Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-38396840

RESUMO

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.


Assuntos
Doença de Crohn , Doenças Inflamatórias Intestinais , Microbiota , Piperidinas , Pirimidinas , Ratos , Animais , Doença de Crohn/tratamento farmacológico , Doença de Crohn/cirurgia , Doença de Crohn/complicações , Ratos Transgênicos , Antígeno HLA-B27 , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/complicações , Gerenciamento Clínico
2.
Gastric Cancer ; 17(2): 235-46, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23703470

RESUMO

BACKGROUND: Helicobacter pylori (Hp), which is one of the causative agents in human gastric adenocarcinoma, is known to interact with mucous gel and alter mucin gene expression. The aim of this work was to study, using an in vitro model of cell infection, the effects of urease, flagellin, and CagA virulence factors on the regulation of the four 11p15 mucin genes (MUC2, MUC5AC, MUC5B, and MUC6). METHODS: KATO-III and AGS gastric cancer cells were infected for 1, 3 or 6 h with Hp wild-type strains (ATCC 43504, N6, and SS1) or corresponding isogenic mutants deficient for urease subunit B, flagellin subunit A, and CagA. mRNA levels of MUC2, MUC5B, MUC5AC and MUC6 were assessed by RT-PCR, and functional activity of their promoters was measured by transient transfection assays. RESULTS: Infection of KATO-III cells with Hp wild-type strains resulted in an early (at 1 h) transient expression of MUC2, MUC5AC, and MUC6 mRNA concomitant with those of interleukin (IL)-1ß, IL-8, and TNF-α cytokines. In these cells, the UreB(-) isogenic mutant induced strong activation of MUC5AC expression, and UreB-responsive elements were located in the -486/-1 region of the promoter. FlaA(-) and CagA(-) mutants had no effect on mucin gene mRNA levels in KATO-III cells. In AGS cells, Hp-responsive elements were identified in all promoters, and overexpression of NF-κB induced upregulation of MUC5AC promoter activity when infected with the UreB(-) isogenic mutant. CONCLUSION: These results indicate that Hp infection of gastric cancer cells alters 11p15 mucin gene transcription and that MUC5AC downregulation is mediated by urease virulence factor.


Assuntos
Flagelina/metabolismo , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/genética , Mucina-5AC/genética , Neoplasias Gástricas/genética , Urease/metabolismo , Western Blotting , Citocinas/genética , Citocinas/metabolismo , Flagelina/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/virologia , Helicobacter pylori/fisiologia , Humanos , Mucina-5AC/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virologia , Células Tumorais Cultivadas , Urease/genética , Virulência
3.
Nat Med ; 13(1): 35-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17159985

RESUMO

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.


Assuntos
Dor Abdominal/fisiopatologia , Intestinos/fisiopatologia , Lactobacillus acidophilus/fisiologia , Receptores de Canabinoides/fisiologia , Receptores Opioides/fisiologia , Dor Abdominal/prevenção & controle , Administração Oral , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Antagonistas de Receptores de Canabinoides , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/fisiopatologia , Relação Dose-Resposta a Droga , Células HT29 , Humanos , Indóis/administração & dosagem , Indóis/farmacologia , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Morfina/administração & dosagem , Morfina/farmacologia , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Probióticos/administração & dosagem , Probióticos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/biossíntese , Receptor CB2 de Canabinoide/fisiologia , Receptores de Canabinoides/biossíntese , Receptores Opioides/biossíntese , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/biossíntese , Receptores Opioides mu/fisiologia , Reto/efeitos dos fármacos , Reto/microbiologia , Reto/fisiopatologia
4.
World J Gastroenterol ; 30(16): 2258-2271, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38690023

RESUMO

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.


Assuntos
Quitina , Colo , Modelos Animais de Doenças , Glucanos , Síndrome do Intestino Irritável , Ratos Sprague-Dawley , Dor Visceral , Animais , Síndrome do Intestino Irritável/tratamento farmacológico , Síndrome do Intestino Irritável/fisiopatologia , Masculino , Humanos , Colo/efeitos dos fármacos , Colo/patologia , Ratos , Dor Visceral/tratamento farmacológico , Dor Visceral/fisiopatologia , Dor Visceral/metabolismo , Dor Visceral/etiologia , Quitina/farmacologia , Glucanos/farmacologia , Glucanos/administração & dosagem , Camundongos , Prebióticos/administração & dosagem , Ácido Trinitrobenzenossulfônico/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Mucosa Intestinal/metabolismo , Colite/tratamento farmacológico , Colite/induzido quimicamente , Colite/fisiopatologia , Colite/patologia , Células HT29
5.
World J Gastrointest Pharmacol Ther ; 15(3): 90757, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38846967

RESUMO

BACKGROUND: Irritable bowel syndrome (IBS), defined according to the Rome IV diagnostic criteria, is a chronic functional gastrointestinal disorder characterized by recurrent abdominal pain related to altered bowel habits. First-line recommended treatments are limited to combining drugs targeting predominant symptoms, particularly pain (antispasmodics), constipation (laxatives), and diarrhea (loperamide), yielding only a limited therapeutic gain. GASTRAP® DIRECT is a class IIa medical formulation composed of a combination of chitin-glucan and simethicone indicated for the symptomatic treatment of gas-related gastrointestinal disorders by combining different mechanisms of action. AIM: To evaluate the efficacy, tolerability, and safety of 4-week GASTRAP® DIRECT treatment in patients with IBS. METHODS: In this prospective, multicenter, open-label trial, 120 patients with IBS received three sticks of GASTRAP® DIRECT (1.5 g/d of chitin-glucan and 0.75 mg/d of simethicone) per day for 4 weeks. The primary endpoint was the responder rate, defined as the number of patients whose abdominal pain score decreased by ≥ 30% from baseline to week (W) 4. The analysis was performed using the per-protocol set. Cardinal symptoms, impact of global symptoms on daily life, change in stool consistency, and improvement in defecatory disorders were evaluated. RESULTS: Overall, 100 patients were evaluated. At W4, 67% (95%CI: 57-75) showed improvement in abdominal pain (score: 5.8 ± 2.4 vs 2.9 ± 2.0, P < 0.0001). Similar improvements were observed for bloating [8.0 ± 1.7 vs 4.7 ± 2.9, P < 0.0001; 60% (95%CI: 50-70) responders], abdominal distension [7.2 ± 2.1 vs 4.4 ± 3.1, P < 0.0001; 53% (95%CI: 43-63) responders], and impact of global symptoms on daily life [7.1 ± 2.0 vs 4.6 ± 2.9, P < 0.0001; 54% (95%CI: 44-64) responders]. Stool consistency improved in most patients (90% and 57% for patients with liquid and hard stools, respectively). Overall, 42% of patients with defecatory disorders reported very much/considerable improvements by W2. No severe adverse event occurred, and tolerability was rated "good" or "very good" by 93% of patients. CONCLUSION: GASTRAP® DIRECT is safe and well tolerated, alleviating IBS symptoms rapidly in 2 weeks. This open-label study suggests that the combination of chitin-glucan and simethicone could be beneficial in patients with IBS.

6.
Carcinogenesis ; 34(11): 2580-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23843037

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Intestinos/efeitos dos fármacos , Mesalamina/farmacologia , PPAR gama/farmacologia , Animais , Azoximetano/toxicidade , Western Blotting , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Técnicas Imunoenzimáticas , Mucosa Intestinal/metabolismo , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos SCID , PPAR gama/antagonistas & inibidores , Células Tumorais Cultivadas
7.
Gut ; 61(1): 78-85, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21940721

RESUMO

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.


Assuntos
Gordura Abdominal/metabolismo , Translocação Bacteriana , Proteína C-Reativa/metabolismo , Doença de Crohn/metabolismo , Escherichia coli/fisiologia , Mesentério/metabolismo , Gordura Abdominal/microbiologia , Adulto , Animais , Linhagem Celular , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Doença de Crohn/microbiologia , Escherichia coli/metabolismo , Feminino , Humanos , Ileíte/metabolismo , Ileíte/microbiologia , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Linfonodos/microbiologia , Masculino , Mesentério/microbiologia , Camundongos , Estudos Prospectivos , Fator de Necrose Tumoral alfa/metabolismo
8.
PLoS One ; 18(8): e0283489, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37639428

RESUMO

The oral cholera vaccine WC-rBS consists of 4 different inactivated strains of Vibrio cholerae (LPS source) admixed with recombinant cholera toxin B subunit. Because of its unique composition and anti-inflammatory properties reported for both CTB and low doses of LPS from other Gram-negative bacteria, we speculated that WC-rBS might have anti-inflammatory potential in a chronic autoimmune disease such as inflammatory bowel diseases. First in vitro endotoxin tolerance experiments showed the surprising WC-rBS potential in the modulation of inflammatory responses on both PBMCs and THP1 cells. WC-rBS was further evaluated in the Dextran Sodium Sulfate colitis mouse model. Administrated orally at different dosages, WC-rBS vaccine was safe and showed immunomodulatory properties when administered in a preventive mode (before and during the induction of DSS colitis) as well as in a curative mode (after colitis induction); with improvement of disease activity index (from 27 to 73%) and histological score (from 65 to 88%). Interestingly, the highest therapeutic effect of WC-rBS vaccine was observed with the lowest dosage, showing even better anti-inflammatory properties than mesalamine; an approved 5-aminosalicylic acid drug for treating IBD patients. In summary, this is the first time that a prophylactic medicine, safe and approved for prevention of an infectious disease, showed a benefit in an inflammatory bowel disease model, potentially offering a novel therapeutic modality for IBD patients.


Assuntos
Vacinas contra Cólera , Colite , Doenças Inflamatórias Intestinais , Vibrio cholerae , Animais , Camundongos , Lipopolissacarídeos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/prevenção & controle , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Mesalamina
9.
J Crohns Colitis ; 16(10): 1617-1627, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-35997152

RESUMO

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.


Assuntos
Bacteriófagos , Colite , Animais , Humanos , Camundongos , Aderência Bacteriana , Colite/patologia , Modelos Animais de Doenças , Disbiose/complicações , Escherichia coli , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Mucosa Intestinal/patologia
10.
J Exp Med ; 201(8): 1205-15, 2005 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-15824083

RESUMO

5-aminosalicylic acid (5-ASA) is an antiinflammatory drug widely used in the treatment of inflammatory bowel diseases. It is known to inhibit the production of cytokines and inflammatory mediators, but the mechanism underlying the intestinal effects of 5-ASA remains unknown. Based on the common activities of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) ligands and 5-ASA, we hypothesized that this nuclear receptor mediates 5-ASA therapeutic action. To test this possibility, colitis was induced in heterozygous PPAR-gamma(+/-) mice and their wild-type littermates, which were then treated with 5-ASA. 5-ASA treatment had a beneficial effect on colitis only in wild-type and not in heterozygous mice. In epithelial cells, 5-ASA increased PPAR-gamma expression, promoted its translocation from the cytoplasm to the nucleus, and induced a modification of its conformation permitting the recruitment of coactivators and the activation of a peroxisome-proliferator response element-driven gene. Validation of these results was obtained with organ cultures of human colonic biopsies. These data identify PPAR-gamma as a target of 5-ASA underlying antiinflammatory effects in the colon.


Assuntos
Anti-Inflamatórios/uso terapêutico , Colite/tratamento farmacológico , Colo/efeitos dos fármacos , Mesalamina/uso terapêutico , PPAR gama/efeitos dos fármacos , Células 3T3-L1 , Animais , Colite/induzido quimicamente , Colo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , PPAR gama/biossíntese , PPAR gama/genética , RNA Mensageiro/biossíntese
11.
PLoS One ; 16(9): e0257281, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34529707

RESUMO

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.


Assuntos
Transdiferenciação Celular , Miofibroblastos/citologia , Propionatos/farmacologia , Fibrose Pulmonar/tratamento farmacológico , Células A549 , Animais , Bleomicina , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Técnicas In Vitro , Inflamação , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo , Fibrose Pulmonar/fisiopatologia , Resultado do Tratamento
12.
J Control Release ; 275: 32-39, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28774843

RESUMO

We have developed a self-assembling polymeric nanocarrier to deliver the potent immunosuppressive drug Cyclosporine A (CsA) to inflammatory lesions in ulcerative colitis (UC) patients. Our nanocarrier has a high drug loading capacity and efficiently targets its CsA payload to the diseased tissue after local administration. Tissue drug levels were several orders of magnitude higher in animals suffering from a trinitrobenzene-sulfonic acid (TNBS) - induced colitis, compared to healthy control animals; no drug was detectable in the plasma, underlining the localized delivery strategy. An efficient reduction in inflammation score was obtained with a CsA dose of 1mg/mL. Therapeutic efficacy was comparable to 5-aminosalicylic acid (5-ASA), the positive control treatment in the TNBS-induced colitis model. Repetitive treatment of healthy animals with CsA nanocarriers for seven days was well tolerated with no alterations in colon histology.


Assuntos
Anti-Inflamatórios/administração & dosagem , Colite Ulcerativa/tratamento farmacológico , Ciclosporina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Imunossupressores/administração & dosagem , Nanopartículas/administração & dosagem , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/patologia , Colo/efeitos dos fármacos , Colo/patologia , Masculino , Camundongos Endogâmicos C57BL , Polímeros/administração & dosagem , Ácido Trinitrobenzenossulfônico
13.
PLoS One ; 13(12): e0209681, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30592734

RESUMO

BACKGROUND: P28GST, a 28Kd glutathione S-transferase enzymatic protein derived from a schistosome helminth prevents experimental colitis when administered subcutaneously in the presence of adjuvant by decreasing pro-inflammatory Th1/Th17 response. Given the antioxidant properties of P28GST, we evaluated its anti-inflammatory potential when administered locally after colitis induction in the absence of adjuvant. METHODS: Colitis was induced in BALB/c mice by rectal administration of TNBS, followed by two intraperitoneal injections of P28GST at day 1 and day 2. Mice were sacrificed 48h after TNBS administration and evaluated for macroscopic and histological scores, myeloperoxidase (MPO) quantification and cytokine messenger RNA expression in the colonic tissues. RESULTS: Both clinical and histological scores significantly decreased in mice treated with P28GST at 5 or 50µg/kg when compared to vehicle- treated mice. A significant reduction of MPO was detected in colonic tissues from P28GST-treated mice, similarly to mice treated with methylprednisolone as the reference treatment. Pro-inflammatory cytokines TNF, IL-1ß, and IL-6, mRNA as well as serum levels were down-regulated in mice colonic tissues treated with P28GST at 5 or 50µg/kg. In addition, a significant decrease of mRNA expression levels of T-bet, and ROR-γ, respective markers of Th1 and Th17 cells was observed. Whereas no significant effect was detected on Gata3 mRNA, a marker of Th2 cells, the Arg/iNOS mRNA levels significantly increased in P28GST-treated mice, suggesting the induction of M2 macrophages. CONCLUSIONS: These findings provide evidence that P28GST injected locally after colitis induction induces a potent decrease of colitis inflammation in mice, associated to downregulation of Th1/Th17 response, and induction of anti-inflammatory alternatively activated macrophages.


Assuntos
Colite/etiologia , Glutationa Transferase/farmacologia , Proteínas de Helminto/farmacologia , Animais , Biomarcadores , Colite/tratamento farmacológico , Colite/metabolismo , Colite/patologia , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Peroxidase/sangue , Peroxidase/metabolismo , Índice de Gravidade de Doença , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
14.
EMBO Mol Med ; 9(11): 1471-1481, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28947679

RESUMO

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.


Assuntos
Intestino Delgado/metabolismo , Lactase/metabolismo , PPAR gama/metabolismo , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Células CACO-2 , Imunoprecipitação da Cromatina , Dieta , Humanos , Lactase/genética , Lactose/metabolismo , Intolerância à Lactose/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , PPAR gama/agonistas , PPAR gama/genética , Fenilpropionatos/farmacologia , Fenilpropionatos/uso terapêutico , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Transcrição Gênica/efeitos dos fármacos
15.
Inflamm Bowel Dis ; 22(2): 279-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26535766

RESUMO

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.


Assuntos
Compostos de Anilina/farmacologia , Colite/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Fibrose/tratamento farmacológico , Inflamação/complicações , Intestinos/efeitos dos fármacos , PPAR gama/metabolismo , Fenilpropionatos/farmacologia , Animais , Western Blotting , Células Cultivadas , Colite/etiologia , Colite/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/etiologia , Fibrose/metabolismo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Inflamação/patologia , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Soc Biol ; 200(2): 121-31, 2006.
Artigo em Francês | MEDLINE | ID: mdl-17151549

RESUMO

PPARgamma has been recently described as being a gene of susceptibility for Intestinal Bowel Diseases (IBD) as NOD2/CARD15 gene. IBD are pathologies due to an abnormal immune response, in genetically predisposed patients, to the bacteria of the intestinal flora. PPARgamma, known for its significant role in adipogenesis, is strongly expressed by the epithelial cells of the colon mucosa. PPARgamma is implicated in the regulation of inflammation. Indeed, agonists of this nuclear receptor decrease strongly the intensity of inflammation during experimental colitis induced by chemical agents. A deficit of PPARgamma in patients with ulcerative colitis has been highlighted, that could in part explain the acute inflammation. In addition, bacteria, including those of the commensal flora, are able to regulate PPARgamma. Toll Like Receptor-4 (TLR-4), responsible for the recognition of bacterial motif as lipopolysaccharide (LPS), is implicated in PPARgamma regulation and its anti-inflammatory properties. All these arguments make of PPARgamma a very interesting therapeutic target for the treatment of IBD.


Assuntos
Doenças Inflamatórias Intestinais/fisiopatologia , PPAR gama/fisiologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Fenômenos Fisiológicos Bacterianos , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/fisiopatologia , Doença de Crohn/tratamento farmacológico , Doença de Crohn/fisiopatologia , Eicosanoides/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Homeostase , Humanos , Inflamação/fisiopatologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/epidemiologia , Resistência à Insulina , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Ligantes , Metabolismo dos Lipídeos , Camundongos , PPAR gama/agonistas , PPAR gama/química , PPAR gama/deficiência , PPAR gama/genética , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Receptor 4 Toll-Like/fisiologia
18.
Infect Immun ; 72(4): 2438-41, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15039377

RESUMO

Rectal instillation of trinitrobenzene sulfonic acid (TNBS) induces acute colitis in the mouse. We tested the efficacy of Yersinia pseudotuberculosis anti-inflammatory components in preventing TNBS-triggered colitis. Animals were orally inoculated with virulence-attenuated Yersinia cells (a phoP mutant) prior to TNBS administration. Under these experimental conditions, colonic lesions and tumor necrosis factor alpha mRNA levels were significantly reduced.


Assuntos
Colite/imunologia , Ácido Trinitrobenzenossulfônico/farmacologia , Fator de Necrose Tumoral alfa/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/imunologia , Animais , Colite/induzido quimicamente , Colite/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Inflamação , Doenças Inflamatórias Intestinais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo , Yersinia pseudotuberculosis/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA