Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
PLoS Negl Trop Dis ; 14(8): e0008282, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817655

RESUMEN

Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, that may be involved in the physiopathological course of the infection. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human primary myoblasts that can be differentiated into myotubes, we found that myoblasts can be productively infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection, suggesting a differentiation-dependent susceptibility. Infection was accompanied by a caspase-independent cytopathic effect, associated with paraptosis-like cytoplasmic vacuolization. Proteomic profiling was performed 24h and 48h post-infection in cells infected with two different isolates. Proteome changes indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, and differentiation-dependent restriction of infection from myoblasts to myotubes. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences in ZIKV infection pathogenesis.


Asunto(s)
Diferenciación Celular , Células Musculares/metabolismo , Células Musculares/virología , Proteómica , Infección por el Virus Zika , Muerte Celular , Línea Celular , Efecto Citopatogénico Viral , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/metabolismo , Células Musculares/patología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virología , Mioblastos/metabolismo , Mioblastos/virología , Proteínas/metabolismo , Células Madre , Replicación Viral , Virus Zika/patogenicidad , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología
3.
Bull Exp Biol Med ; 167(5): 650-652, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31691878

RESUMEN

We studied the sensitivity of domestic proprietary human and animal cell lines from the collection of M. P. Chumakov Federal Scientific Center for Research and Development of Immuneand-Biological Products to infection with different enterovirus 71 strains. A cell system based on domestic proprietary permanent cell line 4647 was for the first time used for reproduction of four enterovirus 71 strains (BrCr, 42266, 42934, and 43374). It was shown that strain 4647 is the optimal cell substrate for enterovirus 71 reproduction. The titers of enterovirus 71 for all four strains considerably (by 2 lgTCID50/ml and more) increased during sequential passages in permanent cell line 4647. The prospects of using permanent cell line 4647 for creation of diagnostic and preventive preparations against 71 was demonstrated.


Asunto(s)
Enterovirus Humano A/fisiología , Células Epiteliales/virología , Células Musculares/virología , Replicación Viral , Animales , Línea Celular , Chlorocebus aethiops , Células Epiteliales/patología , Humanos , Células Musculares/patología , Carga Viral
4.
PLoS One ; 14(9): e0221048, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31498791

RESUMEN

Enterovirus 71 (EV71) induces apoptosis to promote viral particle release. Earlier work showed that EV71 utilizes its 3C protease to induce apoptosis in a caspase-3-dependent pathway, though the mechanism is unknown. However, work from Vagner, Holcik and colleagues showed that host protein heterogeneous ribonucleoprotein A1 (hnRNP A1) binds the IRES of cellular apoptotic peptidase activating factor 1 (apaf-1) mRNA to repress its translation. In this work, we show that apaf-1 expression is essential for EV71-induced apoptosis. EV71 infection or ectopic expression of 3C protease cleaves hnRNP A1, which abolishes its binding to the apaf-1 IRES. This allows IRES-dependent synthesis of apaf-1, activation of caspase-3, and apoptosis. Thus, we reveal a novel mechanism that EV71 utilizes for virus release via a 3C protease-hnRNP A1-apaf-1-caspase-3-apoptosis axis.


Asunto(s)
Factor Apoptótico 1 Activador de Proteasas/genética , Caspasa 3/genética , Cisteína Endopeptidasas/genética , Enterovirus Humano A/genética , Ribonucleoproteína Nuclear Heterogénea A1/genética , Biosíntesis de Proteínas , Proteínas Virales/genética , Proteasas Virales 3C , Animales , Apoptosis/genética , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/metabolismo , Regulación de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Sitios Internos de Entrada al Ribosoma , Células Musculares/metabolismo , Células Musculares/virología , Neuroglía/metabolismo , Neuroglía/virología , Unión Proteica , Proteolisis , Transducción de Señal , Células Vero , Proteínas Virales/metabolismo
5.
J Virol ; 92(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021893

RESUMEN

Infectious bursal disease virus (IBDV), a nonenveloped, double-stranded RNA (dsRNA) virus with a T=13 icosahedral capsid, has a virion assembly strategy that initiates with a precursor particle based on an internal scaffold shell similar to that of tailed double-stranded DNA (dsDNA) viruses. In IBDV-infected cells, the assembly pathway results mainly in mature virions that package four dsRNA segments, although minor viral populations ranging from zero to three dsRNA segments also form. We used cryo-electron microscopy (cryo-EM), cryo-electron tomography, and atomic force microscopy to characterize these IBDV populations. The VP3 protein was found to act as a scaffold protein by building an irregular, ∼40-Å-thick internal shell without icosahedral symmetry, which facilitates formation of a precursor particle, the procapsid. Analysis of IBDV procapsid mechanical properties indicated a VP3 layer beneath the icosahedral shell, which increased the effective capsid thickness. Whereas scaffolding proteins are discharged in tailed dsDNA viruses, VP3 is a multifunctional protein. In mature virions, VP3 is bound to the dsRNA genome, which is organized as ribonucleoprotein complexes. IBDV is an amalgam of dsRNA viral ancestors and traits from dsDNA and single-stranded RNA (ssRNA) viruses.IMPORTANCE Structural analyses highlight the constraint of virus evolution to a limited number of capsid protein folds and assembly strategies that result in a functional virion. We report the cryo-EM and cryo-electron tomography structures and the results of atomic force microscopy studies of the infectious bursal disease virus (IBDV), a double-stranded RNA virus with an icosahedral capsid. We found evidence of a new inner shell that might act as an internal scaffold during IBDV assembly. The use of an internal scaffold is reminiscent of tailed dsDNA viruses, which constitute the most successful self-replicating system on Earth. The IBDV scaffold protein is multifunctional and, after capsid maturation, is genome bound to form ribonucleoprotein complexes. IBDV encompasses numerous functional and structural characteristics of RNA and DNA viruses; we suggest that IBDV is a modern descendant of ancestral viruses and comprises different features of current viral lineages.


Asunto(s)
Infecciones por Birnaviridae/virología , Genoma Viral , Virus de la Enfermedad Infecciosa de la Bolsa/fisiología , ARN Bicatenario/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus , Animales , Infecciones por Birnaviridae/genética , Infecciones por Birnaviridae/metabolismo , Cápside/fisiología , Cápside/ultraestructura , Células Cultivadas , Coturnix/virología , Microscopía por Crioelectrón , Virus de la Enfermedad Infecciosa de la Bolsa/ultraestructura , Células Musculares/virología , Proteínas de Unión al ARN/genética , Proteínas Estructurales Virales/genética , Virión
6.
J Gen Virol ; 99(1): 73-85, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29182509

RESUMEN

Enterovirus A71 (EV-A71) is a positive-strand RNA virus that causes hand-foot-mouth disease and neurological complications in children and infants. Although the underlying mechanisms remain to be further defined, impaired immunity is thought to play an important role. The host zinc-finger antiviral protein (ZAP), an IFN-stimulated gene product, has been reported to specifically inhibit the replication of certain viruses. However, whether ZAP restricts the infection of enteroviruses remains unknown. Here, we report that EV-A71 infection upregulates ZAP mRNA in RD and HeLa cells. Moreover, ZAP overexpression rendered 293 T cells resistant to EV-A71 infection, whereas siRNA-mediated depletion of endogenous ZAP enhanced EV-A71 infection. The EV-A71 infection stimulated site-specific proteolysis of two ZAP isoforms, leading to the accumulation of a 40 kDa N-terminal ZAP fragment in virus-infected cells. We further revealed that the 3C protease (3Cpro) of EV-A71 mediates ZAP cleavage, which requires protease activity. Furthermore, ZAP variants with single amino acid substitutions at Gln-369 were resistant to 3Cpro cleavage, implying that Gln-369 is the sole cleavage site in ZAP. Moreover, although ZAP overexpression inhibited EV-A71 replication, the cleaved fragments did not show this effect. Our results indicate that an equilibrium between ZAP and enterovirus 3Cpro controls viral infection. The findings in this study suggest that viral 3Cpro mediated ZAP cleavage may represent a mechanism to escape host antiviral responses.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/enzimología , Interacciones Huésped-Patógeno , Proteínas de Unión al ARN/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Proteasas Virales 3C , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Enterovirus Humano A/genética , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Células HeLa , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Células Musculares/metabolismo , Células Musculares/virología , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Células Sf9/inmunología , Células Sf9/virología , Transducción de Señal , Spodoptera , Proteínas Virales/genética
7.
Lipids Health Dis ; 16(1): 189, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28969646

RESUMEN

BACKGROUND: Lipoprotein lipase (LPL) deficiency is an autosomal recessive genetic disorder characterized by extreme hypertriglyceridemia, with no cure presently available. The purpose of this study was to test the possibility of using cell therapy to alleviate LPL deficiency. METHODS: The LPL coding sequence was cloned into the MSCV retrovirus vector, after which MSCV-hLPL and MSCV (empty construct without LPL coding sequence) virion suspensions were made using the calcium chloride method. A muscle cell line (C2C12), kidney cell line (HEK293T) and pre-adipocyte cell line (3 T3-L1) were transfected with the virus in order to express recombinant LPL in vitro. Finally, each transfected cell line was injected subcutaneously into nude mice to identify the cell type which could secret recombinant LPL in vivo. Control cells were transfected with the MSCV empty vector. LPL activity was analyzed using a radioimmunoassay. RESULTS: After virus infection, the LPL activity at the cell surface of each cell type was significantly higher than in the control cells, which indicates that all three cell types can be used to generate functional LPL. The transfected cells were injected subcutaneously into nude mice, and the LPL activity of the nearby muscle tissue at the injection site in mice injected with 3 T3-L1 cells was more than 5 times higher at the injection sites than at non-injected control sites. The other two types of cells did not show this trend. CONCLUSION: The subcutaneous injection of adipocytes overexpressing LPL can improve the LPL activity of the adjacent tissue of nude mice. This is a ground-breaking preliminary study for the treatment of LPL deficiency, and lays a good foundation for using cell therapy to correct LPL deficiency.


Asunto(s)
Adipocitos/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Hiperlipoproteinemia Tipo I/terapia , Hipertrigliceridemia/terapia , Lipoproteína Lipasa/genética , Células Musculares/trasplante , Adipocitos/citología , Adipocitos/metabolismo , Adipocitos/virología , Animales , Línea Celular , Modelos Animales de Enfermedad , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/metabolismo , Hiperlipoproteinemia Tipo I/patología , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Hipertrigliceridemia/patología , Inyecciones Subcutáneas , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Desnudos , Células Musculares/citología , Células Musculares/metabolismo , Células Musculares/virología , Células 3T3 NIH , Retroviridae/genética , Retroviridae/metabolismo , Transfección , Triglicéridos/metabolismo
8.
Sci Rep ; 7(1): 8580, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819261

RESUMEN

Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Leucocitos Mononucleares/inmunología , Virus Vaccinia/inmunología , Vacunas Virales/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Hurones , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Macaca fascicularis , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones , Microscopía Confocal , Células Musculares/inmunología , Células Musculares/metabolismo , Células Musculares/virología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/fisiología
9.
J Vet Med Sci ; 79(8): 1394-1397, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28674326

RESUMEN

Attenuated derivative rabies virus Ni-CE replicates in muscle cells less efficiently than does the parental pathogenic strain Nishigahara. To examine the mechanism underlying the less efficient replication of Ni-CE, we compared the activities of Ni-CE and Nishigahara phosphoproteins, viral interferon (IFN) antagonists, to suppress IFN-ß promoter activity in muscle cells and we demonstrated a defect of Ni-CE phosphoprotein in this ability. Treatment with an IFN-ß-neutralizing antibody improved the replication efficiency of Ni-CE in muscle cells, indicating that produced IFN inhibits Ni-CE replication. The results indicate the importance of IFN antagonism of rabies virus phosphoprotein for viral replication in muscle cells.


Asunto(s)
Interferones/antagonistas & inhibidores , Células Musculares/virología , Fosfoproteínas/fisiología , Virus de la Rabia/efectos de los fármacos , Rabia/virología , Proteínas Virales/fisiología , Replicación Viral , Animales , Línea Celular , Ratones , Fosfoproteínas/genética , Virus de la Rabia/genética , Virus de la Rabia/patogenicidad , Proteínas Virales/genética , Virulencia/genética
10.
ACS Infect Dis ; 3(8): 585-594, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28605587

RESUMEN

MDL-860 is a broad-spectrum antipicornavirus compound discovered in 1982 and one of the few promising candidates effective in in vivo virus infection. Despite the effectiveness, the target and the mechanism of action of MDL-860 remain unknown. Here, we have characterized antipoliovirus activity of MDL-860 and identified host phosphatidylinositol-4 kinase III beta (PI4KB) as the target. MDL-860 treatment caused covalent modification and irreversible inactivation of PI4KB. A cysteine residue at amino acid 646 of PI4KB, which locates at the bottom of a surface pocket apart from the active site, was identified as the target site of MDL-860. This work reveals the mechanism of action of this class of PI4KB inhibitors and offers insights into novel allosteric regulation of PI4KB activity.


Asunto(s)
Antivirales/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Células Musculares/efectos de los fármacos , Nitrilos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Regulación Alostérica , Sitio Alostérico , Antivirales/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Expresión Génica , Células HEK293 , Humanos , Cinética , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Modelos Moleculares , Células Musculares/enzimología , Células Musculares/virología , Nitrilos/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Picornaviridae/efectos de los fármacos , Picornaviridae/fisiología , Unión Proteica , Replicación Viral/efectos de los fármacos
11.
Antiviral Res ; 141: 133-139, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28115196

RESUMEN

Oxymatrine is the primary pharmacological component of Sophora flavescens Ait. In the present study, we investigated the protective effect of oxymatrine against Coxsackievirus B3-induced myocarditis in mice. Coxsackievirus B3-infected HeLa cells were treated with oxymatrine and the viral titer, as well as the degree of cellular proliferation were determined. Additionally, BALB/c mice were infected with Coxsackievirus B3 and received differing concentrations of oxymatrine. On days 5 and 12 following treatment, mice were sacrificed, and serum lactate dehydrogenase, creatine kinase-MB isozyme, and tumor necrosis factor-α levels were quantified. The heart index and degree of myocardial tissue inflammation were also assessed. On day 5, the Coxsackievirus B3 TCID50 values of the heart tissue, and the expression of NTR, IFN-γ, and TNF-α genes in the myocardial tissue were measured. Our results showed that oxymatrine exhibits potent antiviral effects on Coxsackievirus B3 as 50% inhibition was achieved at a concentration as low as 0.238 mg/mL. Oxymatrine markedly reduced the viral titer and inhibited cardiac myocyte pathology exhibited in viral myocarditis. Furthermore, oxymatrine treatment reduced the expression of Coxsackievirus B3 NTR and mouse TNF-α genes compared to the controls. Therefore, our findings indicate that oxymatrine is a promising potent antiviral agent against Coxsackievirus B3-induced myocarditis.


Asunto(s)
Alcaloides/farmacología , Alcaloides/uso terapéutico , Antivirales/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , Quinolizinas/farmacología , Quinolizinas/uso terapéutico , Alcaloides/administración & dosificación , Animales , Antivirales/administración & dosificación , Infecciones por Coxsackievirus/complicaciones , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Enterovirus Humano B/efectos de los fármacos , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Células Musculares/efectos de los fármacos , Células Musculares/virología , Miocarditis/virología , Quinolizinas/administración & dosificación , Factor de Necrosis Tumoral alfa/biosíntesis , Carga Viral/efectos de los fármacos
12.
Nucleic Acids Res ; 45(1): 271-287, 2017 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-27899653

RESUMEN

Cells and viruses can utilize internal ribosome entry sites (IRES) to drive translation when cap-dependent translation is inhibited by stress or viral factors. IRES trans-acting factors (ITAFs) are known to participate in such cap-independent translation, but there are gaps in the understanding as to how ITAFs, particularly negative ITAFs, regulate IRES-driven translation. This study found that Lys109, Lys121 and Lys122 represent critical ubiquitination sites for far upstream element-binding protein 2 (KHSRP, also known as KH-type splicing regulatory protein or FBP2), a negative ITAF. Mutations at these sites subsequently reduced KHSRP ubiquitination and abolished its inhibitory effect on IRES-driven translation. We further found that interaction between the Kelch domain of Kelch-like protein 12 (KLHL12) and the C-terminal domain of KHSRP contributed to KHSRP ubiquitination, leading to downregulation of enterovirus IRES-mediated translation in infected cells and increased competition against other positive ITAFs. Together, these results show that ubiquitination can exert control over IRES-driven translation via modification of ITAFs, and to the best of our knowledge, this is the first description of such a regulatory mechanism for IRES-dependent translation.


Asunto(s)
Enterovirus/genética , Interacciones Huésped-Patógeno , Proteínas de Microfilamentos/genética , Células Musculares/metabolismo , Biosíntesis de Proteínas , Proteínas de Unión al ARN/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales , Línea Celular Tumoral , Enterovirus/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Sitios Internos de Entrada al Ribosoma , Lisina/metabolismo , Proteínas de Microfilamentos/metabolismo , Células Musculares/virología , Mutación , Dominios Proteicos , Proteínas de Unión al ARN/metabolismo , Ribosomas/química , Ribosomas/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Ubiquitinación
13.
Elife ; 3: e04531, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25490153

RESUMEN

Mutating RNA virus genomes to alter codon pair (CP) frequencies and reduce translation efficiency has been advocated as a method to generate safe, attenuated virus vaccines. However, selection for disfavoured CPs leads to unintended increases in CpG and UpA dinucleotide frequencies that also attenuate replication. We designed and phenotypically characterised mutants of the picornavirus, echovirus 7, in which these parameters were independently varied to determine which most influenced virus replication. CpG and UpA dinucleotide frequencies primarily influenced virus replication ability while no fitness differences were observed between mutants with different CP usage where dinucleotide frequencies were kept constant. Contrastingly, translation efficiency was unaffected by either CP usage or dinucleotide frequencies. This mechanistic insight is critical for future rational design of live virus vaccines and their safety evaluation; attenuation is mediated through enhanced innate immune responses to viruses with elevated CpG/UpA dinucleotide frequencies rather the viruses themselves being intrinsically defective.


Asunto(s)
Islas de CpG , Fosfatos de Dinucleósidos/metabolismo , Enterovirus Humano B/genética , ARN Viral/genética , Replicación Viral/genética , Emparejamiento Base , Línea Celular Tumoral , Codón , Fosfatos de Dinucleósidos/química , Enterovirus Humano B/metabolismo , Humanos , Células Musculares/metabolismo , Células Musculares/virología , Biosíntesis de Proteínas , ARN Viral/metabolismo , Vacunas Atenuadas , Vacunas Virales/biosíntesis , Vacunas Virales/química
14.
J Virol ; 87(22): 12327-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24027304

RESUMEN

Rabies virus (RABV), which is transmitted via a bite wound caused by a rabid animal, infects peripheral nerves and then spreads to the central nervous system (CNS) before causing severe neurological symptoms and death in the infected individual. Despite the importance of this ability of the virus to spread from a peripheral site to the CNS (neuroinvasiveness) in the pathogenesis of rabies, little is known about the mechanism underlying the neuroinvasiveness of RABV. In this study, to obtain insights into the mechanism, we conducted comparative analysis of two fixed RABV strains, Nishigahara and the derivative strain Ni-CE, which cause lethal and asymptomatic infections, respectively, in mice after intramuscular inoculation. Examination of a series of chimeric viruses harboring the respective genes from Nishigahara in the genetic background of Ni-CE revealed that the Nishigahara phosphoprotein (P) gene plays a major role in the neuroinvasiveness by mediating infection of peripheral nerves. The results obtained from both in vivo and in vitro experiments strongly suggested that the Nishigahara P gene, but not the Ni-CE P gene, is important for stable viral replication in muscle cells. Further investigation based on the previous finding that RABV phosphoprotein counteracts the host interferon (IFN) system demonstrated that the Nishigahara P gene, but not the Ni-CE P gene, functions to suppress expression of the beta interferon (IFN-ß) gene (Ifn-ß) and IFN-stimulated genes in muscle cells. In conclusion, we provide the first data strongly suggesting that RABV phosphoprotein assists viral replication in muscle cells by counteracting the host IFN system and, consequently, enhances infection of peripheral nerves.


Asunto(s)
Células Musculares/virología , Mioblastos/virología , Nervios Periféricos/virología , Fosfoproteínas/metabolismo , Virus de la Rabia/patogenicidad , Rabia/virología , Proteínas Estructurales Virales/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Western Blotting , Células Cultivadas , Femenino , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Interferones/farmacología , Ratones , Chaperonas Moleculares , Células Musculares/metabolismo , Células Musculares/patología , Mioblastos/metabolismo , Mioblastos/patología , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/virología , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Fosfoproteínas/genética , ARN Mensajero/genética , Rabia/genética , Rabia/patología , Virus de la Rabia/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Rabdomiosarcoma/virología , Proteínas Estructurales Virales/genética , Virulencia , Replicación Viral
15.
J Oral Pathol Med ; 42(6): 486-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23445118

RESUMEN

Herpes simplex virus type 1 is one of the most frequent causes of oral infection in humans, especially during early childhood. Several experimental models have been developed to study the pathogenesis of this virus but all of them employed adult animals. In this work, we developed an experimental model that uses mice younger than 4 days old, to more closely resemble human infection. Mice were infected subcutaneously with the prototype strain McIntyre of Herpes simplex-1, and the progression of infection was studied by immunoperoxidase. All animals died within 24-72 h post-infection, while viral antigens were found in the oral epithelium, nerves and brain. The most striking result was the finding of viral antigens in the nucleus and cytoplasm of cells belonging to striated muscles. Organotypic cultures of striated muscles were performed, and viral replication was observed in them by immunocytochemistry, electron microscopy and viral isolation. We conclude that the infection of striated muscles is present from the onset of oral infection and, eventually, could explain some clinical observations in humans.


Asunto(s)
Herpesvirus Humano 1/fisiología , Músculo Estriado/virología , Estomatitis Herpética/virología , Lengua/virología , Animales , Animales Recién Nacidos , Antígenos Virales/análisis , Encéfalo/virología , Causas de Muerte , Núcleo Celular/virología , Chlorocebus aethiops , Citoplasma/virología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Ratones Endogámicos BALB C , Mucosa Bucal/virología , Células Musculares/virología , Músculo Estriado/inervación , Fibras Nerviosas/virología , Neuronas/virología , Organismos Libres de Patógenos Específicos , Estomatitis Herpética/inmunología , Técnicas de Cultivo de Tejidos , Lengua/inervación , Células Vero , Replicación Viral/fisiología
16.
Microsc Res Tech ; 75(7): 986-90, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22419610

RESUMEN

Virus particles and viral inclusions were detected by transmission electron microscopy examination of sections of the seminal vesicles and mucus gland of asymptomatic young drones from colonies of Apis mellifera lightly infested by Varroa mite. In the mucus gland the infection was found in the muscular sheath and epithelium, while in the seminal vesicle in cells of the outer serosa. Isolated viral particles were also observed in the hemolymph occupying the intercellular spaces of the muscular sheath fibers. In the muscle the virus appeared as polygonal crystalloid inclusions, while in the epithelium mainly inside cytoplasmic vesicles. The infected cells apparently are not damaged. The virus particles are present in the hemolymph and forming more mature structures, as crystalloids, in the muscle. This suggests that the virus is liberated in the body fluid and infects the tissues penetrating the cells through endocytosis. The presence of virus in mucus gland epithelial vesicles raise the possibility of its transference to the gland secretion and therefore, to the semen.


Asunto(s)
Abejas/virología , Virus/aislamiento & purificación , Animales , Células Epiteliales/virología , Femenino , Genitales/virología , Hemolinfa/virología , Masculino , Microscopía Electrónica de Transmisión , Células Musculares/virología
17.
Microbes Infect ; 13(10): 862-70, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21612764

RESUMEN

Enterovirus 71 (EV71) infections can usually cause epidemic hand, foot, and mouth disease (HFMD), and occasionally lead to aseptic meningitis, encephalitis, and polio-like illness. Skeletal muscles have been thought to be crucial for the pathogenesis of EV71-related diseases. However, little is known about the virulence of mouse muscle-adapted EV71. The EV71 0805 were subjected to four passages in the mouse muscle to generate a mouse-adapted EV71 strain of 0805a. In comparison with the parental EV71 0805, the mouse muscle-adapted EV71 0805a displayed stronger cytotoxicity against Rhabdomyosarcoma (RD) cells and more efficient replication in RD cells. Furthermore, infection with the EV71 0805a significantly inhibited the gain of body weight, accompanied by increased muscle virus load and multiple tissue distribution in the infected mouse. Histological examinations indicated that infection with the EV71 0805 did not cause obvious pathogenic lesions in mice, while infection with the muscle-adapted 0805a resulted in severe necrotizing myositis in the skeletal and cardio muscles, and intestinitis in mice on day 5 post infection. Further analysis revealed many mutations in different regions of the genome of mouse muscle-adapted virus. Collectively, these data demonstrated the mouse muscle-adapted EV71 0805a with increased virulence in mice.


Asunto(s)
Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Células Musculares/virología , Adaptación Biológica , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Animales Recién Nacidos , Peso Corporal , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos ICR , Miositis , Necrosis , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología , Pase Seriado , Carga Viral , Virulencia
18.
Am J Trop Med Hyg ; 82(4): 696-704, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20348522

RESUMEN

Many West Nile (WN) virus isolates associated with significant outbreaks possess a glycosylation site on the envelope (E) protein. E-protein glycosylated variants of New York (NY) strains of WN virus are more neuroinvasive in mice than the non-glycosylated variants. To determine how E protein glycosylation affects the interactions between WN virus and avian hosts, we inoculated young chicks with NY strains of WN virus containing either glycosylated or non-glycosylated variants of the E protein. The glycosylated variants were more virulent and had higher viremic levels than the non-glycosylated variants. The glycosylation status of the variant did not affect viral multiplication and dissemination in mosquitoes in vivo. Glycosylated variants showed more heat-stable propagation than non-glycosylated variants in mammalian (BHK) and avian (QT6) cells but not in mosquito (C6/36) cells. Thus, E-protein glycosylation may be a requirement for efficient transmission of WN virus from avian hosts to mosquito vectors.


Asunto(s)
Enfermedades de las Aves de Corral/virología , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/fisiología , Fiebre del Nilo Occidental/veterinaria , Virus del Nilo Occidental/fisiología , Animales , Línea Celular , Pollos , Cricetinae , Culex , Femenino , Genes Virales , Glicosilación , Masculino , Células Musculares/patología , Células Musculares/virología , Mutación , Proteínas del Envoltorio Viral/genética , Viremia , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/patogenicidad
19.
J Virol ; 84(8): 3984-92, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20130060

RESUMEN

Intercellular adhesion molecule 1 (ICAM-1) mediates binding and entry of major group human rhinoviruses (HRVs). Whereas the entry pathway of minor group HRVs has been studied in detail and is comparatively well understood, the pathway taken by major group HRVs is largely unknown. Use of immunofluorescence microscopy, colocalization with specific endocytic markers, dominant negative mutants, and pharmacological inhibitors allowed us to demonstrate that the major group virus HRV14 enters rhabdomyosarcoma cells transfected to express human ICAM-1 in a clathrin-, caveolin-, and flotillin-independent manner. Electron microscopy revealed that many virions accumulated in long tubular structures, easily distinguishable from clathrin-coated pits and caveolae. Virus entry was strongly sensitive to the Na(+)/H(+) ion exchange inhibitor amiloride and moderately sensitive to cytochalasin D. Thus, cellular uptake of HRV14 occurs via a pathway exhibiting some, but not all, characteristics of macropinocytosis and is similar to that recently described for adenovirus 3 entry via alpha(v) integrin/CD46 in HeLa cells.


Asunto(s)
Molécula 1 de Adhesión Intercelular/biosíntesis , Células Musculares/virología , Rhinovirus/fisiología , Internalización del Virus , Amilorida/farmacología , Caveolinas/metabolismo , Línea Celular Tumoral , Clatrina/metabolismo , Citocalasina D/farmacología , Humanos , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Bloqueadores de los Canales de Sodio/farmacología
20.
Microbiol Immunol ; 52(5): 277-82, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18557898

RESUMEN

The NS2-3 of BVDV is cleaved in cultured cells infected with cp BVDV but not in those infected with ncp BVDV when tested more than 10 hours post infection. However, it is not known whether cleavage of NS2-3 occurs in vivo. In the present study, cleavage of NS2-3 in cattle persistently infected with BVDV was investigated. All BVDV isolated from PI animals were of the ncp biotype, and NS2-3 proteins were detected in bovine fetal muscular cells infected with these viruses. On the other hand, in the leukocytes of those PI animals, NS3 proteins, products of the cleavage of NS2-3 proteins, were detected. In addition, the NS3 proteins were also detected in leukocytes artificially infected with ncp BVDV. These results reveal that the NS2-3 protein of BVDV is cleaved in leukocytes. Furthermore, NS3 proteins were detected in many tissues of PI cattle, such as lymphoid tissue, brain, thyroid, lung, and kidney. These results suggest that the NS2-3 protein of ncp BVDV cleaves in vivo.


Asunto(s)
Diarrea Mucosa Bovina Viral/virología , Virus de la Diarrea Viral Bovina Tipo 1/fisiología , Proteínas no Estructurales Virales/metabolismo , Animales , Bovinos , Leucocitos/virología , Células Musculares/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...