Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
bioRxiv ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38826450

RESUMEN

Fibrosis drives end-organ damage in many diseases. However, clinical trials targeting individual upstream activators of fibroblasts, such as TGFß, have largely failed. Here, we target the leukemia inhibitory factor receptor (LIFR) as a "master amplifier" of multiple upstream activators of lung fibroblasts. In idiopathic pulmonary fibrosis (IPF), the most common fibrotic lung disease, we found that lung myofibroblasts had high LIF expression. Further, TGFß1, one of the key drivers of fibrosis, upregulated LIF expression in IPF fibroblasts. In vitro anti-LIFR antibody blocking on human IPF lung fibroblasts reduced induction of profibrotic genes downstream of TGFß1, IL-4 and IL-13. Further, siRNA silencing of LIFR in IPF precision cut lung slices reduced expression of fibrotic proteins. Together, we find that LIFR drives an autocrine positive feedback loop that amplifies and sustains pathogenic activation of IPF fibroblasts downstream of multiple external stimuli, implicating LIFR as a therapeutic target in fibrosis. Significance Statement: Fibroblasts have a central role in the pathogenesis of fibrotic diseases. However, due to in part to multiple profibrotic stimuli, targeting a single activator of fibroblasts, like TGFß, has not yielded successful clinical treatments. We hypothesized that a more effective therapeutic strategy is identifying a downstream "master amplifier" of a range of upstream profibrotic stimuli. This study identifies the leukemia inhibitory factor receptor (LIFR) on fibrotic lung fibroblasts amplifies multiple profibrotic stimuli, such as IL-13 and TGFß. Blocking LIFR reduced fibrosis in ex vivo lung tissue from patients with idiopathic pulmonary fibrosis (IPF). LIFR, acting as a master amplifier downstream of fibroblast activation, offers an alternative therapeutic strategy for fibrotic diseases.

2.
Am J Respir Crit Care Med ; 208(11): 1177-1195, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37756440

RESUMEN

Rationale: Despite the importance of inflammation in chronic obstructive pulmonary disease (COPD), the immune cell landscape in the lung tissue of patients with mild-moderate disease has not been well characterized at the single-cell and molecular level. Objectives: To define the immune cell landscape in lung tissue from patients with mild-moderate COPD at single-cell resolution. Methods: We performed single-cell transcriptomic, proteomic, and T-cell receptor repertoire analyses on lung tissue from patients with mild-moderate COPD (n = 5, Global Initiative for Chronic Obstructive Lung Disease I or II), emphysema without airflow obstruction (n = 5), end-stage COPD (n = 2), control (n = 6), or donors (n = 4). We validated in an independent patient cohort (N = 929) and integrated with the Hhip+/- murine model of COPD. Measurements and Main Results: Mild-moderate COPD lungs have increased abundance of two CD8+ T cell subpopulations: cytotoxic KLRG1+TIGIT+CX3CR1+ TEMRA (T effector memory CD45RA+) cells, and DNAM-1+CCR5+ T resident memory (TRM) cells. These CD8+ T cells interact with myeloid and alveolar type II cells via IFNG and have hyperexpanded T-cell receptor clonotypes. In an independent cohort, the CD8+KLRG1+ TEMRA cells are increased in mild-moderate COPD lung compared with control or end-stage COPD lung. Human CD8+KLRG1+ TEMRA cells are similar to CD8+ T cells driving inflammation in an aging-related murine model of COPD. Conclusions: CD8+ TEMRA cells are increased in mild-moderate COPD lung and may contribute to inflammation that precedes severe disease. Further study of these CD8+ T cells may have therapeutic implications for preventing severe COPD.


Asunto(s)
Linfocitos T CD8-positivos , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Animales , Ratones , Modelos Animales de Enfermedad , Proteómica , Pulmón/metabolismo , Inflamación , Receptores de Antígenos de Linfocitos T
3.
Arthritis Res Ther ; 25(1): 130, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37496081

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is a systemic chronic inflammatory disease that leads to joint destruction and functional disability due to the targeting of self-antigens present in the synovium, cartilage, and bone. RA is caused by a number of complex factors, including genetics, environment, dietary habits, and altered intestinal microbial flora. Microorganisms in the gut bind to nod-like receptors and Toll-like receptors to regulate the immune system and produce various metabolites, such as short-chain fatty acids (SCFAs) that interact directly with the host. Faecalibacterium prausnitzii is a representative bacterium that produces butyrate, a well-known immunomodulatory agent in the body, and this microbe exerts anti-inflammatory effects in autoimmune diseases. METHODS: In this study, F. prausnitzii was administered in a mouse model of RA, to investigate RA pathology and changes in the intestinal microbial flora. Using collagen-induced arthritic mice, which is a representative animal model of RA, we administered F. prausnitzii orally for 7 weeks. RESULTS: The arthritis score and joint tissue damage were decreased in the mice administered F. prausnitzii compared with the vehicle-treated group. In addition, administration of F. prausnitzii reduced the abundance of systemic immune cells that secrete the pro-inflammatory cytokine IL-17 and induced changes in SCFA concentrations and the intestinal microbial flora composition. It also resulted in decreased lactate and acetate concentrations, an increased butyrate concentration, and altered compositions of bacteria known to exacerbate or improve RA. CONCLUSION: These results suggest that F. prausnitzii exerts a therapeutic effect on RA by regulation of IL-17 producing cells. In addition, F. prausnitzii modify the microbial flora composition and short chain fatty acids in experimental RA mouse model.


Asunto(s)
Artritis Reumatoide , Faecalibacterium prausnitzii , Ratones , Animales , Faecalibacterium prausnitzii/metabolismo , Interleucina-17/metabolismo , Ácidos Grasos Volátiles/metabolismo , Modelos Animales de Enfermedad , Butiratos , Artritis Reumatoide/tratamiento farmacológico
4.
Thorax ; 77(10): 1041-1044, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35907639

RESUMEN

Although interstitial lung disease (ILD) causes significant morbidity and mortality in rheumatoid arthritis (RA), it is difficult to predict the development or progression of ILD, emphasising the need for improved discovery through minimally invasive diagnostic tests. Aptamer-based proteomic profiling was used to assess 1321 proteins from 159 patients with rheumatoid arthritis with interstitial lung disease (RA-ILD), RA without ILD, idiopathic pulmonary fibrosis and healthy controls. Differential expression and gene set enrichment analyses revealed molecular signatures that are strongly associated with the presence and severity of RA-ILD and provided insight into unexplored pathways of disease. These warrant further study as non-invasive diagnostic tools and future therapeutic targets.


Asunto(s)
Artritis Reumatoide , Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares Intersticiales , Humanos , Proteómica , Enfermedades Pulmonares Intersticiales/complicaciones , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/complicaciones , Artritis Reumatoide/genética , Artritis Reumatoide/complicaciones
5.
Med ; 3(7): 481-518.e14, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35649411

RESUMEN

BACKGROUND: Pro-inflammatory fibroblasts are critical for pathogenesis in rheumatoid arthritis, inflammatory bowel disease, interstitial lung disease, and Sjögren's syndrome and represent a novel therapeutic target for chronic inflammatory disease. However, the heterogeneity of fibroblast phenotypes, exacerbated by the lack of a common cross-tissue taxonomy, has limited our understanding of which pathways are shared by multiple diseases. METHODS: We profiled fibroblasts derived from inflamed and non-inflamed synovium, intestine, lungs, and salivary glands from affected individuals with single-cell RNA sequencing. We integrated all fibroblasts into a multi-tissue atlas to characterize shared and tissue-specific phenotypes. FINDINGS: Two shared clusters, CXCL10+CCL19+ immune-interacting and SPARC+COL3A1+ vascular-interacting fibroblasts, were expanded in all inflamed tissues and mapped to dermal analogs in a public atopic dermatitis atlas. We confirmed these human pro-inflammatory fibroblasts in animal models of lung, joint, and intestinal inflammation. CONCLUSIONS: This work represents a thorough investigation into fibroblasts across organ systems, individual donors, and disease states that reveals shared pathogenic activation states across four chronic inflammatory diseases. FUNDING: Grant from F. Hoffmann-La Roche (Roche) AG.


Asunto(s)
Artritis Reumatoide , Membrana Sinovial , Animales , Artritis Reumatoide/genética , Fibroblastos/metabolismo , Fenotipo , Células del Estroma/metabolismo
6.
Sci Immunol ; 7(68): eabf2846, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35148199

RESUMEN

Macrophages regulate protective immune responses to infectious microbes, but aberrant macrophage activation frequently drives pathological inflammation. To identify regulators of vigorous macrophage activation, we analyzed RNA-seq data from synovial macrophages and identified SLAMF7 as a receptor associated with a superactivated macrophage state in rheumatoid arthritis. We implicated IFN-γ as a key regulator of SLAMF7 expression and engaging SLAMF7 drove a strong wave of inflammatory cytokine expression. Induction of TNF-α after SLAMF7 engagement amplified inflammation through an autocrine signaling loop. We observed SLAMF7-induced gene programs not only in macrophages from rheumatoid arthritis patients but also in gut macrophages from patients with active Crohn's disease and in lung macrophages from patients with severe COVID-19. This suggests a central role for SLAMF7 in macrophage superactivation with broad implications in human disease pathology.


Asunto(s)
Inflamación/inmunología , Activación de Macrófagos/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Transcriptoma/inmunología , Enfermedad Aguda , Adulto , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , COVID-19/genética , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Enfermedad Crónica , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Activación de Macrófagos/genética , RNA-Seq/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Análisis de la Célula Individual/métodos , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Transcriptoma/genética
7.
Front Immunol ; 12: 736196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867956

RESUMEN

The potential therapeutic effects of probiotic bacteria in rheumatoid arthritis (RA) remain controversial. Thus, this study aimed to discover potential therapeutic bacteria based on the relationship between the gut microbiome and rheumatoid factor (RF) in RA. Bacterial genomic DNA was extracted from the fecal samples of 93 RA patients and 16 healthy subjects. Microbiota profiling was conducted through 16S rRNA sequencing and bioinformatics analyses. The effects of Bifidobacterium strains on human peripheral blood mononuclear cells and collagen-induced arthritis (CIA) mice were assessed. Significant differences in gut microbiota composition were observed in patients with different RF levels. The relative abundance of Bifidobacterium and Collinsella was lower in RF-high than in RF-low and RF-negative RA patients, while the relative abundance of Clostridium of Ruminococcaceae family was higher in RF-high than in RF-low and RF-negative patients. Among 10 differentially abundant Bifidobacterium, B. longum RAPO exhibited the strongest ability to inhibit IL-17 secretion. Oral administration of B. longum RAPO in CIA mice, obese CIA, and humanized avatar model significantly reduced RA incidence, arthritis score, inflammation, bone damage, cartilage damage, Th17 cells, and inflammatory cytokine secretion. Additionally, B. longum RAPO significantly inhibited Th17 cells and Th17-related genes-IL-17A, IRF4, RORC, IL-21, and IL-23R-in the PBMCs of rheumatoid arthritis patients. Our findings suggest that B. longum RAPO may alleviate RA by inhibiting the production of IL-17 and other proinflammatory mediators. The safety and efficacy of B. longum RAPO in patients with RA and other autoimmune disorders merit further investigation.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/terapia , Bifidobacterium/inmunología , Bifidobacterium/aislamiento & purificación , Microbioma Gastrointestinal/inmunología , Probióticos/uso terapéutico , Factor Reumatoide/sangre , Adulto , Animales , Artritis Experimental/inmunología , Artritis Experimental/terapia , Bifidobacterium/genética , Biodiversidad , Estudios de Casos y Controles , Femenino , Microbioma Gastrointestinal/genética , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Ratones Obesos , Ratones SCID , Persona de Mediana Edad , Células Th17/inmunología
8.
Rheumatology (Oxford) ; 60(2): 708-716, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32789440

RESUMEN

OBJECTIVE: Patients with RA commonly use gastrointestinal (GI) protective drugs for treatment and prevention of drug-associated GI injuries. However, how these drugs affect the gut microbiota in RA patients remains unknown. The objective of this study was to examine the gut microbiota of RA patients according to use of GI protective drugs such as proton pump inhibitors (PPIs), histamine 2-receptor antagonists and rebamipide. METHODS: Faecal samples were obtained from 15 healthy controls and 32 RA patients who were receiving PPI, histamine 2-receptor antagonist or rebamipide. Bacterial DNA was extracted from the faecal samples and 16S rRNA sequencing was performed. Microbial composition and function were analysed using Quantitative Insights Into Microbial Ecology and Phylogenetic Investigation of Communities by Reconstruction of Unobserved States. RESULTS: RA patients exhibited reduced diversity and altered composition of the gut microbiota compared with healthy controls. The gut microbiota of RA patients receiving acid-suppressing drugs, particularly PPIs, was distinct from that of RA patients receiving rebamipide (PPI vs rebamipide, P = 0.005). Streptococcus was enriched in RA patients receiving PPI, while Clostridium bolteae was enriched in RA patients receiving rebamipide. The gut microbiota of PPI users was abundant with microbial functional pathway involved in the production of virulence factors. This featured microbial function was positively correlated with relative abundance of Streptococcus, the differentially abundant taxa of PPI users. CONCLUSION: The gut microbiota of RA patients receiving PPIs was distinguishable from that of those receiving rebamipide. The enriched virulent function in the gut microbiota of PPI users suggests that inappropriate PPI use may be harmful in RA patients.


Asunto(s)
Alanina/análogos & derivados , Artritis Reumatoide/tratamiento farmacológico , Bacterias/aislamiento & purificación , Enfermedades Gastrointestinales/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Inhibidores de la Bomba de Protones/uso terapéutico , Quinolonas/farmacología , Alanina/farmacología , Artritis Reumatoide/complicaciones , Bacterias/genética , ADN Bacteriano/análisis , Inhibidores Enzimáticos/farmacología , Femenino , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/microbiología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
9.
Microorganisms ; 8(5)2020 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-32466189

RESUMEN

Faecalibacterium prausnitzii is a major commensal bacterium in the human gut. It produces short-chain fatty acids that promote intestinal health. However, the bacterium is extremely oxygen-sensitive, making it difficult to develop as a probiotic. To facilitate practical application of F. prausnitzii, we investigated factors that affect its growth and mammalian gut colonization. We evaluated cross-feeding interactions between F. prausnitzii and seven Bifidobacterium strains, and the anti-inflammatory properties of bacterial metabolites produced in co-culture, in vitro and in vivo. Co-culture of F. prausnitzii and Bifidobacterium catenulatum, with fructooligosaccharides as an energy source, resulted in the greatest viable cell-count and butyrate production increases. Further, the co-culture supernatant reduced the amount of proinflammatory cytokines produced by HT-29 cells and RAW 264.7 macrophages, an effect that was similar to that of butyrate. Furthermore, feeding mice both Faecalibacterium and Bifidobacterium enhanced F. prausnitzii gut colonization. Finally, feeding the co-culture supernatant decreased interleukin 8 levels in the colon and increased butyrate levels in the cecum in the dextran sodium sulfate-induced colitis mouse model. These observations indicate that the Faecalibacterium-Bifidobacterium co-culture exerts an anti-inflammatory effect by promoting F. prausnitzii survival and short-chain fatty acid production, with possible implications for the treatment of inflammatory bowel disease.

10.
Int J Mol Sci ; 21(2)2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-31936703

RESUMEN

Short-chain fatty acids (SCFAs), especially butyrate, produced in mammalian intestinal tracts via fermentation of dietary fiber, are known biofunctional compounds in humans. However, the variability of fermentable fiber consumed on a daily basis and the diversity of gut microbiota within individuals often limits the production of short-chain fatty acids in the human gut. In this study, we attempted to enhance the butyrate levels in human fecal samples by utilizing butyl-fructooligosaccharides (B-FOS) as a novel prebiotic substance. Two major types of B-FOS (GF3-1B and GF3-2B), composed of short-chain fructooligosaccharides (FOS) bound to one or two butyric groups by ester bonds, were synthesized. Qualitative analysis of these B-FOS using Fourier transform infrared (FT-IR) spectroscopy, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS), nuclear magnetic resonance (NMR) and low-resolution fast-atom bombardment mass spectra (LR-FAB-MS), showed that the chemical structure of GF3-1B and GF3-2B were [O-(1-buty-ß-D-fru-(2→1)-O-ß-D-fru-(2→1)-O-ß-D-fru-O-α-D-glu] and [O-(1-buty)-ß-D-fru-(2→1)-O-ß-D-fru-(2→1)-O-(4-buty)-ß-D-fru-O-α-D-glu], respectively. The ratio of these two compounds was approximately 5:3. To verify their biofunctionality as prebiotic oligosaccharides, proliferation and survival patterns of human fecal microbiota were examined in vitro via 16S rRNA metagenomics analysis compared to a positive FOS control and a negative control without a carbon source. B-FOS treatment showed different enrichment patterns on the fecal microbiota community during fermentation, and especially stimulated the growth of major butyrate producing bacterial consortia and modulated specific butyrate producing pathways with significantly enhanced butyrate levels. Furthermore, the relative abundance of Fusobacterium and ammonia production with related metabolic genes were greatly reduced with B-FOS and FOS treatment compared to the control group. These findings indicate that B-FOS differentially promotes butyrate production through the enhancement of butyrate-producing bacteria and their metabolic genes, and can be applied as a novel prebiotic compound in vivo.


Asunto(s)
Butiratos/metabolismo , Oligosacáridos/química , Oligosacáridos/metabolismo , Prebióticos/análisis , Adulto , Amoníaco/análisis , Bacterias/clasificación , Bacterias/metabolismo , Biodiversidad , Fibras de la Dieta , Ácidos Grasos Volátiles/metabolismo , Heces/química , Heces/microbiología , Femenino , Fermentación , Microbioma Gastrointestinal , Humanos , Masculino , Metagenoma , Espectroscopía Infrarroja por Transformada de Fourier , Adulto Joven
11.
Microb Cell Fact ; 18(1): 110, 2019 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-31196110

RESUMEN

BACKGROUND: Lactic acid bacteria (LAB) are known to have a significant ability to colonize the human intestinal tract and adhere to the surface of intestinal epithelial cells. Among the various lactic acid bacteria, exopolysaccharide (EPS) producing strains are known to provide a variety of health benefits for their hosts (e.g. anti-inflammatory, antioxidant, antitumor and stress tolerant effects). Recently, EPSs and EPS-producing lactic acid cultures have gained interest within the food industry and are playing important roles as biothickeners and texturizing agents due to their hydrocolloidal nature. In this study, 156 lactic acid bacterial strains isolated from fecal samples of healthy young children were screened and evaluated for active EPS-production capability. RESULTS: Among the various human origin lactic acid flora isolated, Weissella confusa VP30 showed the highest EPS productivity and its EPS producing properties were characterized under various cultural conditions in this research. To document the safety of W. confusa VP30, antibiotic resistance, hemolytic, and ammonia production properties were evaluated in addition. No significant negative results were observed. The maximum EPS production by W. confusa VP30 was 59.99 ± 0.91 g/l after 48 h of cultivation in media containing 10% sucrose, far exceeding EPS production by other bacterial strains reported elsewhere. Based on gel permeation chromatography (GPC), the molecular weight of EPS produced by W. confusa VP30 was 3.8 × 106 Da. Structural analysis of the released EPS fraction by 13C and 1H nuclear magnetic resonance (NMR) spectroscopy revealed that W. confusa VP30 can produce dextran with glucose units linked with 96.5% α (1 → 6) glycosidic bonds and 3.5% α (1 → 3) branches. CONCLUSION: The high EPS production capability and safety of W. confusa VP30 justify food industry consideration of this cell strain for further evaluation and potential industrial use.


Asunto(s)
Heces/microbiología , Polisacáridos Bacterianos/biosíntesis , Weissella/aislamiento & purificación , Preescolar , ADN Bacteriano/genética , Femenino , Humanos , Lactante , Espectroscopía de Resonancia Magnética , Masculino , Filogenia , Polisacáridos Bacterianos/química , ARN Ribosómico 16S/genética , Sacarosa/metabolismo , Weissella/clasificación , Weissella/genética , Weissella/metabolismo
12.
J Clin Med ; 8(5)2019 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-31100891

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation of the joints and extra-articular manifestations. Recent studies have shown that microorganisms affect RA pathogenesis. However, few studies have examined the microbial distribution of early RA patients, particularly female patients. In the present study, we investigated the gut microbiome profile and microbial functions in early RA female patients, including preclinical and clinically apparent RA cases. Changes in microbiological diversity, composition, and function in each group were analyzed using quantitative insights into microbial ecology (QIIME) and phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt). The results revealed the dysbiosis due to decreased diversity in the early RA patients compared with healthy subjects. There were significant differences in the microbial distribution of various taxa from phylum to genus levels between healthy subjects and early RA patients. Phylum Bacteroidetes was enriched in early RA patients, while Actinobacteria, including the genus Collinsella, was enriched in healthy subjects. Functional analysis based on clusters of orthologous groups revealed that the genes related to the biosynthesis of menaquinone, known to be derived from gram-positive bacteria, were enriched in healthy subjects, while iron transport-related genes were enriched in early RA patients. Genes related to the biosynthesis of lipopolysaccharide, the gram-negative bacterial endotoxin, were enriched in clinically apparent RA patients. The obvious differences in microbial diversity, taxa, and associated functions of the gut microbiota between healthy subjects and early RA patients highlight the involvement of the gut microbiome in the early stages of RA.

13.
Microorganisms ; 7(4)2019 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018530

RESUMEN

Rotavirus infection is the most common diarrheal disease worldwide in children under five years of age, and it often results in death. In recent years, research on the relationship between rotavirus and probiotics has shown that probiotics are effective against diarrhea. A clinical trial has reported that Bifidobacterium longum BORI reduced diarrhea induced by rotavirus. The present work investigated the anti-rotaviral effect of B. longum BORI by cytopathic effect observation and real time cell analyses. Our study found that B. longum BORI showed strong anti-rotaviral effect when incubated with MA104 cells prior to viral infection, suggesting that the probiotic does in fact interfere with the interaction of viruses and host cells. It is believed that the efficacy is due to low-molecular weight and non-protein components derived from B. longum BORI. This discovery can help broaden the industrial application of B. longum BORI, which has been proven to be a safe and effective probiotic.

14.
Sci Rep ; 4: 4980, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24828152

RESUMEN

The Met receptor tyrosine kinase is an attractive target for cancer therapy as it promotes invasive tumor growth. SAIT301 is a novel anti-Met antibody, which induces LRIG1-mediated Met degradation and inhibits tumor growth. However, detailed downstream mechanism by which LRIG1 mediates target protein down-regulation is unknown. In the present study, we discovered that SAIT301 induces ubiquitination of LRIG1, which in turn promotes recruitment of Met and LRIG1 complex to the lysosome through its interaction with Hrs, resulting in concomitant degradation of both LRIG1 and Met. We also identified USP8 as a LRIG1-specific deubiquitinating enzyme, reporting the interaction between USP8 and LRIG1 for the first time. SAIT301 triggers degradation of LRIG1 by inhibiting the interaction of LRIG1 and USP8, which regulates ubiquitin modification and stability of LRIG1. In summary, SAIT301 employs ubiquitination of LRIG1 for its highly effective Met degradation. This unique feature of SAIT301 enables it to function as a fully antagonistic antibody without Met activation. We found that USP8 is involved in deubiquitination of LRIG1, influencing the efficiency of Met degradation. The relation of Met, LRIG1 and USP8 strongly supports the potential clinical benefit of a combination treatment of a USP8 inhibitor and a Met inhibitor, such as SAIT301.


Asunto(s)
Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/fisiología , Línea Celular Tumoral , Humanos , Lisosomas/metabolismo , Proteolisis
15.
Mol Cells ; 34(6): 523-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23180291

RESUMEN

c-Met, the high affinity receptor for hepatocyte growth factor (HGF), is one of the most frequently activated tyrosine kinases in many human cancers and a target for cancer therapy. However, inhibitory targeting of c-Met with antibodies has proven difficult, because most antibodies have intrinsic agonist activity. Therefore, the strategy for reducing the agonism is critical for successful development of cancer therapies based on anti-c-Met antibodies. Here we developed a mechanism-based assay method for rapid screening of anti-c-Met antibodies, involving the determination of Akt phosphorylation and c-Met degradation for agonism and efficacy, respectively. Using the method, we identified an antibody, F46, that binds to human c-Met with high affinity (Kd = 2.56 nM) and specificity, and induces the degradation of c-Met in multiple cancer cells (including MKN45, a gastric cancer cell line) with minimal activation of c-Met signaling. F46 induced c-Met internalization in both HGF-dependent and HGF-independent cells, suggesting that the degradation of c-Met results from antibody-mediated receptor internalization. Furthermore, F46 competed with HGF for binding to c-Met, resulting in the inhibition of both HGF-mediated invasion and angiogenesis. Consistently, F46 inhibited the proliferation of MKN45 cells, in which c-Met is constitutively activated in an HGF-independent manner. Xenograft analysis revealed that F46 markedly inhibits the growth of subcutaneously implanted gastric and lung tumors. These results indicate that F46, identified by a novel mechanism-based assay, induces c-Met degradation with minimal agonism, implicating a potential role of F46 in therapy of human cancers.


Asunto(s)
Isoanticuerpos/química , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Proliferación Celular , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Isoanticuerpos/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Int Immunopharmacol ; 11(4): 511-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21238621

RESUMEN

In the present study, we examined the inhibitive effect of ginsenoside Rh1 on oxazolone-induced atopic dermatitis-like skin lesions in hairless mice. Oral administration of ginsenoside Rh1 improved clinical symptoms, and it was confirmed by histophathological analysis. In ginsenoside Rh1 (20mg/kg) group, ear swellings and ear weights were significantly lower than the control group. Moreover, elevation of IL-6 and total IgE levels in serum were suppressed by ginsenoside Rh1 (20mg/kg). In addition, ginsenoside Rh1 (20mg/kg) significantly increased mRNA expression of IFNγ and Foxp3, and slightly decreased IL-4 expression in draining lymph nodes. The results suggest that ginsenoside Rh1 can alleviate inflammatory symptoms in atopic dermatitis (AD) by reduction of IgE and IL-6 levels in peripheral blood, increase of Foxp3 expression in draining lymph nodes and suppression of inflammation in skin regions. Indeed, ginsenoside Rh1 exhibited therapeutic possibility in immune disorders.


Asunto(s)
Dermatitis Atópica/prevención & control , Ginsenósidos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Oxazolona/inmunología , Piel/efectos de los fármacos , Administración Oral , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/inmunología , Ginsenósidos/administración & dosificación , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Factores Inmunológicos/administración & dosificación , Interferón gamma/biosíntesis , Interleucina-6/biosíntesis , Interleucina-6/sangre , Interleucina-6/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Pelados , Estructura Molecular , Piel/inmunología , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...