Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Emerg Microbes Infect ; 9(1): 2061-2075, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32902370

RESUMEN

Zika virus (ZIKV) infection during pregnancy is associated with congenital brain abnormalities, a finding that highlights the urgent need to understand mother-to-fetus transmission mechanisms. Human umbilical cord mesenchymal stem cells (hUCMSCs) are susceptible to ZIKV infection but the underlying mechanisms of viral susceptibility remain largely unexplored. In this study, we have characterized and compared host mRNA and miRNA expression profiles in hUCMSCs after infection with two lineages of ZIKV, African (MR766) and Asian (PRVABC59). RNA sequencing analysis identified differentially expressed genes involved in anti-viral immunity and mitochondrial dynamics following ZIKV infection. In particular, ZIKV-infected hUCMSCs displayed mitochondrial elongation and the treatment of hUCMSCs with mitochondrial fission inhibitor led to a dose-dependent increase in ZIKV gene expression and decrease in anti-viral signalling pathways. Moreover, small RNA sequencing analysis identified several significantly up- or down-regulated microRNAs. Interestingly, miR-142-5p was significantly downregulated upon ZIKV infection, whereas cellular targets of miR-142-5p, IL6ST and ITGAV, were upregulated. Overexpression of miR-142-5p resulted in the suppression of ZIKV replication. Furthermore, blocking ITGAV expression resulted in a significant suppression of ZIKV binding to cells, suggesting a potential role of ITGAV in ZIKV entry. In conclusion, these results demonstrate both common and specific host responses to African and Asian ZIKV lineages and indicate miR-142-5p as a key regulator of ZIKV replication in the umbilical cords.


Asunto(s)
Perfilación de la Expresión Génica , MicroARNs/genética , Mitocondrias/metabolismo , ARN Mensajero/genética , Infección por el Virus Zika/genética , Células A549 , Animales , Antivirales/inmunología , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Regulación de la Expresión Génica , Células HeLa , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata , Células Madre Mesenquimatosas/virología , Mitocondrias/virología , Análisis de Secuencia de ARN , Transducción de Señal , Cordón Umbilical/virología , Células Vero , Acoplamiento Viral , Internalización del Virus , Replicación Viral , Virus Zika , Infección por el Virus Zika/inmunología
2.
Pathogens ; 9(3)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120897

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that has emerged and caused global outbreaks since 2007. Although ZIKV proteins have been shown to suppress early anti-viral innate immune responses, little is known about the exact mechanisms. This study demonstrates that infection with either the African or Asian lineage of ZIKV leads to a modulated expression of suppressor of cytokine signaling (SOCS) genes encoding SOCS1 and SOCS3 in the following cell models: A549 human lung adenocarcinoma cells; JAr human choriocarcinoma cells; human neural progenitor cells. Studies of viral gene expression in response to SOCS1 or SOCS3 demonstrated that the knockdown of these SOCS proteins inhibited viral NS5 or ZIKV RNA expression, whereas overexpression resulted in an increased expression. Moreover, the overexpression of SOCS1 or SOCS3 inhibited the retinoic acid-inducible gene-I-like receptor-mediated activation of both type I and III interferon pathways. These results imply that SOCS upregulation following ZIKV infection modulates viral replication, possibly via the regulation of anti-viral innate immune responses.

3.
Sci Rep ; 9(1): 4760, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886186

RESUMEN

Hantavax is an inactivated vaccine for hemorrhagic fever with renal syndrome (HFRS). The immunogenic responses have not been elucidated yet. Here we conducted a cohort study in which 20 healthy subjects were administered four doses of Hantavax during 13-months period. Pre- and post- vaccinated peripheral blood mononuclear cells (PBMCs) and sera were analysed by transcriptomic and metabolomic profilings, respectively. Based on neutralizing antibody titers, subjects were subsequently classified into three groups; non responders (NRs), low responders (LRs) and high responders (HRs). Post vaccination differentially expressed genes (DEGs) associated with innate immunity and cytokine pathways were highly upregulated. DEG analysis revealed a significant induction of CD69 expression in the HRs. High resolution metabolomics (HRM) analysis showed that correlated to the antibody response, cholesteryl nitrolinoleate, octanoyl-carnitine, tyrosine, ubiquinone-9, and benzoate were significantly elevated in HRs, while chenodeoxycholic acid and methyl palmitate were upregulated in NRs and LRs, compared with HRs. Additionally, gene-metabolite interaction revealed upregulated gene-metabolite couplings in, folate biosynthesis, nicotinate and nicotinamide, arachidonic acid, thiamine and pyrimidine metabolism in a dose dependent manner in HR group. Collectively, our data provide new insight into the underlying mechanisms of the Hantavax-mediated immunogenicity in humans.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Virus Hantaan/inmunología , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Vacunas Virales/inmunología , Adulto , Anciano , Formación de Anticuerpos/inmunología , Citocinas/sangre , Femenino , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/virología , Humanos , Inmunidad Innata/inmunología , Masculino , Metaboloma/fisiología , Persona de Mediana Edad , Transcriptoma/genética , Vacunación , Vacunología/métodos , Adulto Joven
4.
J Invest Dermatol ; 139(2): 391-399, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30218650

RESUMEN

Zika virus (ZIKV) has emerged as a global pathogen causing significant public health concern. ZIKV infections in humans principally occur via mosquito bites. Thus, host skin cells are permissive to ZIKV infection and are the first line of defense against the virus. Here, we examined the role and mechanisms of antiviral skin immunity against ZIKV infection. ZIKV infection (African lineage MR766) in human dermal fibroblasts, human epidermal keratinocytes, and HaCaT keratinocytes resulted in distinct expression changes in RIG-I-like receptors, such as RIG-I and MDA5. Inhibition of RIG-I using small interfering RNA resulted in increased viral gene expression and reduced induction of IFNs and IFN-stimulated genes. Furthermore, ZIKV NS1 directly interacted with RIG-I or MDA5 and down-regulated RIG-I-like receptor-mediated antiviral signaling pathways. Asian lineage ZIKV (PRVABC59) infection also showed a distinct pattern of antiviral immunity in human skin cells, compared with other ZIKV strains. Additionally, ZIKV infections in human neural progenitor cells induced the robust activation of RIG-I-like receptor-mediated signaling, followed by highly enhanced IFN-stimulated gene expression. Our findings provide important insights into ZIKV tropism and subsequent antiviral signaling pathways that regulate ZIKV replication in human dermal fibroblasts and human epidermal keratinocytes.


Asunto(s)
Inmunidad Innata , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Fibroblastos/inmunología , Fibroblastos/virología , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Queratinocitos/inmunología , Queratinocitos/virología , Receptores Citoplasmáticos y Nucleares/inmunología , Células Vero , Infección por el Virus Zika/virología
5.
Viruses ; 10(2)2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29425176

RESUMEN

Zika virus (ZIKV) has recently emerged as a new public health threat. ZIKV infections have caused a wide spectrum of neurological diseases, such as Guillain-Barré syndrome, myelitis, meningoencephalitis, and congenital microcephaly. No effective therapies currently exist for treating patients infected with ZIKV. Herein, we evaluated the anti-viral activity of favipiravir (T-705) and ribavirin against Asian and African strains of ZIKV using different cell models, including human neuronal progenitor cells (hNPCs), human dermal fibroblasts (HDFs), human lung adenocarcinoma cells (A549) and Vero cells. Cells were treated with favipiravir or ribavirin and effects on ZIKV replication were determined using quantitative real-time PCR and plaque assay. Our results demonstrate that favipiravir or ribavirin treatment significantly inhibited ZIKV replication in a dose-dependent manner. Moreover, favipiravir treatment of ZIKV-infected hNPCs led to reduced cell death, enhanced AKT pathway phosphorylation, and increased expression of anti-apoptotic factor B cell lymphoma 2. In conclusion, our results demonstrate conclusively that favipiravir inhibits ZIKV replication and prevents cell death, and can be a promising intervention for ZIKV-associated disease.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Pirazinas/farmacología , Ribavirina/farmacología , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Humanos , Células Vero
6.
Immunobiology ; 222(11): 979-988, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28734654

RESUMEN

Schlafen (SLFN) proteins have been suggested to play important functions in cell proliferation and immune cell development. In this study, we determined the antiviral activities of putative RNA-helicase domain-containing SLFN14. Murine SLFN14 expression was specifically induced by TLR3-mediated pathways and type I interferon (IFN) in RAW264.7 mouse macrophages. To examine the role of SLFN during viral infection, cells were infected with either wild-type PR8 or delNS1/PR8 virus. SLFN14 expression was specifically induced following influenza virus infection. Overexpression of SLFN14 in A549 cells reduced viral replication, whereas knockdown of SLFN14 in RAW264.7 cells enhanced viral titers. Furthermore, SLFN14 promoted the delay in viral NP translocation from cytoplasm to nucleus and enhanced RIG-I-mediated IFN-ß signaling. In addition, SLFN14 overexpression promoted antiviral activity against varicella zoster virus (VZV), a DNA virus. In conclusion, our data suggest that SLFN14 is a novel antiviral factor for both DNA and RNA viruses.


Asunto(s)
Endorribonucleasas/metabolismo , Células Epiteliales/fisiología , Herpesvirus Humano 3/fisiología , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Macrófagos/fisiología , Infecciones por Orthomyxoviridae/inmunología , ARN Helicasas/genética , Infección por el Virus de la Varicela-Zóster/inmunología , Replicación Viral , Células A549 , Animales , Endorribonucleasas/genética , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Inmunidad , Control de Infecciones , Macrófagos/virología , Ratones , Células RAW 264.7 , ARN Helicasas/metabolismo , ARN Interferente Pequeño/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal
7.
Immune Netw ; 16(5): 286-295, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27799874

RESUMEN

Cellular replicative senescence is a major contributing factor to aging and to the development and progression of aging-associated diseases. In this study, we sought to determine viral replication efficiency of influenza virus (IFV) and Varicella Zoster Virus (VZV) infection in senescent cells. Primary human bronchial epithelial cells (HBE) or human dermal fibroblasts (HDF) were allowed to undergo numbers of passages to induce replicative senescence. Induction of replicative senescence in cells was validated by positive senescence-associated ß-galactosidase staining. Increased susceptibility to both IFV and VZV infection was observed in senescent HBE and HDF cells, respectively, resulting in higher numbers of plaque formation, along with the upregulation of major viral antigen expression than that in the non-senescent cells. Interestingly, mRNA fold induction level of virus-induced type I interferon (IFN) was attenuated by senescence, whereas IFN-mediated antiviral effect remained robust and potent in virus-infected senescent cells. Additionally, we show that a longevity-promoting gene, sirtuin 1 (SIRT1), has antiviral role against influenza virus infection. In conclusion, our data indicate that enhanced viral replication by cellular senescence could be due to senescence-mediated reduction of virus-induced type I IFN expression.

8.
J Microbiol ; 54(10): 695-700, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27687232

RESUMEN

Melanoma differentiation associated gene-7 (mda-7)/interleukin- 24 (IL-24) is a secreted cytokine, which plays an essential role in tumor suppression. Although its role as a multifunctional protein affecting broad types of cancers is well described, functions of IL-24 in host defense against virus infection are yet to be determined. In this study, we explored the anti-viral effect of recombinant IL-24 treatment during influenza infection. Infection of human lung adenocarcinoma cells (A549) with the influenza A virus up-regulated IL-24 mRNA and protein expression in a time-dependent manner. Pre-treatment of A549 cells with recombinant IL-24 protein effectively suppressed viral plaque formation. Furthermore, IL-24 treatment of A549 cells reduced viral non-structural protein 1 (NS1) synthesis, whereas IL-24 knockdown resulted in increased viral replication. Interestingly, IL-24 treatment following influenza A virus infection led to up-regulation of interferon (IFN)-induced antiviral signaling. Taken together, our results suggest that IL-24 exerts a potent suppressive effect on influenza viral replication and can be used in the treatment of influenza infection.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antivirales/farmacología , Virus de la Influenza A/efectos de los fármacos , Interleucinas/farmacología , Replicación Viral/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/genética , Células A549 , Técnicas de Silenciamiento del Gen , Humanos , Virus de la Influenza A/fisiología , Gripe Humana , Interleucinas/genética , Proteínas de Resistencia a Mixovirus/genética , Proteínas Recombinantes/farmacología , Transducción de Señal/genética , Regulación hacia Arriba , Proteínas no Estructurales Virales/genética , Ensayo de Placa Viral
9.
Nutr Res Pract ; 10(1): 3-10, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26865910

RESUMEN

BACKGROUND/OBJECTIVES: Oligonol, mainly found in lychee fruit, is an antioxidant polyphenolic compound which has been shown to have anti-inflammatory and anti-cancer properties. The detailed mechanisms by which oligonol may act as an anti-aging molecule have not been determined. MATERIALS/METHODS: In this study, we evaluated the ability of oligonol to modulate sirtuin (SIRT) expression in human lung epithelial (A549) cells. Oligonol was added to A549 cells and reactive oxygen species production, mitochondrial superoxide formation, and p21 protein levels were measured. Signaling pathways activated upon oligonol treatment were also determined by western blotting. Furthermore, the anti-aging effect of oligonol was evaluated ex vivo in mouse splenocytes and in vivo in Caenorhabditis elegans. RESULTS: Oligonol specifically induced the expression of SIRT1, whose activity is linked to gene expression, metabolic control, and healthy aging. In response to influenza virus infection of A549 cells, oligonol treatment significantly up-regulated SIRT1 expression and down-regulated viral hemagglutinin expression. Oligonol treatment also resulted in the activation of autophagy pathways and the phosphorylation of AMP-activated protein kinase (AMPK). Furthermore, oligonol-treated spleen lymphocytes from old mice showed increased cell proliferation, and mRNA levels of SIRT1 in the lungs of old mice were significantly lower than those in the lungs of young mice. Additionally, in vivo lethality assay revealed that oligonol extended the lifespan of C. elegans infected with lethal Vibrio cholerae. CONCLUSIONS: These data demonstrated that oligonol may act as an anti-aging molecule by modulating SIRT1/autophagy/AMPK pathways.

11.
Sci Rep ; 5: 16512, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26576844

RESUMEN

Emerging outbreaks of newly found, highly pathogenic avian influenza (HPAI) A(H5N8) viruses have been reported globally. Previous studies have indicated that H5N8 pathogenicity in mice is relatively moderate compared with H5N1 pathogenicity. However, detailed mechanisms underlying avian influenza pathogenicity are still undetermined. We used a high-throughput RNA-seq method to analyse host and pathogen transcriptomes in the lungs of mice infected with A/MD/Korea/W452/2014 (H5N8) and A/EM/Korea/W149/2006 (H5N1) viruses. Sequenced numbers of viral transcripts and expression levels of host immune-related genes at 1 day post infection (dpi) were higher in H5N8-infected than H5N1-infected mice. Dual sequencing of viral transcripts revealed that in contrast to the observations at 1 dpi, higher number of H5N1 genes than H5N8 genes was sequenced at 3 and 7 dpi, which is consistent with higher viral titres and virulence observed in infected lungs in vivo. Ingenuity pathway analysis revealed a more significant upregulation of death receptor signalling, driven by H5N1 than with H5N8 infection at 3 and 7 dpi. Early induction of immune response-related genes may elicit protection in H5N8-infected mice, which correlates with moderate pathogenicity in vivo. Collectively, our data provide new insight into the underlying mechanisms of the differential pathogenicity of avian influenza viruses.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Virus de la Influenza A , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/virología , Animales , Análisis por Conglomerados , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Subtipo H5N1 del Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Interferones/metabolismo , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Receptores de Muerte Celular/metabolismo , Transducción de Señal , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA