Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
1.
Stem Cell Reports ; 19(5): 629-638, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38670110

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primarily affects the lung but can also cause gastrointestinal (GI) symptoms. In vitro experiments confirmed that SARS-CoV-2 robustly infects intestinal epithelium. However, data on infection of adult gastric epithelium are sparse and a side-by-side comparison of the infection in the major segments of the GI tract is lacking. We provide this direct comparison in organoid-derived monolayers and demonstrate that SARS-CoV-2 robustly infects intestinal epithelium, while gastric epithelium is resistant to infection. RNA sequencing and proteome analysis pointed to angiotensin-converting enzyme 2 (ACE2) as a critical factor, and, indeed, ectopic expression of ACE2 increased susceptibility of gastric organoid-derived monolayers to SARS-CoV-2. ACE2 expression pattern in GI biopsies of patients mirrors SARS-CoV-2 infection levels in monolayers. Thus, local ACE2 expression limits SARS-CoV-2 expression in the GI tract to the intestine, suggesting that the intestine, but not the stomach, is likely to be important in viral replication and possibly transmission.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Mucosa Gástrica , Mucosa Intestinal , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/fisiología , Humanos , COVID-19/virología , COVID-19/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/virología , Tropismo Viral , Organoides/virología , Organoides/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Replicación Viral , Animales
2.
Am J Vet Res ; 85(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38484465

RESUMEN

OBJECTIVE: To investigate the role of equine herpesvirus-2 (EHV-2) and equine herpesvirus-5 (EHV-5) in equine glandular gastric disease (EGGD) by visualizing and quantifying these gamma herpesviruses in EGGD-affected and normal glandular gastric mucosa of horses. A secondary objective was to describe the histopathological abnormalities in the equine gastric glandular mucosa in horses with EGGD. ANIMALS: 29 horses (n = 21 postmortem and 8 gastroscopy) categorized as normal (11), EGGD (12), or both EGGD and equine squamous gastric disease (6). METHODS: Glandular gastric mucosal samples were collected from horses by gastroscopy or postmortem. Histopathology and in situ hybridization targeting EHV-2 and EHV-5 were performed on grossly normal and abnormal glandular gastric mucosa. The number of in situ hybridization-positive cells per millimeter squared of tissue was calculated. Evaluators were blinded to groups. RESULTS: Glandular gastric tissues from horses without EGGD had higher viral loads in the mucosa than normal or abnormal tissues from EGGD horses. There was no difference in viral loads for EHV-2 or EHV-5 between grossly or endoscopically normal to abnormal gastric tissues within horses with EGGD. Lymphocytic plasmacytic gastritis was the most common histopathological abnormality, with only 3 horses having mucosal disruption (glandular ulcer or erosion). CLINICAL RELEVANCE: Equine gamma herpesviruses are unlikely to play a role in the pathophysiology of EGGD. EGGD is frequently inflammatory with occasional mucosal disruption (ulcer or erosion).


Asunto(s)
Infecciones por Herpesviridae , Enfermedades de los Caballos , Gastropatías , Carga Viral , Animales , Caballos , Enfermedades de los Caballos/virología , Enfermedades de los Caballos/patología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/patología , Carga Viral/veterinaria , Gastropatías/veterinaria , Gastropatías/virología , Gastropatías/patología , Femenino , Masculino , Mucosa Gástrica/virología , Mucosa Gástrica/patología , Gammaherpesvirinae/aislamiento & purificación , Hibridación in Situ/veterinaria
3.
Nutrients ; 14(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35268019

RESUMEN

Helicobacter pylori (H. pylori) causes gastric diseases by increasing reactive oxygen species (ROS) and interleukin (IL)-8 expression in gastric epithelial cells. ROS and inflammatory responses are regulated by the activation of nuclear factor erythroid-2-related factor 2 (Nrf2) and the expression of Nrf2 target genes, superoxide dismutase (SOD) and heme oxygenase-1 (HO-1). We previously demonstrated that Korean red ginseng extract (RGE) decreases H. pylori-induced increases in ROS and monocyte chemoattractant protein 1 in gastric epithelial cells. We determined whether RGE suppresses the expression of IL-8 via Nrf2 activation and the expression of SOD and HO-1 in H. pylori-infected gastric epithelial AGS cells. H. pylori-infected cells were treated with RGE with or without ML385, an Nrf2 inhibitor, or zinc protoporphyrin (ZnPP), a HO-1 inhibitor. Levels of ROS and IL-8 expression; abundance of Keap1, HO-1, and SOD; levels of total, nuclear, and phosphorylated Nrf2; indices of mitochondrial dysfunction (reduction in mitochondrial membrane potential and ATP level); and SOD activity were determined. As a result, RGE disturbed Nrf2-Keap1 interactions and increased nuclear Nrf2 levels in uninfected cells. H. pylori infection decreased the protein levels of SOD-1 and HO-1, as well as SOD activity, which was reversed by RGE treatment. RGE reduced H. pylori-induced increases in ROS and IL-8 levels as well as mitochondrial dysfunction. ML385 or ZnPP reversed the inhibitory effect of RGE on the alterations caused by H. pylori. In conclusion, RGE suppressed IL-8 expression and mitochondrial dysfunction via Nrf2 activation, induction of SOD-1 and HO-1, and reduction of ROS in H. pylori-infected cells.


Asunto(s)
Mucosa Gástrica , Infecciones por Helicobacter , Interleucina-8 , Factor 2 Relacionado con NF-E2 , Panax , Extractos Vegetales , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Infecciones por Helicobacter/virología , Helicobacter pylori , Humanos , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias/química , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Panax/metabolismo , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo
4.
mSphere ; 6(5): e0075121, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34585958

RESUMEN

Persistent coinfection with Helicobacter pylori and Epstein-Barr virus (EBV) promotes aggressive gastric carcinoma (GC). The molecular mechanisms underlying the aggressiveness in H. pylori and EBV-mediated GC are not well characterized. We investigated the molecular mechanism involved in H. pylori- and EBV-driven proliferation of gastric epithelial cells. Results showed that the coinfection is significantly more advantageous to the pathogens as coinfection creates a microenvironment favorable to higher pathogen-associated gene expression. The EBV latent genes ebna1 and ebna3c are highly expressed in the coinfection compared to lone EBV infection at 12 and 24 h. The H. pylori-associated genes 16S rRNA, cagA, and babA were also highly expressed during coinfection compared to H. pylori alone. In addition, upregulation of gankyrin, which is a small oncoprotein, modulates various cell signaling pathways, leading to oncogenesis. Notably, the knockdown of gankyrin decreased the cancer properties of gastric epithelial cells. Gankyrin showed a similar expression pattern as that of ebna3c at both transcript and protein levels, suggesting a possible correlation. Further, EBV and H. pylori created a microenvironment that induced cell transformation and oncogenesis through dysregulation of the cell cycle regulatory (ccnd1, dapk3, pcna, and akt), GC marker (abl1, tff-2, and cdx2), cell migration (mmp3 and mmp7), DNA response (pRB, pten, and p53), and antiapoptotic (bcl2) genes in infected gastric epithelial cells through gankyrin. Our study provides a new insight into the interplay of two oncogenic agents (H. pylori and EBV) that leads to an enhanced carcinogenic activity in gastric epithelial cells through overexpression of gankyrin. IMPORTANCE In the present study, we evaluated the synergistic effects of EBV and H. pylori infection on gastric epithelial cells in various coinfection models. These coinfection models were among the first to depict the exposures of gastric epithelial cells to EBV followed by H. pylori; however, coinfection models exist that narrated the scenario upon exposure to H. pylori followed by that to EBV. We determined that a coinfection by EBV and H. pylori enhanced the expression of oncogenic protein gankyrin. The interplay between EBV and H. pylori promoted the oncogenic properties of AGS cells like elevated focus formation, cell migration, and cell proliferation through gankyrin. EBV and H. pylori mediated an enhanced expression of gankyrin, which further dysregulated cancer-associated genes such as cell migratory, tumor suppressor, DNA damage response, and proapoptotic genes.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Infecciones por Helicobacter/genética , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Proto-Oncogénicas/genética , Neoplasias Gástricas/microbiología , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica , Coinfección/genética , Coinfección/microbiología , Coinfección/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/microbiología , Infecciones por Virus de Epstein-Barr/virología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/virología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
5.
PLoS One ; 16(9): e0256440, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34469459

RESUMEN

Epstein-Barr virus (EBV)-associated gastric carcinoma (EBVaGC) is a distinct molecular subtype of gastric cancer characterized by viral infection and cellular abnormalities, including loss of AT-rich interaction domain 1A (ARID1A) expression (lost ARID1A). To evaluate the significance of lost ARID1A in the development of EBVaGC, we performed in situ hybridization of EBV-encoded RNA (EBER) and immunohistochemistry of ARID1A in the non-neoplastic gastric mucosa and intramucosal cancer tissue of EBVaGC with in vitro infection analysis of ARID1A-knockdown and -knockout gastric cells. Screening of EBER by in situ hybridization revealed a frequency of approximately 0.2% EBER-positive epithelial cells in non-neoplastic gastric mucosa tissue samples. Six small foci of EBV-infected epithelial cells showed two types of histology: degenerated (n = 3) and metaplastic (n = 3) epithelial cells. ARID1A was lost in the former type. In intramucosal EBVaGC, there were ARID1A-lost (n = 5) and -preserved tumors (n = 7), suggesting that ARID1A-lost carcinomas are derived from ARID1A-lost precursor cells in the non-neoplastic mucosa. Lost ARID1A was also observed in non-neoplastic mucosa adjacent to an ARID1A-lost EBVaGC. In vitro experiments using siRNA knockdown and the CRISPR/Cas9-knockout system demonstrated that transient reduction or permanent loss of ARID1A expression markedly increased the efficiency of EBV infection to stomach epithelial cells. Taken together, lost ARID1A plays a role in initiating EBV-driven carcinogenesis in stomach epithelial cells, which develop to a distinct subtype of EBVaGC within the proper mucosal layer. Lost ARID1A is one of the constituents of virus-host interactions in the carcinogenesis of EBVaGC.


Asunto(s)
Carcinogénesis/genética , Carcinoma/genética , Proteínas de Unión al ADN/deficiencia , Infecciones por Virus de Epstein-Barr/genética , Neoplasias Gástricas/genética , Factores de Transcripción/deficiencia , Adulto , Anciano , Carcinogénesis/patología , Carcinoma/patología , Carcinoma/virología , Línea Celular Tumoral , Metilación de ADN , Proteínas de Unión al ADN/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno/genética , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Factores de Transcripción/genética
6.
FEBS Lett ; 595(17): 2257-2270, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34278574

RESUMEN

HIV preferentially infects α4 ß7+ CD4 T cells, forming latent reservoirs that contribute to HIV persistence during antiretroviral therapy. However, the properties of α4 ß7+ CD4 T cells in blood and mucosal compartments remain understudied. Employing two distinct models of HIV infection, HIV-infected humans and simian-human immunodeficiency virus (SHIV)-infected rhesus macaques, we show that α4 ß7+ CD4 T cells in blood are enriched for genes regulating cell cycle progression and cellular metabolism. Unlike their circulating counterparts, rectal α4 ß7+ CD4 T cells exhibited a core tissue-residency gene expression program. These features were conserved across primate species, indicating that the environment influences memory T-cell transcriptional networks. Our findings provide an important molecular foundation for understanding the role of α4 ß7 in HIV infection.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/sangre , Integrinas/metabolismo , Adulto , Animales , COVID-19/sangre , COVID-19/virología , Ciclo Celular , Proliferación Celular , Mucosa Gástrica/citología , Mucosa Gástrica/virología , Regulación de la Expresión Génica , Humanos , Inmunización , Macaca mulatta , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
7.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946994

RESUMEN

The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.


Asunto(s)
Bacteriófagos/fisiología , Dieta , Sistema Nervioso Entérico/fisiología , Mucosa Gástrica/virología , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/fisiología , Mucosa Intestinal/virología , Virus , Defensinas/fisiología , Digestión , Susceptibilidad a Enfermedades , Sistema Nervioso Entérico/virología , Alimentos/virología , Mucosa Gástrica/inmunología , Mucosa Gástrica/inervación , Mucosa Gástrica/metabolismo , Gastroenteritis/virología , Interacciones Microbiota-Huesped/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/inervación , Mucosa Intestinal/metabolismo , Desnutrición/virología , Moco/metabolismo , Moco/virología , Neuronas/virología , Infecciones Oportunistas/virología , Virus de Plantas , Virosis/microbiología , Virosis/fisiopatología
8.
BMC Cancer ; 21(1): 228, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676447

RESUMEN

BACKGROUND: Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) is an important cancer stem cell marker in gastric cancer. However, no detailed studies are available on LGR5 expression in poorly differentiated gastric adenocarcinoma (PD-AC). Therefore, we investigated the relationship between LGR5 expression and clinicopathological data in PD-AC. METHODS: LGR5 mRNA expression levels were quantified in 41 PD-AC specimens using a highly sensitive RNAscope in situ hybridization technique. Epstein-Barr virus (EBV) infection was also detected by EBV in situ hybridization. RESULTS: LGR5 expression levels were measured in 38 of 41 PD-AC cases, and 17 cases were identified as LGR5 high. The frequency of EBV positivity tended to be higher in the LGR5-low group than in the LGR5-high group (P = 0.0764). Furthermore, the frequency of vascular invasion tended to be higher in the LGR5-high group than in the LGR5-low group (P = 0.0764). The overall survival of PD-AC patients in the LGR5-high group was significantly lower than in the LGR5-low group (log-rank test, P = 0.0108). The Cox proportional hazard regression model revealed that the LGR5-low group (HR = 0.29; 95% CI: 0.11-0.74; P = 0.01) showed independently better OS for PD-AC. CONCLUSIONS: Quantifying the levels of LGR5 expression may facilitate defining prognosis in Japanese patients with PD-AC. Further study of LGR5 in this context is warranted.


Asunto(s)
Adenocarcinoma/mortalidad , Biomarcadores de Tumor/metabolismo , Infecciones por Virus de Epstein-Barr/epidemiología , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Gástricas/mortalidad , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Adenocarcinoma/virología , Anciano , Infecciones por Virus de Epstein-Barr/diagnóstico , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Gastrectomía , Mucosa Gástrica/patología , Mucosa Gástrica/cirugía , Mucosa Gástrica/virología , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Japón/epidemiología , Estimación de Kaplan-Meier , Masculino , Pronóstico , Estudios Retrospectivos , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/virología
9.
mBio ; 12(1)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33563833

RESUMEN

The human gastrointestinal mucosal surface consists of a eukaryotic epithelium, a prokaryotic microbiota, and a carbohydrate-rich interface that separates them. In the gastrointestinal tract, the interaction of bacteriophages (phages) and their prokaryotic hosts influences the health of the mammalian host, especially colonization with invasive pathobionts. Antibiotics may be used, but they also kill protective commensals. Here, we report a novel phage whose lytic cycle is enhanced in intestinal environments. The tail fiber gene, whose protein product binds human heparan sulfated proteoglycans and localizes the phage to the epithelial cell surface, positions it near its bacterial host, a type of locational targeting mechanism. This finding offers the prospect of developing mucosal targeting phage to selectively remove invasive pathobiont species from mucosal surfaces.IMPORTANCE Invasive pathobionts or microbes capable of causing disease can reside deep within the mucosal epithelium of our gastrointestinal tract. Targeted effective antibacterial therapies are needed to combat these disease-causing organisms, many of which may be multidrug resistant. Here, we isolated a lytic bacteriophage (phage) that can localize to the epithelial surface by binding heparan sulfated glycans, positioning it near its host, Escherichia coli This targeted therapy can be used to selectively remove invasive pathobionts from the gastrointestinal tract, preventing the development of disease.


Asunto(s)
Bacteriófagos/metabolismo , Mucosa Gástrica/citología , Tracto Gastrointestinal/virología , Proteoglicanos de Heparán Sulfato/metabolismo , Interacciones Microbianas , Polisacáridos/metabolismo , Proteínas de la Cola de los Virus/metabolismo , Animales , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Bacteriófagos/patogenicidad , Técnicas de Cultivo de Célula , Escherichia coli/metabolismo , Femenino , Mucosa Gástrica/virología , Tracto Gastrointestinal/fisiología , Humanos , Masculino , Ratones Endogámicos BALB C , Microbiota , Organoides/citología , Organoides/virología , Organismos Libres de Patógenos Específicos , Simbiosis , Proteínas de la Cola de los Virus/genética
10.
Pathol Int ; 71(1): 42-50, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33084164

RESUMEN

Gastritis cystica profunda (GCP) is a lesion characterized by cystic gastric glands within the submucosa. Some studies have reported that GCP is a precancerous lesion. Here, we investigated the association between GCP and gastric cancer. Gastric cancer specimens were taken from 1432 patients undergoing surgery or endoscopic submucosal resection and were classified as GCP or non-GCP. The clinicopathological features, immunohistochemistry and in situ hybridization expression of p53, Ki-67, KCNE2, Epstein-Barr virus (EBV) and programmed death ligand 1 (PD-L1) were compared between the two groups, as well as between GCPs and normal pyloric glands. One hundred and eighty patients (12.6%) had GCPs. In the GCP group, no cancerous lesions were found within the GCPs, but 13% were linked to GCPs and 60.2% were located above or near GCPs. Aberrant p53 expression, EBV-positive cancer cells and PD-L1 scores were significantly higher in the GCP group. The p53 score and Ki-67 labelling index were significantly higher and the KCNE2 score was significantly lower in GCPs than in pyloric glands. Although we suggest GCP is paracancerous, GCP has high proliferation activity and gastric cancer with GCP is associated with aberrant p53 and EBV. GCP is associated with aberrant p53 expression and EBV.


Asunto(s)
Antígeno B7-H1/análisis , Mucosa Gástrica , Herpesvirus Humano 4/aislamiento & purificación , Neoplasias Gástricas , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Gastritis/patología , Neoplasias Gastrointestinales/patología , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Proteína p53 Supresora de Tumor/análisis
11.
Turk J Gastroenterol ; 31(11): 775-781, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33361040

RESUMEN

BACKGROUND/AIMS: The study aimed to explore the effects of Epstein-Barr virus--encoded BARF1 in human gastric epithelial cells (GES-1). MATERIALS AND METHODS: A eukaryotic expression vector carrying BARF1 gene (pcDNA3.1-BARF1) was constructed. The pcDNA3.1-BARF1 was transfected into GES-1 cells, and they were selected by G418. The GES-1 cells lines that expressed BARF1 (GES-1-BARF1) were obtained. The cycle of GES-1-pcDNA3.1 cells (GES-1 cells transfected with empty vector), GES-1-BARF1 cells (GES-1 cells transfected with BARF1), and TPA-GES-1-BARF1(GES-1-BARF1 cells stimulated by 12-O-tetradecanoylphorbol-13-acetate (TPA) were analyzed by flow cytometry. Colony formation in soft agar and tumorigenicity of the transfected cells in mice with severe combined immunodeficiency (SCID) were also observed. RESULTS: The morphology of GES-1-BARF1 cells were changed from the original shuttle to round, the adhesion between the cells and bottle wall was weakened, and the cells showed overlapping growth. The proliferation rate of GES-1-BARF1 and TPA-GES-1-BARF1 cells were faster than GES-1 and GES-1-pcDNA3.1 cells; the S phase was significantly prolonged for GES-1-BARF1 and TPA-GES-1-BARF1. GES-1-BARF1 and TPA-GES-1-BARF1 cells formed colonies in soft agar, with a cloning rate of 24.2% (58/240) and 40.0% (96/240), respectively; GES-1 and GES-1-pcDNA3.1 cells did not form colonies in soft agar. Tumors were formed in mice with SCID after injecting TPA-GES-1-BARF1 cell groups. Tumor formation did not occur in mice with SCID after injecting GES-1 and GES-1-pcDNA3.1 cell groups, but nodules were formed in the mice with SCID after injecting GES-1-BARF1 cell groups. CONCLUSION: GES-1-BARF1 cells malignant transformation was induced by transfected BARF1 gene and TPA stimulation. This result indicated that tumor formation not only require oncogenes, but also the stimulation of cancer-promoting substance.


Asunto(s)
Carcinogénesis/genética , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Herpesvirus Humano 4/genética , Proteínas Virales/metabolismo , Animales , Carcinogénesis/inducido químicamente , Modelos Animales de Enfermedad , Células Epiteliales/virología , Mucosa Gástrica/virología , Humanos , Ratones , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/virología , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Transfección
12.
BMC Cancer ; 20(1): 925, 2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-32993565

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) and Epstein - Barr virus (EBV) plays a significant role in aggressive gastric cancer (GC). The investigation of genes associated with these pathogens and host kinases may be essential to understand the early and dynamic progression of GC. AIM: The study aimed to demonstrate the coinfection of EBV and H. pylori in the AGS cells through morphological changes, expression of the kinase and the probable apoptotic pathways. METHODS: Genomic DNA isolation of H. pylori and its characterization from clinical samples were performed. RT-qPCR of kinases was applied to scrutinize the gene expression of kinases in co-infected GC in a direct and indirect (separated through insert size 0.45 µm) H. pylori infection set up. Morphological changes in co-infected GC were quantified by measuring the tapering ends of gastric epithelial cells. Gene expression profiling of apoptotic genes was assessed through RT-qPCR. RESULTS: An interleukin-2-inducible T-cell kinase (ITK) showed significant upregulation with indirect H. pylori infection. Moreover, Ephrin type-B receptor six precursors (EPHB6) and Tyrosine-protein kinase Fyn (FYN) showed significant upregulation with direct coinfection. The tapering ends in AGS cells were found to be extended after 12 h. A total of 24 kinase genes were selected, out of which EPHB6, ITK, FYN, and TYK2 showed high expression as early as 12 h. These kinases may lead to rapid morphological changes in co-infected gastric cells. Likewise, apoptotic gene expression such as APAF-1 and Bcl2 family genes such as BAD, BID, BIK, BIM, BAX, AND BAK were significantly down-regulated in co-infected AGS cells. CONCLUSION: All the experiments were performed with novel isolates of H. pylori isolated from central India, for the functional assessment of GC. The effect of coinfection with EBV was more profoundly observed on morphological changes in AGS cells at 12 h as quantified by measuring the tapering of ends. This study also identifies the kinase and apoptotic genes modulated in co-infected cells, through direct and indirect approaches. We report that ITK, EPHB6, TYK2, FYN kinase are enhanced, whereas apoptotic genes such as APAF-1, BIK, FASL, BAX are significantly down-regulated in AGS cells coinfected with EBV and H. pylori.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Infecciones por Helicobacter/genética , Fosfotransferasas/genética , Neoplasias Gástricas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Coinfección/genética , Coinfección/microbiología , Coinfección/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/microbiología , Infecciones por Virus de Epstein-Barr/virología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/virología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Humanos , Fosfotransferasas/clasificación , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/virología
13.
Clin Transl Gastroenterol ; 11(7): e00201, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32764207

RESUMEN

INTRODUCTION: Besides Helicobacter pylori and Epstein-Barr virus, other viruses might play potential roles in gastric carcinogenesis. This systematic review and meta-analysis was conducted to compare the prevalence of the viruses between gastric cancer (GC) and any controls. METHODS: Comprehensive literature was searched up to January 25, 2019, and search was updated on April 6, 2020. The studies that compared the prevalence of viruses other than Epstein-Barr virus between GC and healthy or nonmalignant controls were eligible. Stata 12.0 software was used for heterogeneity tests and meta-analyses. Meanwhile, subgroup analysis, sensitivity analysis, and publication bias evaluation were performed where applicable. The power (1-ß) was estimated by the PASS 11 software for each individual study. RESULTS: A total of 41 eligible studies were included, concerning 11 kinds of viruses. Prevalence were significantly higher in GC for hepatitis B virus (odds ratio [OR] = 1.39, 95% confidence interval [CI] 1.11-1.75), human cytomegalovirus (OR = 2.25, 95% CI 1.14-4.43), human papillomavirus (HPV) (OR = 1.63, 95% CI 1.05-2.54), and John Cunningham virus (OR = 2.52, 95% CI 1.26-5.04). In subgroup analyses, HPV-16 infection was significantly associated with GC (OR = 2.42, 95% CI 1.00-5.83). DISCUSSION: This study demonstrated that hepatitis B virus, human cytomegalovirus, HPV, and John Cunningham virus were more prevalent in GC. However, the causal relationship between their infection and risk of GC remains inconclusive, and further investigations are required.


Asunto(s)
Infecciones por Citomegalovirus/epidemiología , Hepatitis B/epidemiología , Infecciones por Papillomavirus/epidemiología , Infecciones por Polyomavirus/epidemiología , Neoplasias Gástricas/epidemiología , Infecciones Tumorales por Virus/epidemiología , Alphapapillomavirus/aislamiento & purificación , Citomegalovirus/aislamiento & purificación , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Hepatitis B/diagnóstico , Hepatitis B/patología , Hepatitis B/virología , Virus de la Hepatitis B/aislamiento & purificación , Virus de la Hepatitis B/patogenicidad , Humanos , Virus JC/aislamiento & purificación , Virus JC/patogenicidad , Oportunidad Relativa , Infecciones por Papillomavirus/diagnóstico , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Infecciones por Polyomavirus/diagnóstico , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Medición de Riesgo/estadística & datos numéricos , Factores de Riesgo , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Infecciones Tumorales por Virus/diagnóstico , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
14.
Clin Cancer Res ; 26(14): 3784-3790, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32156744

RESUMEN

PURPOSE: We evaluated the association between molecular subtypes of advanced gastric cancer (AGC) and the efficacy of standard chemotherapy or immune checkpoint inhibitors. EXPERIMENTAL DESIGN: Patients with AGC who received systemic chemotherapy from October 2015 to July 2018 with available molecular features were analyzed. We investigated the efficacy of standard first- (fluoropyrimidine + platinum ± trastuzumab) and second-line (taxanes ± ramucirumab) chemotherapy, and subsequent anti-PD-1 therapy in patients with four molecular subtypes: MMR-D (mismatch repair deficient), EBV+, HER2+, and all negative. RESULTS: 410 patients were analyzed: MMR-D 5.9%, EBV+ 4.1%, HER2+ 13.7%, and all negative 76.3%. In 285 patients who received standard first-line chemotherapy, the median progression-free survival (PFS) times were 4.2, 6.0, 7.5, and 7.6 months and the objective response rates (ORR) were 31%, 62%, 60%, and 49% in MMR-D, EBV+, HER2+, and all-negative subtypes, respectively. Multivariate analysis showed shorter PFS in MMR-D versus all-negative patients [HR, 1.97; 95% CIs, 1.09-3.53; P = 0.022]. In second-line setting, there were no significant differences in efficacy. In 110 patients who received anti-PD-1 therapy, median PFS times were 13.0, 3.7, 1.6, and 1.9 months and the ORRs were 58%, 33%, 7%, and 13%, respectively. Twelve patients with MMR-D received subsequent anti-PD-1 therapy and showed longer PFS compared with that in 10 (83%) patients who received earlier-line chemotherapy. CONCLUSIONS: MMR-D might result in shorter PFS with first-line chemotherapy for AGC. Subsequent anti-PD-1 therapy achieved higher ORR and longer PFS than prior chemotherapy in most patients with MMR-D, supporting the earlier use of immune checkpoint inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Mucosa Gástrica/patología , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Reparación de la Incompatibilidad de ADN , Resistencia a Antineoplásicos , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/mortalidad , Infecciones por Virus de Epstein-Barr/virología , Femenino , Estudios de Seguimiento , Mucosa Gástrica/virología , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Receptor ErbB-2/análisis , Receptor ErbB-2/metabolismo , Estudios Retrospectivos , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/virología , Adulto Joven
15.
J Med Virol ; 92(1): 71-77, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31471921

RESUMEN

Gastric cancer (GC) is one of the infection-related cancers. Helicobacter pylori and Epstein-Barr virus (EBV) were established risk factors for GC. Recently, there are several reports showing the inconsistent association between hepatitis B virus (HBV) infection and the development of GC. To explore the relationship between HBV infection and the development of GC, we designed a meta-analysis of previous epidemiological studies, a hospital-based case-control study, followed by an immunohistochemistry (IHC) assay of HBV-exposed GC samples. We found that HBV infection was associated with an increased risk of GC based on the meta-analysis. No significant association between HBV infection and GC was detected according to our hospital-based case-control study. Histological examination showed that the gastric epithelium positive for HBx demonstrated a higher nuclear-cytoplasmic ratio compared to those HBx-negative cells. HBx and HBcAg were expressed more in tumors than those in normal counterparts in HBV-exposed subjects, and PD-L1 was lower in GC tissues from HBV carriers than those in HBV clearances. In conclusion, HBV infection may contribute to a higher risk for GC based on the meta-analysis and to the morphological atypia of gastric epithelium by the histological assessment, and GC patients among HBV carriers showed lower expression of PD-L1 may lose the chance for immune checkpoint blockade therapy.


Asunto(s)
Adenocarcinoma/virología , Mucosa Gástrica/virología , Hepatitis B/complicaciones , Neoplasias Gástricas/virología , Anciano , Antígeno B7-H1/genética , Estudios de Casos y Controles , Femenino , Mucosa Gástrica/patología , Antígenos del Núcleo de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales/genética
16.
Virchows Arch ; 475(6): 757-762, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31673776

RESUMEN

Epstein-Barr virus (EBV) has been associated with about 9% of all gastric carcinomas, but its role in gastric carcinogenesis remains unclear since there is lack of evidence of EBV presence in pre-neoplastic lesions of gastric mucosa. This study intends to determine the prevalence of EBV in gastric dysplasia and superficial neoplasia to clarify whether EBV infection is an early or late event in gastric cancer development. This retrospective study included a total of 242 gastric lesions from 199 consecutive patients who were referred for endoscopic resection. The histological classification of lesions includes 137 low- and high-grade dysplasia and 105 superficial carcinomas. EBV infection was investigated by EBER-ISH. Results showed that EBV was not detected in any epithelial cells of any case with dysplasia or superficial carcinomas, although we observed the presence of a small number of EBV-infected lymphocytes in 2.1% of all lesions. These results showed that EBV is not present in gastric dysplasia neither in superficial carcinomas suggesting that EBV carcinogenesis is a late event in well/moderately differentiated gastric carcinogenesis.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Neoplasias Gástricas/virología , Estómago/virología , Anciano , Carcinoma/patología , Carcinoma/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/patología , Femenino , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Humanos , Hibridación in Situ/métodos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Estómago/patología , Neoplasias Gástricas/patología
17.
J Med Virol ; 91(3): 444-449, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30229949

RESUMEN

The Epstein-Barr virus (EBV) is one of the infectious agents found in stomach tissue. Recently, EBV-associated gastric carcinoma (EBVaGC) was classified as a new subtype of gastric carcinoma. To date, there is a lack of knowledge about the distribution and prevalence of EBV infection in both the normal stomach and various gastric lesions, including EBVaGC, in the Thai population. In this study, we detected EBV in the normal stomach (NS; n = 19), chronic gastritis (CG; n = 36), intestinal metaplasia (IM; n = 40), gastric dysplasia (GD; n = 15), and gastric adenocarcinoma (GC; n = 33) by polymerase chain reaction (PCR) amplification of the latent membrane protein (LMP1) gene of EBV. EBV-PCR amplification was positive in 42.1%, 36.1%, 22.5%, 13.3%, and 33.3% of NS, CG, IM, GD, and GC, respectively. For further clarification in EBVaGC, we performed EBV-encoded small RNA in situ hybridization (EBER-ISH) in PCR-positive cases of GD and GC. Four GC cases were EBER-ISH positive (12.1%), while both GD cases were EBER-ISH negative. In addition, we determined the distribution of the EBV strain (type A or B) based on EBNA3C sequence and EBV variants based on LMP1 variation (wild-type and 30-bp deletion variants; wt-LMP1 or del-LMP1). The results showed that type A and wt-LMP1 were the most prevalent in all lesions. In conclusion, EBV is common in both the NS and gastric lesions, and the frequency of EBVaGC was 12.1% in Thai patients.


Asunto(s)
Infecciones por Virus de Epstein-Barr/epidemiología , Mucosa Gástrica/virología , Neoplasias Gástricas/virología , Estómago/patología , Estómago/virología , Adenocarcinoma/epidemiología , Adenocarcinoma/virología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Virus de Epstein-Barr/virología , Femenino , Mucosa Gástrica/patología , Gastritis/epidemiología , Gastritis/virología , Genotipo , Voluntarios Sanos , Herpesvirus Humano 4/genética , Humanos , Masculino , Persona de Mediana Edad , ARN Viral/genética , Neoplasias Gástricas/epidemiología , Tailandia , Proteínas de la Matriz Viral/genética , Adulto Joven
18.
Dig Endosc ; 30(5): 667-671, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29729036

RESUMEN

Epstein-Barr virus-associated gastric cancer (EBV-GC) accounts for approximately 8% of gastric cancers. However, little is known regarding intramucosal EBV-GC. The present study aimed to evaluate endoscopic and clinicopathological characteristics of intramucosal EBV-GC. Pathological data of 172 patients with 173 intramucosal gastric cancers who received gastrectomy with lymph node dissection were obtained for review. EBV-encoded small RNA in situ hybridization (EBER-ISH) was carried out using a tissue microarray block. Eight intramucosal early gastric cancers (4.6%) were EBER-ISH positive in which no cases had any lymph node metastasis. Macroscopic types were either depressed or flat, dominant histology was mixed type of moderate and poorly differentiated adenocarcinoma. In detail, histological features of "lace pattern" or "lymphocyte infiltration into the stroma or cancer nests" were observed.


Asunto(s)
Adenocarcinoma/virología , Carcinoma in Situ/virología , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/aislamiento & purificación , Neoplasias Gástricas/virología , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Anciano , Carcinoma in Situ/diagnóstico por imagen , Carcinoma in Situ/patología , Carcinoma in Situ/cirugía , Infecciones por Virus de Epstein-Barr/diagnóstico por imagen , Infecciones por Virus de Epstein-Barr/cirugía , Infecciones por Virus de Epstein-Barr/virología , Femenino , Gastrectomía , Mucosa Gástrica/diagnóstico por imagen , Mucosa Gástrica/patología , Mucosa Gástrica/cirugía , Mucosa Gástrica/virología , Gastroscopía , Humanos , Hibridación in Situ , Escisión del Ganglio Linfático , Metástasis Linfática , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía
19.
Virus Res ; 244: 173-180, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29169830

RESUMEN

Epstein-Barr virus (EBV), a human herpesvirus, is linked to both epithelial and lymphoid malignancies. Induction of EBV reactivation is a potential therapeutic strategy for EBV-associated tumors. In this study, we assessed the effects of rapamycin on EBV reactivation in gastric carcinoma cells. We found that rapamycin upregulated expression of EBV lytic proteins and increased the viral proliferation triggered by the EBV lytic inducer sodium butyrate. Reverse transcription-qPCR, luciferase activity assays, chromatin immunoprecipitation and western blotting were employed to explore the mechanism by which rapamycin promotes EBV reactivation. Our results showed that rapamycin treatment resulted in increased mRNA levels of EBV immediate-early genes. Rapamycin also enhanced the transcriptional activities of the EBV immediate-early lytic promoters Zp and Rp by strengthening Sp1 binding. Repression of the cellular ataxia telangiectasia-mutated/p53 pathway by siRNA-mediated knockdown of the ataxia telangiectasia-mutated gene significantly abrogated virus reactivation by rapamycin/sodium butyrate treatment, indicating that the ataxia telangiectasia-mutated/p53 pathway is involved in rapamycin-promoted EBV reactivation. Taken together, these findings demonstrate that rapamycin might have the potential to enhance the effectiveness of oncolytic viral therapies developed for EBV-associated malignancies.


Asunto(s)
Mucosa Gástrica/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Sirolimus/farmacología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ácido Butírico/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN Viral/genética , ADN Viral/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/virología , Regulación de la Expresión Génica , Genes Reporteros , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/metabolismo , Humanos , Proteínas Inmediatas-Precoces/agonistas , Proteínas Inmediatas-Precoces/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Viroterapia Oncolítica/métodos , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Oncogene ; 37(7): 884-896, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29059152

RESUMEN

Chronic inflammation is the primary cause of gastric cancer (GC). NLRP3, as an important inflammasome component, has crucial roles in initiating inflammation. However, the potential roles of NLRP3 in GC is unknown. Here, we show that NLRP3 expression is markedly upregulated in GC, which promotes NLRP3 inflammasome activation and interleukin-1ß (IL-1ß) secretion in macrophages. In addition, NLRP3 binds to cyclin-D1 (CCND1) promoter and promotes its transcription in gastric epithelial cells. Consequently, NLRP3 enhances epithelial cells proliferation and GC tumorigenesis. Furthermore, we identify miR-22, which is constitutively expressed in gastric mucosa, as a suppressor of NLRP3. MiR-22 directly targets NLRP3 and attenuates its oncogenic effects in vitro and in vivo. However, Helicobacter pylori (H. pylori) infection suppresses miR-22 expression, while enhances NLRP3 expression, and that triggers uncontrolled proliferation of epithelial cells and the emergence of GC. Thus, our research describes a mechanism by which miR-22 suppresses NLRP3 and maintains homeostasis of gastric microenvironments and suggests miR-22 as a potential target for the intervention of GC.


Asunto(s)
Carcinogénesis/patología , Mucosa Gástrica/patología , Infecciones por Helicobacter/complicaciones , Inflamación/patología , MicroARNs/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neoplasias Gástricas/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Proliferación Celular , Estudios de Seguimiento , Mucosa Gástrica/metabolismo , Mucosa Gástrica/virología , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/virología , Helicobacter pylori/aislamiento & purificación , Humanos , Inflamasomas , Inflamación/metabolismo , Inflamación/virología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Pronóstico , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA