Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Oncology ; 102(4): 354-365, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37812924

RESUMEN

INTRODUCTION: Gastric cancer (GC) remains a common health concern worldwide and is the third leading cause of death in Japan. It can be broadly classified into gastric and intestinal mucin phenotypes using immunohistochemistry. We previously reported numerous associations of kinesin family member (KIF) genes and mucin phenotypes with GC. However, no previous studies have reported on the importance of KIF18B in GC using immunostaining. Thus, in this study, we investigated the expression and functions of KIF18B, which is highly expressed in gastric mucin phenotype GC. METHODS: We performed RNA-seq of gastric and intestinal mucin type GCs, and clinicopathological studies of the KIF18B we found were performed using 96 GC cases. We also performed functional analysis using GC-derived cell lines. RESULT: RNA-seq showed the upregulation of matrisome-associated genes in gastric mucin phenotype GC and a high expression of KIF18B. KIF18B was detected in 52 of the 96 GC cases (54%) through immunohistochemistry. Low KIF18B expression was significantly associated with poor overall survival (p < 0.01). Other molecules that were significantly associated with KIF18B were MUC5AC and claudin 18; these were also significantly associated with the gastric mucin phenotype. KIF18B small interfering RNA (siRNA)-transfected GC cells showed greater growth and spheroid colony formation than the negative control siRNA-transfected cells. Furthermore, expression of snail family transcriptional repressor 1 and cadherin 2 was significantly increased and that of cadherin 1 was significantly decreased in KIF18B siRNA-transfected GC cells. CONCLUSION: These findings not only suggest that KIF18B may be a useful prognostic marker, but also provide insight into the pathogenesis of the GC phenotype.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Cinesinas/genética , Cinesinas/metabolismo , Mucinas Gástricas/genética , Mucinas Gástricas/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , ARN Interferente Pequeño , Transición Epitelial-Mesenquimal/genética , Fenotipo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
2.
Mol Cell Proteomics ; 21(11): 100421, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36182101

RESUMEN

Helicobacter pylori colonizes the stomach of half of the human population. Most H. pylori are located in the mucus layer, which is mainly comprised by glycosylated mucins. Using mass spectrometry, we identified 631 glycans (whereof 145 were fully characterized and the remainder assigned as compositions) on mucins isolated from 14 Helicobacter spp.-infected and 14 Helicobacter spp.-noninfected stomachs. Only six identified glycans were common to all individuals, from a total of 60 to 189 glycans in each individual. An increased number of unique glycan structures together with an increased intraindividual diversity and larger interindividual variation were identified among O-glycans from Helicobacter spp.-infected stomachs compared with noninfected stomachs. H. pylori strain J99, which carries the blood group antigen-binding adhesin (BabA), the sialic acid-binding adhesin (SabA), and the LacdiNAc-binding adhesin, bound both to Lewis b (Leb)-positive and Leb-negative mucins. Among Leb-positive mucins, H. pylori J99 binding was higher to mucins from Helicobacter spp.-infected individuals than noninfected individuals. Statistical correlation analysis, binding experiments with J99 wt, and J99ΔbabAΔsabA and inhibition experiments using synthetic glycoconjugates demonstrated that the differences in H. pylori-binding ability among these four groups were governed by BabA-dependent binding to fucosylated structures. LacdiNAc levels were lower in mucins that bound to J99 lacking BabA and SabA than in mucins that did not, suggesting that LacdiNAc did not significantly contribute to the binding. We identified 24 O-glycans from Leb-negative mucins that correlated well with H. pylori binding whereof 23 contained α1,2-linked fucosylation. The large and diverse gastric glycan library identified, including structures that correlated with H. pylori binding, could be used to select glycodeterminants to experimentally investigate further for their importance in host-pathogen interactions and as candidates to develop glycan-based therapies.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Mucinas Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Helicobacter pylori/metabolismo , Polisacáridos/metabolismo
3.
Cancer Sci ; 113(11): 3852-3863, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35959971

RESUMEN

Gastric cancer is the second leading cause of cancer deaths worldwide, and more understanding of its molecular basis is urgently needed. Gastric gland mucin secreted from pyloric gland cells, mucous neck cells, and cardiac gland cells of the gastric mucosa harbors unique O-glycans carrying terminal α1,4-linked N-acetylglucosamine (αGlcNAc) residues. We previously reported that αGlcNAc loss correlated positively with poor outcomes for patients with differentiated-type gastric cancer. However, the molecular mechanisms underlying these outcomes remained poorly understood. Here, we examined the effects of upregulated αGlcNAc expression on malignant phenotypes of the differentiated-type gastric cancer cell lines, AGS and MKN7. Upregulation of αGlcNAc following ectopic expression of its biosynthetic enzyme attenuated cell proliferation, motility, and invasiveness of AGS and MKN7 cells in vitro. Moreover, AGS cell tumorigenicity was significantly suppressed by αGlcNAc overexpression in a xenograft model. To define the molecular mechanisms underlying these phenotypes, we investigated αGlcNAc binding proteins in AGS cells and identified Mucin-1 (MUC1) and podocalyxin. Both proteins were colocalized with αGlcNAc on human gastric cancer cells. We also found that αGlcNAc was bound to MUC1 in murine normal gastric mucosa. When we assessed the effects of αGlcNAc binding to MUC1, we found that αGlcNAc blocked galectin-3 binding to MUC1, phosphorylation of the MUC1 C-terminus, and recruitment of Src and ß-catenin to that C-terminus. These results suggest that αGlcNAc regulates cancer cell phenotypes by dampening MUC1 signal transduction.


Asunto(s)
Adenocarcinoma , Neoplasias Gástricas , Humanos , Ratones , Animales , Neoplasias Gástricas/patología , Acetilglucosamina/metabolismo , Mucina 6/metabolismo , Mucina-1/genética , Adenocarcinoma/patología , Mucinas Gástricas/metabolismo , Mucosa Gástrica/patología , Transducción de Señal
4.
Histochem Cell Biol ; 157(6): 671-684, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35353213

RESUMEN

Gastric gland mucin consists of core protein MUC6 with residues heavily glycosylated by unique O-glycans carrying α1,4-linked N-acetylglucosamine (αGlcNAc). αGlcNAc-glycosylated MUC6 protein is seen in normal gastric and duodenal glands. Decreased αGlcNAc glycosylation on MUC6-positive tumor cells is often observed in premalignant lesions of the stomach, pancreas, and bile duct, and decreased MUC6 expression is seen in invasive cancer of these organs. Lung cancer (LC) is the most common cause of cancer death worldwide. Recently, the adenocarcinoma subtype has become the most common histological subtype of LC, and one of its invasive forms is invasive mucinous adenocarcinoma (IMA). Currently, prognostic markers of LC IMA are unknown. Here, we analyzed MUC5AC, MUC6, and αGlcNAc expression in 54 IMA LC cases. MUC5AC was positively expressed in 50 (93%), MUC6 in 38 (70%), and αGlcNAc in 19 (35%). Each expression level was scored from 0 to 3. The αGlcNAc expression score was significantly decreased relative to MUC6 (P < 0.001). Interestingly, disease-free survival was significantly higher in MUC6-positive than MUC6-negative cases based on the log-rank test (P = 0.021). For in vitro analysis, we ectopically expressed MUC6 in A549 cells, derived from LC and harboring a KRAS mutation. MUC6-expressing A549 cells showed significantly lower proliferation, motility, and invasiveness than control cells. Finally, F-actin staining in MUC6-expressing cells revealed a decrease or loss of filopodia associated with decreased levels of FSCN transcripts, which encodes an actin-bundling protein fascin1 necessary for cell migration. We conclude that MUC6 expression is a preferable prognostic biomarker in IMA LC.


Asunto(s)
Adenocarcinoma Mucinoso , Adenocarcinoma , Adenocarcinoma/metabolismo , Mucinas Gástricas/metabolismo , Humanos , Pulmón/metabolismo , Mucina 5AC/metabolismo , Mucina 6/análisis , Mucina 6/metabolismo , Pronóstico
5.
BMC Cancer ; 21(1): 1138, 2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34688251

RESUMEN

BACKGROUND: Compared to conventional adenocarcinoma (CA), mucin-producing adenocarcinoma (MPA) is an uncommon histological subtype and is usually separated from other histological types and has been evaluated separately. The objective was to compare the clinicopathological characteristics and survivals of MPA with CA. METHODS: We retrospectively analyzed 1515 MPA patients in SEER database. Log-rank tests and KM survival curves were applied to determine the differences in overall survival (OS) and cancer specific survival (CSS) time. RESULTS: No significant differences were noted in OS and CSS time. The MPA patients who were treated with surgery and chemotherapy exhibited longer OS and CSS time periods than those without treatment. MPA patients treated with radiotherapy exhibited similar OS and CSS time with those without radiotherapy. MPA was not a prognostic factor of survival. CONCLUSIONS: MPA was a rare histological type of gastric cancer. Patients with MPA exhibited similar prognosis with those with CA. Surgery and chemotherapy were effective treatments for patients with MPA.


Asunto(s)
Adenocarcinoma/mortalidad , Adenocarcinoma/fisiopatología , Mucinas Gástricas/metabolismo , Programa de VERF/normas , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Resultado del Tratamiento
6.
Molecules ; 26(22)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34833889

RESUMEN

The aim of this study was to investigate binding interactions between ß-lactoglobulin (BLG) and two different mucins, bovine submaxillary mucins (BSM) and porcine gastric mucin (PGM), using intrinsic and extrinsic fluorescence spectroscopies. Intrinsic fluorescence spectra showed an enhanced decrease of fluorescence intensity of BLG at all pH conditions when BLG was mixed with PGM rather than with BSM. We propose that, unlike BSM, the tertiary structure of PGM changes and the hydrophobic regions are exposed at pH 3 due to protonation of negatively charged residues. Results suggest that PGM also facilitated the structural unfolding of BLG and its binding with PGM by a hydrophobic interaction, especially at acidic pH, which was further supported by extrinsic fluorescence spectroscopy. Hydrophobic interaction is suggested as the dominant interaction mechanism between BLG and PGM at pH 3, whereas electrostatic interaction is the dominant one between BLG and BSM.


Asunto(s)
Mucinas Gástricas/metabolismo , Lactoglobulinas/metabolismo , Mucinas/metabolismo , Adsorción , Animales , Bovinos , Mucinas Gástricas/química , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Mucosa Intestinal/metabolismo , Lactoglobulinas/química , Mucinas/química , Espectrometría de Fluorescencia/métodos , Glándula Submandibular/metabolismo , Porcinos
7.
Curr Top Microbiol Immunol ; 421: 179-207, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31123890

RESUMEN

The human stomach is a harsh and fluctuating environment for bacteria with hazards such as gastric acid and flow through of gastric contents into the intestine. H. pylori gains admission to a stable niche with nutrient access from exudates when attached to the epithelial cells under the mucus layer, whereof adherence to glycolipids and other factors provides stable and intimate attachment. To reach this niche, H. pylori must overcome mucosal defence mechanisms including the continuously secreted mucus layer, which provides several layers of defence: (1) mucins in the mucus layer can bind H. pylori and transport it away from the gastric niche with the gastric emptying, (2) mucins can inhibit H. pylori growth, both via glycans that can have antibiotic like function and via an aggregation-dependent mechanism, (3) antimicrobial peptides (AMPs) have antimicrobial activity and are retained in a strategic position in the mucus layer and (4) underneath the mucus layer, the membrane-bound mucins provide a second barrier, and can function as releasable decoys. Many of these functions are dependent on H. pylori interactions with host glycan structures, and both the host glycosylation and concentration of antimicrobial peptides change with infection and inflammation, making these interactions dynamic. Here, we review our current understanding of mucin glycan and antimicrobial peptide-dependent host defence mechanisms against H. pylori infection.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Mucinas Gástricas/metabolismo , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Antibacterianos/química , Antibacterianos/metabolismo , Mucinas Gástricas/química , Mucosa Gástrica/química , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/microbiología , Humanos
8.
J Pathol ; 247(1): 35-47, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30168144

RESUMEN

Chronic inflammation and intestinal metaplasia are strongly associated with gastric carcinogenesis. Kras activation and Pten deletion are observed in intestinal-type gastric cancer, and Cdh1 mutation is associated with diffuse-type gastric cancer. Although various mouse models of gastric carcinogenesis have been reported, few mouse lines enable gene manipulation selectively in the stomach. Here we established a Tff1-Cre bacterial artificial chromosome transgenic mouse line in an attempt to induce gene modification specifically in the gastric pit lineage. In the stomach, Tff1-Cre-mediated recombination was most evident in the pit lineage in the corpus and in entire antral glands; recombination was also observed in a few gastric chief and parietal cells. Outside the stomach, recombination was patchy throughout the intestines, and particularly frequently in the duodenum (Brunner glands), cecum, and proximal colon. In the stomachs of Tff1-Cre;LSL-KrasG12D mice, proliferating cell clusters expanded throughout the corpus glands, with foveolar cell expansion with ectopic Alcian blue-positive mucins, oxyntic atrophy, and pseudopyloric changes with spasmolytic polypeptide-expressing metaplasia; however, gastric cancer was not observed even at 12 months of age. Corpus-derived organoids from Tff1-Cre;LSL-KrasG12D mice exhibited accelerated growth and abnormal differentiation with a loss of chief and parietal cell markers. Tff1-Cre;Ptenflox/flox mice displayed similar changes to those seen in Tff1-Cre;LSL-KrasG12D mice, both with aberrant ERK activation within 3 months. In contrast, Tff1-Cre;Cdh1flox/flox mice initially showed signet ring-like cells that were rapidly lost with disruption of the mucosal surface, and later developed gastric epithelial shedding with hyperproliferation and loss of normal gastric lineages. Eventually, the glandular epithelium in Tff1-Cre;Cdh1flox/flox mice was completely replaced by squamous epithelium which expanded from the forestomach. Tff1-Cre mice offer an additional useful tool for studying gastric carcinogenesis both in vivo and in vitro. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Cadherinas/deficiencia , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Mucosa Gástrica/enzimología , Gastritis/enzimología , Fosfohidrolasa PTEN/deficiencia , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Gástricas/enzimología , Animales , Cadherinas/genética , Linaje de la Célula , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromosomas Artificiales Bacterianos , Mucinas Gástricas/genética , Mucinas Gástricas/metabolismo , Mucosa Gástrica/patología , Gastritis/genética , Gastritis/patología , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Integrasas/genética , Metaplasia , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Técnicas de Cultivo de Tejidos , Factor Trefoil-1/genética
9.
Appl Microbiol Biotechnol ; 104(13): 5943-5957, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32399588

RESUMEN

The first step in the development of Helicobacter pylori pathogenicity is the receptor-mediated adhesion to the gastric epithelium. Inhibition of outer membrane proteins of H. pylori (e.g. BabA) by antiadhesive drugs will contribute to reduced recolonization and infection. Pectin from apple inhibits the BabA and LPS-mediated adhesion of H. pylori to human stomach cells. Pectin-coated liposomes with encapsulated amoxicillin were characterized for polydispersity, zeta potential, encapsulation efficiency, stability, and amoxicillin release. Coated liposomes did not influence the viability of AGS and HT29-MTX cells up to 100 µg/mL but exert cytotoxicity against H. pylori at 10 µg/mL. Pectin-coating of liposomes provoked direct interaction and subsequent binding of the particles to surface structures of H. pylori, and interaction with mucus from porcine stomach and mucus secreted by HT29-MTX cells. Laser scanning microscopy of H. pylori and AGS cells together with liposomes indicated co-aggregation. The mucoadhesive effect seems interesting as stomach cells are covered by a mucus layer. H. pylori is able to penetrate and cross the mucin rapidly to reach pH-neutral epithelium to escape the acidic environment, followed by interaction with epithelial cells. In summary, all experimental evidence is consistent with a specific interaction of pectin-coated liposomes with mucins and surface structures of H. pylori. As the coated liposomes show mucoadhesion to the negatively charged mucins, docking to stomach mucin, mucus penetration, and recognition of and adhesion to H. pylori, they can be considered a novel type of multifunctional drug carriers for local antibiotic therapy against H. pylori. KEY POINTS: • Smart, multifunctional mucoadhesive liposomes • Specific targeting against BabA/LPS of Helicobacter pylori • Inhibition of bacterial adhesion of H. pylori to human host cells • Release of antibiotic cargo.


Asunto(s)
Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Helicobacter pylori/efectos de los fármacos , Liposomas/química , Pectinas/química , Adhesinas Bacterianas/metabolismo , Amoxicilina/química , Amoxicilina/farmacología , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Línea Celular , Mucinas Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Helicobacter pylori/metabolismo , Humanos , Lipopolisacáridos/metabolismo , Liposomas/metabolismo , Porcinos
10.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30745328

RESUMEN

Enteropathogenic Bacillus cereus causes foodborne infections due to the production of pore-forming enterotoxins in the intestine. Before that, spores have to be ingested, survive the stomach passage, and germinate. Thus, before reaching epithelial cells, B. cereus comes in contact with the intestinal mucus layer. In the present study, different aspects of this interaction were analyzed. Total RNA sequencing revealed major transcriptional changes of B. cereus strain F837/76 upon incubation with porcine gastric mucin (PGM), comprising genes encoding enterotoxins and further putative virulence factors, as well as proteins involved in adhesion to and degradation of mucin. Indeed, PGM was partially degraded by B. cereus via secreted, EDTA-sensitive proteases. The amount of enterotoxins detectable in culture media supplemented with PGM was also clearly increased. Tests of further strains revealed that enhancement of enterotoxin production upon contact with PGM is broadly distributed among B. cereus strains. Interestingly, evidence was found that PGM can also strain-specifically trigger germination of B. cereus spores and that vegetative cells actively move toward mucin. Overall, our data suggest that B. cereus is well adapted to the host environment due to massive transcriptome changes upon contact with PGM, attributing mucin an important and, thus far, neglected role in pathogenesis.


Asunto(s)
Bacillus cereus/metabolismo , Enterotoxinas/metabolismo , Enfermedades Transmitidas por los Alimentos/microbiología , Mucinas Gástricas/metabolismo , Mucosa Intestinal/microbiología , Animales , Bacillus cereus/genética , Bacillus cereus/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Esporas Bacterianas/genética , Esporas Bacterianas/crecimiento & desarrollo , Esporas Bacterianas/metabolismo , Porcinos
11.
Mol Cell Proteomics ; 16(5): 743-758, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28461410

RESUMEN

The mucin O-glycosylation of 10 individuals with and without gastric disease was examined in depth in order to generate a structural map of human gastric glycosylation. In the stomach, these mucins and their O-glycosylation protect the epithelial surface from the acidic gastric juice and provide the first point of interaction for pathogens such as Helicobacter pylori, reported to cause gastritis, gastric and duodenal ulcers and gastric cancer. The rational of the present study was to map the O-glycosylation that the pathogen may come in contact with. An enormous diversity in glycosylation was found, which varied both between individuals and within mucins from a single individual: mucin glycan chain length ranged from 2-13 residues, each individual carried 34-103 O-glycan structures and in total over 258 structures were identified. The majority of gastric O-glycans were neutral and fucosylated. Blood group I antigens, as well as terminal α1,4-GlcNAc-like and GalNAcß1-4GlcNAc-like (LacdiNAc-like), were common modifications of human gastric O-glycans. Furthemore, each individual carried 1-14 glycan structures that were unique for that individual. The diversity and alterations in gastric O-glycosylation broaden our understanding of the human gastric O-glycome and its implications for gastric cancer research and emphasize that the high individual variation makes it difficult to identify gastric cancer specific structures. However, despite the low number of individuals, we could verify a higher level of sialylation and sulfation on gastric O-glycans from cancerous tissue than from healthy stomachs.


Asunto(s)
Mucinas Gástricas/química , Polisacáridos/química , Antígenos de Grupos Sanguíneos/química , Cromatografía Liquida , Epítopos/metabolismo , Mucinas Gástricas/metabolismo , Humanos , Mucina 5AC/química , Mucina 5AC/metabolismo , Polisacáridos/metabolismo , Espectrometría de Masas en Tándem
12.
Int J Mol Sci ; 20(18)2019 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-31500233

RESUMEN

Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) is a small trefoil protein co-expressed with the gastric mucin MUC5AC in surface foveolar cells and co-secreted with MUC5AC into gastric mucus. Helicobacter pylori binds with greater avidity to TFF1 dimer, which is present in gastric mucus, than to TFF1 monomer. Binding of H. pylori to TFF1 is mediated by the core oligosaccharide subunit of H. pylori lipopolysaccharide at pH 5.0-6.0. Treatment of H. pylori lipopolysaccharide with mannosidase or glucosidase inhibits its interaction with TFF1. Both TFF1 and H. pylori have a propensity for binding to mucins with terminal non-reducing α- or ß-linked N-acetyl-d-glucosamine or α-(2,3) linked sialic acid or Gal-3-SO42-. These findings are strong evidence that TFF1 has carbohydrate-binding properties that may involve a conserved patch of aromatic hydrophobic residues on the surface of its trefoil domain. The pH-dependent lectin properties of TFF1 may serve to locate H. pylori deep in the gastric mucus layer close to the epithelium rather than at the epithelial surface. This restricted localisation could limit the interaction of H. pylori with epithelial cells and the subsequent host signalling events that promote inflammation.


Asunto(s)
Helicobacter pylori/fisiología , Lipopolisacáridos/metabolismo , Estómago/microbiología , Factor Trefoil-1/metabolismo , Mucinas Gástricas/metabolismo , Glucosidasas/farmacología , Helicobacter pylori/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Lipopolisacáridos/química , Manosidasas/farmacología , Mucina 5AC/metabolismo , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/metabolismo , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Factor Trefoil-1/química , Tropismo
13.
Chembiochem ; 19(24): 2598-2608, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30371971

RESUMEN

Mucous gels (mucus) cover internal body surfaces. The secretory mucins MUC5AC and MUC6 and the protective peptide TFF2 are characteristic constituents of gastric mucus; TFF2 is co-secreted with MUC6. Herein, we investigated two commercial mucin preparations by FPLC and proteomics, because they are model systems for studying the rheology of gastric mucins. One preparation is also used as a saliva substitute, for example, after radiation therapy. We show that both preparations contain TFF2 (≈0.6 to 1.1 %, w/w). The majority of TFF2 is strongly bound noncovalently to mucin in a manner that is resistant to boiling in SDS. First overlay assays with 125 I-labeled porcine TFF2 revealed that mucin binding is modulated by Ca2+ and can be blocked by the lectin GSA-II and the antibody HIK1083, both recognizing the peripheral GlcNAcα1→4Galß1→R moiety of MUC6. TFF2 binding was also inhibited in the presence of Me-ß-Gal but less so by the α anomer. TFF2 may play a role in the oligomerization and secretion of MUC6, the rheology of gastric mucus, and the adherence of gastric microbiota. TFF2 in artificial saliva may be of benefit. TFF2 might also interact with the sugar moiety of various receptors.


Asunto(s)
Mucinas Gástricas/análisis , Péptidos/aislamiento & purificación , Saliva Artificial/análisis , Factor Trefoil-2/aislamiento & purificación , Animales , Anticuerpos Monoclonales/metabolismo , Western Blotting , Calcio/metabolismo , Mucinas Gástricas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Radioisótopos de Yodo , Espectrometría de Masas , Mucina 6/metabolismo , Péptidos/química , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Proteómica , Saliva Artificial/metabolismo , Estómago/química , Porcinos , Factor Trefoil-2/química , Factor Trefoil-2/metabolismo
14.
Histochem Cell Biol ; 149(6): 569-575, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29658052

RESUMEN

Gastric gland mucin secreted from pyloric gland cells, mucous neck cells, and cardiac gland cells of the gastric mucosa harbors unique O-glycans carrying terminal α1,4-linked N-acetylglucosamine residues (αGlcNAc), which are primarily attached to the scaffold mucin core protein MUC6. αGlcNAc acts as an antibiotic against Helicobacter pylori (H. pylori), a microbe causing gastric cancer. In addition, mice deficient in A4gnt, which encodes the enzyme α1,4-N-acetylglucosaminyltransferase (α4GnT) that catalyzes αGlcNAc biosynthesis, spontaneously develop gastric differentiated-type adenocarcinoma, even if not infected by H. pylori. Thus, αGlcNAc prevents gastric cancer as both an antibiotic and a tumor suppressor (Nakayama in Acta Histochem Cytochem 47:1-9, 2014b). Indeed, in humans αGlcNAc loss on MUC6 in differentiated-type adenocarcinoma is closely associated with poor patient prognosis (Shiratsu et al. in Cancer Sci 105:126-133, 2014). Recently, we reported reduced αGlcNAc expression on MUC6 in both pyloric gland adenoma of the stomach and chronic atrophic gastritis, in Barrett's esophagus, and in pancreatic intraductal papillary-mucinous neoplasm (IPMN)/pancreatic intraepithelial neoplasia (PanIN), all potentially premalignant conditions. This review discusses whether relatively reduced levels of αGlcNAc in these lesions could serve as a biomarker to predict malignant potential and cancer progression.


Asunto(s)
Acetilglucosamina/metabolismo , Adenocarcinoma/metabolismo , Esófago de Barrett/metabolismo , Biomarcadores de Tumor/metabolismo , Mucinas Gástricas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patología , Animales , Esófago de Barrett/patología , Glicosilación , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Gástricas/patología
15.
BMC Gastroenterol ; 18(1): 189, 2018 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-30587141

RESUMEN

BACKGROUND: Intramucosal, histologically mixed-type, low-grade (LG), well-differentiated gastric tubular adenocarcinomas (tub1s; LG-tub1s) have larger mean diameters and exhibit a higher frequency of the gastric mucin phenotype (G-phenotype) than pure LG-tub1s. In proportion to their increases in diameter, G-phenotype differentiated-type early gastric cancer (EGC) tumours reportedly grow to eventually contain (an) undifferentiated-type component(s) and LG-tub1s, which are included in differentiated-type EGCs, reportedly exhibit changes in their glandular architectural and cytological atypia grades from LG to high-grade (HG) and can grow to contain a moderately differentiated tubular adenocarcinoma (tub2) component and undifferentiated components. Because they generally show a higher frequency of malignancy relative to tumours with a higher atypia grade and lower differentiation degree, it is suggested that, among mixed-type LG-tub1s, G-phenotype LG-tub1s containing an HG-tub2 component (LG-tub1s > HG-tub2) with undifferentiated components might lead to late-onset metastasis to lymph nodes even after a successful endoscopic submucosal dissection (ESD). We aimed to clarify the endoscopic and clinicopathological features of these G-phenotype LG-tub1s > HG-tub2. METHODS: Of the 13,217 oesophagogastroduodenoscopies performed at our institutions between September 2008 and March 2016, 185 EGC lesions were evaluated in this retrospective observational study. Among these EGC lesions, 60 intramucosal LG-tub1s were divided into 53 tub1 (44 pure LG-tub1s and nine LG-tub1s containing HG-tub1) lesions and seven LG-tub1 > tub2 (LG-tub1 containing LG- and HG-tub2) lesions. RESULTS: The frequencies of the superficial depressed type (P = 0.026), reddish colour (P = 0.006), HG of contained tub2s (P = 0.006), and G-phenotype (P = 0.028) were significantly higher in the LG-tub1 > tub2 group than those in the tub1 group. However, the largest lesion of the LG-tub1 > tub2 group had a superficial flat appearance, an isochromatic colour, an HG-tub2 and an undifferentiated component, and a large diameter greater than 30 mm, and it exhibited a G-phenotype. CONCLUSIONS: Intramucosal G-phenotype LG-tub1s > HG-tub2 are potential premalignant stomach neoplasms that may have specific endoscopic and clinicopathological features. However, G-phenotype LG-tub1s > HG-tub2 with undifferentiated component, which potentially show higher malignancy than those without undifferentiated components might change from a reddish to isochromatic colour. Accurately diagnosing, treating, and following-up G-phenotype LG-tub1s > HG-tub2 might decrease the number of patients who experience late-onset metastasis after ESD.


Asunto(s)
Adenocarcinoma/patología , Metástasis Linfática , Neoplasias Gástricas/patología , Adenocarcinoma/microbiología , Adenocarcinoma/cirugía , Anciano , Anciano de 80 o más Años , Resección Endoscópica de la Mucosa , Endoscopía del Sistema Digestivo , Femenino , Mucinas Gástricas/metabolismo , Helicobacter pylori/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Fenotipo , Estudios Retrospectivos , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/cirugía
16.
Am J Physiol Gastrointest Liver Physiol ; 313(6): G599-G612, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28882824

RESUMEN

It is generally accepted that the gastric mucus layer provides a protective barrier between the lumen and the mucosa, shielding the mucosa from acid and digestive enzymes and preventing autodigestion of the stomach epithelium. However, the precise mechanisms that contribute to this protective function are still up for debate. In particular, it is not clear what physical processes are responsible for transporting hydrogen protons, secreted within the gastric pits, across the mucus layer to the lumen without acidifying the environment adjacent to the epithelium. One hypothesis is that hydrogen may be bound to the mucin polymers themselves as they are convected away from the mucosal surface and eventually degraded in the stomach lumen. It is also not clear what mechanisms prevent hydrogen from diffusing back toward the mucosal surface, thereby lowering the local pH. In this work we investigate a physics-based model of ion transport within the mucosal layer based on a Nernst-Planck-like equation. Analysis of this model shows that the mechanism of transporting protons bound to the mucus gel is capable of reproducing the trans-mucus pH gradients reported in the literature. Furthermore, when coupled with ion exchange at the epithelial surface, our analysis shows that bicarbonate secretion alone is capable of neutralizing the epithelial pH, even in the face of enormous diffusive gradients of hydrogen. Maintenance of the pH gradient is found to be robust to a wide array of perturbations in both physiological and phenomenological model parameters, suggesting a robust physiological control mechanism.NEW & NOTEWORTHY This work combines modeling techniques based on physical principles, as well as novel numerical simulations to test the plausibility of one hypothesized mechanism for proton transport across the gastric mucus layer. Results show that this mechanism is able to maintain the extreme pH gradient seen in in vivo experiments and suggests a highly robust regulation mechanism to maintain this gradient in the face of dynamic lumen composition.


Asunto(s)
Simulación por Computador , Mucinas Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Modelos Biológicos , Moco/metabolismo , Animales , Humanos , Concentración de Iones de Hidrógeno , Intercambio Iónico , Cinética , Potenciales de la Membrana , Análisis Numérico Asistido por Computador
17.
Nutr Cancer ; 69(2): 289-298, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28094544

RESUMEN

Processed meat intake is carcinogenic to humans. We have shown that intake of a workshop-made cured meat with erythorbate promotes colon carcinogenesis in rats. We speculated that polyphenols could inhibit this effect by limitation of endogenous lipid peroxidation and nitrosation. Polyphenol-rich plant extracts were added to the workshop-made cured meat and given for 14 days to rats and 100 days to azoxymethane-induced rats to evaluate the inhibition of preneoplastic lesions. Colons of 100-d study were scored for precancerous lesions (mucin-depleted foci, MDF), and biochemical end points of peroxidation and nitrosation were measured in urinary and fecal samples. In comparison with cured meat-fed rats, dried red wine, pomegranate extract, α-tocopherol added at one dose to cured meat and withdrawal of erythorbate significantly decreased the number of MDF per colon (but white grape and rosemary extracts did not). This protection was associated with the full suppression of fecal excretion of nitrosyl iron, suggesting that this nitroso compound might be a promoter of carcinogenesis. At optimized concentrations, the incorporation of these plant extracts in cured meat might reduce the risk of colorectal cancer associated with processed meat consumption.


Asunto(s)
Lythraceae/química , Carne/efectos adversos , Extractos Vegetales/farmacología , Lesiones Precancerosas/dietoterapia , Vino , Animales , Biomarcadores/orina , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/prevención & control , Heces , Mucinas Gástricas/metabolismo , Peroxidación de Lípido , Masculino , Carne/análisis , Lesiones Precancerosas/inducido químicamente , Ratas Endogámicas F344 , alfa-Tocoferol/farmacología
18.
Jpn J Clin Oncol ; 46(6): 502-6, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27004903

RESUMEN

OBJECTIVE: To determine whether a subgroup of gastric cancer patients might benefit from epidermal growth factor receptor-tyrosine kinase inhibitors. METHODS: A total of 103 gastric cancer samples were collected for this study. High-resolution melting and deoxyribonucleic acid sequencing were used to detect epidermal growth factor receptor mutations in exons 19 and 21. RESULTS: Polymerase chain reaction-high-resolution melting was successfully performed on all 103 samples. Aberrant melting curves were found in only one sample. Sanger sequencing revealed a 15 bp deletion (c.2235_2249del; p.Glu746_Ala750del) in epidermal growth factor receptor exon 19. The sample was from a male patient, and the pathological diagnosis was a mucin-producing gastric cancer with lymph node metastasis. To date, this is the first report on epidermal growth factor receptor exon 19 mutation in gastric cancer. CONCLUSIONS: An epidermal growth factor receptor mutation in exon 19 was identified in mucin-producing gastric cancer sample from a male patient. This mutation indicates that the small subgroup of patients with mucin-producing gastric cancer might benefit from epidermal growth factor receptor-tyrosine kinase inhibitors.


Asunto(s)
Pueblo Asiatico/genética , Receptores ErbB/genética , Mucinas Gástricas/metabolismo , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , China , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/aislamiento & purificación , ADN de Neoplasias/metabolismo , Exones , Femenino , Eliminación de Gen , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/genética , Temperatura de Transición
19.
J Basic Microbiol ; 56(4): 347-57, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26663202

RESUMEN

α-L-Fucosidases are key enzymes for the degradation of intestinal glycans by gut microbes. In this work, three putative α-L-fucosidases (Afc1, Afc2, and Afc3) genes from Clostridium perfringens ATCC 13124 were cloned and expressed in Escherichia coli. Afc1 had the α-L-fucosidase domain of glycoside hydrolase (GH) 29 family but showed no enzyme activity toward all the substrates examined. The putative acid/base residue of Afc1, Ser205, was replaced by a glutamic acid which is conserved in GH29-B α-L-fucosidases. However, the mutant Afc1-S205E still failed to show enzyme activity. Afc2 and Afc3 were determined to be 1,3-1,4-α-L-fucosidase of GH29-B subfamily and 1,2-α-L-fucosidase of GH95 family, respectively, and both of them could release fucose from porcine gastric mucin (PGM). When C. perfringens ATCC 13124 grew with the presence of PGM, the transcription of afc1 decreased slightly, while those of afc2 and afc3 increased to 2.2-fold and 1.4-fold, respectively, and the enzyme activities of Afc2 and Afc3 in the culture increased to 2.2-fold and 2.6-fold, respectively. These results suggest that Afc2 and Afc3 are involved in the degradation of intestinal fucosyl glycans by C. perfringens ATCC 13124.


Asunto(s)
Clostridium perfringens/enzimología , Clostridium perfringens/genética , alfa-L-Fucosidasa/biosíntesis , alfa-L-Fucosidasa/genética , Animales , Clonación Molecular , Activación Enzimática , Escherichia coli/genética , Fucosa/metabolismo , Mucinas Gástricas/metabolismo , Ácido Glutámico/metabolismo , Mutagénesis Sitio-Dirigida , Polisacáridos/metabolismo , Especificidad por Sustrato , Porcinos
20.
Infect Immun ; 83(4): 1610-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25644008

RESUMEN

Brachyspira hyodysenteriae colonizes the pig colon, resulting in mucohemorrhagic diarrhea and growth retardation. Fecal mucus is a characteristic feature of swine dysentery; therefore, we investigated how the mucin environment changes in the colon during infection with B. hyodysenteriae and how these changes affect this bacterium's interaction with mucins. We isolated and characterized mucins, the main component of mucus, from the colon of experimentally inoculated and control pigs and investigated B. hyodysenteriae binding to these mucins. Fluorescence microscopy revealed a massive mucus induction and disorganized mucus structure in the colon of pigs with swine dysentery. Quantitative PCR (qPCR) and antibody detection demonstrated that the mucus composition of pigs with swine dysentery was characterized by de novo expression of MUC5AC and increased expression of MUC2 in the colon. Mucins from the colon of inoculated and control pigs were isolated by two steps of isopycnic density gradient centrifugation. The mucin densities of control and inoculated pigs were similar, whereas the mucin quantity was 5-fold higher during infection. The level of B. hyodysenteriae binding to mucins differed between pigs, and there was increased binding to soluble mucins isolated from pigs with swine dysentery. The ability of B. hyodysenteriae to bind, measured in relation to the total mucin contents of mucus in sick versus healthy pigs, increased 7-fold during infection. Together, the results indicate that B. hyodysenteriae binds to carbohydrate structures on the mucins as these differ between individuals. Furthermore, B. hyodysenteriae infection induces changes to the mucus niche which substantially increase the amount of B. hyodysenteriae binding sites in the mucus.


Asunto(s)
Adhesión Bacteriana/fisiología , Brachyspira hyodysenteriae/patogenicidad , Mucinas Gástricas/metabolismo , Infecciones por Bacterias Gramnegativas/microbiología , Mucina 5AC/biosíntesis , Mucina 2/biosíntesis , Enfermedades de los Porcinos/microbiología , Animales , Colon/microbiología , Moco/metabolismo , Unión Proteica , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA