Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 13(9): 832-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22842346

RESUMEN

The mechanisms by which tumor microenvironments modulate nucleic acid-mediated innate immunity remain unknown. Here we identify the receptor TIM-3 as key in circumventing the stimulatory effects of nucleic acids in tumor immunity. Tumor-associated dendritic cells (DCs) in mouse tumors and patients with cancer had high expression of TIM-3. DC-derived TIM-3 suppressed innate immune responses through the recognition of nucleic acids by Toll-like receptors and cytosolic sensors via a galectin-9-independent mechanism. In contrast, TIM-3 interacted with the alarmin HMGB1 to interfere with the recruitment of nucleic acids into DC endosomes and attenuated the therapeutic efficacy of DNA vaccination and chemotherapy by diminishing the immunogenicity of nucleic acids released from dying tumor cells. Our findings define a mechanism whereby tumor microenvironments suppress antitumor immunity mediated by nucleic acids.


Asunto(s)
Células Dendríticas/inmunología , Proteína HMGB1/inmunología , Inmunidad Innata , Neoplasias/inmunología , Ácidos Nucleicos/inmunología , Receptores Virales/inmunología , Microambiente Tumoral/inmunología , Animales , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteína HMGB1/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Immunoblotting , Vigilancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente , Neoplasias/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
BMC Gastroenterol ; 23(1): 55, 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36882697

RESUMEN

BACKGROUND: The effectiveness of Helicobacter pylori (H. pylori) eradication depends on the treatment protocol. This study investigates the H. pylori eradication rate in Africa using the best available evidence from databases. METHODS: Databases were searched and results were pooled together. Heterogeneity between studies was assessed using I2 test statistics. Stata version 13 software was employed to compute the pooled eradication rate. In the subgroup analysis comparison, the finding is considered significant when the confidence intervals did not overlap. RESULTS: Twenty-two studies from 9 African countries with a total population of 2,163 were included in this study. The pooled eradication rate of H. pylori was 79% (95% CI: 75%-82%), heterogeneity (I2 = 93.02%). In the subgroup analysis by study design, a higher eradication rate was reported from observational studies (85%, 95% CI: 79%-90%), compared to randomized control trials (77%, 95% CI: 73%-82%); by the duration of therapy, higher eradication rate was reported in 10-days regimen (88%, 95% CI: 84%-92%), compared to 7-days regimen (66%, 95% CI: 55%-77%); by country, the highest eradication rate was found in Ethiopia (90%; 95% CI: 87%-93%) and the lowest eradication rate was reported in Ivory Coast (22.3%; 95% CI:15%-29%); by type of H. pylori test, the highest eradication rate was reported when rapid urease test coupled with histology (88%, 95% CI: 77%-96%), and the lowest eradication rate was reported with histology alone (22.3%; 95% CI:15%-29%). Significant heterogeneity was observed with pooled prevalence (I2 = 93.02%, P < 0.000). CONCLUSIONS: In Africa, the first-line therapy showed a variable eradication rate for H. pylori. This study demonstrates the necessity to optimize current H. pylori treatment regimens in each country, taking into account the antibiotic susceptibility. Future RCT studies with standardized regimens are warranted.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Infecciones por Helicobacter/tratamiento farmacológico , Etiopía , Bases de Datos Factuales , Tiempo de Protrombina , Ensayos Clínicos Controlados Aleatorios como Asunto
3.
Int J Mol Sci ; 24(18)2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37762374

RESUMEN

Epstein-Barr virus (EBV) is associated with various human malignancies. An association between EBV infection and oral squamous cell carcinoma (OSCC) has recently been reported. We established EBV-positive OSCC cells and demonstrated that EBV infection promoted OSCC progression. However, the mechanisms by which EBV promotes OSCC progression remain poorly understood. Therefore, we performed metabolic analyses of EBV-positive OSCC cells and established a xenograft model to investigate the viral contribution to OSCC progression. Here, we demonstrated that EBV infection induced mitochondrial stress by reducing the number of mitochondrial DNA (mtDNA) copies. Microarray data from EBV-positive OSCC cells showed altered expression of glycolysis-related genes, particularly the upregulation of key genes involved in the Warburg effect, including LDHA, GLUT1, and PDK1. Furthermore, lactate production and LDH activity were elevated in EBV-positive OSCC cells. EBV infection significantly upregulated the expression levels of cancer stem cell (CSC) markers such as CD44 and CD133 in the xenograft model. In this model, tumor growth was significantly increased in EBV-positive SCC25 cells compared with that in uninfected cells. Furthermore, tumorigenicity increased after serial passages of EBV-positive SCC25 tumors. This study revealed the oncogenic role of EBV in OSCC progression by inducing the Warburg effect and cancer stemness.


Asunto(s)
Carcinoma de Células Escamosas , Infecciones por Virus de Epstein-Barr , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Herpesvirus Humano 4/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Neoplasias de la Boca/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones
4.
Immunity ; 39(6): 1070-81, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24315994

RESUMEN

Phagocytosis of apoptotic cells by myeloid cells has been implicated in the maintenance of immune homeostasis. In this study, we found that T cell immunoglobulin- and mucin domain-containing molecule-4 (TIM-4) repressed tumor-specific immunity triggered by chemotherapy-induced tumor cell death. TIM-4 was found to be highly expressed on tumor-associated myeloid cells such as macrophages (TAMs) and dendritic cells (TADCs) and danger-associated molecular patterns (DAMPs) released from chemotherapy-damaged tumor cells induced TIM-4 on tumor-associated myeloid cells recruited from bone marrow-derived precursors. TIM-4 directly interacted with AMPKα1 and activated autophagy-mediated degradation of ingested tumors, leading to reduced antigen presentation and impaired CTL responses. Consistently, blockade of the TIM-4-AMPKα1-autophagy pathway augmented the antitumor effect of chemotherapeutics by enhancing tumor-specific CTL responses. Our finding provides insight into the immune tolerance mediated by phagocytosis of dying cells, and targeting of the TIM-4-AMPKα1 interaction constitutes a unique strategy for augmenting antitumor immunity and improving cancer chemotherapy.


Asunto(s)
Presentación de Antígeno/inmunología , Autofagia/inmunología , Tolerancia Inmunológica/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Dendríticas/inmunología , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Ratones , Neoplasias/fisiopatología , Células Tumorales Cultivadas
5.
Int J Mol Sci ; 23(12)2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35743080

RESUMEN

PAR1b is a cytoplasmic serine/threonine kinase that controls cell polarity and cell-cell interaction by regulating microtubule stability while mediating cytoplasmic-to-nuclear translocation of BRCA1. PAR1b is also a cellular target of the CagA protein of Helicobacter pylori, which leads to chronic infection causatively associated with the development of gastric cancer. The CagA-PAR1b interaction inactivates the kinase activity of PAR1b and thereby dampens PAR1b-mediated BRCA1 phosphorylation, which reduces the level of nuclear BRCA1 and thereby leads to BRCAness and BRCAness-associated genome instability underlying gastric carcinogenesis. While PAR1b can multimerize within the cells, little is known about the mechanism and functional role of PAR1b multimerization. We found in the present study that PAR1b was multimerized in vitro by binding with nucleic acids (both single- and double-stranded DNA/RNA) via the spacer region in a manner independent of nucleic-acid sequences, which markedly potentiated the kinase activity of PAR1b. Consistent with these in vitro observations, cytoplasmic introduction of double-stranded DNA or expression of single-stranded RNA increased the PAR1b kinase activity in the cells. These findings indicate that the cytoplasmic DNA/RNA contribute to nuclear accumulation of BRCA1 by constitutively activating/potentiating cytoplasmic PAR1b kinase activity, which is subverted in gastric epithelial cells upon delivery of H. pylori CagA oncoprotein.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Ácidos Nucleicos , Antígenos Bacterianos/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Humanos , Ácidos Nucleicos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , ARN/metabolismo
6.
Int J Mol Sci ; 23(18)2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36142506

RESUMEN

DNA methylation of both viral and host DNA is one of the major mechanisms involved in the development of Epstein-Barr virus-associated gastric carcinoma (EBVaGC); thus, epigenetic treatment using demethylating agents would seem to be promising. We have verified the effect of MC180295, which was discovered by screening for demethylating agents. MC180295 inhibited cell growth of the EBVaGC cell lines YCCEL1 and SNU719 in a dose-dependent manner. In a cell cycle analysis, growth arrest and apoptosis were observed in both YCCEL1 and SNU719 cells treated with MC180295. MKN28 cells infected with EBV were sensitive to MC180295 and showed more significant inhibition of cell growth compared to controls without EBV infection. Serial analysis of gene expression analysis showed the expression of genes belonging to the role of BRCA1 in DNA damage response and cell cycle control chromosomal replication to be significantly reduced after MC180295 treatment. We confirmed with quantitative PCR that the expression levels of BRCA2, FANCM, RAD51, TOP2A, and CDC45 were significantly decreased by MC180295. LMP1 and BZLF1 are EBV genes with expression that is epigenetically regulated, and MC180295 could up-regulate their expression. In conclusion, MC180295 inhibited the growth of EBVaGC cells by suppressing DNA repair and the cell cycle.


Asunto(s)
Carcinoma , Infecciones por Virus de Epstein-Barr , Neoplasias Gástricas , Carcinoma/patología , Ciclo Celular/genética , ADN Helicasas/metabolismo , Reparación del ADN , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/genética , Humanos , Neoplasias Gástricas/patología
7.
Cancer Sci ; 112(11): 4799-4811, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34449934

RESUMEN

Epstein-Barr virus (EBV)-associated gastric cancer belongs to 1 of the 4 subtypes of gastric cancer and accounts for 10% of total gastric cancers. However, most cases of gastric cancer have a history of Helicobacter pylori infection. Therefore, we investigated the possibility that H. pylori infection promotes the development of EBV-associated gastric cancer. H. pylori was exposed to principal EBV receptor, CD21, negative gastric epithelial cells, and then infected with EBV recombinant expressing enhanced green fluorescent protein. Changes in EBV infectivity due to prior H. pylori exposure were analyzed using flow cytometry. The treatment of gastric epithelial cells with H. pylori increased the efficiency of EBV infection. An increase was also observed when CagA-deficient, VacA-deficient, and FlaA-deficient H. pylori strains were used, but not when cag pathogenicity island-deficient H. pylori was used. The treatment of epithelial cells with H. pylori induced the expression of accessory EBV receptors, EphA2 and NMHC-IIA, and increased the efficiency of EBV infection depending on their expression levels. When gastric epithelial cells were treated with EPHA2 or NMHC-IIA siRNA, EBV infection via H. pylori attachment was decreased. The adhesion of H. pylori induced the expression of accessory EBV receptors in gastric epithelial cells and increased the efficiency of EBV infection.


Asunto(s)
Infecciones por Virus de Epstein-Barr/etiología , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/fisiología , Herpesvirus Humano 4 , Neoplasias Gástricas/virología , Antígenos Bacterianos/metabolismo , Sitios de Ligazón Microbiológica/fisiología , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Proteínas Fluorescentes Verdes/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/patogenicidad , Humanos , Hidroliasas/deficiencia , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Oxidorreductasas/deficiencia , ARN Interferente Pequeño/farmacología , Receptor EphA2/genética , Receptor EphA2/metabolismo , Receptores de Complemento 3d/metabolismo , Neoplasias Gástricas/microbiología
8.
J Biol Chem ; 294(38): 14043-14054, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31366735

RESUMEN

Adenosine deaminases acting on RNA-1 (ADAR1) involves adenosine to inosine RNA editing and microRNA processing. ADAR1 is known to be involved in the replication of various viruses, including hepatitis C and D. However, the role of ADAR1 in hepatitis B virus (HBV) infection has not yet been elucidated. Here, for the first time, we demonstrated ADAR1 antiviral activity against HBV. ADAR1 has two splicing isoforms in human hepatocytes: constitutive p110 protein and interferon-α (IFN-α)-responsive p150 protein. We found that overexpression of ADAR1 decreased HBV RNA in an HBV culture model. A catalytic-site mutant ADAR1 also decreased HBV RNA levels, whereas another adenosine deaminases that act on the RNA (ADAR) family protein, ADAR2, did not. Moreover, the induction of ADAR1 by stimulation with IFN-α also reduced HBV RNA levels. Decreases in endogenous ADAR1 expression by knock-down or knock-out increased HBV RNA levels. A major hepatocyte-specific microRNA, miRNA-122, was found to be positively correlated with ADAR1 expression, and exogenous miRNA-122 decreased both HBV RNA and DNA, whereas, conversely, transfection with a miRNA-122 inhibitor increased them. The reduction of HBV RNA by ADAR1 expression was abrogated by p53 knock-down, suggesting the involvement of p53 in the ADAR1-mediated reduction of HBV RNA. This study demonstrated, for the first time, that ADAR1 plays an antiviral role against HBV infection by increasing the level of miRNA-122 in hepatocytes.


Asunto(s)
Adenosina Desaminasa/metabolismo , Virus de la Hepatitis B/fisiología , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Adenosina Desaminasa/genética , Línea Celular , Virus de la Hepatitis B/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , MicroARNs/metabolismo , Isoformas de Proteínas , Edición de ARN , Empalme del ARN , Proteínas de Unión al ARN/genética
9.
Biochem Biophys Res Commun ; 520(2): 373-378, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31606205

RESUMEN

Epstein-Barr virus (EBV) encodes BamHIA rightward transcript (BART) microRNAs (miRNAs). These miRNAs are expressed at high levels in epithelial tumors, such as nasopharyngeal carcinoma (NPC). BART miRNAs play important roles in EBV-associated malignancies, however, the reason for their high expression in NPC is unclear. We performed multiple sequence alignment of six completely sequenced EBV strains: Akata, YCCEL1, SNU719, C666-1, Mutu I, and M81. A single-nucleotide deletion was identified at the promoter region of BART. The luciferase assay suggested that this single-nucleotide polymorphism (SNP) significantly increased BART promoter activity. In addition to deletion, substitution at the same site also increased BART promoter activity. Analysis of the 170 EBV genome sequences from NPC and EBV-associated gastric cancers revealed that the frequency of this SNP was associated with NPC incidence and this SNP was found to be accumulated in the BART promoter region. Overall, our results suggested that this SNP should enhance BART promoter activity and thus, might contribute to the development of EBV-associated epithelial malignancies.


Asunto(s)
Herpesvirus Humano 4/genética , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/virología , Polimorfismo de Nucleótido Simple , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/patogenicidad , Humanos , MicroARNs/genética , Regiones Promotoras Genéticas
10.
Gastric Cancer ; 22(3): 486-496, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30264329

RESUMEN

BACKGROUND: Epstein-Barr virus (EBV) is an oncogenic human herpesvirus involved in the development of around 10% of gastric cancers. The overexpression of PD-L1 is one of the features of EBV-associated gastric cancer (EBVaGC); however, the function of PD-L1 has not been studied in EBVaGC. METHODS: We used three EBVaGC cell lines, SNU719 cells, NCC24 cells, and YCCEL1 cells, to evaluate the PD-L1 expression and function in EBVaGC. Jurkat T-lymphocytes expressing PD-1 were co-cultured with NCC24 and YCCEL1 cells and the cell cycles were analyzed. To study the regulatory mechanism for PD-L1 expression, the 3'UTR of PD-L1 was sequenced, and the effect of inhibitors of the IFN-γ signaling pathway was evaluated. RESULTS: All of the EBVaGC cell lines expressed PD-L1, and its expression was further enhanced by stimulation with IFN-γ. In Jurkat T-cells co-cultured with IFN-γ-stimulated NCC24 and YCCEL1 cells, the number of cells in the G0/G1 phase was significantly increased. This G0/G1 arrest was partially released by administration of anti-PD-L1 antibody. We found SNPs in PD-L1 3'UTR nucleotide sequences that were located at seed regions for microRNAs. Treatment of EBVaGC cell lines with JAK2-inhibitor, PI3K-inhibitor, and mTOR inhibitor reduced the level of PD-L1 expression to the same level as cells without IFN-γ stimulation. CONCLUSIONS: EBVaGC cells expressing high levels of PD-L1 suppress T-cell proliferation, and the IFN-γ signaling pathway is involved in the expression of PD-L1.


Asunto(s)
Antígeno B7-H1/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 4/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Gástricas/inmunología , Linfocitos T/inmunología , Apoptosis , Antígeno B7-H1/genética , Ciclo Celular , Proliferación Celular , Inhibidores Enzimáticos/farmacología , Infecciones por Virus de Epstein-Barr/virología , Humanos , Polimorfismo de Nucleótido Simple , Receptor de Muerte Celular Programada 1/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Células Tumorales Cultivadas
11.
J Virol ; 90(8): 3873-3889, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26819314

RESUMEN

UNLABELLED: Latent membrane protein 1 (LMP1) is a major oncogene essential for primary B cell transformation by Epstein-Barr virus (EBV). Previous studies suggested that some transcription factors, such as PU.1, RBP-Jκ, NF-κB, and STAT, are involved in this expression, but the underlying mechanism is unclear. Here, we identified binding sites for PAX5, AP-2, and EBF in the proximal LMP1 promoter (ED-L1p). We first confirmed the significance of PU.1 and POU domain transcription factor binding for activation of the promoter in latency III. We then focused on the transcription factors AP-2 and early B cell factor (EBF). Interestingly, among the three AP-2-binding sites in the LMP1 promoter, two motifs were also bound by EBF. Overexpression, knockdown, and mutagenesis in the context of the viral genome indicated that AP-2 plays an important role in LMP1 expression in latency II in epithelial cells. In latency III B cells, on the other hand, the B cell-specific transcription factor EBF binds to the ED-L1p and activates LMP1 transcription from the promoter. IMPORTANCE: Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is crucial for B cell transformation and oncogenesis of other EBV-related malignancies, such as nasopharyngeal carcinoma and T/NK lymphoma. Its expression is largely dependent on the cell type or condition, and some transcription factors have been implicated in its regulation. However, these previous reports evaluated the significance of specific factors mostly by reporter assay. In this study, we prepared point-mutated EBV at the binding sites of such transcription factors and confirmed the importance of AP-2, EBF, PU.1, and POU domain factors. Our results will provide insight into the transcriptional regulation of the major oncogene LMP1.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Transactivadores/metabolismo , Factor de Transcripción AP-2/metabolismo , Proteínas de la Matriz Viral/genética , Sitios de Unión , Transformación Celular Viral , Células HEK293 , Células HeLa , Humanos , Mutación , Oncogenes , Factores del Dominio POU/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo
12.
Int J Mol Sci ; 18(8)2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28757548

RESUMEN

The Epstein-Barr virus (EBV) is detected in about 10% of gastric carcinoma cases throughout the world. In EBV-associated gastric carcinoma (EBVaGC), all tumor cells harbor the clonal EBV genome. The expression of latent EBV genes is strictly regulated through the methylation of EBV DNA. The methylation of viral DNA regulates the type of EBV latency, and methylation of the tumor suppressor genes is a key abnormality in EBVaGC. The methylation frequencies of several tumor suppressor genes and cell adhesion molecules are significantly higher in EBVaGC than in control cases. EBV-derived microRNAs repress translation from viral and host mRNAs. EBV regulates the expression of non-coding RNA in gastric carcinoma. With regard to the clinical application of demethylating agents against EBVaGC, we investigated the effects of decitabine against the EBVaGC cell lines. Decitabine inhibited the cell growth of EBVaGC cells. The promoter regions of p73 and Runt-related transcription factor 3(RUNX3) were demethylated, and their expression was upregulated by the treatment. We review the role of epigenetic regulation in the development and maintenance of EBVaGC and discuss the therapeutic application of DNA demethylating agents for EBVaGC.


Asunto(s)
Metilación de ADN , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/patogenicidad , MicroARNs/genética , Neoplasias Gástricas/virología , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Azacitidina/farmacología , Azacitidina/uso terapéutico , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Islas de CpG/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , ADN Viral/genética , Decitabina , Epigénesis Genética/efectos de los fármacos , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/genética , Humanos , ARN Viral/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Proteína Tumoral p73/genética
13.
J Gen Virol ; 97(11): 2989-3006, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27655016

RESUMEN

Epstein-Barr virus (EBV) establishes a lifelong latent infection in B lymphocytes and often is found in epithelial cells. Several lines of evidence indicate that viral transmission mediated by cell-to-cell contact is the dominant mode of infection by EBV for epithelial cells. However, its detailed molecular mechanism has not been fully elucidated. We investigated the role of host membrane trafficking machinery in this process. We have found that adhesion molecules critical for this process are expressed in EBV-positive and -negative Burkitt's lymphoma (BL) cells and multiple epithelial cell lines. Treatment with blocking antibodies against ß1 and ß2 integrin families and their ligands suppressed EBV transmission in a dose-dependent manner. We also confirmed that adhesion molecules are upregulated in co-cultured BL cells. Immunofluorescence staining revealed that the intracellular adhesion molecule 1 (ICAM-1) distributed to the cell surface and partially co-localized with recycling endosomes in co-cultured BL cells. Moreover, cell-to-cell EBV transmission was inhibited upon blocking endocytic recycling by expression of a dominant-negative form of a small GTPase Rab11 or by knockdown of Rab11, supporting the notion that the endocytic pathway-dependent trafficking of ICAM-1 to the cell surface of BL cells contributes to viral transmission by stabilizing cell-to-cell contact between the donor cells and recipient cells. Finally, we demonstrated that co-cultivation upregulated clathrin-mediated endocytosis in the recipient cells, allowing EBV to be internalized. Taken together, our findings demonstrate that EBV exploits host endocytic machinery in both donor and recipient cells, a process which is facilitated by cell-to-cell contact, thereby promoting successful viral transmission.


Asunto(s)
Endocitosis , Infecciones por Virus de Epstein-Barr/fisiopatología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Sinapsis/virología , Linfoma de Burkitt , Células Epiteliales/metabolismo , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Humanos , Molécula 1 de Adhesión Intercelular , Sinapsis/metabolismo
14.
Cancer Sci ; 105(2): 211-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24238277

RESUMEN

Measles virus (MV) is one of the candidates for the application of oncolytic virotherapy (OVT). Although an advanced clinical study has been reported on a T-cell lymphoma, the potential of MV OVT against B-cell lymphomas remains to be clarified. We found that an EBV-transformed B lymphoblastoid cell line, a model for diffuse large B-cell lymphoma, and EBV-positive Burkitt's lymphoma cells bearing type III latency were highly susceptible to the cytolysis induced by an MV vaccine strain CAM-70. As analyzed by EBV-positive and -negative counterparts of the same cytogenetic background, type III EBV latency, not type I, was shown to augment the susceptibility of B lymphoma cells to MV-induced cytolysis. Cell surface levels of CD150/signaling lymphocytic activation molecule, a receptor of MV, were upregulated in B lymphoma cell lines with type III EBV latency by 3.8-fold, on average. The cytolytic activity of CD150-tropic WT MV was akin to that of CD46- and CD150-tropic CAM-70, suggesting that CD150 is critical for the susceptibility to MV-induced cytolysis. Among EBV-encoded genes, latent membrane protein 1 was responsible for the CD150 upregulation. It was notable that the majority of B lymphoma cell lines of type III EBV latency showed higher susceptibility to the non-Edmonston-derived CAM-70 than to the Edmonston-derived Schwarz strain. This is the first report indicating the potential of non-Edmonston MV strain for the application of OVT. Furthermore, a cellular regulator of MV replication was implicated that functions in a vaccine strain-specific fashion. Altogether, the MV OVT should serve as an alternative therapy against EBV-positive diffuse large B-cell lymphoma with type III EBV latency.


Asunto(s)
Antígenos CD/metabolismo , Herpesvirus Humano 4/fisiología , Linfoma de Células B/metabolismo , Linfoma de Células B/virología , Virus del Sarampión/fisiología , Receptores de Superficie Celular/metabolismo , Antígenos CD/genética , Antígenos Virales/genética , Antígenos Virales/metabolismo , Línea Celular Tumoral , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Humanos , Período de Latencia Psicosexual , Activación de Linfocitos , Linfoma de Células B/genética , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/virología , Virus del Sarampión/genética , Virus del Sarampión/metabolismo , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Regulación hacia Arriba , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Latencia del Virus , Replicación Viral/genética
15.
J Virol ; 87(18): 10334-47, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864627

RESUMEN

Epstein-Barr virus (EBV), a human gammaherpesvirus, establishes a lifelong latent infection in B lymphocytes and epithelial cells following primary infection. Several lines of evidence suggest that exosomes derived from EBV-infected cells are internalized and transfer viral factors, including EBV-encoded latent membrane protein and microRNAs, to the recipient cells. However, the detailed mechanism by which exosomes are internalized and their physiological impact on the recipient cells are still poorly understood. In this study, we visualized the internalization of fluorescently labeled exosomes derived from EBV-uninfected and EBV-infected B cells of type I and type III latency into EBV-negative epithelial cells. In this way, we demonstrated that exosomes derived from all three cell types were internalized into the target cells in a similar fashion. Internalization of exosomes was significantly suppressed by treatment with an inhibitor of dynamin and also by the knockdown of caveolin-1. Labeled exosomes were colocalized with caveolae and subsequently trafficked through endocytic pathways. Moreover, we observed that exosomes derived from type III latency cells upregulated proliferation and expression of intercellular adhesion molecule 1 (ICAM-1) in the recipient cells more significantly than did those derived from EBV-negative and type I latency cells. We also identified the EBV latent membrane protein 1 (LMP1) gene as responsible for induction of ICAM-1 expression. Taken together, our data indicate that exosomes released from EBV-infected B cells are internalized via caveola-dependent endocytosis, which, in turn, contributes to phenotypic changes in the recipient cells through transferring one or more viral factors.


Asunto(s)
Linfocitos B/virología , Caveolas/metabolismo , Endocitosis , Células Epiteliales/virología , Exosomas/metabolismo , Herpesvirus Humano 4/crecimiento & desarrollo , Linfocitos B/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Humanos
16.
J Infect Chemother ; 20(3): 169-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24462438

RESUMEN

In the stomach of a gastric ulcer patient who had been administered an anti-acid, a gram-negative and urease-negative bacillus similar in size to Helicobacter pylori was infected together with H. pylori. According to biochemical test and 16S rRNA gene analysis, the urease-negative bacterium was identified as Kingella denitrificans, a human nasopharyngeal commensal. In contrast to the standard strain of K. denitrificans, the isolate showed catalase activity, did not produce acid from glucose, and exhibited acid tolerance. Acid tolerance of H. pylori was increased by cocultivation with the K. denitrificans isolate, but not with other isolates of K. denitrificans. Disruption of physiological and immunological niche by dysbiotic colonization of bacterium may provide pathological attributes to human stomach. Collectively, a careful administration of anti-acids to the elderly, especially those with atrophic gastritis, is necessary to avoid repression of the gastric barrier to bacteria.


Asunto(s)
Portador Sano/microbiología , Kingella/aislamiento & purificación , Infecciones por Neisseriaceae/microbiología , Úlcera Gástrica/microbiología , Anciano , Antiulcerosos/efectos adversos , Antiulcerosos/uso terapéutico , Biopsia , Portador Sano/diagnóstico , Mucosa Gástrica/citología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/microbiología , Helicobacter pylori , Humanos , Concentración de Iones de Hidrógeno , Kingella/fisiología , Masculino , Viabilidad Microbiana , Infecciones por Neisseriaceae/diagnóstico , Ranitidina/efectos adversos , Ranitidina/uso terapéutico , Úlcera Gástrica/tratamiento farmacológico
17.
Proc Natl Acad Sci U S A ; 108(30): 12425-30, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21746895

RESUMEN

Recent evidence has unveiled the critical role of tumor cells with stem cell activities in tumorigenicity and drug resistance, but how tumor microenvironments regulate cancer stem/initiating cells (CSCs) remains unknown. We clarified the role of tumor-associated macrophages (TAMs) and their downstream factor milk-fat globule-epidermal growth factor-VIII (MFG-E8) in the regulation of CSC activities. Bone marrow chimeric systems and adoptive cell transfers elucidated the importance of MFG-E8 from TAMs in conferring to CSCs with the ability to promote tumorigenicity and anticancer drug resistance. MFG-E8 mainly activates signal transducer and activator of transcription-3 (Stat3) and Sonic Hedgehog pathways in CSCs and further amplifies their anticancer drug resistance in cooperation with IL-6. Thus, the pharmacological targeting of key factors derived from tumor-associated inflammation provides a unique strategy to eradicate therapy-resistant tumors by manipulating CSC activities.


Asunto(s)
Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Traslado Adoptivo , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Proteínas Hedgehog/inmunología , Humanos , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas de la Leche/genética , Proteínas de la Leche/inmunología , Modelos Inmunológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología
18.
Viruses ; 16(4)2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38675863

RESUMEN

One of the methods to inactivate viruses is to denature viral proteins using released ions. However, there have been no reports detailing the effects of changes in humidity or contamination with body fluids on the inactivation of viruses. This study investigated the effects of humidity changes and saliva contamination on the efficacy of SARS-CoV-2 inactivation with ions using multiple viral strains. Virus solutions with different infectious titers were dropped onto a circular nitrocellulose membrane and irradiated with ions from 10 cm above the membrane. After the irradiation of ions for 60, 90, and 120 min, changes in viral infectious titers were measured. The effect of ions on virus inactivation under different humidity conditions was also examined using virus solutions containing 90% mixtures of saliva collected from 10 people. A decrease in viral infectivity was observed over time for all strains, but ion irradiation further accelerated the decrease in viral infectivity. Ion irradiation can inactivate all viral strains, but at 80% humidity, the effect did not appear until 90 min after irradiation. The presence of saliva protected the virus from drying and maintained infectiousness for a longer period compared with no saliva. In particular, the Omicron strain retained its infectivity titer longer than the other strains. Ion irradiation demonstrated a consistent reduction in the number of infectious viruses when compared to the control across varying levels of humidity and irradiation periods. This underscores the notable effectiveness of irradiation, even when the reduction effect is as modest as 50%, thereby emphasizing its crucial role in mitigating the rapid dissemination of SARS-CoV-2.


Asunto(s)
COVID-19 , Humedad , SARS-CoV-2 , Saliva , Inactivación de Virus , SARS-CoV-2/efectos de la radiación , SARS-CoV-2/fisiología , Saliva/virología , Humanos , Inactivación de Virus/efectos de la radiación , COVID-19/virología , COVID-19/prevención & control , Iones , Animales , Células Vero , Chlorocebus aethiops
19.
Artículo en Inglés | MEDLINE | ID: mdl-38825404

RESUMEN

BACKGROUND: Periodontal disease is the leading cause of tooth loss, and an association between periodontal disease and non-oral systemic diseases has been shown. Formation of biofilm by periodontal pathogens such as Fusobacterium nucleatum, Porphyromonas gingivalis, and Streptococcus mutans and their resistance to antimicrobial agents are at the root of persistent and chronic bacterial infections. METHODS: The bactericidal effect of far-ultraviolet (F-UV) light irradiation at 222 nm on periodontal bacteria was assessed qualitatively and quantitatively. The effect of biofilm disruption by F-UV light on periodontal bacteria was examined by crystal violet staining, and the morphologic changes of the biofilm after F-UV irradiation were explored by confocal laser microscopy and scanning electron microscopy. We developed a thin fiber-type 222 nm F-UV irradiator and studied its safety and effect of reducing bacteria in rodent models. RESULTS: F-UV light at 222 nm had a bactericidal effect on F. nucleatum, P. gingivalis, and S. mutans. Irradiation with F-UV light reduced the biofilm formed by the bacteria and sterilized them from within. Confocal laser microscopy showed a clear reduction in biofilm thickness, and scanning electron microscopy confirmed disintegration of the biofilm architecture. F-UV irradiation was less damaging to DNA and less cytotoxic than deep-ultraviolet light, and it reduced bacterial counts on the tooth surface. CONCLUSION: F-UV irradiation has the potential to destroy biofilm and act as a bactericide against pathogenic bacteria in the biofilm.

20.
Cancer Immunol Immunother ; 62(4): 629-37, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23143694

RESUMEN

Cancer vaccines have been developed to instruct the endogenous immune responses to autologous tumors and to generate durable clinical responses. However, the therapeutic benefits of cancer vaccines remain insufficient due to the multiple immunosuppressive signals delivered by tumors. Thus, to improve the clinical efficacy of cancer immunotherapy, it is important to develop new modalities to overcome immunosuppressive tumor microenvironments and elicit effective antitumor immune responses. In this study, we show that novel monoclonal antibodies (mAbs) specifically targeting either T cell immunoglobulin mucin protein-3 (TIM-3) or T cell immunoglobulin mucin protein-4 (TIM-4) enhance the therapeutic effects of vaccination against established B16 murine melanomas. This is true for vaccination with irradiated B16 melanoma cells engineered to express the flt3 ligand gene (FVAX). More importantly, combining anti-TIM-3 and anti-TIM-4 mAbs markedly increased vaccine-induced antitumor responses against established B16 melanoma. TIM-3 blockade mainly stimulated antitumor effector activities via natural killer cell-dependent mechanisms, while CD8(+) T cells served as the main effectors induced by anti-TIM-4 mAb. Our findings reveal that therapeutic manipulation of TIM-3 and TIM-4 may provide a novel strategy for improving the clinical efficacy of cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Vacunas contra el Cáncer/farmacología , Melanoma Experimental/terapia , Proteínas de la Membrana/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Sinergismo Farmacológico , Femenino , Receptor 2 Celular del Virus de la Hepatitis A , Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores Virales/inmunología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA