Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
World J Gastroenterol ; 30(16): 2258-2271, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38690023

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is one of the most frequent and debilitating conditions leading to gastroenterological referrals. However, recommended treatments remain limited, yielding only limited therapeutic gains. Chitin-glucan (CG) is a novel dietary prebiotic classically used in humans at a dosage of 1.5-3.0 g/d and is considered a safe food ingredient by the European Food Safety Authority. To provide an alternative approach to managing patients with IBS, we performed preclinical molecular, cellular, and animal studies to evaluate the role of chitin-glucan in the main pathophysiological mechanisms involved in IBS. AIM: To evaluate the roles of CG in visceral analgesia, intestinal inflammation, barrier function, and to develop computational molecular models. METHODS: Visceral pain was recorded through colorectal distension (CRD) in a model of long-lasting colon hypersensitivity induced by an intra-rectal administration of TNBS [15 milligrams (mg)/kilogram (kg)] in 33 Sprague-Dawley rats. Intracolonic pressure was regularly assessed during the 9 wk-experiment (weeks 0, 3, 5, and 7) in animals receiving CG (n = 14) at a human equivalent dose (HED) of 1.5 g/d or 3.0 g/d and compared to negative control (tap water, n = 11) and positive control (phloroglucinol at 1.5 g/d HED, n = 8) groups. The anti-inflammatory effect of CG was evaluated using clinical and histological scores in 30 C57bl6 male mice with colitis induced by dextran sodium sulfate (DSS) administered in their drinking water during 14 d. HT-29 cells under basal conditions and after stimulation with lipopolysaccharide (LPS) were treated with CG to evaluate changes in pathways related to analgesia (µ-opioid receptor (MOR), cannabinoid receptor 2 (CB2), peroxisome proliferator-activated receptor alpha, inflammation [interleukin (IL)-10, IL-1b, and IL-8] and barrier function [mucin 2-5AC, claudin-2, zonula occludens (ZO)-1, ZO-2] using the real-time PCR method. Molecular modelling of CG, LPS, lipoteichoic acid (LTA), and phospholipomannan (PLM) was developed, and the ability of CG to chelate microbial pathogenic lipids was evaluated by docking and molecular dynamics simulations. Data were expressed as the mean ± SEM. RESULTS: Daily CG orally-administered to rats or mice was well tolerated without including diarrhea, visceral hypersensitivity, or inflammation, as evaluated at histological and molecular levels. In a model of CRD, CG at a dosage of 3 g/d HED significantly decreased visceral pain perception by 14% after 2 wk of administration (P < 0.01) and reduced inflammation intensity by 50%, resulting in complete regeneration of the colonic mucosa in mice with DSS-induced colitis. To better reproduce the characteristics of visceral pain in patients with IBS, we then measured the therapeutic impact of CG in rats with TNBS-induced inflammation to long-lasting visceral hypersensitivity. CG at a dosage of 1.5 g/d HED decreased visceral pain perception by 20% five weeks after colitis induction (P < 0.01). When the CG dosage was increased to 3.0 g/d HED, this analgesic effect surpassed that of the spasmolytic agent phloroglucinol, manifesting more rapidly within 3 wk and leading to a 50% inhibition of pain perception (P < 0.0001). The underlying molecular mechanisms contributing to these analgesic and anti-inflammatory effects of CG involved, at least in part, a significant induction of MOR, CB2 receptor, and IL-10, as well as a significant decrease in pro-inflammatory cytokines IL-1b and IL-8. CG also significantly upregulated barrier-related genes including muc5AC, claudin-2, and ZO-2. Molecular modelling of CG revealed a new property of the molecule as a chelator of microbial pathogenic lipids, sequestering gram-negative LPS and gram-positive LTA bacterial toxins, as well as PLM in fungi at the lowesr energy conformations. CONCLUSION: CG decreased visceral perception and intestinal inflammation through master gene regulation and direct binding of microbial products, suggesting that CG may constitute a new therapeutic strategy for patients with IBS or IBS-like symptoms.


Subject(s)
Chitin , Colon , Disease Models, Animal , Glucans , Irritable Bowel Syndrome , Rats, Sprague-Dawley , Visceral Pain , Animals , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/physiopathology , Male , Humans , Colon/drug effects , Colon/pathology , Rats , Visceral Pain/drug therapy , Visceral Pain/physiopathology , Visceral Pain/metabolism , Visceral Pain/etiology , Chitin/pharmacology , Glucans/pharmacology , Glucans/administration & dosage , Mice , Prebiotics/administration & dosage , Trinitrobenzenesulfonic Acid/toxicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/physiopathology , Colitis/pathology , HT29 Cells
2.
Int J Mol Sci ; 25(4)2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38396840

ABSTRACT

The therapeutic management of Crohn's disease (CD), a chronic relapsing-remitting inflammatory bowel disease (IBD), is highly challenging. Surgical resection is sometimes a necessary procedure even though it is often associated with postoperative recurrences (PORs). Tofacitinib, an orally active small molecule Janus kinase inhibitor, is an anti-inflammatory drug meant to limit PORs in CD. Whereas bidirectional interactions between the gut microbiota and the relevant IBD drug are crucial, little is known about the impact of tofacitinib on the gut microbiota. The HLA-B27 transgenic rat is a good preclinical model used in IBD research, including for PORs after ileocecal resection (ICR). In the present study, we used shotgun metagenomics to first delineate the baseline composition and determinants of the fecal microbiome of HLA-B27 rats and then to evaluate the distinct impact of either tofacitinib treatment, ileocecal resection or the cumulative effect of both interventions on the gut microbiota in these HLA-B27 rats. The results confirmed that the microbiome of the HLA-B27 rats was fairly different from their wild-type littermates. We demonstrated here that oral treatment with tofacitinib does not affect the gut microbial composition of HLA-B27 rats. Of note, we showed that ICR induced an intense loss of bacterial diversity together with dramatic changes in taxa relative abundances. However, the oral treatment with tofacitinib neither modified the alpha-diversity nor exacerbated significant modifications in bacterial taxa induced by ICR. Collectively, these preclinical data are rather favorable for the use of tofacitinib in combination with ICR to address Crohn's disease management when considering microbiota.


Subject(s)
Crohn Disease , Inflammatory Bowel Diseases , Microbiota , Piperidines , Pyrimidines , Rats , Animals , Crohn Disease/drug therapy , Crohn Disease/surgery , Crohn Disease/complications , Rats, Transgenic , HLA-B27 Antigen , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/complications , Disease Management
3.
World J Gastroenterol ; 29(5): 851-866, 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36816618

ABSTRACT

BACKGROUND: Postoperative recurrence (POR) after ileocecal resection (ICR) affects most Crohn's disease patients within 3-5 years after surgery. Adherent-invasive Escherichia coli (AIEC) typified by the LF82 strain are pathobionts that are frequently detected in POR of Crohn's disease and have a potential role in the early stages of the disease pathogenesis. Saccharomyces cerevisiae CNCM I-3856 is a probiotic yeast reported to inhibit AIEC adhesion to intestinal epithelial cells and to favor their elimination from the gut. AIM: To evaluate the efficacy of CNCM I-3856 in preventing POR induced by LF82 in an HLA-B27 transgenic (TgB27) rat model. METHODS: Sixty-four rats [strain F344, 38 TgB27, 26 control non-Tg (nTg)] underwent an ICR at the 12th wk (W12) of life and were sacrificed at the 18th wk (W18) of life. TgB27 rats were challenged daily with oral administration of LF82 (109 colony forming units (CFUs)/day (d), n = 8), PBS (n = 5), CNCM I-3856 (109 CFUs/d, n = 7) or a combination of LF82 and CNCM I-3856 (n = 18). nTg rats receiving LF82 (n = 5), PBS (n = 5), CNCM I-3856 (n = 7) or CNCM I-3856 and LF82 (n = 9) under the same conditions were used as controls. POR was analyzed using macroscopic (from 0 to 4) and histologic (from 0 to 6) scores. Luminal LF82 quantifications were performed weekly for each animal. Adherent LF82 and inflammatory/regulatory cytokines were quantified in biopsies at W12 and W18. Data are expressed as the median with the interquartile range. RESULTS: nTg animals did not develop POR. A total of 7/8 (87%) of the TgB27 rats receiving LF82 alone had POR (macroscopic score ≥ 2), which was significantly prevented by CNCM I-3856 administration [6/18 (33%) TgB27 rats, P = 0.01]. Macroscopic lesions were located 2 cm above the anastomosis in the TgB27 rats receiving LF82 alone and consisted of ulcerations with a score of 3.5 (2 - 4). Seven out of 18 TgB27 rats (39%) receiving CNCM I-3856 and LF82 had no macroscopic lesions. Compared to untreated TgB27 animals receiving LF82 alone, coadministration of CNCM I-3856 and LF82 significantly reduced the macroscopic [3.5 (2 - 4) vs 1 (0 - 3), P = 0.002] and histological lesions by more than 50% [4.5 (3.3 - 5.8) vs 2 (1.3 - 3), P = 0.003]. The levels of adherent LF82 were correlated with anastomotic macroscopic scores in TgB27 rats (r = 0.49, P = 0.006), with a higher risk of POR in animals having high levels of luminal LF82 (71.4% vs 25%, P = 0.02). Administration of CNCM I-3856 significantly reduced the levels of luminal and adherent LF82, increased the production of interleukin (IL)-10 and decreased the production of IL-23 and IL-17 in TgB27 rats. CONCLUSION: In a reliable model of POR induced by LF82 in TgB27 rats, CNCM I-3856 prevents macroscopic POR by decreasing LF82 infection and gut inflammation.


Subject(s)
Crohn Disease , Escherichia coli Infections , Rats , Animals , Crohn Disease/pathology , Escherichia coli , Saccharomyces cerevisiae , Rats, Transgenic , HLA-B27 Antigen , Intestinal Mucosa/pathology , Rats, Inbred F344 , Bacterial Adhesion
4.
J Crohns Colitis ; 16(10): 1617-1627, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35997152

ABSTRACT

BACKGROUND AND AIMS: Adherent invasive Escherichia coli [AIEC] are recovered with a high frequency from the gut mucosa of Crohn's disease patients and are believed to contribute to the dysbiosis and pathogenesis of this inflammatory bowel disease. In this context, bacteriophage therapy has been proposed for specifically targeting AIEC in the human gut with no deleterious impact on the commensal microbiota. METHODS: The in vitro efficacy and specificity of a seven lytic phage cocktail [EcoActive™] was assessed against [i] 210 clinical AIEC strains, and [ii] 43 non-E. coli strains belonging to the top 12 most common bacterial genera typically associated with a healthy human microbiome. These data were supported by in vivo safety and efficacy assays conducted on healthy and AIEC-colonized mice, respectively. RESULTS: The EcoActive cocktail was effective in vitro against 95% of the AIEC strains and did not lyse any of the 43 non-E. coli commensal strains, in contrast to conventional antibiotics. Long-term administration of the EcoActive cocktail to healthy mice was safe and did not induce dysbiosis according to metagenomic data. Using a murine model of induced colitis of animals infected with the AIEC strain LF82, we found that a single administration of the cocktail failed to alleviate inflammatory symptoms, while mice receiving the cocktail twice a day for 15 days were protected from clinical and microscopical manifestations of inflammation. CONCLUSIONS: Collectively, the data support the approach of AIEC-targeted phage therapy as safe and effective treatment for reducing AIEC levels in the gut of IBD patients.


Subject(s)
Bacteriophages , Colitis , Animals , Humans , Mice , Bacterial Adhesion , Colitis/pathology , Disease Models, Animal , Dysbiosis/complications , Escherichia coli , Escherichia coli Infections/complications , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Intestinal Mucosa/pathology
5.
PLoS One ; 16(9): e0257281, 2021.
Article in English | MEDLINE | ID: mdl-34529707

ABSTRACT

The development of more effective, better tolerated drug treatments for progressive pulmonary fibrosis (of which idiopathic pulmonary fibrosis is the most common and severe form) is a research priority. The peroxisome proliferator-activated receptor gamma (PPAR-γ) is a key regulator of inflammation and fibrosis and therefore represents a potential therapeutic target. However, the use of synthetic PPAR-γ agonists may be limited by their potentially severe adverse effects. In a mouse model of bleomycin (BLM)-induced pulmonary fibrosis, we have demonstrated that the non-racemic selective PPAR-γ modulator GED-0507 is able to reduce body weight loss, ameliorate clinical and histological features of pulmonary fibrosis, and increase survival rate without any safety concerns. Here, we focused on the biomolecular effects of GED-0507 on various inflammatory/fibrotic pathways. We demonstrated that preventive and therapeutic administration of GED-0507 reduced the BLM-induced mRNA expression of several markers of fibrosis, including transforming growth factor (TGF)-ß, alpha-smooth muscle actin, collagen and fibronectin as well as epithelial-to-mesenchymal transition (EMT) and expression of mucin 5B. The beneficial effect of GED-0507 on pulmonary fibrosis was confirmed in vitro by its ability to control TGFß-induced myofibroblast activation in the A549 human alveolar epithelial cell line, the MRC-5 lung fibroblast line, and primary human lung fibroblasts. Compared with the US Food and Drug Administration-approved antifibrotic drugs pirfenidone and nintedanib, GED-0507 displayed greater antifibrotic activity by controlling alveolar epithelial cell dysfunction, EMT, and extracellular matrix remodeling. In conclusion, GED-0507 demonstrated potent antifibrotic properties and might be a promising drug candidate for the treatment of pulmonary fibrosis.


Subject(s)
Cell Transdifferentiation , Myofibroblasts/cytology , Propionates/pharmacology , Pulmonary Fibrosis/drug therapy , A549 Cells , Animals , Bleomycin , Cell Line , Cell Survival/drug effects , Humans , In Vitro Techniques , Inflammation , Lung/drug effects , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , Pulmonary Fibrosis/physiopathology , Treatment Outcome
7.
Cell Mol Gastroenterol Hepatol ; 7(1): 185-196, 2019.
Article in English | MEDLINE | ID: mdl-30534582

ABSTRACT

Background & Aims: Irritable bowel syndrome (IBS) is a multifactorial disease arising from a complex interplay between genetic predisposition and environmental influences. To date, environmental triggers are not well known. Aluminum is commonly present in food, notably by its use as food additive. We investigated the effects of aluminum ingestion in rodent models of visceral hypersensitivity, and the mechanisms involved. Methods: Visceral hypersensitivity was recorded by colorectal distension in rats administered with oral low doses of aluminum. Inflammation was analyzed in the colon of aluminum-treated rats by quantitative PCR for cytokine expression and by immunohistochemistry for immune cells quantification. Involvement of mast cells in the aluminum-induced hypersensitivity was determined by cromoglycate administration of rats and in mast cell-deficient mice (KitW-sh/W-sh). Proteinase-activated receptor-2 (PAR2) activation in response to aluminum was evaluated and its implication in aluminum-induced hypersensitivity was assessed in PAR2 knockout mice. Results: Orally administered low-dose aluminum induced visceral hypersensitivity in rats and mice. Visceral pain induced by aluminum persisted over time even after cessation of treatment, reappeared and was amplified when treatment resumed. As observed in humans, female animals were more sensitive than males. Major mediators of nociception were up-regulated in the colon by aluminum. Activation of mast cells and PAR2 were required for aluminum-induced hypersensitivity. Conclusions: These findings indicate that oral exposure to aluminum at human dietary level reproduces clinical and molecular features of IBS, highlighting a new pathway of prevention and treatment of visceral pain in some susceptible patients.


Subject(s)
Aluminum/toxicity , Colon/pathology , Hypersensitivity/pathology , Rectum/pathology , Administration, Oral , Aluminum/administration & dosage , Animals , Colon/drug effects , Female , Inflammation/pathology , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Nociception/drug effects , Rats, Sprague-Dawley , Receptor, PAR-2/metabolism , Rectum/drug effects , Visceral Pain/metabolism , Visceral Pain/pathology
8.
Eur J Histochem ; 62(3)2018 Jul 31.
Article in English | MEDLINE | ID: mdl-30064196

ABSTRACT

A concomitant action of multiple profibrotic mediators appears crucial in the development and progression of fibrosis. Sphingosine kinase/sphingosine 1 phosphate and transforming growth factor-ß/Smads pathways are both involved in pathogenesis of fibrosis in several organs by controlling differentiation of fibroblasts to myofibroblasts and the epithelial to-mesenchymal transition. However, their direct involvement in chronic colitis-associated fibrosis it is not yet known. In this study we evaluated the immunohistochemical expression of some proteins implicated in sphingosine kinase/sphingosine 1 phosphate and transforming growth factor-ß/Smads pathways in Dextrane Sodium Sulphate (DSS)-induced colorectal fibrosis in mice. Compared to control mice, DSS-induced chronic colitis mice developed a marked intestinal fibrosis associated with a concomitant overexpression of TGF-ß, p-Smad3, α-SMA, collagen I-III, SPHK1, RhoA, PI3K, Akt, p-Akt, p-mTOR. This study highlights the relationship between the two pathways and the possible role of SPHK1 in the intestinal fibrosis.  These results, if confirmed by in vitro studies, may have important clinical implications in the development of new therapeutical approaches in inflammatory bowel disease.


Subject(s)
Colitis/immunology , Colitis/physiopathology , Intestines/pathology , Lysophospholipids/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Smad3 Protein/physiology , Sphingosine/analogs & derivatives , Transforming Growth Factor beta/physiology , Animals , Disease Models, Animal , Fibrosis/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Reference Standards , Signal Transduction , Sphingosine/physiology
9.
J Bioenerg Biomembr ; 50(3): 213-222, 2018 06.
Article in English | MEDLINE | ID: mdl-29524020

ABSTRACT

The hexosamine biosynthetic pathway (HBP) integrates glucose, amino acids, fatty acids and nucleotides metabolisms for uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) synthesis. UDP-GlcNAc is the nucleotide sugar donor for O-linked ß-N-acetylglucosaminylation (O-GlcNAcylation) processes. O-GlcNAc transferase (OGT) is the enzyme which transfers the N-acetylglucosamine (O-GlcNAc) residue onto target proteins. Several studies previously showed that glucose metabolism dysregulations associated with obesity, diabetes or cancer correlated with an increase of OGT expression and global O-GlcNAcylation levels. Moreover, these diseases present an increased activation of the nutrient sensing mammalian target of rapamycin (mTOR) pathway. Other works demonstrate that mTOR regulates protein O-GlcNAcylation in cancer cells through stabilization of OGT. In this context, we studied the cross-talk between these two metabolic sensors in vivo in obese mice predisposed to diabetes and in vitro in normal and colon cancer cells. We report that levels of OGT and O-GlcNAcylation are increased in obese mice colon tissues and colon cancer cells and are associated with a higher activation of mTOR signaling. In parallel, treatments with mTOR regulators modulate OGT and O-GlcNAcylation levels in both normal and colon cancer cells. However, deregulation of O-GlcNAcylation affects mTOR signaling activation only in cancer cells. Thus, a crosstalk exists between O-GlcNAcylation and mTOR signaling in contexts of metabolism dysregulation associated to obesity or cancer.


Subject(s)
Acetylglucosamine/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Colonic Neoplasms/metabolism , Glycosylation , Mice , Mice, Obese , N-Acetylglucosaminyltransferases/metabolism , Obesity/metabolism , Receptor Cross-Talk
10.
EMBO Mol Med ; 9(11): 1471-1481, 2017 11.
Article in English | MEDLINE | ID: mdl-28947679

ABSTRACT

Lactase (LCT) deficiency affects approximately 75% of the world's adult population and may lead to lactose malabsorption and intolerance. Currently, the regulation of LCT gene expression remains poorly known. Peroxisome proliferator activator receptorγ (PPARγ) is a key player in carbohydrate metabolism. While the intestine is essential for carbohydrate digestion and absorption, the role of PPARγ in enterocyte metabolic functions has been poorly investigated. This study aims at characterizing PPARγ target genes involved in intestinal metabolic functions. In microarray analysis, the LCT gene was the most upregulated by PPARγ agonists in Caco-2 cells. We confirmed that PPARγ agonists were able to increase the expression and activity of LCT both in vitro and in vivo in the proximal small bowel of rodents. The functional response element activated by PPARγ was identified in the promoter of the human LCT gene. PPARγ modulation was able to improve symptoms induced by lactose-enriched diet in weaned rats. Our results demonstrate that PPARγ regulates LCT expression, and suggest that modulating intestinal PPARγ activity might constitute a new therapeutic strategy for lactose malabsorption.


Subject(s)
Intestine, Small/metabolism , Lactase/metabolism , PPAR gamma/metabolism , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Caco-2 Cells , Chromatin Immunoprecipitation , Diet , Humans , Lactase/genetics , Lactose/metabolism , Lactose Intolerance/drug therapy , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mutagenesis, Site-Directed , PPAR gamma/agonists , PPAR gamma/genetics , Phenylpropionates/pharmacology , Phenylpropionates/therapeutic use , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
11.
PLoS One ; 12(2): e0171093, 2017.
Article in English | MEDLINE | ID: mdl-28207769

ABSTRACT

BACKGROUND: Intestinal fibrosis is characterized by abnormal production and deposition of extracellular matrix (ECM) proteins by activated myofibroblasts. The main progenitor cells of activated myofibroblasts are the fibroblasts and the epithelial cells, the latter through the epithelial-mesenchymal transition (EMT). AIM: To evaluate the action of the new PPAR-γ modulator, GED-0507-34 Levo (GED) on the expression of EMT associated and regulatory proteins such as TGF-ß, Smad3, E-cadherin, Snail, ZEB1, ß-catenin, and GSK-3ß, in a mouse model of DSS-induced intestinal fibrosis. METHODS: Chronic colitis and fibrosis were induced by oral administration of 2.5% DSS (w/v) for 6 weeks. GW9662 (GW), a selective PPAR-γ inhibitor, was also administered by intraperitoneal injection at the dose of 1 mg/kg/day combined with GED treatment. All drugs were administered at the beginning of the second cycle of DSS (day 12). 65 mice were randomly divided into five groups (H2O as controls n = 10, H2O+GED n = 10, DSS n = 15, DSS+GED n = 15, DSS+GED+GW n = 15). The colon was excised for macroscopic examination and histological and morphometric analyses. The level of expression of molecules involved in EMT and fibrosis, like TGF-ß, Smad3, E-cadherin, Snail, ZEB1, ß-catenin, GSK-3ß and PPAR-γ, was assessed by immunohistochemistry, immunofluorescence, western blot and Real Time PCR. RESULTS: GED improved the DSS-induced chronic colitis and fibrosis. GED was able to reduce the expression of the main fibrosis markers (α-SMA, collagen I-III and fibronectin) as well as the pivotal pro-fibrotic molecules IL-13, TGF-ß and Smad3, while it increased the anti-fibrotic PPAR-γ. All these GED effects were nullified by co-administration of GW with GED. Furthermore, GED was able to normalize the expression levels of E-cadherin and ß-catenin and upregulated GSK-3ß, that are all known to be involved both in EMT and fibrosis. CONCLUSIONS: The DSS-induced intestinal fibrosis was improved by the new PPAR-γ modulator GED-0507-34 Levo through the modulation of EMT mediators and pro-fibrotic molecules and through GSK-3ß induction.


Subject(s)
Colitis/pathology , Dextran Sulfate/toxicity , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Glycogen Synthase Kinase 3 beta/metabolism , PPAR gamma/metabolism , Rectum/pathology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , Rectum/drug effects , Rectum/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Inflamm Bowel Dis ; 22(2): 279-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26535766

ABSTRACT

BACKGROUND: Intestinal fibrosis is mainly associated with Crohn's disease and is defined as a progressive and excessive deposition of extracellular matrix components. No specific antifibrotic therapies are available. In this study, we evaluate the antifibrotic effect of a novel 5-ASA analog able to activate the peroxisome proliferator-activated receptor γ, named GED-0507-34 Levo. METHODS: Colonic fibrosis was induced in 110 C57BL/6 mice by 3 cycles of 2.5% (wt/vol) dextran sulfate sodium administration for 6 weeks. The preventive effects of oral daily GED (30 mg · kg(-1) · d(-1)) administration were evaluated using a macroscopic and histological score and also through biological endpoints. Expression of main markers of myofibroblasts activation was determined in transforming growth factor (TGF-ß)-stimulated intestinal fibroblasts and epithelial cells. RESULTS: GED improved macroscopic and microscopic intestinal lesions in dextran sulfate sodium-treated animals and reduced the profibrotic gene expression of Acta2, COL1a1, and Fn1 by 1.48-folds (P < 0.05), 1.93-folds (P < 0.005), and 1.03-fold (P < 0.05), respectively. It reduced protein levels of main markers of fibrosis (α-SMA and Collagen I-II) and the main TGF-ß/Smad pathway components. GED also decreased the interleukin-13 and connective tissue growth factor expression by 1.89-folds (P < 0.05) and 2.2-folds (P < 0.005), respectively. GED inhibited TGF-ß-induced activation of both fibroblast and intestinal epithelial cell lines, by regulating mRNA expression of α-SMA and fibronectin, and restoring the TGF-ß-induced loss of intestinal epithelial cell markers. GED treatment also reduced the TGF-ß and ACTA1 expression in primary human intestinal fibroblasts from ulcerative colitis patients. CONCLUSIONS: GED ameliorates intestinal fibrosis in dextran sulfate sodium-induced chronic colitis in mice and regulates major profibrotic cellular and molecular mechanisms.


Subject(s)
Aniline Compounds/pharmacology , Colitis/drug therapy , Fibroblasts/drug effects , Fibrosis/drug therapy , Inflammation/complications , Intestines/drug effects , PPAR gamma/metabolism , Phenylpropionates/pharmacology , Animals , Blotting, Western , Cells, Cultured , Colitis/etiology , Colitis/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/etiology , Fibrosis/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestines/pathology , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G274-85, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24875097

ABSTRACT

The glucagon-like peptide 2 (GLP-2) is an intestinotrophic hormone with growth promoting and anti-inflammatory actions. However, the full biological functions of GLP-2 and the localization of its receptor (GLP-2R) remain controversial. Among cell lines tested, the expression of GLP-2R transcript was detected in human colonic myofibroblasts (CCD-18Co) and in primary culture of rat enteric nervous system but not in intestinal epithelial cell lines, lymphocytes, monocytes, or endothelial cells. Surprisingly, GLP-2R was expressed in murine (GLUTag), but not human (NCI-H716) enteroendocrine cells. The screening of GLP-2R mRNA in mice organs revealed an increasing gradient of GLP-2R toward the distal gut. An unexpected expression was detected in the mesenteric fat, mesenteric lymph nodes, bladder, spleen, and liver, particularly in hepatocytes. In two mice models of trinitrobenzene sulfonic acid (TNBS)- and dextran sulfate sodium (DSS)-induced colitis, the colonic expression of GLP-2R mRNA was decreased by 60% compared with control mice. Also, GLP-2R mRNA was significantly downregulated in intestinal tissues of inflammatory bowel disease patients. Therapeutically, GLP-2 showed a weak restorative effect on intestinal inflammation during TNBS-induced colitis as assessed by macroscopic score and inflammatory markers. Finally, GLP-2 treatment accelerated mouse liver regeneration following partial hepatectomy as assessed by histological and molecular analyses. In conclusion, the limited therapeutic effect of GLP-2 on colonic inflammation dampens its utility in the management of severe inflammatory intestinal disorders. However, the role of GLP-2 in liver regeneration is a novelty that might introduce GLP-2 into the management of liver diseases and emphasizes on the importance of elucidating other extraintestinal functions of GLP-2.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Colon/drug effects , Gastrointestinal Agents/pharmacology , Glucagon-Like Peptide 2/pharmacology , Liver Regeneration/drug effects , Liver/drug effects , Peptide Fragments/pharmacology , Receptors, Glucagon/agonists , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Gene Expression Regulation , Glucagon-Like Peptide-2 Receptor , HT29 Cells , Hep G2 Cells , Hepatectomy , Humans , Jurkat Cells , Liver/metabolism , Liver/surgery , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Rats , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Recombinant Proteins/pharmacology , Time Factors , Trinitrobenzenesulfonic Acid
14.
Carcinogenesis ; 34(11): 2580-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23843037

ABSTRACT

Epidemiological evidences suggested that 5-aminosalicylic acid (5-ASA) therapy may prevent the development of colorectal cancer in inflammatory bowel disease patients. Our aim is to investigate whether peroxisome proliferator-activated receptor-γ (PPARγ) mediates the antineoplastic effects of 5-ASA. HT-29 and Caco-2 cells were treated by 5-ASA, rosiglitazone (PPARγ ligand) or etoposide (anticarcinogenic drug). Epithelial cell growth, proliferation and apoptosis were assessed by cell count, Ki-67 staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, respectively. The antineoplastic effect of 5-ASA was evaluated in a xenograft tumor model in SCID mice and in azoxymethane (AOM)-induced colon carcinogenesis in A/JOlaHsd mice. The role of PPARγ was examined by administration of PPARγ antagonist, GW9662 and in PPAR knockdown cells. Compared with untreated cells, treatment of HT-29 cells by 5-ASA inhibited significantly cell growth and cell proliferation (respectively, 60% and 63%) and induced apoptosis in 75% of cells. These effects were abolished by co-treatment with GW9662 and blunted in PPAR knockdown cells. Contrarily to etoposide, similar inhibitory effects of GW9662 were obtained in HT-29 cells treated with rosiglitazone. In the xenograft model, GW9662 abolished the therapeutic effect of 5-ASA, which decreased tumor weight and volume by 80% in SCID mice compared with untreated mice. In A/JOlaHsd mice, 5-ASA suppressed colon carcinogenesis by decreasing the number of aberrant crypt foci (75%) and aberrant crypts (22%) induced by AOM treatment with an absence of 5-ASA response after GW9662 administration. In conclusion, 5-ASA exerts potent antineoplastic effects that are mediated through PPARγ. These data provide new rational for designing more effective and safe antineoplastic PPARγ ligands with topical effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Intestines/drug effects , Mesalamine/pharmacology , PPAR gamma/pharmacology , Animals , Azoxymethane/toxicity , Blotting, Western , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Intestines/pathology , Male , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, SCID , PPAR gamma/antagonists & inhibitors , Tumor Cells, Cultured
15.
Gut ; 61(1): 78-85, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21940721

ABSTRACT

OBJECTIVE: Mesenteric fat hyperplasia is a hallmark of Crohn's disease (CD), and C reactive protein (CRP) is correlated with disease activity. The authors investigated whether mesenteric adipocytes may be a source of CRP in CD and whether inflammatory and bacterial triggers may stimulate its production by adipocytes. DESIGN: CRP expression in the mesenteric and subcutaneous fats of patients with CD and the correlation between CRP plasma concentrations and mesenteric messenger RNA (mRNA) levels were assessed. The impact of inflammatory and bacterial challenges on CRP synthesis was tested using an adipocyte cell line. Bacterial translocation to mesenteric fat was studied in experimental models of colitis and ileitis and in patients with CD. RESULTS: CRP expression was increased in the mesenteric fat of patients with CD, with mRNA levels being 80 ± 40 (p<0.05) and 140 ± 65 (p=0.04) times higher than in the mesenteric fat of patients with ulcerative colitis and in the subcutaneous fat of the same CD subjects, respectively, and correlated with plasma levels. Escherichia coli (1230 ± 175-fold, p<0.01), lipopolysaccharide (26 ± 0.5-fold, p<0.01), tumour necrosis factor α (15 ± 0.3-fold, p<0.01) and interleukin-6 (10 ± 0.7-fold, p<0.05) increased CRP mRNA levels in adipocyte 3T3-L1 cells. Bacterial translocation to mesenteric fat occurred in 13% and 27% of healthy and CD subjects, respectively, and was increased in experimental colitis and ileitis. Human mesenteric adipocytes constitutively expressed mRNA for TLR2, TLR4, NOD1 and NOD2. CONCLUSION: Mesenteric fat is an important source of CRP in CD. CRP production by mesenteric adipocytes may be triggered by local inflammation and bacterial translocation to mesenteric fat, providing a mechanism whereby mesenteric fat hyperplasia may contribute to inflammatory response in CD.


Subject(s)
Abdominal Fat/metabolism , Bacterial Translocation , C-Reactive Protein/metabolism , Crohn Disease/metabolism , Escherichia coli/physiology , Mesentery/metabolism , Abdominal Fat/microbiology , Adult , Animals , Cell Line , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Crohn Disease/microbiology , Escherichia coli/metabolism , Female , Humans , Ileitis/metabolism , Ileitis/microbiology , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lymph Nodes/microbiology , Male , Mesentery/microbiology , Mice , Prospective Studies , Tumor Necrosis Factor-alpha/metabolism
16.
Gut ; 56(7): 974-81, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17299060

ABSTRACT

BACKGROUND AND AIMS: The detrimental impact of opioid agonist on the clinical management of inflammatory diseases remains elusive. Given the anti-inflammatory properties of the mu-opioid receptor (MOR) agonists at the intestinal barrier, we hypothesised that MOR activation might also dampen acute hepatic inflammation and cell death-major determinants in the pathogenesis of liver diseases. PATIENTS AND METHODS: The expression of MOR in liver biopsy specimens and peripheral blood mononuclear cells of untreated patients with chronic hepatitis C virus infection and controls, primary hepatocytes and cell lines was determined by quantitative PCR, immunoblotting and/or immunohistochemistry. The effects of peripheral MOR agonist (d-Ala2,NMe-Phe4,Gly5-ol (DAMGO)) and/or antagonist (naloxone methiodide) were explored in two models of acute hepatitis in mice. MOR-deficient mice were used to evaluate the essential regulatory role of MOR during carbon tetrachloride (CCl(4))-induced hepatitis. The role of DAMGO in cell death was investigated using terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling (TUNEL) analysis and quantification of lactate dehydrogenase release. RESULTS: The key role of MOR in the prevention of acute hepatic inflammation and cell death in vivo and in vitro is reported. Whereas MOR gene expression increased transiently in the model of acute liver injury and TNFalpha-treated HepG2 cells, an impaired expression of MOR mRNA in human chronic hepatitis C samples was found. Furthermore, preventive administration of the selective MOR agonist DAMGO enhanced hepatoprotective-signalling pathways in vivo that were blocked by using naloxone methiodide. Consistently, genetic and pharmacological inhibition of MOR enhanced the severity associated with experimental hepatotoxin-induced hepatitis. Finally, treatment with DAMGO was shown to prevent cell death in vitro in HepG2 cells in a MOR-dependent manner and to prevent concanavalin A- and CCl(4)-induced cell death in vivo, providing a possible explanation for the anti-inflammatory role of MOR activation in the liver. CONCLUSIONS: The results indicate that MOR agonists may prevent acute hepatitis and hold promising therapeutic use to maintain remission in both chronic inflammatory bowel and liver diseases.


Subject(s)
Hepatitis/prevention & control , Receptors, Opioid, mu/physiology , Acute Disease , Animals , Biopsy , Carbon Tetrachloride , Cell Death , Concanavalin A , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use , Gene Expression , Hepatitis/metabolism , Hepatitis C, Chronic/metabolism , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hepatitis, Animal/prevention & control , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
17.
Nat Med ; 13(1): 35-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17159985

ABSTRACT

Abdominal pain is common in the general population and, in patients with irritable bowel syndrome, is attributed to visceral hypersensitivity. We found that oral administration of specific Lactobacillus strains induced the expression of mu-opioid and cannabinoid receptors in intestinal epithelial cells, and mediated analgesic functions in the gut-similar to the effects of morphine. These results suggest that the microbiology of the intestinal tract influences our visceral perception, and suggest new approaches for the treatment of abdominal pain and irritable bowel syndrome.


Subject(s)
Abdominal Pain/physiopathology , Intestines/physiopathology , Lactobacillus acidophilus/physiology , Receptors, Cannabinoid/physiology , Receptors, Opioid/physiology , Abdominal Pain/prevention & control , Administration, Oral , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Cannabinoid Receptor Antagonists , Colon/drug effects , Colon/microbiology , Colon/physiopathology , Dose-Response Relationship, Drug , HT29 Cells , Humans , Indoles/administration & dosage , Indoles/pharmacology , Intestines/drug effects , Intestines/microbiology , Male , Mice , Mice, Inbred BALB C , Morphine/administration & dosage , Morphine/pharmacology , Naloxone/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Probiotics/administration & dosage , Probiotics/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/physiology , Receptors, Cannabinoid/biosynthesis , Receptors, Opioid/biosynthesis , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/physiology , Rectum/drug effects , Rectum/microbiology , Rectum/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...