Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Lupus ; 32(4): 461-470, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36896485

ABSTRACT

Lupus erythematosus is a complex autoimmune disease characterized by skin and/or systemic involvement. Among systemic disorders, half of the patients will experience non-specific digestive symptoms, usually due to drug medication or transitory infections. In rare cases, lupus enteritis can be observed, and its diagnosis may precede the disease and/or be associated with an inflammatory bowel disease (IBD). Among the underlying mechanisms explaining the digestive damages observed in systemic lupus erythematosus (SLE) and the intestinal barrier function (IBF), increased intestinal permeability, microbiota dysbiosis, and intestinal immune system dysregulations are described in numerous murine and human studies. New therapeutic approaches in addition to conventional treatments are evoked in order to better control the IBF disruption and maybe prevent the onset or worsening of the disease. Thus, the aims of this review are to present the alterations of the digestive tract in SLE patients and the link between SLE and IBD as well as how the different elements of the IBF could participate in SLE pathogenesis.


Subject(s)
Enteritis , Inflammatory Bowel Diseases , Lupus Erythematosus, Systemic , Microbiota , Humans , Animals , Mice , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/complications , Skin/pathology
2.
Part Fibre Toxicol ; 20(1): 45, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37996842

ABSTRACT

BACKGROUND: Perinatal exposure to titanium dioxide (TiO2), as a foodborne particle, may influence the intestinal barrier function and the susceptibility to develop inflammatory bowel disease (IBD) later in life. Here, we investigate the impact of perinatal foodborne TiO2 exposure on the intestinal mucosal function and the susceptibility to develop IBD-associated colitis. Pregnant and lactating mother mice were exposed to TiO2 until pups weaning and the gut microbiota and intestinal barrier function of their offspring was assessed at day 30 post-birth (weaning) and at adult age (50 days). Epigenetic marks was studied by DNA methylation profile measuring the level of 5-methyl-2'-deoxycytosine (5-Me-dC) in DNA from colic epithelial cells. The susceptibility to develop IBD has been monitored using dextran-sulfate sodium (DSS)-induced colitis model. Germ-free mice were used to define whether microbial transfer influence the mucosal homeostasis and subsequent exacerbation of DSS-induced colitis. RESULTS: In pregnant and lactating mice, foodborne TiO2 was able to translocate across the host barriers including gut, placenta and mammary gland to reach embryos and pups, respectively. This passage modified the chemical element composition of foetus, and spleen and liver of mothers and their offspring. We showed that perinatal exposure to TiO2 early in life alters the gut microbiota composition, increases the intestinal epithelial permeability and enhances the colonic cytokines and myosin light chain kinase expression. Moreover, perinatal exposure to TiO2 also modifies the abilities of intestinal stem cells to survive, grow and generate a functional epithelium. Maternal TiO2 exposure increases the susceptibility of offspring mice to develop severe DSS-induced colitis later in life. Finally, transfer of TiO2-induced microbiota dysbiosis to pregnant germ-free mice affects the homeostasis of the intestinal mucosal barrier early in life and confers an increased susceptibility to develop colitis in adult offspring. CONCLUSIONS: Our findings indicate that foodborne TiO2 consumption during the perinatal period has negative long-lasting consequences on the development of the intestinal mucosal barrier toward higher colitis susceptibility. This demonstrates to which extent environmental factors influence the microbial-host interplay and impact the long-term mucosal homeostasis.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Pregnancy , Female , Animals , Mice , Dysbiosis/chemically induced , Lactation , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Mice, Inbred C57BL , Disease Models, Animal
3.
Gut ; 70(6): 1088-1097, 2021 06.
Article in English | MEDLINE | ID: mdl-32978245

ABSTRACT

OBJECTIVE: Data from clinical research suggest that certain probiotic bacterial strains have the potential to modulate colonic inflammation. Nonetheless, these data differ between studies due to the probiotic bacterial strains used and the poor knowledge of their mechanisms of action. DESIGN: By mass-spectrometry, we identified and quantified free long chain fatty acids (LCFAs) in probiotics and assessed the effect of one of them in mouse colitis. RESULTS: Among all the LCFAs quantified by mass spectrometry in Escherichia coli Nissle 1917 (EcN), a probiotic used for the treatment of multiple intestinal disorders, the concentration of 3-hydroxyoctadecaenoic acid (C18-3OH) was increased in EcN compared with other E. coli strains tested. Oral administration of C18-3OH decreased colitis induced by dextran sulfate sodium in mice. To determine whether other bacteria composing the microbiota are able to produce C18-3OH, we targeted the gut microbiota of mice with prebiotic fructooligosaccharides (FOS). The anti-inflammatory properties of FOS were associated with an increase in colonic C18-3OH concentration. Microbiota analyses revealed that the concentration of C18-3OH was correlated with an increase in the abundance in Allobaculum, Holdemanella and Parabacteroides. In culture, Holdemanella biformis produced high concentration of C18-3OH. Finally, using TR-FRET binding assay and gene expression analysis, we demonstrated that the C18-3OH is an agonist of peroxisome proliferator activated receptor gamma. CONCLUSION: The production of C18-3OH by bacteria could be one of the mechanisms implicated in the anti-inflammatory properties of probiotics. The production of LCFA-3OH by bacteria could be implicated in the microbiota/host interactions.


Subject(s)
Colitis/drug therapy , Intestinal Mucosa/metabolism , PPAR gamma/metabolism , Stearates/metabolism , Stearates/therapeutic use , Animals , Bacteroidetes , Caco-2 Cells , Cell Membrane Permeability , Chemokine CXCL1/genetics , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , Epithelial Cells/physiology , Escherichia coli/metabolism , Firmicutes/metabolism , Gastrointestinal Microbiome/physiology , Gene Expression/drug effects , Humans , Interleukin-1beta/genetics , Mass Spectrometry , Mice , Oligosaccharides/pharmacology , PPAR gamma/genetics , Pancreatitis-Associated Proteins/genetics , Permeability , Peyer's Patches , Prebiotics , Probiotics/chemistry , Stearates/analysis , Zonula Occludens-1 Protein/genetics
4.
Part Fibre Toxicol ; 18(1): 26, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330311

ABSTRACT

The gastrointestinal tract is a complex interface between the external environment and the immune system. Its ability to control uptake across the mucosa and to protect the body from damage of harmful substances from the lumen is defined as the intestinal barrier function (IBF). The IBF involves four elements: the intestinal microbiota, the mucus layer, the epithelium and the immune system. Its dysfunction is linked with human diseases including inflammatory, metabolic, infectious, autoimmune and neurologic disorders. Most of these diseases are complex and involve genetic, psychological and environmental factors. Over the past 10 years, many genetic polymorphisms predisposing to inflammatory bowel disease (IBD) have been identified. Yet, it is now clear that they are insufficient to explain the onset of these chronic diseases. Although it has been evidenced that some environmental factors such as cigarette smoking or carbohydrate intake are associated with IBD, other environmental factors also present potential health risks such as ingestion of food additives introduced in the human diet, including those composed of mineral particles, by altering the four elements of the intestinal barrier function. The aim of this review is to provide a critical opinion on the potential of TiO2 particles, especially when used as a food additive, to alter the four elements of the intestinal barrier function, and consequently to evaluate if this additive would likely play a role in the development and/or exacerbation of IBD.


Subject(s)
Colorectal Neoplasms , Inflammatory Bowel Diseases , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Diet/adverse effects , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa , Titanium
5.
Int J Mol Sci ; 22(20)2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34681921

ABSTRACT

Inflammatory bowel diseases (IBDs), which include Crohn's disease and ulcerative colitis, are multifactorial diseases that involve in particular a modification of the gut microbiota, known as dysbiosis. The initial sets of metataxonomic and metagenomic data first made it possible to approximate the microbiota profile in IBD. In addition, today the new 'omics' techniques have enabled us to draw up a functional and integrative map of the microbiota. The key concern in IBD is to develop biomarkers that allow us to assess the activity of the disease and predict the complications and progression, while also guiding the therapeutic care so as to develop personalized medicine. In this review, we present all of the latest discoveries on the microbiota provided by "omics" and we outline the benefits of these techniques in developing new diagnostic, prognostic and therapeutic tools.


Subject(s)
Bacteria/classification , Inflammatory Bowel Diseases/microbiology , Metagenomics/methods , Bacteria/isolation & purification , Disease Progression , Gastrointestinal Microbiome , Humans , Phylogeny , Precision Medicine
6.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919747

ABSTRACT

Crohn's disease is an inflammatory bowel disease whose prevalence is increasing worldwide. Among medical strategies, dietary therapy with exclusive enteral nutrition is recommended as a first-line option, at least for children, because it induces clinical remission and mucosal healing. Modulen®, a polymeric TGF-ß2 enriched formula, has good palatability and is widely used. For the first time in the literature, this review outlines and discusses the clinical outcomes obtained with this therapy, as well as the potential mechanisms of action of its compounds. It can be explained by its TGF-ß2 content, but also by its protein and lipid composition. Further well-designed studies are required to improve our knowledge and to optimize therapeutic strategies.


Subject(s)
Crohn Disease/pathology , Polymers/pharmacology , Crohn Disease/drug therapy , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Models, Biological , Remission Induction , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factor beta2/therapeutic use
7.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299013

ABSTRACT

Mucosal CD4+ T lymphocytes display a potent opioid-mediated analgesic activity in interleukin (IL)-10 knockout mouse model of inflammatory bowel diseases (IBD). Considering that endogenous opioids may also exhibit anti-inflammatory activities in the periphery, we examined the consequences of a peripheral opioid receptor blockade by naloxone-methiodide, a general opioid receptor antagonist unable to cross the blood-brain barrier, on the development of piroxicam-accelerated colitis in IL-10-deficient (IL-10-/-) mice. Here, we show that IL-10-deficient mice treated with piroxicam exhibited significant alterations of the intestinal barrier function, including permeability, inflammation-related bioactive lipid mediators, and mucosal CD4+ T lymphocyte subsets. Opioid receptor antagonization in the periphery had virtually no effect on colitis severity but significantly worsened epithelial cell apoptosis and intestinal permeability. Thus, although the endogenous opioid tone is not sufficient to reduce the severity of colitis significantly, it substantially contributes to the protection of the physical integrity of the epithelial barrier.


Subject(s)
Colitis/metabolism , Interleukin-10/genetics , Intestinal Mucosa/drug effects , Naloxone/analogs & derivatives , Narcotic Antagonists/administration & dosage , Piroxicam/pharmacology , Receptors, Opioid/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , CD4-Positive T-Lymphocytes/drug effects , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/pharmacology , Permeability/drug effects , Quaternary Ammonium Compounds/pharmacology , Severity of Illness Index
8.
PLoS Pathog ; 13(3): e1006177, 2017 03.
Article in English | MEDLINE | ID: mdl-28253332

ABSTRACT

Nucleotide-binding oligomerization domain 2 (NOD2) is an intracellular pattern recognition receptor that senses bacterial peptidoglycan (PGN)-conserved motifs in cytosol and stimulates host immune response. The association of NOD2 mutations with a number of inflammatory pathologies, including Crohn disease (CD), Graft-versus-host disease (GVHD), and Blau syndrome, highlights its pivotal role in host-pathogen interactions and inflammatory response. Stimulation of NOD2 by its ligand (muramyl dipeptide) activates pro-inflammatory pathways such as nuclear factor-κB (NF-κB), mitogen-activated protein kinases (MAPKs), and Caspase-1. A loss of NOD2 function may result in a failure in the control of microbial infection, thereby initiating systemic responses and aberrant inflammation. Because the ligand of Nod2 is conserved in both gram-positive and gram-negative bacteria, NOD2 detects a wide variety of microorganisms. Furthermore, current literature evidences that NOD2 is also able to control viruses' and parasites' infections. In this review, we present and discuss recent developments about the role of NOD2 in shaping the gut commensal microbiota and pathogens, including bacteria, viruses, and parasites, and the mechanisms by which Nod2 mutations participate in disease occurrence.


Subject(s)
Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Intestines/immunology , Nod2 Signaling Adaptor Protein/immunology , Animals , Humans
11.
Part Fibre Toxicol ; 11: 13, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24666995

ABSTRACT

BACKGROUND: TiO2 particles are commonly used as dietary supplements and may contain up to 36% of nano-sized particles (TiO2-NPs). Still impact and translocation of NPs through the gut epithelium is poorly documented. RESULTS: We show that, in vivo and ex vivo, agglomerates of TiO2-NPs cross both the regular ileum epithelium and the follicle-associated epithelium (FAE) and alter the paracellular permeability of the ileum and colon epithelia. In vitro, they accumulate in M-cells and mucus-secreting cells, much less in enterocytes. They do not cause overt cytotoxicity or apoptosis. They translocate through a model of FAE only, but induce tight junctions remodeling in the regular ileum epithelium, which is a sign of integrity alteration and suggests paracellular passage of NPs. Finally we prove that TiO2-NPs do not dissolve when sequestered up to 24 h in gut cells. CONCLUSIONS: Taken together these data prove that TiO2-NPs would possibly translocate through both the regular epithelium lining the ileum and through Peyer's patches, would induce epithelium impairment, and would persist in gut cells where they would possibly induce chronic damage.


Subject(s)
Epithelium/metabolism , Gastrointestinal Tract/metabolism , Nanoparticles/metabolism , Nanoparticles/toxicity , Titanium/pharmacokinetics , Titanium/toxicity , Animals , Biological Transport , Cell Line , Cell Survival/drug effects , Humans , Intestinal Absorption , Mice , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Real-Time Polymerase Chain Reaction , Spectrometry, X-Ray Emission , Suspensions , Titanium/chemistry , X-Ray Absorption Spectroscopy
12.
Gastroenterology ; 142(5): 1097-1099.e4, 2012 May.
Article in English | MEDLINE | ID: mdl-22285936

ABSTRACT

Autophagy-related 16 like-1 (ATG16L-1), immunity-related GTPase-M (IRGM), and nucleotide-binding oligomerization domain-containing 2 (NOD2) regulate autophagy, and variants in these genes have been associated with predisposition to Crohn's disease (CD). However, little is known about the role of autophagy in CD. Intestinal biopsies from untreated pediatric patients with CD, celiac disease, or ulcerative colitis were analyzed by immunohistochemistry and electron microscopy. We observed that autophagy was specifically activated in Paneth cells from patients with CD, independently of mucosal inflammation or disease-associated variants of ATG16L1 or IRGM. In these cells, activation of autophagy was associated with a significant decrease in number of secretory granules and features of crinophagy. These observations might account for the disorganization of secretory granules previously reported in Paneth cells from patients with CD.


Subject(s)
Autophagy/physiology , Crohn Disease/pathology , Paneth Cells/ultrastructure , Secretory Vesicles/physiology , Autophagy-Related Proteins , Carrier Proteins/genetics , Child , Female , GTP-Binding Proteins/genetics , Humans , Infant , Male , Microtubule-Associated Proteins/analysis , Nod2 Signaling Adaptor Protein/genetics , Polymorphism, Single Nucleotide
13.
Gastroenterology ; 143(1): 122-32.e15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22446194

ABSTRACT

BACKGROUND & AIMS: Cell adhesion is one function regulated by cellular prion protein (PrP(c)), a ubiquitous, glycosylphosphatidylinositol-anchored glycoprotein. PrP(c) is located in cell-cell junctions and interacts with desmosome proteins in the intestinal epithelium. We investigated its role in intestinal barrier function. METHODS: We analyzed permeability and structure of cell-cell junctions in intestine tissues from PrP(c) knockout (PrP(c-/-)) and wild-type mice. PrP(c) expression was knocked down in cultured human Caco-2/TC7 enterocytes using small hairpin RNAs. We analyzed colon samples from 24 patients with inflammatory bowel disease (IBD). RESULTS: Intestine tissues from PrP(c-/-) mice had greater paracellular permeability than from wild-type mice (105.9 ± 13.4 vs 59.6 ± 10.1 mg/mL fluorescein isothiocyanate-dextran flux; P < .05) and impaired intercellular junctions. PrP(c-/-) mice did not develop spontaneous disease but were more sensitive than wild-type mice to induction of colitis with dextran sulfate (32% mortality vs 4%, respectively; P = .0033). Such barrier defects were observed also in Caco-2/TC7 enterocytes following PrP(c) knockdown; the cells had increased paracellular permeability (1.5-fold over 48 hours; P < .001) and reduced transepithelial electrical resistance (281.1 ± 4.9 vs 370.6 ± 5.7 Ω.cm(2); P < .001). Monolayer shape and cell-cell junctions were altered in cultures of PrP(c) knockdown cells; levels of E-cadherin, desmoplakin, plakoglobin, claudin-4, occludin, zonula occludens 1, and tricellulin were decreased at cell contacts. Cell shape and junctions were restored on PrP(c) re-expression. Levels of PrP(c) were decreased at cell-cell junctions in colonic epithelia from patients with Crohn's disease or ulcerative colitis. CONCLUSIONS: PrP(c) regulates intestinal epithelial cell-cell junctions and barrier function. Its localization is altered in colonic epithelia from patients with IBD, supporting the concept that disrupted barrier function contributes to this disorder.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Intercellular Junctions/metabolism , Intestinal Mucosa/metabolism , PrPC Proteins/metabolism , Animals , Cell Membrane Permeability/physiology , Cells, Cultured , Colon/metabolism , Enterocytes/metabolism , Humans , Mice , Mice, Knockout
14.
Commun Biol ; 6(1): 1168, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37968381

ABSTRACT

Opioid-dependent immune-mediated analgesic effects have been broadly reported upon inflammation. In preclinical mouse models of intestinal inflammatory diseases, the local release of enkephalins (endogenous opioids) by colitogenic T lymphocytes alleviate inflammation-induced pain by down-modulating gut-innervating nociceptor activation in periphery. In this study, we wondered whether this immune cell-derived enkephalin-mediated regulation of the nociceptor activity also operates under steady state conditions. Here, we show that chimeric mice engrafted with enkephalin-deficient bone marrow cells exhibit not only visceral hypersensitivity but also an increase in both epithelial paracellular and transcellular permeability, an alteration of the microbial topography resulting in increased bacteria-epithelium interactions and a higher frequency of IgA-producing plasma cells in Peyer's patches. All these alterations of the intestinal homeostasis are associated with an anxiety-like behavior despite the absence of an overt inflammation as observed in patients with irritable bowel syndrome. Thus, our results show that immune cell-derived enkephalins play a pivotal role in maintaining gut homeostasis and normal behavior in mice. Because a defect in the mucosal opioid system remarkably mimics some major clinical symptoms of the irritable bowel syndrome, its identification might help to stratify subgroups of patients.


Subject(s)
Irritable Bowel Syndrome , Humans , Animals , Mice , Analgesics, Opioid , Enkephalins/genetics , Inflammation , Pain
15.
Gut ; 59(2): 207-17, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19837677

ABSTRACT

Nucleotide oligomerisation domain 2 (NOD2) mutations are associated with susceptibility to Crohn's disease and graft-versus-host disease, two human disorders related with dysfunctions of Peyer's patches (PPs). In Nod2(-/-) mice transcellular permeability and bacterial translocation are increased in PPs. In this study, we show that both anti-CD4(+) and anti-interferon gamma (anti-IFNgamma) monoclonal antibodies abrogate this phenotype and reduce the expression of tumour necrosis factor (TNF) receptor 2 and the long isoform of myosin light chain kinase, thus demonstrating that immune T cells influence the epithelial functions. In turn, intraperitoneal injection of ML-7 (a myosin light chain kinase inhibitor) normalises the values of CD4(+) T cells, IFNgamma and TNFalpha. This reciprocal cross-talk is under the control of the gut microflora as shown by the normalisation of all parameters after antibiotic treatment. Toll-like receptor 2 (TLR2) and TLR4 expression were increased in Nod2(-/-) mice under basal conditions and TLR2 and TLR4 agonists induced an increased transcellular permeability in Nod2(+/+) mice. Muramyldipeptide (a Nod2 agonist) or ML-7 was able to reverse this phenomenon. It thus appears that Nod2 modulates the cross-talk between CD4(+) T cells and the epithelium recovering PP and that it downregulates the pro-inflammatory effect driven by the ileal microflora, likely by inhibiting the TLR pathways.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Intestinal Absorption/immunology , Nod2 Signaling Adaptor Protein/immunology , Peyer's Patches/immunology , Peyer's Patches/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Translocation , CD4-Positive T-Lymphocytes/drug effects , Diffusion Chambers, Culture , Ileum/microbiology , Interferon-gamma/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/physiopathology , Mice , Mice, Inbred C57BL , Permeability , Peyer's Patches/physiopathology , Reverse Transcriptase Polymerase Chain Reaction/methods , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/physiology
16.
J Crohns Colitis ; 15(4): 678-686, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-32949122

ABSTRACT

Crohn's disease [CD] is an inflammatory bowel disease of unknown aetiology. During recent decades, significant technological advances led to development of -omic datasets allowing a detailed description of the disease. Unfortunately these have not, to date, resolved the question of the aetiology of CD. Thus, it may be necessary to [re]consider hypothesis-driven approaches to resolve the aetiology of CD. According to the cold chain hypothesis, the development of industrial and domestic refrigeration has led to frequent exposure of human populations to bacteria capable of growing in the cold. These bacteria, at low levels of exposure, particularly those of the genus Yersinia, are believed to be capable of inducing exacerbated inflammation of the intestine in genetically predisposed subjects. We discuss the consistency of this working hypothesis in light of recent data from epidemiological, clinical, pathological, microbiological, and molecular studies.


Subject(s)
Crohn Disease/microbiology , Food Microbiology , Refrigeration , Yersinia/pathogenicity , Causality , Crohn Disease/genetics , Genetic Predisposition to Disease , Humans
17.
J Crohns Colitis ; 15(9): 1528-1541, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-33609354

ABSTRACT

BACKGROUND AND AIMS: Intestinal epithelial cells [IECs] from inflammatory bowel disease [IBD] patients exhibit an excessive induction of endoplasmic reticulum stress [ER stress] linked to altered intestinal barrier function and inflammation. Colonic tissues and the luminal content of IBD patients are also characterized by increased serine protease activity. The possible link between ER stress and serine protease activity in colitis-associated epithelial dysfunctions is unknown. We aimed to study the association between ER stress and serine protease activity in enterocytes and its impact on intestinal functions. METHODS: The impact of ER stress induced by Thapsigargin on serine protease secretion was studied using either human intestinal cell lines or organoids. Moreover, treating human intestinal cells with protease-activated receptor antagonists allowed us to investigate ER stress-resulting molecular mechanisms that induce proteolytic activity and alter intestinal epithelial cell biology. RESULTS: Colonic biopsies from IBD patients exhibited increased epithelial trypsin-like activity associated with elevated ER stress. Induction of ER stress in human intestinal epithelial cells displayed enhanced apical trypsin-like activity. ER stress-induced increased trypsin activity destabilized intestinal barrier function by increasing permeability and by controlling inflammatory mediators such as C-X-C chemokine ligand 8 [CXCL8]. The deleterious impact of ER stress-associated trypsin activity was specifically dependent on the activation of protease-activated receptors 2 and 4. CONCLUSIONS: Excessive ER stress in IECs caused an increased release of trypsin activity that, in turn, altered intestinal barrier function, promoting the development of inflammatory process.


Subject(s)
Colitis, Ulcerative/pathology , Crohn Disease/pathology , Endoplasmic Reticulum Stress/physiology , Enterocytes/physiology , Intestinal Absorption/physiology , Trypsin/metabolism , Cell Culture Techniques , Cell Line , Colitis, Ulcerative/etiology , Colitis, Ulcerative/metabolism , Crohn Disease/etiology , Crohn Disease/metabolism , Humans , Organoids , Thapsigargin
18.
J Crohns Colitis ; 14(8): 1149-1161, 2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32090263

ABSTRACT

Crohn's disease [CD] and ulcerative colitis [UC] are the two main forms of inflammatory bowel disease [IBD]. Previous studies reported increased levels of proteolytic activity in stool and tissue samples from IBD patients, whereas the re-establishment of the proteolytic balance abrogates the development of experimental colitis. Furthermore, recent data suggest that IBD occurs in genetically predisposed individuals who develop an abnormal immune response to intestinal microbes once exposed to environmental triggers. In this review, we highlight the role of proteases in IBD pathophysiology, and we showcase how the main cellular pathways associated with IBD influence proteolytic unbalance and how functional proteomics are allowing the unambiguous identification of dysregulated proteases in IBD, paving the way to the development of new protease inhibitors as a new potential treatment.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Immunity, Mucosal/genetics , Peptide Hydrolases/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Crohn Disease/drug therapy , Crohn Disease/enzymology , Crohn Disease/genetics , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Pharmacogenetics , Protease Inhibitors/pharmacology , Risk Factors
19.
Curr Opin Pharmacol ; 54: 51-58, 2020 10.
Article in English | MEDLINE | ID: mdl-32947075

ABSTRACT

Moderate to severe inflammatory bowel disease patients can fail to respond to conventional therapy and/or to biologic treatment. In the era of TNFα antagonists and other non-anti-TNF biologic drugs, it is important to review the literature on biologic treatment failure, which could be defined as primary non-response, secondary loss of response and intolerance. Therapeutic drug monitoring (TDM), that is, drug trough level and antidrug antibodies, should enable to determine the mechanisms of treatment failure and to optimize drug efficacy. There is a consensus on reactive TDM at the time of loss of response. Proactive TDM could be of interest during induction and/or maintenance, but randomized controlled trials are required.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biological Products/therapeutic use , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ustekinumab/therapeutic use , Drug Monitoring , Drug Resistance , Humans , Treatment Failure
20.
J Crohns Colitis ; 14(5): 669-679, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-31784737

ABSTRACT

BACKGROUND AND AIMS: Nucleotide oligomerization domain 2 [NOD2] mutations are key risk factors for Crohn's disease [CD]. NOD2 contributes to intestinal homeostasis by regulating innate and adaptive immunity together with intestinal epithelial function. However, the exact roles of NOD2 in CD and other NOD2-associated disorders remain poorly known. METHODS: We initially observed that NOD2 expression was increased in epithelial cells away from inflamed areas in CD patients. To explore this finding, Nod2 mRNA expression, inflammation, and cytokines expression were examined in the small bowel of wild-type [WT], Nod2 knockout and Nod2 mutant mice after rectal instillation of 2,4,6-trinitrobenzene sulphonic acid [TNBS]. RESULTS: In WT mice, Nod2 upregulation upstream to rectal injury was associated with pro-inflammatory cytokine expression but no overt histological inflammatory lesions. Conversely, in Nod2-deficient mice the inflammation spread from colitis to ileum and duodenum. CONCLUSIONS: Nod2 protects the gut from colitis spreading to small intestine.


Subject(s)
Colitis/genetics , Duodenitis/genetics , Ileitis/genetics , Intestinal Mucosa/metabolism , Nod2 Signaling Adaptor Protein/genetics , RNA, Messenger/metabolism , Animals , Cecum/metabolism , Cecum/pathology , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Duodenitis/chemically induced , Duodenitis/metabolism , Duodenitis/pathology , Duodenum/metabolism , Duodenum/pathology , Gene Expression , Humans , Ileitis/chemically induced , Ileitis/metabolism , Ileitis/pathology , Ileum/metabolism , Ileum/pathology , Interferon-gamma/metabolism , Interleukin-12/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Nod2 Signaling Adaptor Protein/metabolism , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL