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








Base de dados
Intervalo de ano de publicação
1.
mBio ; 13(6): e0169822, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36346228

RESUMO

The lipid composition of the host cell membrane is one of the key determinants of the entry of enveloped viruses into cells. To elucidate the detailed mechanisms behind the cell entry of rubella virus (RuV), one of the enveloped viruses, we searched for host factors involved in such entry by using CRISPR/Cas9 genome-wide knockout screening, and we found sphingomyelin synthase 1 (SMS1), encoded by the SGMS1 gene, as a candidate. RuV growth was strictly suppressed in SGMS1-knockout cells and was completely recovered by the overexpression of enzymatically active SMS1 and partially recovered by that of SMS2, another member of the SMS family, but not by that of enzymatically inactive SMS1. An entry assay using pseudotyped vesicular stomatitis virus possessing RuV envelope proteins revealed that sphingomyelin generated by SMSs is crucial for at least RuV entry. In SGMS1-knockout cells, lipid mixing between the RuV envelope membrane and the membrane of host cells occurred, but entry of the RuV genome from the viral particles into the cytoplasm was strongly inhibited. This indicates that sphingomyelin produced by SMSs is essential for the formation of membrane pores after hemifusion occurs during RuV entry. IMPORTANCE Infection with rubella virus during pregnancy causes congenital rubella syndrome in infants. Despite its importance in public health, the detailed mechanisms of rubella virus cell entry have only recently become somewhat clearer. The E1 protein of rubella virus is classified as a class II fusion protein based on its structural similarity, but it has the unique feature that its activity is dependent on calcium ion binding in the fusion loops. In this study, we found another unique feature, as cellular sphingomyelin plays a critical role in the penetration of the nucleocapsid into the cytoplasm after hemifusion by rubella virus. This provides important insight into the entry mechanism of rubella virus. This study also presents a model of hemifusion arrest during cell entry by an intact virus, providing a useful tool for analyzing membrane fusion, a biologically important phenomenon.


Assuntos
Vírus da Rubéola , Rubéola (Sarampo Alemão) , Gravidez , Feminino , Humanos , Vírus da Rubéola/metabolismo , Esfingomielinas , Internalização do Vírus , Membrana Celular/metabolismo , Proteínas do Envelope Viral/genética , Citoplasma/metabolismo , Vírion/metabolismo , Nucleocapsídeo/metabolismo
2.
Front Microbiol ; 12: 751909, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867872

RESUMO

Many efforts have been dedicated to the discovery of antiviral drug candidates against the mumps virus (MuV); however, no specific drug has yet been approved. The development of efficient screening methods is a key factor for the discovery of antiviral candidates. In this study, we evaluated a screening method using an Aequorea coerulescens green fluorescent protein-expressing MuV infectious molecular clone. The application of this system to screen for active compounds against MuV replication revealed that CD437, a retinoid acid receptor agonist, has anti-MuV activity. The point of antiviral action was a late step(s) in the MuV life cycle. The replication of other paramyxoviruses was also inhibited by CD437. The induction of retinoic acid-inducible gene (RIG)-I expression is a reported mechanism for the antiviral activity of retinoids, but our results indicated that CD437 did not stimulate RIG-I expression. Indeed, we observed antiviral activity despite the absence of RIG-I, suggesting that CD437 antiviral activity does not require RIG-I induction.

3.
Jpn J Infect Dis ; 73(4): 304-307, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32074516

RESUMO

During the emergence of novel coronavirus 2019 (nCoV) outbreak in Wuhan city, China at the end of 2019, there was movement of many airline travelers between Wuhan and Japan, suggesting that the Japanese population was at high risk of infection by the virus. Hence, we urgently developed diagnostic systems for detection of 2019 nCoV. Two nested RT-PCR and two real-time RT-PCR assays were adapted for use in Japan. As of February 8, 2020, these assays have successfully detected 25 positive cases of infection in Japan.


Assuntos
Betacoronavirus/genética , Técnicas de Laboratório Clínico/métodos , Infecções por Coronavirus/diagnóstico , Pneumonia Viral/diagnóstico , RNA Viral/análise , COVID-19 , Teste para COVID-19 , Vacinas contra COVID-19 , Humanos , Japão , Pandemias , Poliproteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/genética , Proteínas Virais/genética
4.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31484751

RESUMO

Two viral nonstructural proteins, p150 and p90, are expressed in rubella virus (RUBV)-infected cells and mediate viral genome replication, presumably using various host machineries. Molecular chaperones are critical host factors for the maintenance of cellular proteostasis, and certain viral proteins use this chaperone system. The RUBV p150 and p90 proteins are generated from a precursor polyprotein, p200, via processing by the protease activity of its p150 region. This processing is essential for RUBV genome replication. Here we show that heat shock protein 90 (HSP90), a molecular chaperone, is an important host factor for RUBV genome replication. The treatment of RUBV-infected cells with the HSP90 inhibitors 17-allylamino-17-desmethoxygeldanamycin (17-AAG) and ganetespib suppressed RUBV genome replication. HSP90α physically interacted with p150, but not p90. Further analyses into the mechanism of action of the HSP90 inhibitors revealed that HSP90 activity contributes to p150 functional integrity and promotes p200 processing. Collectively, our data demonstrate that RUBV p150 is a client of the HSP90 molecular chaperone and that HSP90 functions as a key host factor for RUBV replication.IMPORTANCE Accumulating evidence indicates that RNA viruses use numerous host factors during replication of their genomes. However, the host factors involved in rubella virus (RUBV) genome replication are largely unknown. In this study, we demonstrate that the HSP90 molecular chaperone is needed for the efficient replication of the RUBV genome. Further, we reveal that HSP90 interacts with RUBV nonstructural protein p150 and its precursor polyprotein, p200. HSP90 contributes to the stability of p150 and the processing of p200 via its protease domain in the p150 region. We conclude that the cellular molecular chaperone HSP90 is a key host factor for functional maturation of nonstructural proteins for RUBV genome replication. These findings provide novel insight into this host-virus interaction.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Vírus da Rubéola/metabolismo , Proteínas não Estruturais Virais/metabolismo , Células A549 , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Proteínas de Choque Térmico HSP90/fisiologia , Humanos , Chaperonas Moleculares/metabolismo , Proteólise , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Rubéola (Sarampo Alemão)/virologia , Células Vero , Proteínas não Estruturais Virais/genética , Replicação Viral/genética , Replicação Viral/fisiologia
5.
Proc Natl Acad Sci U S A ; 116(24): 11587-11589, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31138700

RESUMO

Mononegaviruses are promising tools as oncolytic vectors and transgene delivery vectors for gene therapy and regenerative medicine. By using the Magnet proteins, which reversibly heterodimerize upon blue light illumination, photocontrollable mononegaviruses (measles and rabies viruses) were generated. The Magnet proteins were inserted into the flexible domain of viral polymerase, and viruses showed strong replication and oncolytic activities only when the viral polymerases were activated by blue light illumination.


Assuntos
Vírus do Sarampo/genética , Vírus Oncolíticos/genética , Vírus da Raiva/genética , Animais , Linhagem Celular Tumoral , RNA Polimerases Dirigidas por DNA/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Luz , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Viral Oncolítica/métodos , Transgenes/genética , Replicação Viral/genética
6.
PLoS Pathog ; 15(5): e1007749, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31121004

RESUMO

The regulation of paramyxovirus RNA synthesis by host proteins is poorly understood. Here, we identified a novel regulation mechanism of paramyxovirus RNA synthesis by the Hsp90 co-chaperone R2TP complex. We showed that the R2TP complex interacted with the paramyxovirus polymerase L protein and that silencing of the R2TP complex led to uncontrolled upregulation of mumps virus (MuV) gene transcription but not genome replication. Regulation by the R2TP complex was critical for MuV replication and evasion of host innate immune responses. The R2TP complex also regulated measles virus (MeV) RNA synthesis, but its function was inhibitory and not beneficial to MeV, as MeV evaded host innate immune responses in the absence of the R2TP complex. The identification of the R2TP complex as a critical host factor sheds new light on the regulation of paramyxovirus RNA synthesis.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Caxumba/genética , Caxumba/genética , RNA Viral/biossíntese , Proteínas Virais/metabolismo , Replicação Viral , Células A549 , Proteínas de Choque Térmico HSP90/genética , Humanos , Caxumba/virologia , Proteínas Virais/genética
7.
PLoS One ; 14(4): e0215822, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31013314

RESUMO

Human metapneumovirus (HMPV) has been a notable etiological agent of acute respiratory infection in humans, but it was not discovered until 2001, because HMPV replicates only in a limited number of cell lines and the cytopathic effect (CPE) is often mild. To promote the study of HMPV, several groups have generated green fluorescent protein (GFP)-expressing recombinant HMPV strains (HMPVGFP). However, the growing evidence has complicated the understanding of cell line specificity of HMPV, because it seems to vary notably among HMPV strains. In addition, unique A2b clade HMPV strains with a 180-nucleotide duplication in the G gene (HMPV A2b180nt-dup strains) have recently been detected. In this study, we re-evaluated and compared the cell line specificity of clinical isolates of HMPV strains, including the novel HMPV A2b180nt-dup strains, and six recombinant HMPVGFP strains, including the newly generated recombinant HMPV A2b180nt-dup strain, MG0256-EGFP. Our data demonstrate that VeroE6 and LLC-MK2 cells generally showed the highest infectivity with any clinical isolates and recombinant HMPVGFP strains. Other human-derived cell lines (BEAS-2B, A549, HEK293, MNT-1, and HeLa cells) showed certain levels of infectivity with HMPV, but these were significantly lower than those of VeroE6 and LLC-MK2 cells. Also, the infectivity in these suboptimal cell lines varied greatly among HMPV strains. The variations were not directly related to HMPV genotypes, cell lines used for isolation and propagation, specific genome mutations, or nucleotide duplications in the G gene. Thus, these variations in suboptimal cell lines are likely intrinsic to particular HMPV strains.


Assuntos
Linhagem Celular/virologia , Efeito Citopatogênico Viral/genética , Metapneumovirus/crescimento & desenvolvimento , Infecções Respiratórias/virologia , Células A549 , Proteínas de Fluorescência Verde/genética , Células HEK293 , Células HeLa , Humanos , Metapneumovirus/genética , Metapneumovirus/patogenicidade , Infecções Respiratórias/genética , Infecções Respiratórias/prevenção & controle
8.
J Virol ; 91(6)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28053100

RESUMO

Paramyxoviral RNAs are synthesized by a viral RNA-dependent RNA polymerase (RdRp) consisting of the large (L) protein and its cofactor phosphoprotein (P protein). The L protein is a multifunctional protein that catalyzes RNA synthesis, mRNA capping, and mRNA polyadenylation. Growing evidence shows that the stability of several paramyxovirus L proteins is regulated by heat shock protein 90 (Hsp90). In this study, we demonstrated that Hsp90 activity was important for mumps virus (MuV) replication. The Hsp90 activity was required for L-protein stability and activity because an Hsp90-specific inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), destabilized the MuV L protein and suppressed viral RNA synthesis. However, once the L protein formed a mature polymerase complex with the P protein, Hsp90 activity was no longer required for the stability and activity of the L protein. When the Hsp90 activity was inhibited, the MuV L protein was degraded through the CHIP (C terminus of Hsp70-interacting protein)-mediated proteasomal pathway. High concentrations of 17-AAG showed strong cytotoxicity to certain cell types, but combined use of an Hsp70 inhibitor, VER155008, potentiated degradation of the L protein, allowing a sufficient reduction of 17-AAG concentration to block MuV replication with minimum cytotoxicity. Regulation of the L protein by Hsp90 and Hsp70 chaperones was also demonstrated for another paramyxovirus, the measles virus. Collectively, our data show that the Hsp90/Hsp70 chaperone machinery assists in the maturation of the paramyxovirus L protein and thereby in the formation of a mature RdRp complex and efficient viral replication.IMPORTANCE Heat shock protein 90 (Hsp90) is nearly universally required for viral protein homeostasis. Here, we report that Hsp90 activity is required for efficient propagation of mumps virus (MuV). Hsp90 functions in the maintenance of the catalytic subunit of viral polymerase, the large (L) protein, prior to formation of a mature polymerase complex with the polymerase cofactor of L, phosphoprotein. Hsp70 collaborates with Hsp90 to regulate biogenesis of the MuV L protein. The functions of these chaperones on the viral polymerase may be common among paramyxoviruses because the L protein of measles virus is also similarly regulated. Our data provide important insights into the molecular mechanisms of paramyxovirus polymerase maturation as well as a basis for the development of novel antiviral drugs.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Caxumba/fisiologia , RNA Polimerase Dependente de RNA/metabolismo , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/virologia , Humanos , Vírus do Sarampo/fisiologia , Estabilidade Proteica , Proteólise
9.
J Virol ; 89(23): 12026-34, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26378159

RESUMO

UNLABELLED: Mumps virus (MuV) is an airborne virus that causes a systemic infection in patients. In vivo, the epithelium is a major replication site of MuV, and thus, the mode of MuV infection of epithelial cells is a subject of interest. Our data in the present study showed that MuV entered polarized epithelial cells via both the apical and basolateral surfaces, while progeny viruses were predominantly released from the apical surface. In polarized cells, intracellular transport of viral ribonucleoprotein (vRNP) complexes was dependent on Rab11-positive endosomes, and vRNP complexes were transported to the apical membrane. Expression of a dominant negative form of Rab11 (Rab11S25N) reduced the progeny virus release in polarized cells but not in nonpolarized cells. Although in this way these effects were correlated with cell polarity, Rab11S25N did not modulate the direction of virus release from the apical surface. Therefore, our data suggested that Rab11 is not a regulator of selective apical release of MuV, although it acts as an activator of virus release from polarized epithelial cells. In addition, our data and previous studies on Sendai virus, respiratory syncytial virus, and measles virus suggested that selective apical release from epithelial cells is used by many paramyxoviruses, even though they cause either a systemic infection or a local respiratory infection. IMPORTANCE: Mumps virus (MuV) is the etiological agent of mumps and causes a systemic infection. However, the precise mechanism by which MuV breaks through the epithelial barriers and achieves a systemic infection remains unclear. In the present study, we show that the entry of MuV is bipolar, while the release is predominantly from the apical surface in polarized epithelial cells. In addition, the release of progeny virus was facilitated by a Rab11-positive recycling endosome and microtubule network. Our data provide important insights into the mechanism of transmission and pathogenesis of MuV.


Assuntos
Endossomos/virologia , Células Epiteliais/virologia , Vírus da Caxumba/fisiologia , Liberação de Vírus/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Chlorocebus aethiops , Cães , Endossomos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Microscopia de Fluorescência , Plasmídeos/genética
10.
J Virol ; 89(6): 3188-99, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552722

RESUMO

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.


Assuntos
Proteínas de Choque Térmico HSP72/metabolismo , Vírus da Caxumba/metabolismo , Caxumba/enzimologia , Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitinas/metabolismo , Proteínas Virais/metabolismo , Proteínas de Choque Térmico HSP72/genética , Humanos , Corpos de Inclusão Viral/metabolismo , Corpos de Inclusão Viral/virologia , Caxumba/genética , Caxumba/virologia , Vírus da Caxumba/genética , Fosfoproteínas/genética , Ligação Proteica , Proteólise , Proteínas Virais/genética
11.
PLoS One ; 9(4): e94999, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24722368

RESUMO

Inhibitor of κB kinase ε (IKKε) and TANK binding kinase 1 (TBK1), so-called non-canonical IKKs or IKK-related kinases, are involved in the cellular innate immunity by inducing type I IFNs. Two kinases commonly phosphorylate transcription factors IRF3 and IRF7 in type I IFN production pathway. In contrast to TBK1, underlying mechanisms of IKKε activation and regions required for activation of downstream molecules are poorly understood. In this study, we investigated regions of IKKε required for the activation of type I IFN promoter specially, by focusing on the C-terminal region. To show the functional significance of the IKKε C-terminal region on type I IFN production, we employed various mutant forms of IKKε and compared to corresponding region of TBK1. We identified the specific regions and residues of IKKε involved in the activation of downstream signaling. Interestingly, corresponding region and residues are not required for activation of downstream signaling by TBK1. The results highlight the importance of the C-terminal region in the functional activity of IKKε in innate immune response and also the difference in activation mechanisms between IKKε and the closely related TBK1.


Assuntos
Quinase I-kappa B/metabolismo , Interferon Tipo I/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Células HEK293 , Humanos , Quinase I-kappa B/genética , Interferon Tipo I/genética , Células L , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética
12.
J Virol ; 88(10): 5608-16, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24600012

RESUMO

UNLABELLED: Proteolytic cleavage of the hemagglutinin (HA) protein is essential for influenza A virus (IAV) to acquire infectivity. This process is mediated by a host cell protease(s) in vivo. The type II transmembrane serine protease TMPRSS2 is expressed in the respiratory tract and is capable of activating a variety of respiratory viruses, including low-pathogenic (LP) IAVs possessing a single arginine residue at the cleavage site. Here we show that TMPRSS2 plays an essential role in the proteolytic activation of LP IAVs, including a recently emerged H7N9 subtype, in vivo. We generated TMPRSS2 knockout (KO) mice. The TMPRSS2 KO mice showed normal reproduction, development, and growth phenotypes. In TMPRSS2 KO mice infected with LP IAVs, cleavage of HA was severely impaired, and consequently, the majority of LP IAV progeny particles failed to gain infectivity, while the viruses were fully activated proteolytically in TMPRSS2+/+ wild-type (WT) mice. Accordingly, in contrast to WT mice, TMPRSS2 KO mice were highly tolerant of challenge infection by LP IAVs (H1N1, H3N2, and H7N9) with ≥1,000 50% lethal doses (LD50) for WT mice. On the other hand, a high-pathogenic H5N1 subtype IAV possessing a multibasic cleavage site was successfully activated in the lungs of TMPRSS2 KO mice and killed these mice, as observed for WT mice. Our results demonstrate that recently emerged H7N9 as well as seasonal IAVs mainly use the specific protease TMPRSS2 for HA cleavage in vivo and, thus, that TMPRSS2 expression is essential for IAV replication in vivo. IMPORTANCE: Influenza A virus (IAV) is a leading pathogen that infects and kills many humans every year. We clarified that the infectivity and pathogenicity of IAVs, including a recently emerged H7N9 subtype, are determined primarily by a host protease, TMPRSS2. Our data showed that TMPRSS2 is the key host protease that activates IAVs in vivo through proteolytic cleavage of their HA proteins. Hence, TMPRSS2 is a good target for the development of anti-IAV drugs. Such drugs could also be effective for many other respiratory viruses, including the recently emerged Middle East respiratory syndrome (MERS) coronavirus, because they are also activated by TMPRSS2 in vitro. Consequently, the present paper could have a large impact on the battle against respiratory virus infections and contribute greatly to human health.


Assuntos
Interações Hospedeiro-Patógeno , Subtipo H7N9 do Vírus da Influenza A/fisiologia , Serina Endopeptidases/metabolismo , Replicação Viral , Animais , Modelos Animais de Doenças , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Dose Letal Mediana , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Serina Endopeptidases/deficiência , Análise de Sobrevida
13.
PLoS One ; 8(12): e82343, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24358174

RESUMO

Canine distemper virus (CDV) becomes able to use human receptors through a single amino acid substitution in the H protein. In addition, CDV strains possessing an intact C protein replicate well in human epithelial H358 cells. The present study showed that CDV strain 007Lm, which was isolated from lymph node tissue of a dog with distemper, failed to replicate in H358 cells, although it possessed an intact C protein. Sequence analyses suggested that a cysteine-to-tyrosine substitution at position 267 of the V protein caused this growth defect. Analyses using H358 cells constitutively expressing the CDV V protein showed that the V protein with a cysteine, but not that with a tyrosine, at this position effectively blocked the interferon-stimulated signal transduction pathway, and supported virus replication of 007Lm in H358 cells. Thus, the V protein as well as the C protein appears to be functional and essential for CDV replication in human epithelial cells.


Assuntos
Vírus da Cinomose Canina/metabolismo , Cinomose/virologia , Células Epiteliais/virologia , Replicação Viral/fisiologia , Animais , Linhagem Celular , Cães , Células Epiteliais/metabolismo , Humanos
14.
J Virol ; 87(8): 4683-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23408617

RESUMO

Many viruses use the host trafficking system at a variety of their replication steps. Measles virus (MV) possesses a nonsegmented negative-strand RNA genome that encodes three components of the ribonucleoprotein (RNP) complex (N, P, and L), two surface glycoproteins, a matrix protein, and two nonstructural proteins. A subset of immune cells and polarized epithelial cells are in vivo targets of MV, and MV is selectively released from the apical membrane of polarized epithelial cells. However, the molecular mechanisms for the apical release of MV remain largely unknown. In the present study, the localization and trafficking mechanisms of the RNP complex of MV were analyzed in detail using recombinant MVs expressing fluorescent protein-tagged L proteins. Live cell imaging analyses demonstrated that the MV RNP complex was transported in a manner dependent on the microtubule network and together with Rab11A-containing recycling endosomes. The RNP complex was accumulated at the apical membrane and the apical recycling compartment. The accumulation and shedding of infectious virions were severely impaired by expression of a dominant negative form of Rab11A. On the other hand, recycling endosome-mediated RNP transport was totally dispensable for virus production in nonpolarized cells. These data provide the first demonstration of the regulated intracellular trafficking events of the MV RNP complex that define the directional viral release from polarized epithelial cells.


Assuntos
Endossomos/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus do Sarampo/fisiologia , Ribonucleoproteínas/metabolismo , Liberação de Vírus , Animais , Fusão Gênica Artificial , Transporte Biológico , Linhagem Celular , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , RNA Viral/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Virais/metabolismo
15.
Virology ; 435(2): 485-92, 2013 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-23174504

RESUMO

Recent outbreaks in monkeys have proven that canine distemper virus (CDV) causes diseases in a wide range of mammals. CDV uses SLAM and nectin4 as receptors to replicate in susceptible animals. Here, we show that human nectin4, but not human SLAM, is fully functional as a CDV receptor. The CDV Ac96I strain hardly replicated in nectin4-expressing human epithelial NCI-H358 cells, but readily adapted to grow in them. Unsurprisingly, no amino acid change in the H protein was required for the adaptation. The original Ac96I strain possessed a truncated C protein, and a subpopulation possessing the intact C protein was selected after growth in NCI-H358 cells. Other CDV strains possessing the intact C protein showed significantly higher growth abilities in NCI-H358 cells than the Ac96I strain with the truncated C protein. These findings suggest that the C protein is functional in human epithelial cells and critical for CDV replication in them.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus da Cinomose Canina/fisiologia , Células Epiteliais/virologia , Receptores Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Moléculas de Adesão Celular/genética , Chlorocebus aethiops , Cinomose/virologia , Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/metabolismo , Cães , Humanos , Dados de Sequência Molecular , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Virais/genética , Análise de Sequência de DNA , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero
16.
J Virol ; 85(22): 11871-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917959

RESUMO

Subacute sclerosing panencephalitis (SSPE) is a fatal sequela associated with measles and is caused by persistent infection of the brain with measles virus (MV). The SI strain was isolated in 1976 from a patient with SSPE and shows neurovirulence in animals. Genome nucleotide sequence analyses showed that the SI strain genome possesses typical genome alterations for SSPE-derived strains, namely, accumulated amino acid substitutions in the M protein and cytoplasmic tail truncation of the F protein. Through the establishment of an efficient reverse genetics system, a recombinant SI strain expressing a green fluorescent protein (rSI-AcGFP) was generated. The infection of various cell types with rSI-AcGFP was evaluated by fluorescence microscopy. rSI-AcGFP exhibited limited syncytium-forming activity and spread poorly in cells. Analyses using a recombinant MV possessing a chimeric genome between those of the SI strain and a wild-type MV strain indicated that the membrane-associated protein genes (M, F, and H) were responsible for the altered growth phenotype of the SI strain. Functional analyses of viral glycoproteins showed that the F protein of the SI strain exhibited reduced fusion activity because of an E300G substitution and that the H protein of the SI strain used CD46 efficiently but used the original MV receptors on immune and epithelial cells poorly because of L482F, S546G, and F555L substitutions. The data obtained in the present study provide a new platform for analyses of SSPE-derived strains as well as a clear example of an SSPE-derived strain that exhibits altered receptor specificity and limited fusion activity.


Assuntos
Genoma Viral , Vírus do Sarampo/isolamento & purificação , Vírus do Sarampo/patogenicidade , Panencefalite Esclerosante Subaguda/virologia , Internalização do Vírus , Substituição de Aminoácidos , Análise Mutacional de DNA , Humanos , Vírus do Sarampo/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto , RNA Viral/genética , Análise de Sequência de DNA , Proteínas Virais/genética , Tropismo Viral , Virulência
17.
J Virol ; 84(1): 372-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846522

RESUMO

Measles virus (MV), a member of the family Paramyxoviridae, is a nonsegmented negative-strand RNA virus. The RNA helicases retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are differentially involved in the detection of cytoplasmic viral RNAs and induction of alpha/beta interferon (IFN-alpha/beta). RIG-I is generally believed to play a major role in the recognition of paramyxoviruses, whereas many viruses of this family produce V proteins that can inhibit MDA5. To determine the individual roles of MDA5 and RIG-I in IFN induction after MV infection, small interfering RNA-mediated knockdown of MDA5 or RIG-I was performed in the human epithelial cell line H358, which is susceptible to wild-type MV isolates. The production of IFN-beta mRNA in response to MV infection was greatly reduced in RIG-I knockdown clones compared to that in H358 cells, confirming the importance of RIG-I in the detection of MV. The IFN-beta mRNA levels were also moderately reduced in MDA5 knockdown clones, even though these clones retained fully functional RIG-I. A V protein-deficient recombinant MV (MVDeltaV) induced higher amounts of IFN-beta mRNA at the early stage of infection in H358 cells compared to the parental virus. The reductions in the IFN-beta mRNA levels in RIG-I knockdown clones were less pronounced after infection with MVDeltaV than after infection with the parental virus. Taken together, the present results indicate that RIG-I and MDA5 both contribute to the recognition of MV and that the V protein promotes MV growth at least partly by inhibiting the MDA5-mediated IFN responses.


Assuntos
RNA Helicases DEAD-box/fisiologia , Interferon Tipo I/genética , Vírus do Sarampo/imunologia , Ativação Transcricional , Linhagem Celular , Proteína DEAD-box 58 , Células Epiteliais/virologia , Humanos , Helicase IFIH1 Induzida por Interferon , Interferon-alfa/genética , Interferon beta/genética , RNA Mensageiro/análise , Receptores Imunológicos , Proteínas Virais/fisiologia
18.
Viral Immunol ; 22(6): 389-95, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19951175

RESUMO

In bovine Mx1, only an amino acid substitution between Ile and Met at position 120 was detected by the nucleotide sequence and mismatched PCR-RFLP technique. The Ile variant was assumed to distribute mainly in the bovine population since the gene frequency was 79.3%. Furthermore, we cloned water buffalo Mx1 cDNA, which showed 51 nucleotide and 20 amino acid substitutions in comparison with that of the cow. Another kind of Mx1 cDNA, bovine Mx1B cDNA, was found and it was deduced to cause 27 amino acid substitutions at the N-terminus compared to the original Mx1 by alternative splicing. However, no variation was detected in 27 amino acids specific for Mx1B among 29 cows and a water buffalo. We established four kinds of mRNA-expressing 3T3 cells and Vero cells. When infection experiments were performed using recombinant vesicular stomatitis virus (VSVDeltaG*-G), bovine Ile and Met types and water buffalo Mx1 mRNA-expressing cell lines showed equally positive antiviral activities (p < 0.05). On the other hand, bovine Mx1B mRNA-expressing cell lines did not have activity against VSVDeltaG*-G. Intracellular localization of bovine Mx1 and Mx1B proteins was examined by a transiently GFP-fused expression system in 3T3 cells. Bovine Mx1 was localized in the cytoplasm, while bovine Mx1B was mainly localized in the nucleus. An arginine-rich nuclear localization signal was found in 27 amino acids specific for Mx1B. N-terminus-deleted Mx1B was only localized in the cytoplasm, and the deleted Mx1B-expressing cell lines showed significantly positive antiviral activities (p < 0.05) against VSVDeltaG*-G.


Assuntos
Búfalos/genética , Bovinos/genética , Proteínas de Ligação ao GTP/genética , Vesiculovirus/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células 3T3 BALB , Núcleo Celular/química , Citoplasma/química , DNA Complementar/genética , Feminino , Proteínas de Ligação ao GTP/análise , Proteínas de Ligação ao GTP/fisiologia , Camundongos , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus , Isoformas de Proteínas/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Transfecção
19.
J Virol ; 83(22): 11996-2001, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726523

RESUMO

The P, V, and C proteins of measles virus are encoded in overlapping reading frames of the P gene, which makes it difficult to analyze the functions of the individual proteins in the context of virus infection. We established a system to analyze the C protein independently from the P and V proteins by placing its gene in an additional transcription unit between the H and L genes. Analyses with this system indicated that a highly attenuated Edmonston lineage vaccine strain encodes a fully functional C protein, and the P and/or V protein is involved in the attenuated phenotype.


Assuntos
Vacina contra Sarampo/genética , Vacinas Atenuadas/genética , Proteínas não Estruturais Virais/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Regulação Viral da Expressão Gênica/genética , Genes Virais/genética , Humanos , Vírus do Sarampo/genética , Fenótipo , Células Vero , Proteínas não Estruturais Virais/fisiologia , Replicação Viral/fisiologia
20.
J Virol ; 83(20): 10374-83, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656884

RESUMO

The genome of measles virus (MV) is encapsidated by the nucleocapsid (N) protein and associates with RNA-dependent RNA polymerase to form the ribonucleoprotein complex. The matrix (M) protein is believed to play an important role in MV assembly by linking the ribonucleoprotein complex with envelope glycoproteins. Analyses using a yeast two-hybrid system and coimmunoprecipitation in mammalian cells revealed that the M protein interacts with the N protein and that two leucine residues at the carboxyl terminus of the N protein (L523 and L524) are critical for the interaction. In MV minigenome reporter gene assays, the M protein inhibited viral RNA synthesis only when it was able to interact with the N protein. The N protein colocalized with the M protein at the plasma membrane when the proteins were coexpressed in plasmid-transfected or MV-infected cells. In contrast, the N protein formed small dots in the perinuclear area when it was expressed without the M protein, or it was incapable of interacting with the M protein. Furthermore, a recombinant MV possessing a mutant N protein incapable of interacting with the M protein grew much less efficiently than the parental virus. Since the M protein has an intrinsic ability to associate with the plasma membrane, it may retain the ribonucleoprotein complex at the plasma membrane by binding to the N protein, thereby stopping viral RNA synthesis and promoting viral particle production. Consequently, our results indicate that the M protein regulates MV RNA synthesis and assembly via its interaction with the N protein.


Assuntos
Regulação Viral da Expressão Gênica , Vírus do Sarampo/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , RNA Viral/metabolismo , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Células HeLa , Humanos , Vírus do Sarampo/genética , Proteínas do Nucleocapsídeo/genética , RNA Viral/genética , Técnicas do Sistema de Duplo-Híbrido , Células Vero , Proteínas da Matriz Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA