Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959732

RESUMEN

Chikungunya virus (CHIKV) and Zika virus (ZIKV) are emerging arboviruses that pose a worldwide threat to human health. Currently, neither vaccine nor antiviral treatment to control their infections is available. As the skin is a major viral entry site for arboviruses in the human host, we determined the global proteomic profile of CHIKV and ZIKV infections in human skin fibroblasts using Stable Isotope Labelling by Amino acids in Cell culture (SILAC)-based mass-spectrometry analysis. We show that the expression of the interferon-stimulated proteins MX1, IFIT1, IFIT3 and ISG15, as well as expression of defense response proteins DDX58, STAT1, OAS3, EIF2AK2 and SAMHD1 was significantly up-regulated in these cells upon infection with either virus. Exogenous expression of IFITs proteins markedly inhibited CHIKV and ZIKV replication which, accordingly, was restored following the abrogation of IFIT1 or IFIT3. Overexpression of SAMHD1 in cutaneous cells, or pretreatment of cells with the virus-like particles containing SAMHD1 restriction factor Vpx, resulted in a strong increase or inhibition, respectively, of both CHIKV and ZIKV replication. Moreover, silencing of SAMHD1 by specific SAMHD1-siRNA resulted in a marked decrease of viral RNA levels. Together, these results suggest that IFITs are involved in the restriction of replication of CHIKV and ZIKV and provide, as yet unreported, evidence for a proviral role of SAMHD1 in arbovirus infection of human skin cells.


Asunto(s)
Virus Chikungunya/fisiología , Fibroblastos/metabolismo , Fibroblastos/virología , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Piel/patología , Replicación Viral/fisiología , Virus Zika/fisiología , Línea Celular , Fiebre Chikungunya/virología , Humanos , Anotación de Secuencia Molecular , Mapas de Interacción de Proteínas , Proteolisis , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias Virales/metabolismo , Infección por el Virus Zika/virología
2.
Am J Pathol ; 186(4): 938-51, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26896692

RESUMEN

The single nucleotide polymorphism located within the IFNL3 (also known as IL28B) promoter is one of the host factors associated with hepatitis C virus (HCV) clearance by interferon (IFN)-α therapy; however the mechanism remains unknown. We investigated how IL28B gene polymorphism influences HCV clearance with infected primary human hepatocytes, liver biopsies, and hepatoma cell lines. Our study confirms that the rs12979860-T/T genotype has a strong correlation with ss469415590-ΔG/ΔG single nucleotide polymorphism that produces IFN-λ4 protein. We found that IFN-α and IFN-λ1 antiviral activity against HCV was impaired in IL28B T/T infected hepatocytes compared with C/C genotype. Western blot analysis showed that IL28B TT genotype hepatocytes expressed higher levels of IFN-λ proteins (IL28B, IL-29), preactivated IFN-stimulated gene (ISG) expression, and impaired Stat phosphorylation when stimulated with either IFN-α or IFN-λ1. Furthermore, we showed that silencing IFN-λ1 in T/T cell line reduced basal ISG expression and improved antiviral activity. Likewise, overexpression of IFN-λ (1 to 4) in C/C cells induced basal ISG expression and prevented IFN-α antiviral activity. We showed that IFN-λ4, produced at low level only in T/T cells induced expression of IL28B and IL-29 and prevented IFN-α antiviral activity in HCV cell culture. Our results suggest that IFN-λ4 protein expression associated with the IL28B-T/T variant preactivates the Janus kinase-Stat signaling, leading to impaired HCV clearance by both IFN-α and IFN-λ.


Asunto(s)
Hepatitis C Crónica/tratamiento farmacológico , Interleucinas/genética , Polimorfismo de Nucleótido Simple/genética , Antivirales/farmacología , Genotipo , Hepacivirus/genética , Hepacivirus/aislamiento & purificación , Hepatitis C Crónica/virología , Hepatocitos/metabolismo , Humanos , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interferones , Neoplasias Hepáticas/metabolismo
3.
J Virol ; 89(1): 626-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339775

RESUMEN

UNLABELLED: Ribavirin (RBV) continues to be an important component of interferon-free hepatitis C treatment regimens, as RBV alone does not inhibit hepatitis C virus (HCV) replication effectively; the reason for this ineffectiveness has not been established. In this study, we investigated the RBV resistance mechanism using a persistently HCV-infected cell culture system. The antiviral activity of RBV against HCV was progressively impaired in the persistently infected culture, whereas interferon lambda 1 (IFN-λ1), a type III IFN, showed a strong antiviral response and induced viral clearance. We found that HCV replication in persistently infected cultures induces an autophagy response that impairs RBV uptake by preventing the expression of equilibrative nucleoside transporter 1 (ENT1). The Huh-7.5 cell line treated with an autophagy inducer, Torin 1, downregulated membrane expression of ENT1 and terminated RBV uptake. In contrast, the autophagy inhibitors hydroxychloroquine (HCQ), 3-methyladenine (3-MA), and bafilomycin A1 (BafA1) prevented ENT1 degradation and enhanced RBV antiviral activity. The HCV-induced autophagy response, as well as treatment with Torin 1, degrades clathrin heavy chain expression in a hepatoma cell line. Reduced expression of the clathrin heavy chain by HCV prevents ENT1 recycling to the plasma membrane and forces ENT1 to the lysosome for degradation. This study provides a potential mechanism for the impairment of RBV antiviral activity in persistently HCV-infected cell cultures and suggests that inhibition of the HCV-induced autophagy response could be used as a strategy for improving RBV antiviral activity against HCV infection. IMPORTANCE: The results from this work will allow a review of the competing theories of antiviral therapy development in the field of HCV virology. Ribavirin (RBV) remains an important component of interferon-free hepatitis C treatment regimens. The reason why RBV alone does not inhibit HCV replication effectively has not been established. This study provides a potential mechanism for why RBV antiviral activity is impaired in persistently HCV-infected cell cultures and suggests that inhibition of the HCV-induced autophagy response could be used as a strategy to increase RBV antiviral activity against HCV infection. Therefore, it is anticipated that this work would generate a great deal of interest, not only among virologists but also among the general public.


Asunto(s)
Antivirales/metabolismo , Clatrina/metabolismo , Resistencia a Medicamentos , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Hepacivirus/efectos de los fármacos , Ribavirina/metabolismo , Línea Celular , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Transporte de Proteínas
4.
Cell Mol Life Sci ; 70(7): 1297-306, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23184194

RESUMEN

Like most positive-strand RNA viruses, hepatitis C virus (HCV) forms a membrane-associated replication complex consisting of replicating RNA, viral and host proteins anchored to altered cell membranes. We used a combination of qualitative and quantitative electron microscopy (EM), immuno-EM, and the 3D reconstruction of serial EM sections to analyze the host cell membrane alterations induced by HCV. Three different types of membrane alteration were observed: vesicles in clusters (ViCs), contiguous vesicles (CVs), and double-membrane vesicles (DMVs). The main ultrastructural change observed early in infection was the formation of a network of CVs surrounding the lipid droplets. Later stages in the infectious cycle were characterized by a large increase in the number of DMVs, which may be derived from the CVs. These DMVs are thought to constitute the membranous structures harboring the viral replication complexes in which viral replication is firmly and permanently established and to protect the virus against double-stranded RNA-triggered host antiviral responses.


Asunto(s)
Membrana Celular/ultraestructura , Membrana Celular/virología , Hepacivirus/fisiología , Hepatitis C/patología , Interacciones Huésped-Patógeno/fisiología , Membrana Celular/metabolismo , Membrana Celular/patología , Hepacivirus/genética , Hepacivirus/metabolismo , Humanos , Imagenología Tridimensional , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestructura , Membranas Intracelulares/virología , Microscopía Electrónica , Modelos Biológicos , Unión Proteica , ARN Viral/metabolismo , Células Tumorales Cultivadas , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Replicación Viral/fisiología
5.
J Hepatocell Carcinoma ; 11: 839-855, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38741679

RESUMEN

Introduction: Sorafenib, an FDA-approved drug for advanced hepatocellular carcinoma (HCC) treatment, encounters resistance in many patients. Deciphering the mechanisms underlying sorafenib resistance is crucial for devising alternative strategies to overcome it. Aim: This study aimed to investigate sorafenib resistance mechanisms using a diverse panel of HCC cell lines. Methods: HCC cell lines were subjected to continuous sorafenib treatment, and stable cell lines (Huh 7.5 and Huh 7PX) exhibiting sustained growth in its presence were isolated. The investigation of drug resistance mechanisms involved a comparative analysis of drug-targeted signal transduction pathways (EGFR/RAF/MEK/ERK/Cyclin D), sorafenib uptake, and membrane expression of the drug uptake transporter. Results: HCC cell lines (Huh 7.5 and Huh 7PX) with a higher IC50 (10µM) displayed a more frequent development of sorafenib resistance compared to those with a lower IC50 (2-4.8µM), indicating a potential impact of IC50 variation on initial treatment response. Our findings reveal that activated overexpression of Raf1 kinases and impaired sorafenib uptake, mediated by reduced membrane expression of organic cation transporter-1 (OCT1), contribute to sorafenib resistance in HCC cultures. Stable expression of the drug transporter OCT1 through cDNA transfection or adenoviral delivery of OCT1 mRNA increased sorafenib uptake and successfully overcame sorafenib resistance. Additionally, consistent with sorafenib resistance in HCC cultures, cirrhotic liver-associated human HCC tumors often exhibited impaired membrane expression of OCT1 and OCT3. Conclusion: Intrinsic differences among HCC cell clones, affecting sorafenib sensitivity at the expression level of Raf kinases, drug uptake, and OCT1 transporters, were identified. This study underscores the potential of HCC tumor targeted OCT1 expression to enhance sorafenib treatment response.

6.
Infect Genet Evol ; 95: 105066, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34487865

RESUMEN

Managing emerging infectious diseases is a current challenge in the fields of microbiology and epidemiology. Indeed, among other environmental and human-related factors, climate change and global warming favor the emergence of new pathogens. The recent Zika virus (ZIKV) epidemic, of which the large and rapid spread surprised the scientific community, is a reminder of the importance to study viruses currently responsible for sporadic infections. Increasing our knowledge of key factors involved in emerging infections is essential to implement specific monitoring that can be oriented according to the pathogen, targeted population, or at-risk environment. Recent technological developments, such as high-throughput sequencing, genome-wide association studies and CRISPR screenings have allowed the identification of human single nucleotide polymorphisms (SNPs) involved in infectious disease outcome. This review focuses on the human genetic host factors that have been identified and shown to be associated with the pathogenesis of ZIKV infection and candidate SNP targets.


Asunto(s)
Enfermedades Transmisibles Emergentes/genética , Infección por el Virus Zika/genética , Virus Zika/genética , Enfermedades Transmisibles Emergentes/virología , Humanos , Infección por el Virus Zika/virología
7.
Viruses ; 13(3)2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799906

RESUMEN

Mayaro virus (MAYV) and chikungunya virus (CHIKV) are known for their arthrotropism, but accumulating evidence shows that CHIKV infections are occasionally associated with serious neurological complications. However, little is known about the capacity of MAYV to invade the central nervous system (CNS). We show that human neural progenitors (hNPCs), pericytes and astrocytes are susceptible to MAYV infection, resulting in the production of infectious viral particles. In primary astrocytes, MAYV, and to a lesser extent CHIKV, elicited a strong antiviral response, as demonstrated by an increased expression of several interferon-stimulated genes, including ISG15, MX1 and OAS2. Infection with either virus led to an enhanced expression of inflammatory chemokines, such as CCL5, CXCL10 and CXCL11, whereas MAYV induced higher levels of IL-6, IL-12 and IL-15 in these cells. Moreover, MAYV was more susceptible than CHIKV to the antiviral effects of both type I and type II interferons. Taken together, this study shows that although MAYV and CHIKV are phylogenetically related, they induce different types of antiviral responses in astrocytes. This work is the first to evaluate the potential neurotropism of MAYV and shows that brain cells and particularly astrocytes and hNPCs are permissive to MAYV, which, consequently, could lead to MAYV-induced neuropathology.


Asunto(s)
Infecciones por Alphavirus/inmunología , Alphavirus/inmunología , Astrocitos/inmunología , Astrocitos/virología , Encéfalo/inmunología , 2',5'-Oligoadenilato Sintetasa/metabolismo , Infecciones por Alphavirus/patología , Animales , Encéfalo/virología , Línea Celular , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocina CXCL11/metabolismo , Fiebre Chikungunya/inmunología , Virus Chikungunya/inmunología , Chlorocebus aethiops , Citocinas/metabolismo , Humanos , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Proteínas de Resistencia a Mixovirus/metabolismo , Células-Madre Neurales/virología , Pericitos/virología , Ubiquitinas/metabolismo , Células Vero
8.
J Gen Virol ; 91(Pt 9): 2230-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20484561

RESUMEN

Like most other positive-strand RNA viruses, hepatitis C virus (HCV) induces changes in the host cell's membranes, resulting in a membranous web. The non-structural proteins of the viral replication complex are thought to be associated with these newly synthesized membranes. We studied this phenomenon, using a Huh7.5 cell clone displaying high levels of replication of a subgenomic replicon of the JFH-1 strain. Electron microscopy of ultrathin sections of these cells revealed the presence of numerous double membrane vesicles (DMVs), resembling those observed for other RNA viruses such as poliovirus and coronavirus. Some sections had more discrete multivesicular units consisting of circular concentric membranes organized into clusters surrounded by a wrapping membrane. These structures were highly specific to HCV as they were not detected in naive Huh7.5 cells. Preparations enriched in these structures were separated from other endoplasmic reticulum-derived membranes by cell cytoplasm homogenization and ultracentrifugation on a sucrose gradient. They were found to contain the non-structural NS3 and NS5A HCV proteins, HCV RNA and LC3-II, a specific marker of autophagic membranes. By analogy to other viral models, HCV may induce DMVs by activating the autophagy pathway. This could represent a strategy to conceal the viral RNA and help the virus to evade double-stranded RNA-triggered host antiviral responses. More detailed characterization of these virus-cell interactions may facilitate the development of new treatments active against HCV and other RNA viruses that are dependent on newly synthesized cellular membranes for replication.


Asunto(s)
Membrana Celular/virología , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno/fisiología , Línea Celular , Membrana Celular/química , Membrana Celular/ultraestructura , Hepacivirus/fisiología , Hepacivirus/ultraestructura , Humanos , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , ARN Viral/química , Proteínas Virales/química , Replicación Viral
9.
Microbes Infect ; 21(8-9): 353-360, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31158508

RESUMEN

Since the ZIKV outbreak in Brazil in 2015, the scientific community has joined efforts to gather more information on the epidemiology, clinical features and pathogenicity of the virus. Here, we summarize the most important advances made recently and discuss promising, innovative approaches to understand and control ZIKV infection.


Asunto(s)
Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/virología , Virus Zika/fisiología , Virus Zika/patogenicidad , Animales , Antivirales/farmacología , Arbovirus/patogenicidad , Arbovirus/fisiología , Brasil/epidemiología , Humanos , Inmunidad Innata , Mosquitos Vectores/virología , Vacunas Virales/inmunología , Virus Zika/efectos de los fármacos , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/inmunología
10.
Emerg Microbes Infect ; 8(1): 1003-1016, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31282298

RESUMEN

Zika virus (ZIKV) is a mosquito-borne Flavivirus that causes Zika disease with particular neurological complications, including Guillain-Barré Syndrome and congenital microcephaly. Although ZIKV has been shown to directly infect human neural progenitor cells (hNPCs), thereby decreasing their viability and growth, it is as yet unknown which of the cellular pathways involved in the disruption of neurogenesis are affected following ZIKV infection. By comparing the effect of two ZIKV strains in vitro on hNPCs, the differentiation process of the latter cells was found to lead to a decreased susceptibility to infection and cell death induced by each of the ZIKV strains, which was associated with an earlier and stronger antiviral innate immune response in infected, differentiated hNPCs, as compared to undifferentiated cells. Moreover, ZIKV modulated, both in hNPCs and in vivo in fetal brain in an experimental mouse model, the expression of the Notch pathway which is involved in cellular proliferation, apoptosis and differentiation during neurogenesis. These results show that the differentiation state of hNPCs is a significant factor contributing to the outcome of ZIKV infection and furthermore suggest that ZIKV infection might initiate early activation of the Notch pathway resulting in an abnormal differentiation process, implicated in ZIKV-induced brain injury.


Asunto(s)
Células-Madre Neurales/virología , Neurogénesis , Receptor Notch1/metabolismo , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Apoptosis , Femenino , Humanos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptor Notch1/genética , Transducción de Señal , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/fisiopatología
11.
Viruses ; 11(9)2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31470617

RESUMEN

Mayaro virus (MAYV) is an emerging arthritogenic alphavirus belonging to the Togaviridae family. Infection leads to a dengue-like illness accompanied by severe polyarthralgia. However, the molecular and cellular mechanisms of arthritis as a result of MAYV infection remain poorly understood. In the present study, we assess the susceptibility of human chondrocytes (HC), fibroblast-like synoviocytes and osteoblasts that are the major cell types involved in osteoarthritis, to infection with MAYV. We show that these cells are highly permissive to MAYV infection and that viral RNA copy number and viral titers increase over time in infected cells. Knowing that HC are the primary cells in articular cartilage and are essential for maintaining the cartilaginous matrix, gene expression studies were conducted in MAYV-infected primary HC using polymerase chain reaction (PCR) arrays. The infection of the latter cells resulted in an induction in the expression of several matrix metalloproteinases (MMP) including MMP1, MMP7, MMP8, MMP10, MMP13, MMP14 and MMP15 which could be involved in the destruction of articular cartilage. Infected HC were also found to express significantly increased levels of various IFN-stimulated genes and arthritogenic mediators such as TNF-α and IL-6. In conclusion, MAYV-infected primary HC overexpress arthritis-related genes, which may contribute to joint degradation and pathogenesis.


Asunto(s)
Infecciones por Alphavirus/virología , Alphavirus/fisiología , Artritis/genética , Condrocitos/virología , Alphavirus/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/inmunología , Adhesión Celular/genética , Supervivencia Celular , Células Cultivadas , Condrocitos/inmunología , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular/genética , Perfilación de la Expresión Génica , Humanos , Metaloproteinasas de la Matriz/genética , Osteoblastos/virología , ARN Viral/metabolismo , Sinoviocitos/virología
13.
PLoS One ; 13(10): e0206093, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30359409

RESUMEN

Zika virus (ZIKV) is an emerging arbovirus of the Flaviviridae family. Although infection with ZIKV generally leads to mild disease, its recent emergence in the Americas has been associated with an increase in the development of the Guillain-Barré syndrome in adults, as well as with neurological complications, in particular congenital microcephaly, in new-borns. To date, little information is available on neuroinflammation induced by ZIKV, notably in microglial cells in the context of their metabolic activity, a series of chemical transformations that are essential for their growth, reproduction, structural maintenance and environmental responses. Therefore, in the present study we investigated the metabolomic profile of ZIKV-infected microglia. Microglial cells were exposed to ZIKV at different time points and were analyzed by a Liquid Chromatography-High Resolution mass spectrometry-based metabolomic approach. The results show that ZIKV infection in microglia leads to modulation of the expression of numerous metabolites, including lysophospholipids, particulary Lysophosphatidylcholine, and phospholipids such as Phosphatidylcholine, Phosphatidylserine, Ceramide and Sphingomyelin, and carboxylicic acids as Undecanedioic and Dodecanedioic acid. Some of these metabolites are involved in neuronal differentiation, regulation of apoptosis, virion architecture and viral replication. ZIKV infection was associated with concomitant secretion of inflammatory mediators linked with central nervous system inflammation such as IL-6, TNF-α, IL-1ß, iNOS and NO. It also resulted in the upregulation of the expression of the gene encoding CX3CR1, a chemokine receptor known to regulate functional synapse plasticity and signaling between microglial cells. These findings highlight an important role for microglia and their metabolites in the process of neuroinflammation that occurs during ZIKV pathogenesis.


Asunto(s)
Metaboloma/fisiología , Microglía/metabolismo , Infección por el Virus Zika/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops , Culicidae , Feto/citología , Feto/virología , Humanos , Metabolómica , Microcefalia/metabolismo , Microcefalia/patología , Microglía/patología , Células Vero , Replicación Viral/fisiología , Virus Zika/fisiología , Infección por el Virus Zika/patología
14.
Open Biol ; 7(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28100662

RESUMEN

Zika virus (ZIKV) causes congenital microcephaly. Although ZIKV can impair cell cycle progression and provoke apoptosis, which probably contributes to disease aetiology through depletion of neural progenitor cells, additional cellular mechanisms may be important. Here, we investigated whether ZIKV infection alters centrosome number and spindle positioning, because such defects are thought to be at the root of inherited primary autosomal recessive microcephaly (MCPH). In addition to HeLa cells, in which centrosome number and spindle positioning can be well monitored, we analysed retinal epithelial cells (RPE-1), as well as brain-derived microglial (CHME-5) and neural progenitor (ReN) cells, using immunofluorescence. We established that ZIKV infection leads to supernumerary foci containing centriolar proteins that in some cases drive multipolar spindle assembly, as well as spindle positioning defects in HeLa, RPE-1 and CHME-5 cells, but not in ReN cells. We uncovered similar phenotypes in HeLa cells upon infection with dengue virus (DENV-2), another flavivirus that does not target brain cells and does not cause microcephaly. We conclude that infection with Flaviviridae can increase centrosome numbers and impair spindle positioning, thus potentially contributing to microcephaly in the case of Zika.


Asunto(s)
Centrosoma/metabolismo , Huso Acromático/metabolismo , Infección por el Virus Zika/metabolismo , Virus Zika/patogenicidad , Animales , Línea Celular , Chlorocebus aethiops , Células HeLa , Humanos , Microglía/citología , Microglía/metabolismo , Microglía/virología , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Neuronas/virología , Retina/citología , Retina/metabolismo , Retina/virología , Células Vero , Infección por el Virus Zika/virología
15.
Infect Genet Evol ; 55: 68-70, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28866137

RESUMEN

Chikungunya virus (CHIKV) transmission occurs through the bite of an infected Aedes mosquito which injects virus-containing saliva into the skin of the human host during blood feeding. In the present study, we have determined the effect of Aedes aegypti saliva on CHIKV replication in human skin fibroblasts, a major cell type for viral entry, which mimics the events that occur during natural transmission. A significant increase in the expression of viral transcripts and infectious viral particles was observed in fibroblasts infected with CHIKV in the presence of saliva, as compared with those infected with virus alone. CHIKV-infected human fibroblasts were found to express significantly increased levels of various type I IFN-responsive genes, as demonstrated by specific PCR array analysis. In contrast, the expression of these genes was markedly decreased in cells infected with CHIKV in the presence of mosquito saliva. Moreover, Western blotting analysis revealed that STAT2 and its phosphorylated form were down-regulated in the presence of mosquito saliva. Our data demonstrate for the first time the significance of Aedes aegypti saliva in promoting CHIKV infection via down-regulation of several type I IFN-responsive genes in infected human skin fibroblasts via the JAK-STAT signaling pathway.


Asunto(s)
Aedes/virología , Fiebre Chikungunya/metabolismo , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Interferón Tipo I/metabolismo , Saliva/virología , Transducción de Señal , Replicación Viral , Animales , Células Cultivadas , Fiebre Chikungunya/genética , Fiebre Chikungunya/transmisión , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica , Humanos
16.
Infect Genet Evol ; 49: 134-137, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28095299

RESUMEN

ZIKA virus (ZIKV) is a newly emerging arbovirus. Since its discovery 60years ago in Uganda, it has spread throughout the Pacific, Latin America and the Caribbean, emphasizing the capacity of ZIKV to spread to non-endemic regions worldwide. Although infection with ZIKV often leads to mild disease, its recent emergence in the Americas has coincided with an increase in adults developing Guillain-Barré syndrome and neurological complications in new-borns, such as congenital microcephaly. Many questions remain unanswered regarding the complications caused by different primary isolates of ZIKV. Here, we report the permissiveness of primary human astrocytes for two clinically relevant, Asian and African ZIKV strains and show that both isolates strongly induce antiviral immune responses in these cells albeit with markedly different kinetics. This study describes for the first time the specific antiviral gene expression in infected primary human astrocytes, the major glial cells within the central nervous system.


Asunto(s)
Astrocitos/inmunología , Proteína 58 DEAD Box/inmunología , Interacciones Huésped-Patógeno , Proteínas NLR/inmunología , Receptores Toll-Like/inmunología , Astrocitos/virología , Proteína 58 DEAD Box/genética , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Proteínas NLR/genética , Cultivo Primario de Células , ARN Viral/biosíntesis , ARN Viral/genética , Receptores Inmunológicos , Factores de Tiempo , Receptores Toll-Like/genética , Carga Viral/inmunología , Replicación Viral/inmunología , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
17.
Sci Rep ; 7(1): 3145, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28600536

RESUMEN

Chikungunya virus (CHIKV) is an emerging arbovirus of the Togaviridae family that poses a present worldwide threat to human in the absence of any licensed vaccine or antiviral treatment to control viral infection. Here, we show that compounds interfering with intracellular cholesterol transport have the capacity to inhibit CHIKV replication in human skin fibroblasts, a major viral entry site in the human host. Pretreatment of these cells with the class II cationic amphiphilic compound U18666A, or treatment with the FDA-approved antidepressant drug imipramine resulted in a near total inhibition of viral replication and production at the highest concentration used without any cytotoxic effects. Imipramine was found to affect both the fusion and replication steps of the viral life cycle. The key contribution of cholesterol availability to the CHIKV life cycle was validated further by the use of fibroblasts from Niemann-Pick type C (NPC) patients in which the virus was unable to replicate. Interestingly, imipramine also strongly inhibited the replication of several Flaviviridae family members, including Zika, West Nile and Dengue virus. Together, these data show that this compound is a potential drug candidate for anti-arboviral treatment.


Asunto(s)
Virus Chikungunya/efectos de los fármacos , Colesterol/metabolismo , Imipramina/farmacología , Piel/virología , Androstenos/farmacología , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Fibroblastos/citología , Fibroblastos/virología , Humanos , Enfermedad de Niemann-Pick Tipo C/patología , Piel/citología , Piel/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
18.
Curr Opin Virol ; 21: 139-145, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27770704

RESUMEN

Arboviruses such as Dengue, Chikungunya, and Zika viruses represent a major public health problem due to globalization and propagation of susceptible vectors worldwide. Arthropod vector-derived salivary factors have the capacity to modulate human cells function by enhancing or suppressing viral replication and, therefore, modify the establishment of local and systemic viral infection. Here, we discuss how mosquito saliva may interfere with Dengue virus (DENV) infection in humans. Identification of saliva factors that enhance infectivity will allow the production of vector-based vaccines and therapeutics that would interfere with viral transmission by targeting arthropod saliva components. Understanding the role of salivary proteins in DENV transmission will provide tools to control not only Dengue but also other arboviral diseases transmitted by the same vectors.


Asunto(s)
Culicidae/virología , Virus del Dengue/patogenicidad , Dengue/transmisión , Transmisión de Enfermedad Infecciosa , Interacciones Huésped-Patógeno , Saliva/metabolismo , Animales , Humanos
19.
Viruses ; 8(5)2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27223299

RESUMEN

Hepatitis C virus (HCV) infection frequently leads to chronic liver disease, liver cirrhosis and hepatocellular carcinoma (HCC). The molecular mechanisms by which HCV infection leads to chronic liver disease and HCC are not well understood. The infection cycle of HCV is initiated by the attachment and entry of virus particles into a hepatocyte. Replication of the HCV genome inside hepatocytes leads to accumulation of large amounts of viral proteins and RNA replication intermediates in the endoplasmic reticulum (ER), resulting in production of thousands of new virus particles. HCV-infected hepatocytes mount a substantial stress response. How the infected hepatocyte integrates the viral-induced stress response with chronic infection is unknown. The unfolded protein response (UPR), an ER-associated cellular transcriptional response, is activated in HCV infected hepatocytes. Over the past several years, research performed by a number of laboratories, including ours, has shown that HCV induced UPR robustly activates autophagy to sustain viral replication in the infected hepatocyte. Induction of the cellular autophagy response is required to improve survival of infected cells by inhibition of cellular apoptosis. The autophagy response also inhibits the cellular innate antiviral program that usually inhibits HCV replication. In this review, we discuss the physiological implications of the HCV-induced chronic ER-stress response in the liver disease progression.


Asunto(s)
Autofagia , Estrés del Retículo Endoplásmico , Hepacivirus/fisiología , Hepatitis C/patología , Interacciones Huésped-Patógeno , Respuesta de Proteína Desplegada , Replicación Viral , Animales , Humanos , Ratones SCID
20.
PLoS One ; 10(5): e0125962, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25961570

RESUMEN

BACKGROUND: Hepatic steatosis is a risk factor for both liver disease progression and an impaired response to interferon alpha (IFN-α)-based combination therapy in chronic hepatitis C virus (HCV) infection. Previously, we reported that free fatty acid (FFA)-treated HCV cell culture induces hepatocellular steatosis and impairs the expression of interferon alpha receptor-1 (IFNAR1), which is why the antiviral activity of IFN-α against HCV is impaired. AIM: To investigate the molecular mechanism by which IFNAR1 expression is impaired in HCV cell culture with or without free fatty acid-treatment. METHOD: HCV-infected Huh 7.5 cells were cultured with or without a mixture of saturated (palmitate) and unsaturated (oleate) long-chain free fatty acids (FFA). Intracytoplasmic fat accumulation in HCV-infected culture was visualized by oil red staining. Clearance of HCV in FFA cell culture treated with type I IFN (IFN-α) and Type III IFN (IFN-λ) was determined by Renilla luciferase activity, and the expression of HCV core was determined by immunostaining. Activation of Jak-Stat signaling in the FFA-treated HCV culture by IFN-α alone and IFN-λ alone was examined by Western blot analysis and confocal microscopy. Lysosomal degradation of IFNAR1 by chaperone-mediated autophagy (CMA) in the FFA-treated HCV cell culture model was investigated. RESULTS: FFA treatment induced dose-dependent hepatocellular steatosis and lipid droplet accumulation in HCV-infected Huh-7.5 cells. FFA treatment of infected culture increased HCV replication in a concentration-dependent manner. Intracellular lipid accumulation led to reduced Stat phosphorylation and nuclear translocation, causing an impaired IFN-α antiviral response and HCV clearance. Type III IFN (IFN-λ), which binds to a separate receptor, induces Stat phosphorylation, and nuclear translocation as well as antiviral clearance in FFA-treated HCV cell culture. We show here that the HCV-induced autophagy response is increased in FFA-treated cell culture. Pharmacological inhibitors of lysosomal degradation, such as ammonium chloride and bafilomycin, prevented IFNAR1 degradation in FFA-treated HCV cell culture. Activators of chaperone-mediated autophagy, including 6-aminonicotinamide and nutrient starvation, decreased IFNAR1 levels in Huh-7.5 cells. Co-immunoprecipitation, colocalization and siRNA knockdown experiments revealed that IFNAR1 but not IFNLR1 interacts with HSC70 and LAMP2A, which are core components of chaperone-mediated autophagy (CMA). CONCLUSION: Our study presents evidence indicating that chaperone-mediated autophagy targets IFNAR1 degradation in the lysosome in FFA-treated HCV cell culture. These results provide a mechanism for why HCV induced autophagy response selectively degrades type I but not the type III IFNAR1.


Asunto(s)
Autofagia/efectos de los fármacos , Ácidos Grasos no Esterificados/metabolismo , Lisosomas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Antivirales/farmacología , Línea Celular , Células Cultivadas , Ácidos Grasos no Esterificados/farmacología , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/virología , Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Humanos , Interferón Tipo I/farmacología , Interferón gamma/farmacología , Quinasas Janus/metabolismo , Fosforilación , Unión Proteica , Proteolisis/efectos de los fármacos , Receptor de Interferón alfa y beta/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA