Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38928402

RESUMEN

The gut microbial and metabolic characteristics of intestinal Behçet's disease (BD), a condition sharing many clinical similarities with ulcerative colitis (UC) and Crohn's disease (CD), are largely unexplored. This study investigated the gut microbial and metabolic characteristics of intestinal BD as well as potential biomarkers, comparing them with those in UC, CD, and healthy controls. Colon tissue and stool samples from 100 patients (35 UC, 30 CD, and 35 intestinal BD) and 41 healthy volunteers were analyzed using 16S ribosomal RNA sequencing to assess microbial diversity, taxonomic composition, and functional profiling. Plasma metabolomic analyses were performed using gas chromatography and ultra-performance liquid chromatography-mass spectrometry. Results indicated reduced microbial diversity in CD but not in intestinal BD, with intestinal BD showing fewer changes compared to controls yet distinct taxonomic features from UC, CD, and controls. Common alterations across all diseases included a reduction in beneficial bacteria producing short-chain fatty acids. Intestinal BD-specific changes featured a decreased abundance of Bacteroides fragilis. Metabolomic profiles in intestinal BD were similar to those in CD but distinct from those in UC, displaying significant changes in energy metabolism and genetic information processing. This integrative analysis revealed both shared and unique profiles in intestinal BD compared with UC, CD, and controls, advancing our understanding of the distinctive features of these diseases.


Asunto(s)
Síndrome de Behçet , Microbioma Gastrointestinal , Metaboloma , Humanos , Síndrome de Behçet/microbiología , Síndrome de Behçet/metabolismo , Masculino , Femenino , Adulto , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/metabolismo , Metabolómica/métodos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/metabolismo , Biomarcadores , Heces/microbiología , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/metabolismo , Estudios de Casos y Controles
2.
Cancer Gene Ther ; 30(5): 694-703, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36577836

RESUMEN

Development of colorectal cancer (CRC) is regulated by a series of genetic and microenvironmental alterations. Olfactomedin 4 (OLFM4) is a secreted glycoprotein that is highly expressed in the gastrointestinal tract and modulates inflammation. However, the role of OLFM4 in CRC is uncertain. Here we aimed to explore the function of OLFM4 in CRC in vivo and in vitro. The mRNA expression of OLFM4 was up-regulated in precursor lesions with dysplasia or ulcerative colitis but was reduced in CRC. OLFM4 neutralizing antibody suppressed inflammation-mediated early-stage CRC formation in an AOM/DSS colitis-associated cancer model. OLFM4 knockdown cells exhibited increased cell proliferation and motility in vitro and in vivo. Ablation of OLFM4 increased tumor growth and metastasis in xenograft experiments. In addition, OLFM4 knockdown cells showed elevated expression of colon cancer stem cell markers including CD133, resulting in increased metastasis via epithelial-mesenchymal transition signaling. This study demonstrated that OLFM4 regulates inflammation and cancer progression differently; ablation of OLFM4 promotes cancer metastasis via stemness and epithelial-mesenchymal transition. These results suggest a new route for controlling cancer progression and metastasis.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Neoplasias del Colon/genética , Células Madre Neoplásicas/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proliferación Celular , Inflamación , Metástasis de la Neoplasia , Transición Epitelial-Mesenquimal/genética
4.
Gut Liver ; 17(5): 766-776, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36167345

RESUMEN

Background/Aims: The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function. Methods: Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay. Results: Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner. Conclusions: The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Factor de Necrosis Tumoral alfa , Humanos , Células CACO-2 , Histona Desacetilasa 6/metabolismo , Histona Desacetilasa 6/farmacología , Histona Desacetilasa 6/uso terapéutico , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Lipopolisacáridos/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Cadherinas/metabolismo , Cadherinas/farmacología , Cadherinas/uso terapéutico , ARN Mensajero/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
5.
Front Immunol ; 13: 870817, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693794

RESUMEN

Inflammatory bowel disease (IBD) is a chronic immune-mediated disorder characterized by prolonged inflammation of the gastrointestinal tract. IBD can result from gut barrier dysfunction, altered gut microbiota, and abnormal intestinal immunity induced by environmental factors in genetically susceptible individuals. Proton pump inhibitors (PPIs) such as rabeprazole are frequently employed for gastric acid inhibition. However, long-term PPI administration can alter the intestinal microbiome composition, possibly worsening IBD severity. The present study revealed that tegoprazan, a potassium-competitive acid blocker, significantly improved colitis in mice and enhanced the intestinal epithelial barrier function. Tegoprazan alleviated gut microbiota dysbiosis and enhanced the growth of Bacteroides vulgatus. In turn, B. vulgatus alleviated intestinal inflammation by inhibiting epithelial adhesion of pathogenic bacteria. Unlike rabeprazole, tegoprazan did not induce gut dysbiosis. Our findings provide novel insights into the potential role of tegoprazan as an intestinal protectant for IBD and as a therapeutic agent for gastric acid-related diseases.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Derivados del Benceno , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Disbiosis/microbiología , Imidazoles , Inflamación , Ratones , Potasio , Inhibidores de la Bomba de Protones/farmacología , Rabeprazol/efectos adversos
7.
Free Radic Biol Med ; 174: 110-120, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34358646

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract whose occurrence is attributed to various factors, including genetic factors, immune response, microbial changes, and oxidative stress. Microbial-targeted therapy has emerged as an alternative to immunosuppressive therapy for IBD. METHODS: The effects of an atypical commensal Escherichia coli strain harboring an additional catalase gene (compared to typical E. coli strain) on dextran sulfate sodium (DSS)-induced colitis were explored in mice. RESULTS: The atypical E. coli (atEc) significantly restored body weight, reduced disease activity score, and improved histological scores in mice with colitis. Hydrogen peroxide levels in colitis mice were noticeably decreased when the mice were administered atEc. The proinflammatory cytokine levels were decreased and regulatory T cell numbers were increased after the administration of atEc. The abundance of Firmicutes was significantly recovered, while that of Proteobacteria decreased in atEc -treated mice compared with that in vehicle-treated wild-type mice. To investigate the role of interleukin (IL)-17A in mediating the anti-inflammatory effects of the atEc, IL-17A‒knockout mice were orally administered atEc. Clinical and immune responses and microbial composition were significantly reduced in IL-17A‒knockout mice compared with those in wild-type mice. CONCLUSIONS: atEc ameliorates colonic inflammation by controlling hydrogen peroxide levels, immune responses (including regulatory T cells and IL-17A), and microbial composition. atEc could be a novel candidate of probiotic for IBD treatment.


Asunto(s)
Colitis , Linfocitos T Reguladores , Animales , Catalasa , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Citocinas , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Escherichia coli/genética , Peróxido de Hidrógeno , Interleucina-17 , Ratones , Ratones Noqueados
8.
Front Immunol ; 12: 650864, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767714

RESUMEN

Triggering receptor expressed on myeloid cell-1 (TREM-1) signaling is expressed on neutrophils and monocytes that is necessary for the successful antimicrobial response and resolution of inflammation in the gut. In this study, we determined the effect of an anti-TREM-1 agonistic antibody (α-TREM-1) on colitis and identify its underlying mechanism of action. Administration of α-TREM-1 alleviated colitis in mice and resolved dysbiosis, which required TLR4/Myd88 signaling. α-TREM-1 increased the production of neutrophil extracellular traps and interleukin-22 by CD177+ neutrophils, which led to pathogen clearance and protection of the intestinal barrier. TREM-1 activation using an α-TREM-1 antibody protects against colitis by rebalancing the microbiota and protecting the epithelium against the immune response as well as modulates the function of neutrophils and macrophages. These results highlight the importance of the TREM-1 pathway in intestinal homeostasis and suggest that α-TREM-1 treatment may be an effective therapeutic strategy for inflammatory bowel disease.


Asunto(s)
Anticuerpos/farmacología , Proteínas Ligadas a GPI/inmunología , Inflamación/prevención & control , Intestinos/efectos de los fármacos , Isoantígenos/inmunología , Neutrófilos/efectos de los fármacos , Receptores de Superficie Celular/inmunología , Receptor Activador Expresado en Células Mieloides 1/agonistas , Animales , Anticuerpos/inmunología , Colitis/inmunología , Colitis/metabolismo , Colitis/prevención & control , Disbiosis/microbiología , Disbiosis/prevención & control , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/inmunología , Proteínas Ligadas a GPI/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/prevención & control , Interleucinas/inmunología , Interleucinas/metabolismo , Intestinos/inmunología , Intestinos/patología , Isoantígenos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptor Activador Expresado en Células Mieloides 1/inmunología , Interleucina-22
9.
Intest Res ; 19(3): 349-353, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33120440

RESUMEN

BACKGROUND/AIMS: The safety and effectiveness of adalimumab was demonstrated in a phase 3 trial in Japanese patients with intestinal Behçet's disease. The aim of this study was to evaluate the long-term safety and effectiveness of adalimumab in Japanese patients with intestinal Behçet's disease.

10.
J Nanobiotechnology ; 18(1): 133, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32933548

RESUMEN

BACKGROUND: Infliximab (IFX), a TNF-α blocking chimeric monoclonal antibody, induces clinical response and mucosal healing in patients with inflammatory bowel disease (IBD). However, systemic administration of this agent causes unwanted side effects. Oral delivery of antibody therapeutics might be an effective treatment strategy for IBD compared to intravenous administration. RESULTS: All three carriers had a high encapsulation efficiency, narrow size distribution, and minimal systemic exposure. There was a higher interaction between nanocomposite carriers and monocytes compared to lymphocytes in the PBMC of IBD patients. Orally administered nanocomposite carriers targeted to inflamed colitis minimized systemic exposure. All IFX delivery formulations with nanocomposite carriers had a significantly less colitis-induced body weight loss, colon shortening and histomorphological score, compared to the DSS-treated group. AC-IFX-L and EAC-IFX-L groups showed significantly higher improvement of the disease activity index, compared to the DSS-treated group. In addition, AC-IFX-L and EAC-IFX-L alleviated pro-inflammatory cytokine expressions (Tnfa, Il1b, and Il17). CONCLUSION: We present orally administered antibody delivery systems which improved efficacy in murine colitis while reducing systemic exposure. These oral delivery systems suggest a promising therapeutic approach for treating IBD.


Asunto(s)
Colitis/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Infliximab/farmacología , Nanocompuestos/administración & dosificación , Nanocompuestos/química , Administración Oral , Animales , Anticuerpos Monoclonales , Colitis/patología , Colon/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Leucocitos Mononucleares , Liposomas , Linfocitos , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Factor de Necrosis Tumoral alfa/efectos de los fármacos
11.
Int J Med Microbiol ; 310(2): 151391, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32007342

RESUMEN

Lactobacillus plantarum has been identified as a probiotic bacterium owing to its role in immune regulation and maintenance of intestinal permeability. Here, we investigated the anti-colitic effects and mechanism of L. plantarum CBT LP3 (LP3). This in vivo study was performed using dextran sodium sulfate (DSS) to induce colitis in mice. Mice were randomly divided into three groups: a control supplied with normal drinking water, a DSS-treated group followed by oral administration of vehicle, and a DSS-treated group gavaged with LP3 daily for 7 days following DSS administration. An analysis of macrophages and T cell subsets harvesting from peritonium cavity cells and splenocytes was performed using a flow cytometric assay. Gene expression and cytokine profiles were measured using quantitative reverse transcriptase polymerase chain reaction. The administration of LP3 significantly attenuated disease activity and histolopathology compared to control. LP3 had anti-inflammatory effects, with increased induction of regulatory T cells and type 2 helper T cells in splenocytes and restoration of goblet cells accompanied by suppression of proinflammatory cytokine expressions. These findings suggest that L. plantarum CBT LP3 can be used as a potent immunomodulator, which has significant implications for IBD treatment.


Asunto(s)
Colitis/inmunología , Colitis/terapia , Lactobacillus plantarum , Probióticos/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Animales , Colitis/inducido químicamente , Citocinas/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad , Factores Inmunológicos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Células Th2/inmunología
12.
FASEB J ; 34(2): 3289-3304, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31916636

RESUMEN

The enzyme glutathione S-transferase theta 1 (GSTT1) is involved in detoxifying chemicals, including reactive oxygen species (ROS). Here, we provide a significant insight into the role of GSTT1 in inflammatory bowel disease (IBD). We identified decreased expression of GSTT1 in inflamed colons from IBD patients compared to controls. We intrarectally or intraperitoneally delivered Gstt1 gene to mice with dextran sodium sulfate (DSS)-induced colitis and noted attenuation of colitis through gene transfer of Gstt1 via an IL-22 dependent pathway. Downregulation of GSTT1 by pathogen-associated molecular patterns (PAMPs) of microbes reduced innate defense responses and goblet cell differentiation. The GSTT1 mutation in intestinal epithelial cells (IECs) and IBD patients decreased its dimerization, which was connected to insufficient phosphorylation of signal transducer and activator of transcription-3 and p38/mitogen-activated protein kinase by their common activator, IL-22. GSTT1 ameliorated colitis and contributed as a modulator of goblet cells through sensing pathogens and host immune responses. Its mutations are linked to chronic intestinal inflammation due to its insufficient dimerization. Our results provide new insights into GSTT1 mutations that are linked to chronic intestinal inflammation due to its insufficient dimerization and their functional consequences in IBDs.


Asunto(s)
Diferenciación Celular , Colitis Ulcerosa/metabolismo , Glutatión Transferasa/metabolismo , Células Caliciformes/metabolismo , Interleucinas/metabolismo , Animales , Células Cultivadas , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Enterocitos/citología , Enterocitos/metabolismo , Femenino , Glutatión Transferasa/genética , Células Caliciformes/citología , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Multimerización de Proteína , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células THP-1 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Interleucina-22
13.
J Gastroenterol Hepatol ; 34(1): 178-185, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29933526

RESUMEN

BACKGROUND AND AIM: Nuclear factor kappa B (NF-κB) activation and endoplasmic reticulum (ER) stress signaling play significant roles in the pathogenesis of inflammatory bowel disease (IBD). Thus, we evaluated whether new therapeutic probiotics have anti-colitic effects, and we investigated their mechanisms related to NF-κB and ER-stress pathways. METHODS: Luciferase, nitric oxide, and cytokine assays using HT-29 or RAW264.7 cells were conducted. Mouse colitis was induced using dextran sulfate sodium and confirmed by disease activity index and histology. Macrophages and T-cell subsets in isolated peritoneal cavity cells and splenocytes were analyzed by flow cytometry. Gene and cytokine expression profiles were determined using reverse-transcription polymerase chain reaction. RESULTS: Lactobacillus acidophilus (LA1) and Pediococcus pentosaceus inhibited nitric oxide production in RAW264.7 cells, but only LA1 inhibited Tnfa and induced Il10 expression. LA1 increased the lifespan of dextran sulfate sodium-treated mice and attenuated the severity of colitis by inducing M2 macrophages in peritoneal cavity cells and Th2 and Treg cells in splenocytes. The restoration of goblet cells in the colon was accompanied by the induction of Il10 expression and the suppression of pro-inflammatory cytokines. Additionally, we found that LA1 exerts an anti-colitic effect by improving ER stress in HT-29 cells as well as in vivo. CONCLUSIONS: We showed that LA1 significantly interferes with ER stress and suppresses NF-κB activation. Our findings suggest that LA1 can be used as a potent immunomodulator in IBD treatment, and the regulation of ER stress may have significant implications in treating IBD.


Asunto(s)
Colitis/inmunología , Colitis/terapia , Estrés del Retículo Endoplásmico , Mucosa Intestinal/inmunología , Lactobacillus acidophilus , Probióticos/farmacología , Animales , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Células Caliciformes , Células HT29 , Humanos , Interleucina-10/metabolismo , Mucosa Intestinal/patología , Macrófagos , Masculino , Ratones , FN-kappa B , Óxido Nítrico/antagonistas & inhibidores , Pediococcus pentosaceus , Cavidad Peritoneal/citología , Células RAW 264.7 , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
14.
Inflamm Bowel Dis ; 24(8): 1706-1717, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29733354

RESUMEN

Background: Genome-wide association studies and meta-analyses have revealed the genetic background of ulcerative colitis (UC) by identifying common variants. However, these variants do not fully explain the disease variance in UC. To identify novel variants, we performed deep resequencing of UC-associated genes in Korean UC patients and subsequently investigated the functional roles of identified susceptibility genes. Methods: We performed targeted deep resequencing of 108 genes in 24 Korean UC patients and then performed association analysis with data from 126 healthy controls. We validated these variants using 2-stage replication studies including 793 UC patients and 783 controls. We performed in silico and pathway analyses and functional analyses. Results: The combined analysis including 2 replication studies identified 6 novel susceptibility loci and reconfirmed 10 previously reported loci. Among the novel single nucleotide variants (SNVs), rs10035653 in C5orf55 (P = 2.08 × 10-3; OR = 1.50), rs41417449 in BTNL2 (P = 1.27 × 10-2; OR = 1.32), rs3117099 in HCG23 (P = 9.98 × 10-6; OR = 1.40), rs7192 in HLA-DRA (P = 6.95 × 10-9; OR = 1.57), and rs3744246 in ORMDL3 (P = 2.21 × 10-2; OR = 1.21) were identified as causal variants, whereas rs713669 in IL17REL (P = 2.69 × 10-2; OR = 0.84) as a protective variant for UC. When correcting multiple testing, 3 novel SNVs (rs41417449 in BTNL2, rs3744246 in ORMDL3, and rs713669 in IL17REL) and 4 previously reported SNVs did not reach a statistical significance. Functional study suggested that SNVs of BTNL2 and C5orf55 exacerbated the inflammatory response both in vitro and in vivo. Conclusions: This study identified 3 novel susceptibility loci and validated 6 previously reported SNVs for UC through deep resequencing in Koreans and revealed the functional roles of BTNL2 and C5orf55.


Asunto(s)
Butirofilinas/genética , Colitis Ulcerosa/genética , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intracelular/genética , Adulto , Estudios de Casos y Controles , Femenino , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , República de Corea , Adulto Joven
15.
Sci Rep ; 7(1): 851, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28404987

RESUMEN

Interleukin 33 (IL-33) that signals through the ST2 receptor has emerged as a critical modulator in several inflammatory disorders, including inflammatory bowel disease (IBD). However, the precise mechanisms by which IL-33 modulates IBD are controversial. The aim of this study was thus to clarify the role of IL-33 in IBD. The plasma levels of IL-33 were significantly decreased, but soluble ST2 levels were increased in patients with IBD compared to healthy individuals. Moreover, IL-33 restored goblet cell numbers and induced macrophage switching from the M1 to the M2 phenotype. These effects were sufficient to ameliorate colitis in dextran sodium sulfate, trinitrobenzene sulfonic acid, and peritoneal cavity cell transfer models. IL-33 facilitated goblet cell restoration via modulating macrophages toward the M2 phenotype. In addition, wound healing was significantly faster in IL-33-treated human monocyte-derived macrophages than in control cells, which could be attributed to increased polarisation into M2 macrophages. We found that patients with IBD show decreased serum levels of IL-33 compared with healthy individuals and that IL-33 can attenuate colitis and aid tissue repair in mice. The mechanism by which IL-33 exerts these effects appears to involve the stimulation of differentiation of goblet cells and M2 macrophages.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-33/sangre , Mucosa Intestinal/metabolismo , Macrófagos/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Células Cultivadas , Células Caliciformes/efectos de los fármacos , Células HT29 , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/farmacología , Interleucina-33/uso terapéutico , Intestinos/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA