Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4189, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760379

RESUMEN

The viral polymerase complex, comprising the large protein (L) and phosphoprotein (P), is crucial for both genome replication and transcription in non-segmented negative-strand RNA viruses (nsNSVs), while structures corresponding to these activities remain obscure. Here, we resolved two L-P complex conformations from the mumps virus (MuV), a typical member of nsNSVs, via cryogenic-electron microscopy. One conformation presents all five domains of L forming a continuous RNA tunnel to the methyltransferase domain (MTase), preferably as a transcription state. The other conformation has the appendage averaged out, which is inaccessible to MTase. In both conformations, parallel P tetramers are revealed around MuV L, which, together with structures of other nsNSVs, demonstrates the diverse origins of the L-binding X domain of P. Our study links varying structures of nsNSV polymerase complexes with genome replication and transcription and points to a sliding model for polymerase complexes to advance along the RNA templates.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Parotiditis , Proteínas Virales , Virus de la Parotiditis/genética , Virus de la Parotiditis/ultraestructura , Virus de la Parotiditis/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura , Proteínas Virales/química , Proteínas Virales/genética , Modelos Moleculares , ARN Viral/metabolismo , ARN Viral/ultraestructura , ARN Viral/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/ultraestructura , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , Dominios Proteicos , Fosfoproteínas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/ultraestructura , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/ultraestructura , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Replicación Viral , Transcripción Genética , Conformación Proteica
2.
J Virol ; 97(4): e0035923, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37017528

RESUMEN

Mumps is a highly contagious viral disease that can be prevented by vaccination. In the last decade, we have encountered repeated outbreaks of mumps in highly vaccinated populations, which call into question the effectiveness of available vaccines. Animal models are crucial for understanding virus-host interactions, and viruses such as mumps virus (MuV), whose only natural host is the human, pose a particular challenge. In our study, we examined the interaction between MuV and the guinea pig. Our results present the first evidence that guinea pigs of the Hartley strain can be infected in vivo after intranasal and intratesticular inoculation. We observed a significant viral replication in infected tissues up to 5 days following infection and induction of cellular and humoral immune responses as well as histopathological changes in infected lungs and testicles, without clinical signs of disease. Transmission of the infection through direct contact between animals was not possible. Our results demonstrate that guinea pigs and guinea pig primary cell cultures represent a promising model for immunological and pathogenetic studies of the complex MuV infection. IMPORTANCE Understanding of mumps virus (MuV) pathogenesis and the immune responses against MuV infection is limited. One of the reasons is the lack of relevant animal models. This study explores the interaction between MuV and the guinea pig. We demonstrated that all tested guinea pig tissue homogenates and primary cell cultures are highly susceptible to MuV infection and that α2,3-sialylated glycans (MuV cellular receptors) are being abundantly expressed at their surface. The virus remains in the guinea pig lungs and trachea for up to 4 days following intranasal infection. Although asymptomatic, MuV infection strongly activates both humoral and cellular immune response in infected animals and provides protection against virus challenge. Infection of the lungs and testicles after intranasal and intratesticular inoculation, respectively, is also supported by histopathological changes in these organs. Our findings give perspective for application of guinea pigs in research on MuV pathogenesis, antiviral response, and vaccine development and testing.


Asunto(s)
Virus de la Parotiditis , Paperas , Animales , Cobayas , Humanos , Paperas/inmunología , Paperas/fisiopatología , Paperas/virología , Virus de la Parotiditis/metabolismo , Replicación Viral , Células Cultivadas , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Pulmón/virología , Testículo/virología
3.
Commun Biol ; 4(1): 833, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215847

RESUMEN

Mumps virus (MuV) is a highly contagious human pathogen and frequently causes worldwide outbreaks despite available vaccines. Similar to other mononegaviruses such as Ebola and rabies, MuV uses a single-stranded negative-sense RNA as its genome, which is enwrapped by viral nucleoproteins into the helical nucleocapsid. The nucleocapsid acts as a scaffold for genome condensation and as a template for RNA replication and transcription. Conformational changes in the MuV nucleocapsid are required to switch between different activities, but the underlying mechanism remains elusive due to the absence of high-resolution structures. Here, we report two MuV nucleoprotein-RNA rings with 13 and 14 protomers, one stacked-ring filament and two nucleocapsids with distinct helical pitches, in dense and hyperdense states, at near-atomic resolutions using cryo-electron microscopy. Structural analysis of these in vitro assemblies indicates that the C-terminal tail of MuV nucleoprotein likely regulates the assembly of helical nucleocapsids, and the C-terminal arm may be relevant for the transition between the dense and hyperdense states of helical nucleocapsids. Our results provide the molecular mechanism for structural plasticity among different MuV nucleocapsids and create a possible link between structural plasticity and genome condensation.


Asunto(s)
Microscopía por Crioelectrón/métodos , Virus de la Parotiditis/metabolismo , Nucleocápside/ultraestructura , Nucleoproteínas/ultraestructura , Proteínas Virales/ultraestructura , Virión/metabolismo , Humanos , Modelos Moleculares , Virus de la Parotiditis/genética , Conformación de Ácido Nucleico , Nucleocápside/química , Nucleoproteínas/química , Conformación Proteica , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Virales/química , Virión/genética
4.
Uirusu ; 71(1): 71-78, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-35526997

RESUMEN

Mumps virus (MuV) is the causative agent of mumps, a common childhood illness characterized by fever and swelling of the salivary glands. Like other viral infections, a number of host proteins are thought to involve in MuV infection. We have shown the function of several host factors in MuV infection. The chaperone proteins, heat shock protein 70 (Hsp70) and Hsp90, interact with the P and L proteins that form the polymerase complex and function in the protein quality control of these viral proteins, and thus they are essential host factors in MuV RNA synthesis. The R2TP complex is a host factor that contributes to effective viral propagation by precise regulation of viral RNA synthesis and evasion of host immune responses, and Rab11 is a host factor involved in viral RNP trafficking to the plasma membrane. This article summarizes the functions of host factors involved in MuV infection based on our researches.


Asunto(s)
Virus de la Parotiditis , Paperas , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Interacciones Huésped-Patógeno , Humanos , Paperas/genética , Virus de la Parotiditis/genética , Virus de la Parotiditis/metabolismo , ARN Viral/metabolismo , Proteínas Virales/metabolismo
5.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32907974

RESUMEN

The mumps virus (MuV) fusion protein (F) plays a crucial role for the entry process and spread of infection by mediating fusion between viral and cellular membranes as well as between infected and neighboring cells, respectively. The fusogenicity of MuV differs depending on the strain and might correlate with the virulence; however, it is unclear which mechanisms contribute to the differentiated fusogenicity. The cleavage motif of MuV F is highly conserved among all strains, except the amino acid residue at position 8 (P8) that shows a certain variability with a total of four amino acid variants (leucine [L], proline [P], serine [S], and threonine [T]). We demonstrate that P8 affects the proteolytic processing and the fusogenicity of MuV F. The presence of L or S at P8 resulted in a slower proteolysis of MuV F by furin and a reduced ability to mediate cell-cell fusion. However, virus-cell fusion was more efficient for F proteins harboring L or S at P8, suggesting that P8 contributes to the mechanism of viral spread: P and T enable a rapid spread of infection by cell-to-cell fusion, whereas viruses harboring L or S at P8 spread preferentially by the release of infectious viral particles. Our study provides novel insights into the fusogenicity of MuV and its influence on the mechanisms of virus spread within infected tissues. Assuming a correlation between MuV fusogenicity and virulence, sequence information on the amino acid residue at P8 might be helpful to estimate the virulence of circulating and emerging strains.IMPORTANCE Mumps virus (MuV) is the causative agent of the highly infectious disease mumps. Mumps is mainly associated with mild symptoms, but severe complications such as encephalitis, meningitis, or orchitis can also occur. There is evidence that the virulence of different MuV strains and variants might correlate with the ability of the fusion protein (F) to mediate cell-to-cell fusion. However, the relation between virulence and fusogenicity or the mechanisms responsible for the varied fusogenicity of different MuV strains are incompletely understood. Here, we focused on the amino acid residue at position 8 (P8) of the proteolytic cleavage site of MuV F, because this amino acid residue shows a striking variability depending on the genotype of MuV. The P8 residue has a significant effect on the proteolytic processing and fusogenicity of MuV F and might thereby determine the route of viral spread within infected tissues.


Asunto(s)
Aminoácidos/química , Virus de la Parotiditis/metabolismo , Proteolisis , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Animales , Fusión Celular , Chlorocebus aethiops , Furina/metabolismo , Genotipo , Células HEK293 , Humanos , Cinética , Paperas/virología , Virus de la Parotiditis/genética , Homología de Secuencia de Aminoácido , Células Vero , Proteínas Virales de Fusión/genética , Internalización del Virus
6.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295904

RESUMEN

Mumps virus (MuV), an enveloped RNA virus of the Paramyxoviridae family and the causative agent of mumps, affects the salivary glands and other glandular tissues as well as the central nervous system. The virus enters the cell by inducing the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by MuV envelope proteins: the hemagglutinin-neuraminidase and fusion (F) protein. Cleavage of the MuV F protein (MuV-F) into two subunits by the cellular protease furin is a prerequisite for fusion and virus infectivity. Here, we show that 293T (a derivative of HEK293) cells do not produce syncytia upon expression of MuV envelope proteins or MuV infection. This failure is caused by the inefficient MuV-F cleavage despite the presence of functional furin in 293T cells. An expression cloning strategy revealed that overexpression of lysosome-associated membrane proteins (LAMPs) confers on 293T cells the ability to produce syncytia upon expression of MuV envelope proteins. The LAMP family comprises the ubiquitously expressed LAMP1 and LAMP2, the interferon-stimulated gene product LAMP3, and the cell type-specific proteins. The expression level of the LAMP3 gene, but not of LAMP1 and LAMP2 genes, differed markedly between 293T and HEK293 cells. Overexpression of LAMP1, LAMP2, or LAMP3 allowed 293T cells to process MuV-F efficiently. Furthermore, these LAMPs were found to interact with both MuV-F and furin. Our results indicate that LAMPs support the furin-mediated cleavage of MuV-F and that, among them, LAMP3 may be critical for the process, at least in certain cells.IMPORTANCE The cellular protease furin mediates proteolytic cleavage of many host and pathogen proteins and plays an important role in viral envelope glycoprotein maturation. MuV, an enveloped RNA virus of the Paramyxoviridae family and an important human pathogen, enters the cell through the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by the viral attachment protein and the F protein. Cleavage of MuV-F into two subunits by furin is a prerequisite for fusion and virus infectivity. Here, we show that LAMPs support the furin-mediated cleavage of MuV-F. Expression levels of LAMPs affect the processing of MuV-F and MuV-mediated membrane fusion. Among LAMPs, the interferon-stimulated gene product LAMP3 is most critical in certain cells. Our study provides potential targets for anti-MuV therapeutics.


Asunto(s)
Furina/genética , Interacciones Huésped-Patógeno/genética , Proteínas de Membrana de los Lisosomas/genética , Lisosomas/virología , Virus de la Parotiditis/genética , Proteínas de Neoplasias/genética , Proteínas Virales de Fusión/genética , Células A549 , Animales , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Furina/metabolismo , Regulación de la Expresión Génica , Células Gigantes/química , Células Gigantes/metabolismo , Células HEK293 , Proteína HN/genética , Proteína HN/metabolismo , Células HeLa , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Virus de la Parotiditis/metabolismo , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteolisis , Transducción de Señal , Células Vero , Proteínas Virales de Fusión/metabolismo , Internalización del Virus
7.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295907

RESUMEN

Mumps virus (MuV) caused the most viral meningitis before mass immunization. Unfortunately, MuV has reemerged in the United States in the past several years. MuV is a member of the genus Rubulavirus, in the family Paramyxoviridae, and has a nonsegmented negative-strand RNA genome. The viral RNA-dependent RNA polymerase (vRdRp) of MuV consists of the large protein (L) and the phosphoprotein (P), while the nucleocapsid protein (NP) encapsulates the viral RNA genome. These proteins make up the replication and transcription machinery of MuV. The P protein is phosphorylated by host kinases, and its phosphorylation is important for its function. In this study, we performed a large-scale small interfering RNA (siRNA) screen targeting host kinases that regulated MuV replication. The human kinase ribosomal protein S6 kinase beta-1 (RPS6KB1) was shown to play a role in MuV replication and transcription. We have validated the role of RPS6KB1 in regulating MuV using siRNA knockdown, an inhibitor, and RPS6KB1 knockout cells. We found that MuV grows better in cells lacking RPS6KB1, indicating that it downregulates viral growth. Furthermore, we detected an interaction between the MuV P protein and RPS6KB1, suggesting that RPS6KB1 directly regulates MuV replication and transcription.IMPORTANCE Mumps virus is an important human pathogen. In recent years, MuV has reemerged in the United State, with outbreaks occurring in young adults who have been vaccinated. Our work provides insight into a previously unknown mumps virus-host interaction. RPS6KB1 negatively regulates MuV replication, likely through its interaction with the P protein. Understanding virus-host interactions can lead to novel antiviral drugs and enhanced vaccine production.


Asunto(s)
Genoma Viral , Virus de la Parotiditis/genética , Proteínas de la Nucleocápside/genética , Fosfoproteínas/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Virales/genética , Animales , Chlorocebus aethiops , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Virus de la Parotiditis/metabolismo , Proteínas de la Nucleocápside/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Células Vero , Proteínas Virales/metabolismo , Replicación Viral
8.
Sci Rep ; 10(1): 1589, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005959

RESUMEN

Mumps virus is one of the main cause of respiratory illnesses in humans, especially children. Among the viral surface glycoproteins, the hemagglutinin - neuraminidase, MuV-HN, plays key roles in virus entry into host cells and infectivity, thus representing an ideal target for the design of novel inhibitors. Here we report the detailed analysis of the molecular recognition of host cell surface sialylated glycans by the viral glycoprotein MuV-HN. By a combined use of NMR, docking, molecular modelling and CORCEMA-ST, the structural features of sialoglycans/MuV-HN complexes were revealed. Evidence for a different enzyme activity toward longer and complex substrates compared to unbranched ligands was also examined by an accurate NMR kinetic analysis. Our results provide the basis for the structure-based design of effective drugs against mumps-induced diseases.


Asunto(s)
Hemaglutininas/metabolismo , Virus de la Parotiditis/metabolismo , Neuraminidasa/metabolismo , Polisacáridos/metabolismo , Proteínas Estructurales Virales/metabolismo , Sitios de Unión , Humanos , Cinética , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Conformación Proteica
9.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31619562

RESUMEN

Mumps virus (MuV), an enveloped negative-strand RNA virus belonging to the family Paramyxoviridae, enters the host cell through membrane fusion mediated by two viral envelope proteins, an attachment protein hemagglutinin-neuraminidase (MuV-HN) and a fusion (F) protein. However, how the binding of MuV-HN to glycan receptors triggers membrane fusion is not well understood. The crystal structure of the MuV-HN head domain forms a tetramer (dimer of dimers) like other paramyxovirus attachment proteins. In the structure, a sulfate ion (SO42-) was found at the interface between two dimers, which may be replaced by a hydrogen phosphate ion (HPO42-) under physiological conditions. The anion is captured by the side chain of a positively charged arginine residue at position 139 of one monomer each from both dimers. Substitution of alanine or lysine for arginine at this position compromised the fusion support activity of MuV-HN without affecting its cell surface expression, glycan-receptor binding, and interaction with the F protein. Furthermore, the substitution appeared to affect the tetramer formation of the head domain as revealed by blue native-PAGE analysis. These results, together with our previous similar findings with the measles virus attachment protein head domain, suggest that the dimer-dimer interaction within the tetramer may play an important role in triggering membrane fusion during paramyxovirus entry.IMPORTANCE Despite the use of effective live vaccines, mumps outbreaks still occur worldwide. Mumps virus (MuV) infection typically causes flu-like symptoms and parotid gland swelling but sometimes leads to orchitis, oophoritis, and neurological complications, such as meningitis, encephalitis, and deafness. MuV enters the host cell through membrane fusion mediated by two viral proteins, a receptor-binding attachment protein, and a fusion protein, but its detailed mechanism is not fully understood. In this study, we show that the tetramer (dimer of dimers) formation of the MuV attachment protein head domain is supported by an anion located at the interface between two dimers and that the dimer-dimer interaction plays an important role in triggering the activation of the fusion protein and causing membrane fusion. These results not only further our understanding of MuV entry but provide useful information about a possible target for antiviral drugs.


Asunto(s)
Fusión de Membrana , Virus de la Parotiditis/metabolismo , Multimerización de Proteína , Proteínas Virales de Fusión/metabolismo , Acoplamiento Viral , Internalización del Virus , Sustitución de Aminoácidos , Células HEK293 , Humanos , Virus de la Parotiditis/genética , Mutación Missense , Fosfatos/metabolismo , Dominios Proteicos , Sulfatos/metabolismo , Proteínas Virales de Fusión/genética
10.
Viruses ; 11(11)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683999

RESUMEN

Mumps virus (MuV) is a neurotropic non-segmented, negative-stranded, enveloped RNA virus in the Paramyxovirus family. The 15.4 kb genome encodes seven genes, including the V/P, which encodes, among other proteins, the V protein. The MuV V protein has been shown to target the cellular signal transducer and activator of transcription proteins STAT1 and STAT3 for proteasome-mediated degradation. While MuV V protein targeting of STAT1 is generally accepted as a means of limiting innate antiviral responses, the consequence of V protein targeting of STAT3 is less clear. Further, since the MuV V protein targets both STAT1 and STAT3, specifically investigating viral antagonism of STAT3 targeting is challenging. However, a previous study reported that a single amino acid substitution in the MuV V protein (E95D) inhibits targeting of STAT3, but not STAT1. This provided us with a unique opportunity to examine the specific role of STAT 3 in MuV virulence in an in vivo model. Here, using a clone of a wild type MuV strain expressing the E95D mutant V protein, we present data linking inhibition of STAT3 targeting with the accelerated clearance of the virus and reduced neurovirulence in vivo, suggesting its role in promoting antiviral responses. These data suggest a rational approach to virus attenuation that could be exploited for future vaccine development.


Asunto(s)
Virus de la Parotiditis/patogenicidad , Paperas/virología , Factores de Transcripción STAT/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Virus de la Parotiditis/genética , Virus de la Parotiditis/crecimiento & desarrollo , Virus de la Parotiditis/metabolismo , Mutación , Unión Proteica , Ratas , Proteínas Virales/genética , Virulencia/genética
11.
FASEB J ; 33(11): 12528-12540, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31450968

RESUMEN

Mumps virus (MuV) has high tropism to the testis and may lead to male infertility. Sertoli cells are the major targets of MuV infection. However, the mechanisms by which MuV infection impairs male fertility and Sertoli cell function remain unclear. The present study elucidated the effect of MuV infection on the blood-testis barrier (BTB). The transepithelial electrical resistance of MuV-infected mouse Sertoli cells was monitored, and the expression of major proteins of the BTB was examined. We demonstrated that MuV infection disrupted the BTB by reducing the levels of occludin and zonula occludens 1. Sertoli cells derived from Tlr2-/- and Tnfa-/- mice were analyzed for mediating MuV-induced impairment. TLR2-mediated TNF-α production by Sertoli cells in response to MuV infection impaired BTB integrity. MuV-impaired BTB was not observed in Tlr2-/- and Tnfa-/- Sertoli cells. Moreover, an inhibitor of TNF-α, pomalidomide, prevents the disruption of BTB in response to MuV infection. FITC-labeled biotin tracing assay confirmed that BTB permeability and spermatogenesis were transiently impaired by MuV infection in vivo. These findings suggest that the disruption of the BTB could be one of the mechanisms underlying MuV-impaired male fertility, in which TNF-α could play a critical role.-Wu, H., Jiang, X., Gao, Y., Liu, W., Wang, F., Gong, M., Chen, R., Yu, X., Zhang, W., Gao, B., Song, C., Han, D. Mumps virus infection disrupts blood-testis barrier through the induction of TNF-α in Sertoli cells.


Asunto(s)
Barrera Hematotesticular/metabolismo , Virus de la Parotiditis/metabolismo , Paperas/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Barrera Hematotesticular/patología , Barrera Hematotesticular/virología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/virología , Masculino , Ratones , Ratones Noqueados , Paperas/genética , Paperas/patología , Virus de la Parotiditis/genética , Células de Sertoli/patología , Células de Sertoli/virología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/genética , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
12.
Proc Natl Acad Sci U S A ; 113(41): 11579-11584, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671656

RESUMEN

Mumps virus (MuV) remains an important pathogen worldwide, causing epidemic parotitis, orchitis, meningitis, and encephalitis. Here we show that MuV preferentially uses a trisaccharide containing α2,3-linked sialic acid in unbranched sugar chains as a receptor. Crystal structures of the MuV attachment protein hemagglutinin-neuraminidase (MuV-HN) alone and in complex with the α2,3-sialylated trisaccharide revealed that in addition to the interaction between the MuV-HN active site residues and sialic acid, other residues, including an aromatic residue, stabilize the third sugar of the trisaccharide. The importance of the aromatic residue and the third sugar in the MuV-HN-receptor interaction was confirmed by computational energy calculations, isothermal titration calorimetry studies, and glycan-binding assays. Furthermore, MuV-HN was found to bind more efficiently to unbranched α2,3-sialylated sugar chains compared with branched ones. Importantly, the strategically located aromatic residue is conserved among the HN proteins of sialic acid-using paramyxoviruses, and alanine substitution compromised their ability to support cell-cell fusion. These results suggest that not only the terminal sialic acid but also the adjacent sugar moiety contribute to receptor function for mumps and these paramyxoviruses. The distribution of structurally different sialylated glycans in tissues and organs may explain in part MuV's distinct tropism to glandular tissues and the central nervous system. In the crystal structure, the epitopes for neutralizing antibodies are located around the α-helices of MuV-HN that are not well conserved in amino acid sequences among different genotypes of MuV. This may explain the fact that MuV reinfection sometimes occurs.


Asunto(s)
Virus de la Parotiditis/metabolismo , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Receptores Virales/metabolismo , Trisacáridos/química , Trisacáridos/metabolismo , Animales , Anticuerpos Neutralizantes/química , Fusión Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Cristalografía por Rayos X , Epítopos/química , Células HEK293 , Humanos , Lactosa/química , Lactosa/metabolismo , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Dominios Proteicos , Receptores Virales/química , Termodinámica , Células Vero , Proteínas Virales/química , Proteínas Virales/metabolismo
13.
Eur J Immunol ; 46(4): 919-28, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26763072

RESUMEN

Tumor-associated macrophages (TAMs) are known to regulate tumor response to many anti-cancer therapies, including oncolytic virotherapy. Oncolytic virotherapy employing oncolytic paramyxoviruses, such as attenuated measles (MeV) and mumps (MuV) viruses, has demonstrated therapeutic potential against various malignancies. However, the response of TAMs to oncolytic paramyxoviruses and the consequent effect on virotherapeutic efficacy remains to be characterized. Here, we demonstrate that the presence of human monocyte-derived macrophages (MDMs), irrespective of initial polarization state, enhances the virotherapeutic effect of MeV and MuV on breast cancer cells. Notably, our finding contrasts those of several studies involving other oncolytic viruses, which suggest that TAMs negatively impact virotherapeutic efficacy by impeding virus replication and dissemination. We found that the enhanced virotherapeutic effect in the presence of MDMs was due to slightly delayed proliferation and significantly elevated cell death that was not a result of increased virus replication. Instead, we found that the enhanced virotherapeutic effect involved several macrophage-associated anti-tumor mediators, and was associated with the modulation of MDMs towards an anti-tumor phenotype. Our findings present an alternative view on the role of TAMs in oncolytic virotherapy, and highlight the immunotherapeutic potential of oncolytic paramyxoviruses; possibly contributing towards the overall efficacy of oncolytic virotherapy.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/terapia , Macrófagos/inmunología , Virus del Sarampión/metabolismo , Virus de la Parotiditis/metabolismo , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Microambiente Tumoral , Replicación Viral
14.
J Virol ; 89(21): 11002-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311887

RESUMEN

UNLABELLED: The mumps virus (MuV) genome encodes a phosphoprotein (P) that is important for viral RNA synthesis. P forms the viral RNA-dependent RNA polymerase with the large protein (L). P also interacts with the viral nucleoprotein (NP) and self-associates to form a homotetramer. The P protein consists of three domains, the N-terminal domain (P(N)), the oligomerization domain (P(O)), and the C-terminal domain (P(C)). While P(N) is known to relax the NP-bound RNA genome, the roles of P(O) and P(C) are not clear. In this study, we investigated the roles of P(O) and P(C) in viral RNA synthesis using mutational analysis and a minigenome system. We found that P(N) and P(C) functions can be trans-complemented. However, this complementation requires P(O), indicating that P(O) is essential for P function. Using this trans-complementation system, we found that P forms parallel dimers (P(N) to P(N) and P(C) to P(C)). Furthermore, we found that residues R231, K238, K253, and K260 in P(O) are critical for P's functions. We identified P(C) to be the domain that interacts with L. These results provide structure-function insights into the role of MuV P. IMPORTANCE: MuV, a paramyxovirus, is an important human pathogen. The P protein of MuV is critical for viral RNA synthesis. In this work, we established a novel minigenome system that allows the domains of P to be complemented in trans. Using this system, we confirmed that MuV P forms parallel dimers. An understanding of viral RNA synthesis will allow the design of better vaccines and the development of antivirals.


Asunto(s)
Virus de la Parotiditis/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virales/metabolismo , Clonación Molecular , Análisis Mutacional de ADN , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Luciferasas , N-Acetilglucosaminiltransferasas/metabolismo , Polimerizacion , ARN Viral/biosíntesis
15.
J Virol ; 89(6): 3188-99, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552722

RESUMEN

UNLABELLED: Mumps virus (MuV) infection induces formation of cytoplasmic inclusion bodies (IBs). Growing evidence indicates that IBs are the sites where RNA viruses synthesize their viral RNA. However, in the case of MuV infection, little is known about the viral and cellular compositions and biological functions of the IBs. In this study, pulldown purification and N-terminal amino acid sequencing revealed that stress-inducible heat shock protein 70 (Hsp72) was a binding partner of MuV phosphoprotein (P protein), which was an essential component of the IB formation. Immunofluorescence and immunoblotting analyses revealed that Hsp72 was colocalized with the P protein in the IBs, and its expression was increased during MuV infection. Knockdown of Hsp72 using small interfering RNAs (siRNAs) had little, if any, effect on viral propagation in cultured cells. Knockdown of Hsp72 caused accumulation of ubiquitinated P protein and delayed P protein degradation. These results show that Hsp72 is recruited to IBs and regulates the degradation of MuV P protein through the ubiquitin-proteasome pathway. IMPORTANCE: Formation of cytoplasmic inclusion bodies (IBs) is a common characteristic feature in mononegavirus infections. IBs are considered to be the sites of viral RNA replication and transcription. However, there have been few studies focused on host factors recruited to the IBs and their biological functions. Here, we identified stress-inducible heat shock protein 70 (Hsp72) as the first cellular partner of mumps virus (MuV) phosphoprotein (P protein), which is an essential component of the IBs and is involved in viral RNA replication/transcription. We found that the Hsp72 mobilized to the IBs promoted degradation of the MuV P protein through the ubiquitin-proteasome pathway. Our data provide new insight into the role played by IBs in mononegavirus infection.


Asunto(s)
Proteínas del Choque Térmico HSP72/metabolismo , Virus de la Parotiditis/metabolismo , Paperas/enzimología , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinas/metabolismo , Proteínas Virales/metabolismo , Proteínas del Choque Térmico HSP72/genética , Humanos , Cuerpos de Inclusión Viral/metabolismo , Cuerpos de Inclusión Viral/virología , Paperas/genética , Paperas/virología , Virus de la Parotiditis/genética , Fosfoproteínas/genética , Unión Proteica , Proteolisis , Proteínas Virales/genética
16.
PLoS One ; 8(1): e53881, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23342031

RESUMEN

The capacity of a virus to cross species barriers is determined by the development of bona fide interactions with cellular components of new hosts, and in particular its ability to block IFN-α/ß antiviral signaling. Tioman virus (TioV), a close relative of mumps virus (MuV), has been isolated in giant fruit bats in Southeast Asia. Nipah and Hendra viruses, which are present in the same bat colonies, are highly pathogenic in human. Despite serological evidences of close contacts between TioV and human populations, whether TioV is associated to some human pathology remains undetermined. Here we show that in contrast to the V protein of MuV, the V protein of TioV (TioV-V) hardly interacts with human STAT2, does not degrade STAT1, and cannot block IFN-α/ß signaling in human cells. In contrast, TioV-V properly binds to human STAT3 and MDA5, and thus interferes with IL-6 signaling and IFN-ß promoter induction in human cells. Because STAT2 binding was previously identified as a host restriction factor for some Paramyxoviridae, we established STAT2 sequence from giant fruit bats, and binding to TioV-V was tested. Surprisingly, TioV-V interaction with STAT2 from giant fruit bats is also extremely weak and barely detectable. Altogether, our observations question the capacity of TioV to appropriately control IFN-α/ß signaling in both human and giant fruit bats that are considered as its natural host.


Asunto(s)
Interferón Tipo I/metabolismo , Rubulavirus/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Animales , Quirópteros/virología , ARN Helicasas DEAD-box/metabolismo , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1 , Virus de la Parotiditis/metabolismo , Virus de la Parotiditis/fisiología , ARN Helicasas/metabolismo , Rubulavirus/fisiología , Factor de Transcripción STAT3/metabolismo
17.
Microb Cell Fact ; 11: 132, 2012 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-22992433

RESUMEN

BACKGROUND: Plasmid DNA (pDNA) is a promising molecule for therapeutic applications. pDNA is produced by Escherichia coli in high cell-density cultivations (HCDC) using fed-batch mode. The typical limitations of such cultivations, including metabolic deviations like aerobic acetate production due to the existence of substrate gradients in large-scale bioreactors, remain as serious challenges for fast and effective pDNA production. We have previously demonstrated that the substitution of the phosphotransferase system by the over-expressed galactose permease for glucose uptake in E. coli (strain VH33) allows efficient growth, while strongly decreases acetate production. In the present work, additional genetic modifications were made to VH33 to further improve pDNA production. Several genes were deleted from strain VH33: the recA, deoR, nupG and endA genes were inactivated independently and in combination. The performance of the mutant strains was evaluated in shake flasks for the production of a 6.1 kb plasmid bearing an antigen gene against mumps. The best producer strain was cultivated in lab-scale bioreactors using 100 g/L of glucose to achieve HCDC in batch mode. For comparison, the widely used commercial strain DH5α, carrying the same plasmid, was also cultivated under the same conditions. RESULTS: The various mutations tested had different effects on the specific growth rate, glucose uptake rate, and pDNA yields (YP/X). The triple mutant VH33 Δ (recA deoR nupG) accumulated low amounts of acetate and resulted in the best YP/X (4.22 mg/g), whereas YP/X of strain VH33 only reached 1.16 mg/g. When cultivated at high glucose concentrations, the triple mutant strain produced 186 mg/L of pDNA, 40 g/L of biomass and only 2.2 g/L of acetate. In contrast, DH5α produced only 70 mg/L of pDNA and accumulated 9.5 g/L of acetate. Furthermore, the supercoiled fraction of the pDNA produced by the triple mutant was nearly constant throughout the cultivation. CONCLUSION: The pDNA concentration obtained with the engineered strain VH33 Δ (recA deoR nupG) is, to the best of our knowledge, the highest reported for a batch cultivation, and its supercoiled fraction remained close to 80%. Strain VH33 Δ (recA deoR nupG) and its cultivation using elevated glucose concentrations represent an attractive technology for fast and efficient pDNA production and a valuable alternative to fed-batch cultivations of commercial strains.


Asunto(s)
Escherichia coli/metabolismo , Plásmidos/metabolismo , Antígenos/genética , Antígenos/metabolismo , Biomasa , Reactores Biológicos/microbiología , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Técnicas de Inactivación de Genes , Ingeniería Genética , Glucosa/metabolismo , Proteínas de Transporte de Membrana/genética , Virus de la Parotiditis/metabolismo , Plásmidos/genética , Rec A Recombinasas/genética , Proteínas Represoras/genética , Vacunas de ADN/biosíntesis
18.
Biofizika ; 57(2): 264-6, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-22594283

RESUMEN

Interaction of polystirolsulphonate with polymerization degree of 8 (PSS-8) and polyallylamin PAA (molecular mass 60 kilodaltons) with viruses from bloodline of paramixo- and orthomixoviruses by the example of measles virus, parotitis and flu leads to the decreasing of infective activity. The possible mechanism of viral inhibitive action of these chemical compounds is damaging of interfacial antigenic proteins of paramixo- and orthomixoviruses. In this study it was detected the change of surface tension of bilayer lipid membrane in the presence of PSS-8 and PAA. The change of surface tension leads to disorder in viral proteins adsorption in bilayer lipid membrane. This process could lead to disorder of juncture and self-assembly of virions.


Asunto(s)
Membrana Dobles de Lípidos/química , Poliaminas/química , Poliestirenos/química , Subtipo H2N2 del Virus de la Influenza A/química , Subtipo H2N2 del Virus de la Influenza A/metabolismo , Virus del Sarampión/química , Virus del Sarampión/metabolismo , Virus de la Parotiditis/química , Virus de la Parotiditis/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Inactivación de Virus
19.
J Virol ; 86(3): 1768-76, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22090137

RESUMEN

Mumps virus (MuV) causes an acute infection in humans characterized by a wide array of symptoms ranging from relatively mild manifestations, such as parotitis, to more-severe complications, such as meningitis and encephalitis. Widespread mumps vaccination has reduced mumps incidence dramatically; however, outbreaks still occur in vaccinated populations. The V protein of MuV, when expressed in cell culture, blocks interferon (IFN) expression and signaling and interleukin-6 (IL-6) signaling. In this work, we generated a recombinant MuV incapable of expressing the V protein (rMuVΔV). The rescued MuV was derived from a clinical wild-type isolate from a recent outbreak in the United States (MuV(Iowa/US/06), G genotype). Analysis of the virus confirmed the roles of V protein in blocking IFN expression and signaling and IL-6 signaling. We also found that the rMuV(Iowa/US/06)ΔV virus induced high levels of IL-6 expression in vitro, suggesting that V plays a role in reducing IL-6 expression. In vivo, the rMuV(Iowa/US/06)ΔV virus was highly attenuated, indicating that the V protein plays an essential role in viral virulence.


Asunto(s)
Virus de la Parotiditis/patogenicidad , Proteínas Virales/fisiología , Animales , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Virus de la Parotiditis/metabolismo
20.
Microb Cell Fact ; 10: 37, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21595909

RESUMEN

BACKGROUND: The expression of human virus surface proteins, as well as other mammalian glycoproteins, is much more efficient in cells of higher eukaryotes rather than yeasts. The limitations to high-level expression of active viral surface glycoproteins in yeast are not well understood. To identify possible bottlenecks we performed a detailed study on overexpression of recombinant mumps hemagglutinin-neuraminidase (MuHN) and measles hemagglutinin (MeH) in yeast Saccharomyces cerevisiae, combining the analysis of recombinant proteins with a proteomic approach. RESULTS: Overexpressed recombinant MuHN and MeH proteins were present in large aggregates, were inactive and totally insoluble under native conditions. Moreover, the majority of recombinant protein was found in immature form of non-glycosylated precursors. Fractionation of yeast lysates revealed that the core of viral surface protein aggregates consists of MuHN or MeH disulfide-linked multimers involving eukaryotic translation elongation factor 1A (eEF1A) and is closely associated with small heat shock proteins (sHsps) that can be removed only under denaturing conditions. Complexes of large Hsps seem to be bound to aggregate core peripherally as they can be easily removed at high salt concentrations. Proteomic analysis revealed that the accumulation of unglycosylated viral protein precursors results in specific cytosolic unfolded protein response (UPR-Cyto) in yeast cells, characterized by different action and regulation of small Hsps versus large chaperones of Hsp70, Hsp90 and Hsp110 families. In contrast to most environmental stresses, in the response to synthesis of recombinant MuHN and MeH, only the large Hsps were upregulated whereas sHsps were not. Interestingly, the amount of eEF1A was also increased during this stress response. CONCLUSIONS: Inefficient translocation of MuHN and MeH precursors through ER membrane is a bottleneck for high-level expression in yeast. Overexpression of these recombinant proteins induces the UPR's cytosolic counterpart, the UPR-Cyto, which represent a subset of proteins involved in the heat-shock response. The involvement of eEF1A may explain the mechanism by which only large chaperones, but not small Hsps are upregulated during this stress response. Our study highlights important differences between viral surface protein expression in yeast and mammalian cells at the first stage of secretory pathway.


Asunto(s)
Glicoproteínas de Membrana/biosíntesis , Precursores de Proteínas/biosíntesis , Saccharomyces cerevisiae/metabolismo , Respuesta de Proteína Desplegada , Proteínas Virales/biosíntesis , Factor 1 Eucariótico de Iniciación/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/metabolismo , Morbillivirus/metabolismo , Virus de la Parotiditis/enzimología , Virus de la Parotiditis/metabolismo , Neuraminidasa/biosíntesis , Neuraminidasa/genética , Precursores de Proteínas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/fisiología , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...