Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Obes Surg ; 33(12): 4017-4025, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37924465

ABSTRACT

INTRODUCTION: Obesity is associated with low-grade inflammation, including intestinal inflammation based on fecal or serum calprotectin (FC-SC) measurement. Roux-en-Y gastric bypass (RYGB) improves obesity-related parameters. However, the association between FC-SC levels and postoperative course and the link with metabolic and inflammatory phenotypes before and after RYGB remains unclear. METHODS: We determined SC levels in 48 patients before (T0) and 6 months after (T6M) RYGB. We then analyzed postoperative changes in FC-SC levels and the relationship with inflammation and metabolic status. RESULTS: Twenty-three patients (48%) had elevated SC levels (˃2.9 µg/mL) at T0 and T6M. Six of 29 patients (20.7%) had elevated FC concentrations (>50 µg/g) at T0 vs. 16 of 17 patients (94.1%) at T6M (p=0.006). At T0, FC levels correlated with BMI (Rho=0.63; p=0.001) and systemic inflammation (CRP: Rho=0.66, p=0.0006; IL-6: Rho=0.48, p=0.03; haptoglobin: Rho=0.75; p= 0.0006). SC tended to be positively associated with triglyceride levels (Rho=0.34; p=0.08), BMI (Rho=0.34; p=0.08), and inflammatory markers (CRP: Rho=0.33; p=0.09; IL-6: Rho=0.36; p=0.06). FC levels were associated with increased jejunal IL-17+CD8+ T-cell densities (Rho:0.90; p=0.0002). FC and SC were correlated together at T0 (Rho=0.83; p<0.001) but not at T6M. At T6M, SC decreased by 53.6%, whereas FC increased by 79.7%. SC and FC were not associated with any of the variables studied at T6M. CONCLUSION: FC is a surrogate marker of systemic and intestinal inflammation and adiposity, whereas SC only tends to correlate with systemic inflammation. At 6 months after RYGB, SC-based systemic inflammation decreased, whereas FC-based intestinal inflammation increased. FC and SC levels follow different trajectories and are unrelated to improvements following bariatric surgery.


Subject(s)
Gastric Bypass , Obesity, Morbid , Humans , Obesity, Morbid/surgery , Leukocyte L1 Antigen Complex , Prospective Studies , Interleukin-6 , Obesity/surgery , Inflammation
2.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36142548

ABSTRACT

The importance of gut barrier integrity in intestinal homeostasis and the consequences of its alteration in the etiology of human pathologies have been subjects of exponentially growing interest during the last decade [...].


Subject(s)
Intestinal Mucosa , Homeostasis/physiology , Humans
3.
Sci Rep ; 12(1): 9440, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35676403

ABSTRACT

In the gut ecosystem, microorganisms regulate group behaviour and interplay with the host via a molecular system called quorum sensing (QS). The QS molecule 3-oxo-C12:2-HSL, first identified in human gut microbiota, exerts anti-inflammatory effects and could play a role in inflammatory bowel diseases where dysbiosis has been described. Our aim was to identify which signalling pathways are involved in this effect. We observed that 3-oxo-C12:2-HSL decreases expression of pro-inflammatory cytokines such as Interleukine-1ß (- 35%) and Tumor Necrosis Factor-α (TNFα) (- 40%) by stimulated immune RAW264.7 cells and decreased TNF secretion by stimulated PBMC in a dose-dependent manner, between 25 to 100 µM. Transcriptomic analysis of RAW264.7 cells exposed to 3-oxo-C12:2-HSL, in a pro-inflammatory context, highlighted JAK-STAT, NF-κB and TFN signalling pathways and we confirmed that 3-oxo-C12:2-HSL inhibited JAK1 and STAT1 phosphorylation. We also showed through a screening assay that 3-oxo-C12:2-HSL interacted with several human bitter taste receptors. Its anti-inflammatory effect involved TAS2R38 as shown by pharmacologic inhibition and led to an increase in intracellular calcium levels. We thus unravelled the involvement of several cellular pathways in the anti-inflammatory effects exerted by the QS molecule 3-oxo-C12:2-HSL.


Subject(s)
Gastrointestinal Microbiome , Quorum Sensing , 4-Butyrolactone/metabolism , Anti-Inflammatory Agents/metabolism , Ecosystem , Homoserine/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Pseudomonas aeruginosa/physiology , Taste
4.
World J Gastroenterol ; 27(42): 7247-7270, 2021 Nov 14.
Article in English | MEDLINE | ID: mdl-34876787

ABSTRACT

Bacteria are known to communicate with each other and regulate their activities in social networks by secreting and sensing signaling molecules called autoinducers, a process known as quorum sensing (QS). This is a growing area of research in which we are expanding our understanding of how bacteria collectively modify their behavior but are also involved in the crosstalk between the host and gut microbiome. This is particularly relevant in the case of pathologies associated with dysbiosis or disorders of the intestinal ecosystem. This review will examine the different QS systems and the evidence for their presence in the intestinal ecosystem. We will also provide clues on the role of QS molecules that may exert, directly or indirectly through their bacterial gossip, an influence on intestinal epithelial barrier function, intestinal inflammation, and intestinal carcinogenesis. This review aims to provide evidence on the role of QS molecules in gut physiology and the potential shared by this new player. Better understanding the impact of intestinal bacterial social networks and ultimately developing new therapeutic strategies to control intestinal disorders remains a challenge that needs to be addressed in the future.


Subject(s)
Gastrointestinal Microbiome , Quorum Sensing , Bacteria , Dysbiosis , Ecosystem , Humans
5.
Methods Mol Biol ; 2367: 13-26, 2021.
Article in English | MEDLINE | ID: mdl-33730353

ABSTRACT

Paracellular permeability of the intestinal epithelium is a feature of the intestinal barrier, which plays an important role in the physiology of gut and the whole organism. Intestinal paracellular permeability is controlled by complex processes and is involved in the passage of ions and fluids (called pore pathway) and macromolecules (called leak pathway) through tight junctions, which seal the intercellular space. Impairment of intestinal paracellular permeability is associated with several diseases. The identification of a defect in intestinal paracellular permeability may help to understand the implication of gut barrier as a cause or a consequence in human pathology. Here we describe two complementary methods to evaluate alteration of paracellular permeability in cell culture, using the human intestinal cell line Caco-2 and its clone Caco-2/TC7.


Subject(s)
Enterocytes , Caco-2 Cells , Cell Membrane Permeability , Cellulose, Oxidized , Humans , Intestinal Mucosa/metabolism , Permeability , Tight Junctions/metabolism
6.
Methods Mol Biol ; 2367: 1-11, 2021.
Article in English | MEDLINE | ID: mdl-33733391

ABSTRACT

An increased intestinal permeability has been described in many diseases including inflammatory bowel disease and metabolic disorders, and a better understanding of the contribution of intestinal barrier impairment to pathogenesis is needed. In recent years, attention has been paid to the leak pathway, which is the route of paracellular transport allowing the diffusion of macromolecules through the tight junctions of the intestinal epithelial lining. While the passage of macromolecules by this pathway is very restricted under physiological conditions, its amplification is thought to promote an excessive immune activation in the intestinal mucosa. The Ussing chambers have been widely used to measure both active and passive transepithelial fluxes in intact tissues. In this chapter we present how this simple device can be used to measure paracellular permeability to macromolecules in the mouse intestine. We propose a detailed protocol and describe how to best exploit all the possibilities of this technique, correctly interpret the results, and avoid the main pitfalls.


Subject(s)
Intestines , Animals , Colitis , Intestinal Mucosa , Macromolecular Substances , Mice , Permeability , Tight Junctions
7.
Tissue Barriers ; 8(4): 1832877, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33100129

ABSTRACT

The intestine is home to the largest microbiota community of the human body and strictly regulates its barrier function. Tight junctions (TJ) are major actors of the intestinal barrier, which is impaired in inflammatory bowel disease (IBD), along with an unbalanced microbiota composition. With the aim to identify new actors involved in host-microbiota interplay in IBD, we studied N-acyl homoserine lactones (AHL), molecules of the bacterial quorum sensing, which also impact the host. We previously identified in the gut a new and prominent AHL, 3-oxo-C12:2, which is lost in IBD. We investigated how 3-oxo-C12:2 impacts the intestinal barrier function, in comparison to 3-oxo-C12, a structurally close AHL produced by the opportunistic pathogen P. aeruginosa. Using Caco-2/TC7 cells as a model of polarized enterocytes, we compared the effects on paracellular permeability and TJ integrity of these two AHL, separately or combined with pro-inflammatory cytokines, Interferon-γ and Tumor Necrosis Factor-α, known to disrupt the barrier function during IBD. While 3-oxo-C12 increased paracellular permeability and decreased occludin and tricellulin signal at bicellular and tricellular TJ, respectively, 3-oxo-C12:2 modified neither permeability nor TJ integrity. Whereas 3-oxo-C12 potentiated the hyperpermeability induced by cytokines, 3-oxo-C12:2 attenuated their deleterious effects on occludin and tricellulin, and maintained their interaction with their partner ZO-1. In addition, 3-oxo-C12:2 limited the cytokine-induced ubiquitination of occludin and tricellulin, suggesting that this AHL prevented their endocytosis. In conclusion, the role of 3-oxo-C12:2 in maintaining TJ integrity under inflammatory conditions identifies this new AHL as a potential beneficial actor of host-microbiota interactions in IBD.


Subject(s)
Acyl-Butyrolactones/metabolism , Cytokines/metabolism , Quorum Sensing/genetics , Tight Junctions/metabolism , Humans
8.
Article in English | MEDLINE | ID: mdl-31647994

ABSTRACT

The mechanisms leading to the low-grade inflammation observed during obesity are not fully understood. Seeking the initiating events, we tested the hypothesis that the intestine could be damaged by repeated lipid supply and therefore participate in inflammation. In mice, 1-5 palm oil gavages increased intestinal permeability via decreased expression and mislocalization of junctional proteins at the cell-cell contacts; altered the intestinal bacterial species by decreasing the abundance of Akkermansia muciniphila, segmented filamentous bacteria, and Clostridium leptum; and increased inflammatory cytokine expression. This was further studied in human intestinal epithelial Caco-2/TC7 cells using the two main components of palm oil, i.e., palmitic and oleic acid. Saturated palmitic acid impaired paracellular permeability and junctional protein localization, and induced inflammatory cytokine expression in the cells, but unsaturated oleic acid did not. Inhibiting de novo ceramide synthesis prevented part of these effects. Altogether, our data show that short exposure to palm oil or palmitic acid induces intestinal dysfunctions targeting barrier integrity and inflammation. Excessive palm oil consumption could be an early player in the gut alterations observed in metabolic diseases.


Subject(s)
Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Metabolic Syndrome/pathology , Palm Oil/adverse effects , Palmitic Acid/adverse effects , Administration, Oral , Animals , Caco-2 Cells , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Metabolic Syndrome/immunology , Mice , Palm Oil/administration & dosage , Palm Oil/chemistry , Palmitic Acid/administration & dosage , Permeability , Tight Junctions/drug effects
9.
Nutr Rev ; 77(3): 129-143, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30517714

ABSTRACT

The plasticity of a material corresponds to its capacity to change its feature under the effect of an external action. Intestinal plasticity could be defined as the ability of the intestine to modify its size or thickness and intestinal cells to modulate their absorption and secretion functions in response to external or internal cues/signals. This review will focus on intestinal adaptation mechanisms in response to diet and nutritional status. These physiological mechanisms allow a fine and rapid adaptation of the gut to promote absorption of ingested food, but they can also lead to obesity in response to overnutrition. This plasticity could thus become a therapeutic target to treat not only undernutrition but also obesity. How the intestine adapts in response to 2 types of surgical remodeling of the digestive tract-extensive bowel resection leading to intestinal failure and surgical treatment of pathological obesity (ie, bariatric surgeries)-will also be reviewed.


Subject(s)
Adaptation, Physiological , Diet , Digestive System Surgical Procedures , Intestines/physiology , Nutritional Status , Animals , Female , Humans , Intestinal Absorption , Intestines/surgery , Male
10.
PLoS One ; 13(8): e0202587, 2018.
Article in English | MEDLINE | ID: mdl-30157234

ABSTRACT

BACKGROUND AND AIMS: N-acyl homoserine lactones (AHLs), which are autoinducer quorum-sensing molecules involved in the bacterial communication network, also interact with eukaryotic cells. Searching for these molecules in the context of inflammatory bowel disease (IBD) is appealing. The aims of our study were to look for AHL molecules in faecal samples from healthy subjects (HS) and IBD patients to correlate AHL profiles with the microbiome and investigate the effect of AHLs of interest on epithelial cells. METHODS: Using mass spectrometry, we characterised AHL profiles in faecal samples from HS (n = 26) and IBD patients in remission (n = 24) and in flare (n = 25) and correlated the presence of AHLs of interest with gut microbiota composition obtained by real-time qPCR and 16S sequencing. We synthesised AHLs of interest to test the inflammatory response after IL1ß stimulation and paracellular permeability on Caco-2 cells. RESULTS: We observed 14 different AHLs, among which one was prominent. This AHL corresponded to 3-oxo-C12:2 and was found significantly less frequently in IBD patients in flare (16%) and in remission (37.5%) versus HS (65.4%) (p = 0.001). The presence of 3-oxo-C12:2 was associated with significantly higher counts of Firmicutes, especially Faecalbacterium prausnitzii, and lower counts of Escherichia coli. In vitro, 3-oxo-C12:2 exerted an anti-inflammatory effect on Caco-2 cells. Interestingly, although 3-oxo-C12, the well-known AHL from Pseudomonas aeruginosa, increased paracellular permeability, 3-oxo-C12:2 did not. CONCLUSIONS: We identified AHLs in the human gut microbiota and discovered a new and prominent AHL, 3-oxo-C12:2, which correlates with normobiosis and exerts a protective effect on gut epithelial cells.


Subject(s)
Acyl-Butyrolactones/isolation & purification , Gastrointestinal Microbiome/genetics , Inflammatory Bowel Diseases/microbiology , Quorum Sensing/genetics , Acyl-Butyrolactones/chemistry , Acyl-Butyrolactones/metabolism , Caco-2 Cells , Cell Communication/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Feces/microbiology , Gene Expression Regulation, Bacterial , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Signal Transduction
11.
J Pathol ; 246(2): 217-230, 2018 10.
Article in English | MEDLINE | ID: mdl-29984492

ABSTRACT

Obesity and its metabolic complications are characterized by subclinical systemic and tissue inflammation. In rodent models of obesity, inflammation and metabolic impairments are linked with intestinal barrier damage. However, whether intestinal permeability is altered in human obesity remains to be investigated. In a cohort of 122 severely obese and non-obese patients, we analyzed intestinal barrier function combining in vivo and ex vivo investigations. We found tight junction impairments in the jejunal epithelium of obese patients, evidenced by a reduction of occludin and tricellulin. Serum levels of zonulin and LPS binding protein, two markers usually associated with intestinal barrier alterations, were also increased in obese patients. Intestinal permeability per se was assessed in vivo by quantification of urinary lactitol/mannitol (L/M) and measured directly ex vivo on jejunal samples in Ussing chambers. In the fasting condition, L/M ratio and jejunal permeability were not significantly different between obese and non-obese patients, but high jejunal permeability to small molecules (0.4 kDa) was associated with systemic inflammation within the obese cohort. Altogether, these results suggest that intestinal barrier function is subtly compromised in obese patients. We thus tested whether this barrier impairment could be exacerbated by dietary lipids. To this end, we challenged jejunal samples with lipid micelles and showed that a single exposure increased permeability to macromolecules (4 kDa). Jejunal permeability after the lipid load was two-fold higher in obese patients compared to non-obese controls and correlated with systemic and intestinal inflammation. Moreover, lipid-induced permeability was an explicative variable of type 2 diabetes. In conclusion, intestinal barrier defects are present in human severe obesity and exacerbated by a lipid challenge. This paves the way to the development of novel therapeutic approaches to modulate intestinal barrier function or personalize nutrition therapy to decrease lipid-induced jejunal leakage in metabolic diseases. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Inflammation/metabolism , Intestinal Absorption/drug effects , Jejunum/drug effects , Lipids/administration & dosage , Obesity/metabolism , Acute-Phase Proteins , Adult , Aged , Caco-2 Cells , Carrier Proteins/blood , Case-Control Studies , Cholera Toxin/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Female , Haptoglobins , Humans , Inflammation/complications , Inflammation/physiopathology , Jejunum/metabolism , Jejunum/physiopathology , MARVEL Domain Containing 2 Protein/metabolism , Male , Membrane Glycoproteins/blood , Micelles , Middle Aged , Obesity/complications , Obesity/physiopathology , Occludin/metabolism , Permeability , Protein Precursors , Tight Junctions/metabolism , Young Adult
12.
Radiother Oncol ; 120(1): 175-83, 2016 07.
Article in English | MEDLINE | ID: mdl-27406443

ABSTRACT

BACKGROUND & AIM: Despite extensive study of the contribution of cell death and apoptosis to radiation-induced acute intestinal injury, our knowledge of the signaling mechanisms involved in epithelial barrier dysfunction remains inadequate. Because PrP(c) plays a key role in intestinal homeostasis by renewing epithelia, we sought to study its role in epithelial barrier function after irradiation. DESIGN: Histology, morphometry and plasma FD-4 levels were used to examine ileal architecture, wound healing, and intestinal leakage in PrP(c)-deficient (KO) and wild-type (WT) mice after total-body irradiation. Impairment of the PrP(c) Src pathway after irradiation was explored by immunofluorescence and confocal microscopy, with Caco-2/Tc7 cells. Lastly, dasatinib treatment was used to switch off the Src pathway in vitro and in vivo. RESULTS: The decrease in radiation-induced lethality, improved intestinal wound healing, and reduced intestinal leakage promoted by PrP(c) deficiency demonstrate its involvement in acute intestinal damage. Irradiation of Cacao2/Tc7 cells induced PrP(c) to target the nuclei associated with Src activation. Finally, the protective effect triggered by dasatinib confirmed Src involvement in radiation-induced acute intestinal toxicity. CONCLUSION: Our data are the first to show a role for the PrP(c)-Src pathway in acute intestinal response to radiation injury and offer a novel therapeutic opportunity.


Subject(s)
Dasatinib/therapeutic use , Intestines/radiation effects , Prion Proteins/deficiency , Radiation Injuries/prevention & control , src-Family Kinases/antagonists & inhibitors , Animals , CSK Tyrosine-Protein Kinase , Caco-2 Cells , Humans , Mice , Mice, Inbred C57BL , Prion Proteins/physiology , Whole-Body Irradiation , src-Family Kinases/physiology
13.
Prion ; 10(2): 143-52, 2016 03 03.
Article in English | MEDLINE | ID: mdl-27216988

ABSTRACT

The cellular prion protein PrP(c) plays important roles in proliferation, cell death and survival, differentiation and adhesion. The participation of PrP(c) in tumor growth and metastasis was pointed out, but the underlying mechanisms were not deciphered completely. In the constantly renewing intestinal epithelium, our group demonstrated a dual localization of PrP(c), which is targeted to cell-cell junctions in interaction with Src kinase and desmosomal proteins in differentiated enterocytes, but is predominantly nuclear in dividing cells. While the role of PrP(c) in the dynamics of intercellular junctions was confirmed in other biological systems, we unraveled its function in the nucleus only recently. We identified several nuclear PrP(c) partners, which comprise γ-catenin, one of its desmosomal partners, ß-catenin and TCF7L2, the main effectors of the canonical Wnt pathway, and YAP, one effector of the Hippo pathway. PrP(c) up-regulates the activity of the ß-catenin/TCF7L2 complex and its invalidation impairs the proliferation of intestinal progenitors. We discuss how PrP(c) could participate to oncogenic processes through its interaction with Wnt and Hippo pathway effectors, which are controlled by cell-cell junctions and Src family kinases and dysregulated during tumorigenesis. This highlights new potential mechanisms that connect PrP(c) expression and subcellular redistribution to cancer.


Subject(s)
Cell Nucleus/pathology , Intercellular Junctions/pathology , Neoplasms/pathology , PrPC Proteins/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Cell Proliferation , Desmosomes/metabolism , Desmosomes/pathology , Epithelial-Mesenchymal Transition , Hippo Signaling Pathway , Humans , Intercellular Junctions/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Neoplasms/metabolism , PrPC Proteins/analysis , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway
14.
Med Sci (Paris) ; 32(5): 461-9, 2016 May.
Article in French | MEDLINE | ID: mdl-27225918

ABSTRACT

The increasing incidence of obesity and associated metabolic complications is a worldwide public health issue. The role of the gut in the pathophysiology of obesity, with an important part for microbiota, is becoming obvious. In rodent models of diet-induced obesity, the modifications of gut microbiota are associated with an alteration of the intestinal permeability increasing the passage of food or bacterial antigens, which contribute to low-grade inflammation and insulin resistance. In human obesity, intestinal permeability modification, and its role in the crosstalk between gut microbiota changes and inflammation at systemic and tissular levels, are still poorly documented. Hence, further characterization of the triggering mechanisms of such inflammatory responses in obese subjects could enable the development of personalized intervention strategies that will help to reduce the risk of obesity-associated diseases.


Subject(s)
Dysbiosis/complications , Gastrointestinal Microbiome/physiology , Inflammation/etiology , Intestinal Mucosa/metabolism , Obesity/etiology , Animals , Dysbiosis/immunology , Dysbiosis/metabolism , Humans , Inflammation/metabolism , Inflammation/microbiology , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Obesity/immunology , Obesity/metabolism , Obesity/microbiology , Permeability
15.
Mol Biol Cell ; 26(18): 3313-28, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26224313

ABSTRACT

We reported previously that the cellular prion protein (PrP(c)) is a component of desmosomes and contributes to the intestinal barrier function. We demonstrated also the presence of PrP(c) in the nucleus of proliferating intestinal epithelial cells. Here we sought to decipher the function of this nuclear pool. In human intestinal cancer cells Caco-2/TC7 and SW480 and normal crypt-like HIEC-6 cells, PrP(c) interacts, in cytoplasm and nucleus, with γ-catenin, one of its desmosomal partners, and with ß-catenin and TCF7L2, effectors of the canonical Wnt pathway. PrP(c) up-regulates the transcriptional activity of the ß-catenin/TCF7L2 complex, whereas γ-catenin down-regulates it. Silencing of PrP(c) results in the modulation of several Wnt target gene expressions in human cells, with different effects depending on their Wnt signaling status, and in mouse intestinal crypt cells in vivo. PrP(c) also interacts with the Hippo pathway effector YAP, suggesting that it may contribute to the regulation of gene transcription beyond the ß-catenin/TCF7L2 complex. Finally, we demonstrate that PrP(c) is required for proper formation of intestinal organoids, indicating that it contributes to proliferation and survival of intestinal progenitors. In conclusion, PrP(c) must be considered as a new modulator of the Wnt signaling pathway in proliferating intestinal epithelial cells.


Subject(s)
Intestinal Mucosa/metabolism , PrPC Proteins/metabolism , Wnt Signaling Pathway , Animals , COS Cells , Caco-2 Cells , Catenins/metabolism , Cell Proliferation/genetics , Chlorocebus aethiops , Down-Regulation , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Prions/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation , beta Catenin/metabolism
16.
Tissue Barriers ; 1(2): e24377, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-24665391

ABSTRACT

The cellular prion protein was historically characterized owing to its misfolding in prion disease. Although its physiological role remains incompletely understood, PrP(C) has emerged as an evolutionary conserved, multifaceted protein involved in a wide-range of biological processes. PrP(C) is a GPI-anchored protein targeted to the plasma membrane, in raft microdomains, where its interaction with a repertoire of binding partners, which differ depending on cell models, mediates its functions. Among identified PrP(C) partners are cell adhesion molecules. This review will focus on the multiple implications of PrP(C) in cell adhesion processes, mainly the regulation of cell-cell junctions in epithelial and endothelial cells and the consequences on barrier properties. We will show how recent findings argue for a role of PrP(C) in the recruitment of signaling molecules, which in turn control the targeting or the stability of adhesion complexes at the plasma membrane.

17.
J Clin Invest ; 122(6): 2239-51, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22565313

ABSTRACT

Intestinal barrier function requires intricate cooperation between intestinal epithelial cells and immune cells. Enteropathogens are able to invade the intestinal lymphoid tissue known as Peyer's patches (PPs) and disrupt the integrity of the intestinal barrier. However, the underlying molecular mechanisms of this process are poorly understood. In mice infected with Yersinia pseudotuberculosis, we found that PP barrier dysfunction is dependent on the Yersinia virulence plasmid and the expression of TLR-2 by hematopoietic cells, but not by intestinal epithelial cells. Upon TLR-2 stimulation, Y. pseudotuberculosis-infected monocytes activated caspase-1 and produced IL-1ß. In turn, IL-1ß increased NF-κB and myosin light chain kinase activation in intestinal epithelial cells, thus disrupting the intestinal barrier by opening the tight junctions. Therefore, Y. pseudotuberculosis subverts intestinal barrier function by altering the interplay between immune and epithelial cells during infection.


Subject(s)
Hematopoietic Stem Cells/immunology , Intestinal Mucosa/immunology , Peyer's Patches/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/immunology , Animals , Caco-2 Cells , Caspase 1/genetics , Caspase 1/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/microbiology , Monocytes/pathology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Peyer's Patches/microbiology , Peyer's Patches/pathology , Signal Transduction/genetics , Toll-Like Receptor 2/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
18.
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
19.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G235-44, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056766

ABSTRACT

Enterocytes of the intestinal epithelium are continually regenerated. They arise from precursor cells in crypts, migrate along villi, and finally die, 3-4 days later, when they reach the villus apex. Their death is thought to occur by anoikis, a form of apoptosis induced by cell detachment, but the mechanism of this process remains poorly understood. We have previously shown that a key event in the onset of anoikis in normal enterocytes detached from the basal lamina is the disruption of adherens junctions mediated by E-cadherin (Fouquet S, Lugo-Martinez VH, Faussat AM, Renaud F, Cardot P, Chambaz J, Pincon-Raymond M, Thenet S. J Biol Chem 279: 43061-43069, 2004). Here we have further investigated the mechanisms underlying this disassembly of the adherens junctions. We show that disruption of the junctions occurs through endocytosis of E-cadherin and that this process depends on the tyrosine-kinase activity of the epidermal growth factor receptor (EGFR). Activation of EGFR was detected in detached enterocytes before E-cadherin disappearance. Specific inhibition of EGFR by tyrphostin AG-1478 maintained E-cadherin and its cytoplasmic partners beta- and alpha-catenin at cell-cell contacts and decreased anoikis. Finally, EGFR activation was evidenced in the intestinal epithelium in vivo, in rare individual cells, which were shown to lose their interactions with the basal lamina. We conclude that EGFR is activated as enterocytes become detached from the basal lamina, and that this mechanism contributes to the disruption of E-cadherin-dependent junctions leading to anoikis. This suggests that EGFR participates in the physiological elimination of the enterocytes.


Subject(s)
Anoikis , Cadherins/metabolism , Cell Adhesion , Enterocytes/metabolism , ErbB Receptors/metabolism , Intestine, Small/metabolism , Tight Junctions/metabolism , Animals , Anoikis/drug effects , Cell Adhesion/drug effects , Endocytosis , Enterocytes/drug effects , Enterocytes/pathology , ErbB Receptors/antagonists & inhibitors , Intestine, Small/drug effects , Intestine, Small/pathology , Mice , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Quinazolines , Tight Junctions/drug effects , Tight Junctions/pathology , Tyrphostins/pharmacology , alpha Catenin/metabolism , beta Catenin/metabolism
20.
PLoS One ; 3(8): e3000, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18714380

ABSTRACT

BACKGROUND: The physiological function of the ubiquitous cellular prion protein, PrP(c), is still under debate. It was essentially studied in nervous system, but poorly investigated in epithelial cells. We previously reported that PrP(c) is targeted to cell-cell junctions of polarized epithelial cells, where it interacts with c-Src. METHODOLOGY/FINDINGS: We show here that, in cultured human enterocytes and in intestine in vivo, the mature PrP(c) is differentially targeted either to the nucleus in dividing cells or to cell-cell contacts in polarized/differentiated cells. By proteomic analysis, we demonstrate that the junctional PrP(c) interacts with cytoskeleton-associated proteins, such as gamma- and beta-actin, alpha-spectrin, annexin A2, and with the desmosome-associated proteins desmoglein, plakoglobin and desmoplakin. In addition, co-immunoprecipitation experiments revealed complexes associating PrP(c), desmoglein and c-Src in raft domains. Through siRNA strategy, we show that PrP(c) is necessary to complete the process of epithelial cell proliferation and for the sub-cellular distribution of proteins involved in cell architecture and junctions. Moreover, analysis of the architecture of the intestinal epithelium of PrP(c) knock-out mice revealed a net decrease in the size of desmosomal junctions and, without change in the amount of BrdU incorporation, a shortening of the length of intestinal villi. CONCLUSIONS/SIGNIFICANCE: From these results, PrP(c) could be considered as a new partner involved in the balance between proliferation and polarization/differentiation in epithelial cells.


Subject(s)
Cell Division/physiology , Epithelial Cells/cytology , Intercellular Junctions/physiology , PrPC Proteins/physiology , Caco-2 Cells , Cell Polarity , Epithelial Cells/physiology , Humans , Lipids/pharmacology , Plasmids , PrPC Proteins/genetics , RNA, Small Interfering/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...