. Patients who responded to the initial treatment and then relapsed were randomised to receive 2 repeat courses of rifaximin 550 mg TID or placebo for 2 weeks, with each course separated by 10 weeks. Stool samples were collected at the beginning and end of open-label treatment, at the beginning and end of the first double-blind treatment, and at the end of the study. As a secondary analysis to the TARGET 3 trial, the composition and diversity of the gut microbiota were assessed, from a random subset of patients, using variable 4 hypervariable region 16S ribosomal RNA gene sequencing. Samples from 103 patients were included. After open-label rifaximin treatment for 2 weeks, 7 taxa (e.g. Peptostreptococcaceae, Verrucomicrobiaceae, Enterobacteriaceae) had significantly lower relative abundance at a 10% false discovery rate threshold. The effects of rifaximin were generally short-term, as there was little evidence of significantly different changes in taxa relative abundance at the end of the study (up to 46 weeks) versus baseline. The results suggest that rifaximin has a modest, largely transient effect across a broad range of stool microbes. Future research may determine whether the taxa affected by rifaximin are causally linked to IBS-D. ClinicalTrials.gov identifier number: NCT01543178.
- Published
- 2019
- Full Text
- View/download PDF
20. Putative biomarkers of vedolizumab resistance and underlying inflammatory pathways involved in IBD.
- Author
-
Soendergaard C, Seidelin JB, Steenholdt C, and Nielsen OH
- Abstract
Objectives: Characterise the circulating inflammatory cytokine pattern among patients failing consecutive anti-tumour necrosis factor (anti-TNF) and anti-integrin treatments to identify predictors of response., Methods: A retrospective single-centre cohort study of 28 patients with inflammatory bowel disease (IBD) receiving anti-integrin therapy (vedolizumab) subsequent to the failure of anti-TNF treatment was conducted. Blood samples were obtained immediately prior to initiation of vedolizumab therapy, and the response to treatment was evaluated after completion of the 14-week induction regimen. Multiplex ELISA was applied to quantify 47 preselected plasma proteins based on their putative involvement in the inflammatory process in IBD., Results: Anti-TNF and vedolizumab non-responders (n=20) had significantly higher levels of circulating interleukin (IL)-6 than anti-TNF non-responders with subsequent response to vedolizumab (n=8): median 9.5 pg/mL versus 5.9 pg/mL, p<0.05. Following stratification by diagnosis, patients with Crohn's disease who failed vedolizumab therapy (n=7) had higher soluble CD40 ligand (sCD40L) than responders (n=4): 153.0 pg/mL versus 45.5 pg/mL, p<0.01; sensitivity 100% (95% CI 59% to 100%), specificity 100% (95% CI 40% to 100%). Osteocalcin was higher among patients with ulcerative colitis responding to vedolizumab (n=4) compared with those not responding (n=13): 4219 pg/mL versus 2823 pg/mL, p=0.01; sensitivity 85% (95% CI 55% to 98%), specificity 100% (95% CI 40% to 100%)., Conclusions: Patients with IBD failing vedolizumab induction and anti-TNF therapy have persistent IL-6 pathway activity, which could be a potential alternative treatment target. sCD40L, osteocalcin and the IL-6 pathway activity might be predictors for response to vedolizumab., Competing Interests: Competing interests: None declared.
- Published
- 2018
- Full Text
- View/download PDF
21. Gut symbiotic microbes imprint intestinal immune cells with the innate receptor SLAMF4 which contributes to gut immune protection against enteric pathogens.
- Author
-
Cabinian A, Sinsimer D, Tang M, Jang Y, Choi B, Laouar Y, and Laouar A
- Subjects
- Animals, Flow Cytometry, Germ-Free Life, Humans, Intestinal Mucosa microbiology, Mice, Mice, Inbred C57BL, Real-Time Polymerase Chain Reaction, Signal Transduction, Symbiosis, Gastrointestinal Microbiome immunology, Immunity, Mucosal physiology, Intestinal Mucosa metabolism, Signaling Lymphocytic Activation Molecule Family metabolism
- Abstract
Background: Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field., Aim: We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is., Methods: Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFP
tg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium ., Results: SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC)TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death., Conclusions: SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut., Competing Interests: Competing interests: None declared., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2018
- Full Text
- View/download PDF
22. Ceramide-CD300f binding suppresses experimental colitis by inhibiting ATP-mediated mast cell activation.
- Author
-
Matsukawa T, Izawa K, Isobe M, Takahashi M, Maehara A, Yamanishi Y, Kaitani A, Okumura K, Teshima T, Kitamura T, and Kitaura J
- Subjects
- Animals, Disease Models, Animal, Mice, Mice, Inbred C57BL, Adenosine Triphosphate antagonists & inhibitors, Ceramides physiology, Colitis prevention & control, Mast Cells physiology, Receptors, Immunologic physiology
- Abstract
Objective: Extracellular ATP mediates mast cell-dependent intestinal inflammation via P2X7 purinoceptors. We have previously shown that CD300f (also called the leucocyte mono-immunoglobulin-like receptor 3 (LMIR3)) suppresses immunoglobulin E-dependent and mast cell-dependent allergic responses by binding to ceramide. The aim of the present study was to clarify the role of ceramide-LMIR3 interaction in the development of IBD., Design: The dextran sodium sulfate (DSS)-induced colitis model was used in wild-type (WT), LMIR3(-/-), mast cell-deficient Kit(W-sh/W-sh), Kit(W-sh/W-sh)LMIR3(-/-) or Kit(W-sh/W-sh) mice engrafted with WT or LMIR3(-/-) bone marrow-derived mast cells (BMMCs). The severity of colitis was determined by clinical and histological criteria. Lamina propria cell populations were assessed by flow cytometry. Production of chemical mediators from lamina propria cells was measured by real-time reverse transcription PCR. Production of chemical mediators from ATP-stimulated BMMCs in the presence or absence of ceramide was measured by ELISA. The severity of DSS-induced colitis was assessed in mice given either an Fc fusion protein containing an extracellular domain of LMIR3, and anticeramide antibody, or ceramide liposomes., Results: LMIR3 deficiency exacerbated DSS-induced colitis in mice. Kit(W-sh/W-sh) mice harbouring LMIR3(-/-) mast cells exhibited more severe colitis than those harbouring WT mast cells. Ceramide-LMIR3 interaction inhibited ATP-stimulated activation of BMMCs. DSS-induced colitis was aggravated by disrupting the ceramide-LMIR3 interaction, whereas it was suppressed by treating with ceramide liposomes., Conclusions: LMIR3-deficient colonic mast cells were pivotal in the exacerbation of DSS-induced colitis in LMIR3(-/-) mice. Ceramide liposomes attenuated DSS-induced colitis by inhibiting ATP-mediated activation of colonic mast cells through ceraimide-LMIR3 binding., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
23. Presence of commensal house dust mite allergen in human gastrointestinal tract: a potential contributor to intestinal barrier dysfunction.
- Author
-
Tulic MK, Vivinus-Nébot M, Rekima A, Rabelo Medeiros S, Bonnart C, Shi H, Walker A, Dainese R, Boyer J, Vergnolle N, Piche T, and Verhasselt V
- Subjects
- Animals, Humans, Mice, Mice, Inbred BALB C, Antigens, Dermatophagoides isolation & purification, Dermatophagoides pteronyssinus immunology, Gastrointestinal Diseases immunology, Gastrointestinal Tract immunology
- Abstract
Background: Abnormal gut barrier function is the basis of gut inflammatory disease. It is known that house dust mite (HDM) aero-allergens induce inflammation in respiratory mucosa. We have recently reported allergen from Dermatophagoides pteronyssinus (Der p1) to be present in rodent gut., Objective: To examine whether Der p1 is present in human gut and to assess its effect on gut barrier function and inflammation., Design: Colonic biopsies, gut fluid, serum and stool were collected from healthy adults during endoscopy. Der p1 was measured by ELISA. Effect of HDM was assessed on gut permeability, tight-junction and mucin expression, and cytokine production, in presence or absence of cysteine protease inhibitors or serine protease inhibitors. In vivo effect of HDM was examined in mice given oral HDM or protease-neutralised HDM. Role of HDM in low-grade inflammation was studied in patients with IBS., Results: HDM Der p1 was detected in the human gut. In colonic biopsies from healthy patients, HDM increased epithelial permeability (p<0.001), reduced expression of tight-junction proteins and mucus barrier. These effects were associated with increased tumour necrosis factor (TNF)-α and interleukin (IL)-10 production and were abolished by cysteine-protease inhibitor (p<0.01). HDM effects did not require Th2 immunity. Results were confirmed in vivo in mice. In patients with IBS, HDM further deteriorated gut barrier function, induced TNF-α but failed to induce IL-10 secretion (p<0.001)., Conclusions: HDM, a ubiquitous environmental factor, is present in the human gut where it directly affects gut function through its proteolytic activity. HDM may be an important trigger of gut dysfunction and warrants further investigation., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
24. Compartment-specific immunity in the human gut: properties and functions of dendritic cells in the colon versus the ileum.
- Author
-
Mann ER, Bernardo D, English NR, Landy J, Al-Hassi HO, Peake ST, Man R, Elliott TR, Spranger H, Lee GH, Parian A, Brant SR, Lazarev M, Hart AL, Li X, and Knight SC
- Subjects
- Antigens, CD analysis, Colon ultrastructure, Cytokines metabolism, Dendritic Cells cytology, Flow Cytometry, Humans, Ileum ultrastructure, Integrin alpha Chains analysis, Lymphocyte Culture Test, Mixed, Membrane Glycoproteins, Microscopy, Electron, Molecular Imprinting, Receptors, CCR analysis, Receptors, CCR4 analysis, Receptors, CCR7 analysis, Receptors, Cell Surface analysis, Receptors, Immunologic, T-Lymphocytes immunology, T-Lymphocytes, Regulatory immunology, Colon immunology, Dendritic Cells immunology, Ileum immunology
- Abstract
Objective: Dendritic cells (DC) mediate intestinal immune tolerance. Despite striking differences between the colon and the ileum both in function and bacterial load, few studies distinguish between properties of immune cells in these compartments. Furthermore, information of gut DC in humans is scarce. We aimed to characterise human colonic versus ileal DC., Design: Human DC from paired colonic and ileal samples were characterised by flow cytometry, electron microscopy or used to stimulate T cell responses in a mixed leucocyte reaction., Results: A lower proportion of colonic DC produced pro-inflammatory cytokines (tumour necrosis factor-α and interleukin (IL)-1β) compared with their ileal counterparts and exhibited an enhanced ability to generate CD4(+)FoxP3(+)IL-10(+) (regulatory) T cells. There were enhanced proportions of CD103(+)Sirpα(-) DC in the colon, with increased proportions of CD103(+)Sirpα(+) DC in the ileum. A greater proportion of colonic DC subsets analysed expressed the lymph-node-homing marker CCR7, alongside enhanced endocytic capacity, which was most striking in CD103(+)Sirpα(+) DC. Expression of the inhibitory receptor ILT3 was enhanced on colonic DC. Interestingly, endocytic capacity was associated with CD103(+) DC, in particular CD103(+)Sirpα(+) DC. However, expression of ILT3 was associated with CD103(-) DC. Colonic and ileal DC differentially expressed skin-homing marker CCR4 and small-bowel-homing marker CCR9, respectively, and this corresponded to their ability to imprint these homing markers on T cells., Conclusions: The regulatory properties of colonic DC may represent an evolutionary adaptation to the greater bacterial load in the colon. The colon and the ileum should be regarded as separate entities, each comprising DC with distinct roles in mucosal immunity and imprinting., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
25. Changing the narrative on antibiotics.
- Author
-
Shanahan F
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome genetics, Gene Regulatory Networks
- Published
- 2015
- Full Text
- View/download PDF
26. α4β7 integrin: beyond T cell trafficking.
- Author
-
Kempster SL and Kaser A
- Subjects
- Animals, Colitis immunology, Dendritic Cells metabolism, Integrin beta Chains metabolism, Integrins metabolism, Intestinal Mucosa immunology, Macrophages metabolism
- Published
- 2014
- Full Text
- View/download PDF
27. The vagal innervation of the gut and immune homeostasis.
- Author
-
Matteoli G and Boeckxstaens GE
- Subjects
- Acetylcholine physiology, Homeostasis immunology, Humans, Ileus immunology, Ileus prevention & control, Immunity, Mucosal physiology, Inflammation immunology, Inflammatory Bowel Diseases immunology, Signal Transduction immunology, Intestines innervation, Neuroimmunomodulation physiology, Vagus Nerve immunology
- Abstract
The central nervous system interacts dynamically with the immune system to modulate inflammation through humoral and neural pathways. Recently, in animal models of sepsis, the vagus nerve (VN) has been proposed to play a crucial role in the regulation of the immune response, also referred to as the cholinergic anti-inflammatory pathway. The VN, through release of acetylcholine, dampens immune cell activation by interacting with α-7 nicotinic acetylcholine receptors. Recent evidence suggests that the vagal innervation of the gastrointestinal tract also plays a major role controlling intestinal immune activation. Indeed, VN electrical stimulation potently reduces intestinal inflammation restoring intestinal homeostasis, whereas vagotomy has the reverse effect. In this review, we will discuss the current understanding concerning the mechanisms and effects involved in the cholinergic anti-inflammatory pathway in the gastrointestinal tract. Deeper investigation on this counter-regulatory neuroimmune mechanism will provide new insights in the cross-talk between the nervous and immune system leading to the identification of new therapeutic targets to treat intestinal immune disease.
- Published
- 2013
- Full Text
- View/download PDF
28. Bioinformatic and immunological analysis reveals lack of support for measles virus related mimicry in Crohn’s disease
- Author
-
Konstantinos Triantafyllou, Dimitrios Polymeros, Dimitrios P. Bogdanos, Andreas Koutsoumpas, Maria G. Mytilinaiou, Zacharias P. Tsiamoulos, Daniel S. Smyk, and Spiros D. Ladas
- Subjects
Adult ,Male ,Enzyme-Linked Immunosorbent Assay ,Autoimmunity ,Biology ,medicine.disease_cause ,Gastrointestinal immune response ,Measles ,Inflammatory bowel disease ,Epitope ,Diagnosis, Differential ,Measles virus ,03 medical and health sciences ,0302 clinical medicine ,Crohn Disease ,Antigen ,medicine ,Humans ,Antigens, Viral ,Aged ,030304 developmental biology ,Medicine(all) ,Infectious disease ,0303 health sciences ,Computational Biology ,General Medicine ,Middle Aged ,biology.organism_classification ,medicine.disease ,Virology ,3. Good health ,Molecular mimicry ,Case-Control Studies ,Immunology ,biology.protein ,Colitis, Ulcerative ,Female ,030211 gastroenterology & hepatology ,Antibody ,Research Article - Abstract
Background A link between measles virus and Crohn’s disease (CD) has been postulated. We assessed through bioinformatic and immunological approaches whether measles is implicated in CD induction, through molecular mimicry. Methods The BLAST2p program was used to identify amino acid sequence similarities between five measles virus and 56 intestinal proteins. Antibody responses to measles/human mimics were tested by an in-house ELISA using serum samples from 50 patients with CD, 50 with ulcerative colitis (UC), and 38 matched healthy controls (HCs). Results We identified 15 sets of significant (>70%) local amino acid homologies from two measles antigens, hemagglutinin-neuraminidase and fusion-glycoprotein, and ten human intestinal proteins. Reactivity to at least one measles 15-meric mimicking peptide was present in 27 out of 50 (54%) of patients with CD, 24 out of 50 (48%) with UC (CD versus UC, p = 0.68), and 13 out of 38 (34.2%) HCs (CD versus HC, p = 0.08). Double reactivity to at least one measles/human pair was present in four out of 50 (8%) patients with CD, three out of 50 (6%) with UC (p = 0.99), and in three out of 38 (7.9%) HCs (p >0.05 for all). Titration experiments yielded different extinction curves for anti-measles and anti-human intestinal double-reactive antibodies. Epitope prediction algorithms and three-dimensional modeling provided bioinformatic confirmation for the observed antigenicity of the main measles virus epitopic regions. Conclusions Measles sequences mimicking intestinal proteins are frequent targets of antibody responses in patients with CD, but this reactivity lacks disease specificity and does not initiate cross-reactive responses to intestinal mimics. We conclude that there is no involvement of measles/human molecular mimicry in the etiopathogenesis of CD. Electronic supplementary material The online version of this article (doi:10.1186/s12916-014-0139-9) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.