Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Virol ; 90(16): 7219-7230, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27252539

RESUMEN

UNLABELLED: Dengue virus (DENV) is the most common mosquito-borne virus infecting humans and is currently a serious global health challenge. To establish infection in its host cells, DENV must subvert the production and/or antiviral effects of interferon (IFN). The aim of this study was to understand the mechanisms by which DENV suppresses IFN production. We determined that DENV NS4A interacts with mitochondrial antiviral signaling protein (MAVS), which was previously found to activate NF-κB and IFN regulatory factor 3 (IRF3), thus inducing type I IFN in the mitochondrion-associated endoplasmic reticulum membranes (MAMs). We further demonstrated that NS4A is associated with the N-terminal CARD-like (CL) domain and the C-terminal transmembrane (TM) domain of MAVS. This association prevented the binding of MAVS to RIG-I, resulting in the repression of RIG-I-induced IRF3 activation and, consequently, the abrogation of IFN production. Collectively, our findings illustrate a new molecular mechanism by which DENV evades the host immune system and suggest new targets for anti-DENV strategies. IMPORTANCE: Type I interferon (IFN) constitutes the first line of host defense against invading viruses. To successfully establish infection, dengue virus (DENV) must counteract either the production or the function of IFN. The mechanism by which DENV suppresses IFN production is poorly understood and characterized. In this study, we demonstrate that the DENV NS4A protein plays an important role in suppressing interferon production through binding MAVS and disrupting the RIG-I-MAVS interaction in mitochondrion-associated endoplasmic reticulum membranes (MAMs). Our study reveals that MAVS is a novel host target of NS4A and provides a molecular mechanism for DENV evasion of the host innate immune response. These findings have important implications for understanding the pathogenesis of DENV and may provide new insights into using NS4A as a therapeutic and/or prevention target.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus del Dengue/inmunología , Dengue/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Antivirales/metabolismo , Western Blotting , Células Cultivadas , Dengue/metabolismo , Dengue/virología , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Monocitos/citología , Monocitos/metabolismo , Monocitos/virología , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteínas no Estructurales Virales/genética
2.
Cell Microbiol ; 17(1): 105-18, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25131332

RESUMEN

Interferon-inducible transmembrane proteins 1, 2 and 3 (IFITM1, IFITM2 and IFITM3) have recently been identified as potent antiviral effectors that function to suppress the entry of a broad range of enveloped viruses and modulate cellular tropism independent of viral receptor expression. However, the antiviral effect and mechanisms of IFITMs in response to viral infections remain incompletely understood and characterized. In this work, we focused our investigation on the function of the extracellular IFITM3 protein. In cell models of DENV-2 infection, we found that IFITM3 contributed to both the baseline and interferon-induced inhibition of DENV entry. Most importantly, our study for the first time demonstrated the presence of IFITM-containing exosome in the extracellular environment, and identified an ability of cellular exosome to intercellularly deliver IFITM3 and thus transmit its antiviral effect from infected to non-infected cells. Thus, our findings provide new insights in the basic mechanisms underlying the actions of IFITM3, which might lead to future development of exosome-mediated anti-viral strategies using IFITM3 as a therapeutic agent. Conceivably, variations in the basal and inducible levels of IFITMs, as well as in intracellular and extracellular levels of IFITMs, might predict the severity of dengue virus infections among individuals or across species.


Asunto(s)
Antivirales/metabolismo , Virus del Dengue/inmunología , Dengue/inmunología , Exosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Virus del Dengue/fisiología , Internalización del Virus
3.
Mar Drugs ; 11(12): 4961-72, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24335522

RESUMEN

Three new phomoxanthone compounds, phomolactonexanthones A (1), B (2) and deacetylphomoxanthone C (3), along with five known phomoxanthones, including dicerandrol A (4), dicerandrol B (5), dicerandrol (6), deacetylphomoxanthone B (7) and penexanthone A (8), were isolated in the metabolites of the fungus Phomopsis sp. HNY29-2B, which was isolated from the mangrove plants. The structures of compounds 1-3 were established on the basis of spectroscopic analysis. All compounds were evaluated against four human cancer cell lines including human breast MDA-MB-435, human colon HCT-116, human lung Calu-3 and human liver Huh7 by MTT assay. The compounds 4, 5, 7 and 8 showed cytotoxic activities against tested cancer cell lines (IC50 < 10 µM).


Asunto(s)
Hongos/química , Xantonas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Hongos/metabolismo , Células HCT116 , Humanos , Xantonas/farmacología
4.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166789, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37302425

RESUMEN

Immune checkpoint inhibitors (ICIs) have revolutionized the current treatment landscape for cancer, yet the response rates of ICIs remain unmet. Synergistic with immunotherapy, low-dose radiotherapy (LDRT) has been demonstrated to activate anti-tumor immunity - a transition from traditional radiation therapy geared toward local radical treatment to a type of immunological adjuvant. As such, studies utilizing LDRT to enhance the efficacy of immunotherapy have been increasing preclinically and clinically. This paper reviews the recent strategies of using LDRT to overcome the resistance of ICIs, as well as providing potential opportunities in cancer treatment. Despite the potential of LDRT in immunotherapy is recognized, the mechanisms behind this form of treatment remain largely elusive. Thus, we reviewed history, mechanisms and challenges associated with this form of treatment, as well as different modes of its application, to provide relatively accurate practice standards for LDRT as a sensitizing treatment when combined with immunotherapy or radio-immunotherapy.


Asunto(s)
Neoplasias , Humanos , Neoplasias/radioterapia , Inmunoterapia
5.
Mar Drugs ; 10(4): 694-711, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22690138

RESUMEN

Breast cancer remains a major health problem worldwide. While chemotherapy represents an important therapeutic modality against breast cancer, limitations in the clinical use of chemotherapy remain formidable because of chemoresistance. The HER2/PI-3K/Akt pathway has been demonstrated to play a causal role in conferring a broad chemoresistance in breast cancer cells and thus justified to be a target for enhancing the effects of anti-breast cancer chemotherapies, such as adriamycin (ADR). Agents that can either enhance the effects of chemotherapeutics or overcome chemoresistance are urgently needed for the treatment of breast cancer. In this context, SZ-685C, an agent that has been previously shown, as such, to suppress Akt signaling, is expected to increase the efficacy of chemotherapy. Our current study investigated whether SZ-685C can override chemoresistance through inhibiting Akt signaling in human breast cancer cells. ADR-resistant cells derived from human breast cancer cell lines MCF-7, MCF-7/ADR and MCF-7/Akt, were used as models to test the effects of SZ-685C. We found that SZ-685C suppressed the Akt pathway and induced apoptosis in MCF-7/ADR and MCF-7/Akt cells that are resistant to ADR treatment, leading to antitumor effects both in vitro and in vivo. Our data suggest that use of SZ-685C might represent a potentially promising approach to the treatment of ADR-resistant breast cancer.


Asunto(s)
Antraquinonas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Células HL-60 , Humanos , Células K562 , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Res ; 29(8): 641-654, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31213650

RESUMEN

Type I interferon (IFN-I) production is efficiently induced to ensure a potent innate immune response to viral infection. How this response can be enhanced, however, remains to be explored. Here, we identify a new cytoplasmic long non-coding RNA (lncRNA), lncLrrc55-AS, that drives a positive feedback loop to promote interferon regulatory factor 3 (IRF3) signaling and IFN-I production. We show that lncLrrc55-AS is virus-induced in multiple cell types via the IFN-JAK-STAT pathway. LncLrrc55-AS-deficient mice display a weakened antiviral immune response and are more susceptible to viral challenge. Mechanistically, lncLrrc55-AS binds phosphatase methylesterase 1 (PME-1), and promotes the interaction between PME-1 and the phosphatase PP2A, an inhibitor of IRF3 signaling. LncLrrc55-AS supports PME-1-mediated demethylation and inactivation of PP2A, thereby enhancing IRF3 phosphorylation and signaling. Loss of PME-1 phenocopies lncLrrc55-AS deficiency, leading to diminished IRF3 phosphorylation and IFN-I production. We have identified an IFN-induced lncRNA as a positive regulator of IFN-I production, adding mechanistic insight into lncRNA-mediated regulation of signaling in innate immunity and inflammation.


Asunto(s)
Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Rhabdoviridae/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Perros , Femenino , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Interferón Tipo I/inmunología , Células de Riñón Canino Madin Darby , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Células 3T3 NIH , Fosforilación , Células RAW 264.7 , ARN Largo no Codificante , Infecciones por Rhabdoviridae/virología
7.
J Infect ; 70(6): 631-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25499200

RESUMEN

OBJECTIVE: It has been well recognized that microRNA plays a role in the host-pathogen interaction network. The significance of microRNA in the regulation of dengue virus (DENV) replication, however, remains unknown. The objective of our study was to determine the biological function of miR-548g-3p in modulating the replication of dengue virus. METHODS: Here we report that employment of a microRNA target search algorithm to analyze the 5' untranslated region (5'UTR) consensus sequences of DENV (DENV serotypes 1-4) led to a discovery that miR-548g-3p directly targets the stem loop A promoter element within the 5'UTR, a region essential for DENV replication. Real-time PCR was used to measure the expression levels of miR-548g-3p under DENV infection. We performed overexpression and inhibition assays to test the role of miR-548g-3p on DENV replication. The protein and mRNA levels of interferon were measured by ELISA and real-time PCR respectively. RESULT: We found that overexpression of miR-548g-3p suppressed multiplication of DENV 1, 2, 3 and 4, and that miR-548g-3p was also found to interfere with DENV translation, thereby suppressing the expression of viral proteins. CONCLUSION: Our results suggest that miR-548g-3p directly regulates DENV replication and warrant further study to investigate the feasibility of microRNA-based anti-DENV approaches.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Replicación Viral , Regiones no Traducidas 5'/genética , Animales , Línea Celular , Chlorocebus aethiops , Secuencia de Consenso , Expresión Génica , Genes Reporteros , Humanos , Interferones/genética , Interferones/metabolismo , MicroARNs/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Viral/genética , Células Vero , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Int J Infect Dis ; 25: 159-64, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24927663

RESUMEN

BACKGROUND: The World Health Organization (WHO) ranks respiratory tract infection (RTI) as the second leading cause of death worldwide for children under 5 years of age. The aim of this work was to evaluate the epidemiology characteristics of respiratory viruses found in children and adults with RTI from July 2009 to June 2012 in southern China. METHODS: In this work, a total of 14 237 nasopharyngeal swabs (14 237 patients from 25 hospitals) were analyzed, and seven respiratory viruses (influenza virus, respiratory syncytial virus, parainfluenza virus, adenovirus, human metapneumovirus, human coronavirus, human bocavirus) were detected using PCR/RT-PCR from nasopharyngeal swabs. RESULTS: The demographic characteristics, viral prevalence, age distribution, seasonal distribution, and pathogen spectrum of the patients with RTIs were analyzed. Co-infection was observed in 483 specimens, but it was more common in male patients, inpatients, children, and young adults. It varied by season, being more prevalent in the spring and summer and less so in the winter. Human coronavirus and human bocavirus were the most common pathogens, tending to occur in co-infection with other respiratory viruses. CONCLUSIONS: This work adds to our knowledge of the epidemiology characteristics of these seven common respiratory viruses among patients with RTI in southern China. The detection of the specific viral causes of infection provides a useful starting point for an understanding of illness attributable to respiratory infection, and might also provide data relevant to the development of prevention strategies.


Asunto(s)
Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Virus/clasificación , Adolescente , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , Niño , Preescolar , China/epidemiología , Coinfección , Femenino , Humanos , Incidencia , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Prevalencia , Estaciones del Año , Virus/aislamiento & purificación , Adulto Joven
9.
PLoS Negl Trop Dis ; 8(8): e3088, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25122182

RESUMEN

MicroRNAs have been shown to contribute to a repertoire of host-pathogen interactions during viral infection. Our previous study demonstrated that microRNA-30e* (miR-30e*) directly targeted the IκBα 3'-UTR and disrupted the NF-κB/IκBα negative feedback loop, leading to hyperactivation of NF-κB. This current study investigated the possible role of miR-30e* in the regulation of innate immunity associated with dengue virus (DENV) infection. We found that DENV infection could induce miR-30e* expression in DENV-permissive cells, and such an overexpression of miR-30e* upregulated IFN-ß and the downstream IFN-stimulated genes (ISGs) such as OAS1, MxA and IFITM1, and suppressed DENV replication. Furthermore, suppression of IκBα mediates the enhancing effect of miR-30e* on IFN-ß-induced antiviral response. Collectively, our findings suggest a modulatory role of miR-30e* in DENV induced IFN-ß signaling via the NF-κB-dependent pathway. Further investigation is needed to evaluate whether miR-30e* has an anti-DENV effect in vivo.


Asunto(s)
Virus del Dengue , Interferones/metabolismo , MicroARNs/farmacología , FN-kappa B/metabolismo , Replicación Viral/efectos de los fármacos , Aedes/citología , Animales , Línea Celular , Virus del Dengue/efectos de los fármacos , Virus del Dengue/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA