Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(8): e1009838, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34370796

RESUMO

Through evolution, Hepatitis B Virus (HBV) developed highly intricate mechanisms exploiting host resources for its multiplication within a constrained genetic coding capacity. Yet a clear picture of viral hitchhiking of cellular processes with spatial resolution is still largely unsolved. Here, by leveraging bDNA-based fluorescence in situ hybridization (FISH) combined with immunofluorescence, we developed a microscopic approach for multiplex detection of viral nucleic acids and proteins, which enabled us to probe some of the key aspects of HBV life cycle. We confirmed the slow kinetics and revealed the high variability of viral replication at single-cell level. We directly visualized HBV minichromosome in contact with acetylated histone 3 and RNA polymerase II and observed HBV-induced degradation of Smc5/6 complex only in primary hepatocytes. We quantified the frequency of HBV pregenomic RNAs occupied by translating ribosome or capsids. Statistics at molecular level suggested a rapid translation phase followed by a slow encapsidation and maturation phase. Finally, the roles of microtubules (MTs) on nucleocapsid assembly and virion morphogenesis were analyzed. Disruption of MTs resulted in the perinuclear retention of nucleocapsid. Meanwhile, large multivesicular body (MVB) formation was significantly disturbed as evidenced by the increase in number and decrease in volume of CD63+ vesicles, thus inhibiting mature virion secretion. In conclusion, these data provided spatially resolved molecular snapshots in the context of specific subcellular activities. The heterogeneity observed at single-cell level afforded valuable molecular insights which are otherwise unavailable from bulk measurements.


Assuntos
DNA Viral/análise , Vírus da Hepatite B/fisiologia , Hepatite B/virologia , RNA Viral/análise , Frações Subcelulares/virologia , Montagem de Vírus , Replicação Viral , Células Hep G2 , Humanos , Análise Espaço-Temporal , Vírion
2.
Sci Rep ; 9(1): 707, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679542

RESUMO

Hantavirus assembly and budding are governed by the surface glycoproteins Gn and Gc. In this study, we investigated the glycoproteins of Puumala, the most abundant Hantavirus species in Europe, using fluorescently labeled wild-type constructs and cytoplasmic tail (CT) mutants. We analyzed their intracellular distribution, co-localization and oligomerization, applying comprehensive live, single-cell fluorescence techniques, including confocal microscopy, imaging flow cytometry, anisotropy imaging and Number&Brightness analysis. We demonstrate that Gc is significantly enriched in the Golgi apparatus in absence of other viral components, while Gn is mainly restricted to the endoplasmic reticulum (ER). Importantly, upon co-expression both glycoproteins were found in the Golgi apparatus. Furthermore, we show that an intact CT of Gc is necessary for efficient Golgi localization, while the CT of Gn influences protein stability. Finally, we found that Gn assembles into higher-order homo-oligomers, mainly dimers and tetramers, in the ER while Gc was present as mixture of monomers and dimers within the Golgi apparatus. Our findings suggest that PUUV Gc is the driving factor of the targeting of Gc and Gn to the Golgi region, while Gn possesses a significantly stronger self-association potential.


Assuntos
Glicoproteínas/metabolismo , Infecções por Hantavirus/metabolismo , Febre Hemorrágica com Síndrome Renal/metabolismo , Multimerização Proteica , Virus Puumala/fisiologia , Frações Subcelulares/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Membrana Celular/virologia , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Glicoproteínas/química , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Células HEK293 , Infecções por Hantavirus/virologia , Febre Hemorrágica com Síndrome Renal/virologia , Humanos , Proteínas Luminescentes/metabolismo , Frações Subcelulares/virologia , Células Vero , Proteínas do Envelope Viral/química
3.
J Virol ; 91(21)2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28835496

RESUMO

The K15P membrane protein of Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with multiple cellular signaling pathways and is thought to play key roles in KSHV-associated endothelial cell angiogenesis, regulation of B-cell receptor (BCR) signaling, and the survival, activation, and proliferation of BCR-negative primary effusion lymphoma (PEL) cells. Although full-length K15P is ∼45 kDa, numerous lower-molecular-weight forms of the protein exist as a result of differential splicing and poorly characterized posttranslational processing. K15P has been reported to localize to numerous subcellular organelles in heterologous expression studies, but there are limited data concerning the sorting of K15P in KSHV-infected cells. The relationships between the various molecular weight forms of K15P, their subcellular distribution, and how these may differ in latent and lytic KSHV infections are poorly understood. Here we report that a cDNA encoding a full-length, ∼45-kDa K15P reporter protein is expressed as an ∼23- to 24-kDa species that colocalizes with the trans-Golgi network (TGN) marker TGN46 in KSHV-infected PEL cells. Following lytic reactivation by sodium butyrate, the levels of the ∼23- to 24-kDa protein diminish, and the full-length, ∼45-kDa K15P protein accumulates. This is accompanied by apparent fragmentation of the TGN and redistribution of K15P to a dispersed peripheral location. Similar results were seen when lytic reactivation was stimulated by the KSHV protein replication and transcription activator (RTA) and during spontaneous reactivation. We speculate that expression of different molecular weight forms of K15P in distinct cellular locations reflects the alternative demands placed upon the protein in the latent and lytic phases.IMPORTANCE The K15P protein of Kaposi's sarcoma-associated herpesvirus (KSHV) is thought to play key roles in disease, including KSHV-associated angiogenesis and the survival and growth of primary effusion lymphoma (PEL) cells. The protein exists in multiple molecular weight forms, and its intracellular trafficking is poorly understood. Here we demonstrate that the molecular weight form of a reporter K15P molecule and its intracellular distribution change when KSHV switches from its latent (quiescent) phase to the lytic, infectious state. We speculate that expression of different molecular weight forms of K15P in distinct cellular locations reflects the alternative demands placed upon the protein in the viral latent and lytic stages.


Assuntos
Regulação Viral da Expressão Gênica , Linfoma de Efusão Primária/metabolismo , Sarcoma de Kaposi/metabolismo , Frações Subcelulares/metabolismo , Proteínas Virais/metabolismo , Ativação Viral/fisiologia , Latência Viral/fisiologia , Membrana Celular/metabolismo , Membrana Celular/virologia , Herpesvirus Humano 8/fisiologia , Humanos , Linfoma de Efusão Primária/virologia , Mitocôndrias/metabolismo , Mitocôndrias/virologia , Sarcoma de Kaposi/virologia , Frações Subcelulares/virologia , Rede trans-Golgi/metabolismo , Rede trans-Golgi/virologia
4.
Proc Natl Acad Sci U S A ; 109(47): 19327-32, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23134731

RESUMO

Marine viruses are major evolutionary and biogeochemical drivers in marine microbial foodwebs. However, an in-depth understanding of the cellular mechanisms and the signal transduction pathways mediating host-virus interactions during natural bloom dynamics has remained elusive. We used field-based mesocosms to examine the "arms race" between natural populations of the coccolithophore Emiliania huxleyi and its double-stranded DNA-containing coccolithoviruses (EhVs). Specifically, we examined the dynamics of EhV infection and its regulation of cell fate over the course of bloom development and demise using a diverse suite of molecular tools and in situ fluorescent staining to target different levels of subcellular resolution. We demonstrate the concomitant induction of reactive oxygen species, caspase-specific activity, metacaspase expression, and programmed cell death in response to the accumulation of virus-derived glycosphingolipids upon infection of natural E. huxleyi populations. These subcellular responses to viral infection simultaneously resulted in the enhanced production of transparent exopolymer particles, which can facilitate aggregation and stimulate carbon flux. Our results not only corroborate the critical role for glycosphingolipids and programmed cell death in regulating E. huxleyi-EhV interactions, but also elucidate promising molecular biomarkers and lipid-based proxies for phytoplankton host-virus interactions in natural systems.


Assuntos
Linhagem da Célula , Haptófitas/citologia , Haptófitas/virologia , Interações Hospedeiro-Patógeno/fisiologia , Phycodnaviridae/fisiologia , Biopolímeros/biossíntese , Caspases/metabolismo , Ativação Enzimática , Eutrofização , Haptófitas/enzimologia , Noruega , Frações Subcelulares/virologia , Fatores de Tempo
5.
Virology ; 432(2): 495-504, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22832119

RESUMO

A comparative analysis of the intracellular distribution of avian reovirus (ARV) core protein sigmaA in cells of avian and mammalian origin revealed that, whereas the viral protein accumulates in the cytoplasm and nucleolus of avian cells, most sigmaA concentrates in the nucleoplasm of mammalian cells in tight association with the insoluble nuclear matrix fraction. Our results further showed that sigmaA becomes arrested in the nucleoplasm of mammalian cells via association with mammalian cell-specific factors and that this association prevents nucleolar targeting. Inhibition of RNA polymerase II activity, but not of RNA polymerase I activity, in infected mammalian cells induces nucleus-to-cytoplasm sigmaA translocation through a CRM1- and RanGTP-dependent mechanism, yet a heterokaryon assay suggests that sigmaA does not shuttle between the nucleus and cytoplasm. The scarcity of sigmaA in cytoplasmic viral factories of infected mammalian cells could be one of the factors contributing to limited ARV replication in mammalian cells.


Assuntos
Orthoreovirus Aviário/metabolismo , Proteínas de Ligação a RNA/metabolismo , Frações Subcelulares/metabolismo , Proteínas do Core Viral/metabolismo , Animais , Aves/virologia , Linhagem Celular , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Embrião de Galinha/virologia , Chlorocebus aethiops , Citoplasma/metabolismo , Fibroblastos/virologia , Células HeLa/virologia , Humanos , Mamíferos/virologia , Especificidade da Espécie , Frações Subcelulares/virologia , Células Vero/virologia
6.
J Vis Exp ; (64): e4028, 2012 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-22688655

RESUMO

Like all negative-strand RNA viruses, the genome of influenza viruses is packaged in the form of viral ribonucleoprotein complexes (vRNP), in which the single-stranded genome is encapsidated by the nucleoprotein (NP), and associated with the trimeric polymerase complex consisting of the PA, PB1, and PB2 subunits. However, in contrast to most RNA viruses, influenza viruses perform viral RNA synthesis in the nuclei of infected cells. Interestingly, viral mRNA synthesis uses cellular pre-mRNAs as primers, and it has been proposed that this process takes place on chromatin. Interactions between the viral polymerase and the host RNA polymerase II, as well as between NP and host nucleosomes have also been characterized. Recently, the generation of recombinant influenza viruses encoding a One-Strep-Tag genetically fused to the C-terminus of the PB2 subunit of the viral polymerase (rWSN-PB2-Strep) has been described. These recombinant viruses allow the purification of PB2-containing complexes, including vRNPs, from infected cells. To obtain purified vRNPs, cell cultures are infected, and vRNPs are affinity purified from lysates derived from these cells. However, the lysis procedures used to date have been based on one-step detergent lysis, which, despite the presence of a general nuclease, often extract chromatin-bound material only inefficiently. Our preliminary work suggested that a large portion of nuclear vRNPs were not extracted during traditional cell lysis, and therefore could not be affinity purified. To increase this extraction efficiency, and to separate chromatin-bound from non-chromatin-bound nuclear vRNPs, we adapted a step-wise subcellular extraction protocol to influenza virus-infected cells. Briefly, this procedure first separates the nuclei from the cell and then extracts soluble nuclear proteins (here termed the "nucleoplasmic" fraction). The remaining insoluble nuclear material is then digested with Benzonase, an unspecific DNA/RNA nuclease, followed by two salt extraction steps: first using 150 mM NaCl (termed "ch150"), then 500 mM NaCl ("ch500") (Fig. 1). These salt extraction steps were chosen based on our observation that 500 mM NaCl was sufficient to solubilize over 85% of nuclear vRNPs yet still allow binding of tagged vRNPs to the affinity matrix. After subcellular fractionation of infected cells, it is possible to affinity purify PB2-tagged vRNPs from each individual fraction and analyze their protein and RNA components using Western Blot and primer extension, respectively. Recently, we utilized this method to discover that vRNP export complexes form during late points after infection on the chromatin fraction extracted with 500 mM NaCl (ch500).


Assuntos
Western Blotting/métodos , Cromatina/química , Cromatina/virologia , Orthomyxoviridae/química , Ribonucleoproteínas/isolamento & purificação , Proteínas Virais/isolamento & purificação , Células HeLa , Humanos , Influenza Humana/virologia , RNA Viral/química , Ribonucleoproteínas/química , Coloração pela Prata/métodos , Frações Subcelulares/química , Frações Subcelulares/virologia , Proteínas Virais/química
7.
Virology ; 430(2): 81-9, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22608534

RESUMO

The in vivo subcellular localization of Mal de Río Cuarto virus (MRCV, Fijivirus, Reoviridae) non-structural proteins fused to GFP was analyzed by confocal microscopy. P5-1 showed a cytoplasmic vesicular-like distribution that was lost upon deleting its PDZ binding TKF motif, suggesting that P5-1 interacts with cellular PDZ proteins. P5-2 located at the nucleus and its nuclear import was affected by the deletion of its basic C-termini. P7-1 and P7-2 also entered the nucleus and therefore, along with P5-2, could function as regulators of host gene expression. P6 located in the cytoplasm and in perinuclear cloud-like inclusions, was driven to P9-1 viroplasm-like structures and co-localized with P7-2, P10 and α-tubulin, suggesting its involvement in viroplasm formation and viral intracellular movement. Finally, P9-2 was N-glycosylated and located at the plasma membrane in association with filopodia-like protrusions containing actin, suggesting a possible role in virus cell-to-cell movement and spread.


Assuntos
Reoviridae , Spodoptera/virologia , Proteínas não Estruturais Virais/análise , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Membrana Celular/química , Membrana Celular/virologia , Núcleo Celular/química , Núcleo Celular/virologia , Citoplasma/química , Citoplasma/virologia , Citoesqueleto/virologia , Genoma Viral , Proteínas de Fluorescência Verde/genética , Microscopia Confocal , Proteínas Recombinantes de Fusão/análise , Reoviridae/genética , Reoviridae/fisiologia , Spodoptera/ultraestrutura , Frações Subcelulares/química , Frações Subcelulares/virologia , Proteínas não Estruturais Virais/genética
8.
Plant Biotechnol J ; 10(4): 410-21, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22260326

RESUMO

Human papillomavirus 8 (HPV-8), one of the high-risk cutaneous papillomaviruses (cHPVs), is associated with epidermodysplasia verruciformis and nonmelanoma skin cancer in immuno-compromised individuals. Currently, no vaccines against cHPVs have been reported; however, recent studies on cross-neutralizing properties of their capsid proteins (CP) have fostered an interest in vaccine production against these viruses. We examined the potential of producing HPV-8 major CP L1 in Nicotiana benthamiana by agroinfiltration of different transient expression vectors: (i) the binary vector pBIN19 with or without silencing suppressor constructs, (ii) the nonreplicating Cowpea mosaic virus-derived expression vector pEAQ-HT and (iii) a replicating Tobacco mosaic virus (TMV)-based vector alone or with signal peptides. Although HPV-8 L1 was successfully expressed using pEAQ-HT and TMV, a 15-fold increase was obtained with pEAQ-HT. In contrast, no L1 protein could be immune detected using pBIN19 irrespective of whether silencing suppressors were coexpressed, although such constructs were required for identifying L1-specific transcripts. A fourfold yield increase in L1 expression was obtained when 22 C-terminal amino acids were deleted (L1ΔC22), possibly eliminating a nuclear localization signal. Electron microscopy showed that plant-made HPV-8 L1 proteins assembled in appropriate virus-like particles (VLPs) of T = 1 or T = 7 symmetry. Ultrathin sections of L1ΔC22-expressing cells revealed their accumulation in the cytoplasm in the form of VLPs or paracrystalline arrays. These results show for the first time the production and localization of HPV-8 L1 protein in planta and its assembly into VLPs representing promising candidate for potential vaccine production.


Assuntos
Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/isolamento & purificação , Expressão Gênica , Técnicas Genéticas , Nicotiana/metabolismo , Proteínas Oncogênicas Virais/biossíntese , Proteínas Oncogênicas Virais/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Northern Blotting , Proteínas do Capsídeo/ultraestrutura , DNA Bacteriano/genética , Vetores Genéticos/genética , Humanos , Immunoblotting , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Proteínas Mutantes/metabolismo , Proteínas Oncogênicas Virais/ultraestrutura , Exsudatos de Plantas/metabolismo , Proteínas Recombinantes/ultraestrutura , Frações Subcelulares/virologia , Vírion/metabolismo , Vírion/ultraestrutura
9.
Infect Disord Drug Targets ; 12(1): 38-58, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22034934

RESUMO

Vesicular stomatitis virus (VSV) and Rabies Virus (RABV) are the prototypic members of the rhabdovirus family. These viruses have a particularly broad host range, and despite the availability of vaccines, RABV still causes more than 50,000 human deaths a year. Trafficking of the virion or viral particles is important at several stages of the replicative life cycle, including cellular entry, localization into the cytoplasmic inclusion bodies which primarily house the transcription and replication of the viral genome, and migration to the plasma membrane from whence the virus is released by budding. Intriguingly, specific viral proteins, VSV M and RABV P have also been shown to undergo intracellular trafficking independent of the other viral apparatus. These proteins are multifunctional, and play roles in antagonism of host processes, namely the IFN system, and as such enable viral evasion of the innate cellular antiviral response. A body of recent research has been aimed at characterizing the mechanisms by which these proteins are able to shuttle between and localize to various subcellular sites, including the nucleus, which is not required during the cytoplasmic replicative life cycle of the virus. This work has indicated that trafficking of these proteins plays a significant role in determining the ability of the viruses to replicate and cause infection, and as such, represents a viable target for development of a new generation of vaccines and prophylactic therapeutics which are required to battle these pathogens of human and agricultural significance.


Assuntos
Vírus da Raiva/imunologia , Raiva/imunologia , Frações Subcelulares/imunologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Humanos , Evasão da Resposta Imune , Raiva/virologia , Frações Subcelulares/virologia , Estomatite Vesicular/virologia , Proteínas Virais/imunologia
10.
PLoS One ; 6(3): e17818, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21412439

RESUMO

Each year, during winter months, human Metapneumovirus (hMPV) is associated with epidemics of bronchiolitis resulting in the hospitalization of many infants. Bronchiolitis is an acute illness of the lower respiratory tract with a consequent inflammation of the bronchioles. The rapid onset of inflammation suggests the innate immune response may have a role to play in the pathogenesis of this hMPV infection. Since, the matrix protein is one of the most abundant proteins in the Paramyxoviridae family virion, we hypothesized that the inflammatory modulation observed in hMPV infected patients may be partly associated with the matrix protein (M-hMPV) response. By western blot analysis, we detected a soluble form of M-hMPV released from hMPV infected cell as well as from M-hMPV transfected HEK 293T cells suggesting that M-hMPV may be directly in contact with antigen presenting cells (APCs) during the course of infection. Moreover, flow cytometry and confocal microscopy allowed determining that M-hMPV was taken up by dendritic cells (moDCs) and macrophages inducing their activation. Furthermore, these moDCs enter into a maturation process inducing the secretion of a broad range of inflammatory cytokines when exposed to M-hMPV. Additionally, M-hMPV activated DCs were shown to stimulate IL-2 and IFN-γ production by allogeneic T lymphocytes. This M-hMPV-mediated activation and antigen presentation of APCs may in part explain the marked inflammatory immune response observed in pathology induced by hMPV in patients.


Assuntos
Imunidade/imunologia , Inflamação/imunologia , Metapneumovirus/imunologia , Proteínas da Matriz Viral/imunologia , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD1/metabolismo , Apoptose , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Endocitose , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Interferons/biossíntese , Receptores de Lipopolissacarídeos/metabolismo , Ativação Linfocitária/imunologia , Monócitos/citologia , Ligação Proteica , Frações Subcelulares/imunologia , Frações Subcelulares/virologia , Linfócitos T/imunologia , Transfecção
11.
Virology ; 406(1): 138-48, 2010 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-20674956

RESUMO

Human noroviruses are the leading cause of nonbacterial gastroenteritis worldwide and are now recognised as a significant human pathogen. Whereas human noroviruses cannot be cultivated in the laboratory, mouse norovirus 1 (MNV-1) is easily cultivated and has a defined tropism for cells of a mononuclear origin. As such, MNV-1 provides an ideal opportunity to study many aspects of norovirus biology and replication. Previously, we have shown that MNV-1 RNA replication is associated with components of the early and late secretory pathway and that all six open reading frame 1 (ORF1) proteins are associated with the viral dsRNA within the replication complex (RC) during the course of infection. In this study, we further characterise the subcellular localisation of the MNV-1 ORF1 proteins when recombinantly expressed in cells. We show that two MNV-1 proteins, NS1-2 and NS4, associate with the endoplasmic reticulum and endosomes, respectively. Whereas NS6 (the viral protease) appeared to localize within the cytoplasm and to mitochondria, NS7 (the viral polymerase) was observed to localize diffusely within the cytoplasm and within the nucleus, and NS3 localized to discrete foci within the cytoplasm which were of unknown origin. Based on the localization patterns observed we propose a model by which NS1-2 and NS4 may recruit host membranes to the MNV-1 RC during replication.


Assuntos
Norovirus/fisiologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Animais , Chlorocebus aethiops , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos , Modelos Biológicos , Norovirus/genética , Norovirus/patogenicidade , Fases de Leitura Aberta , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares/virologia , Transfecção , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética , Replicação Viral/genética
12.
J Biomol Struct Dyn ; 28(2): 175-86, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20645651

RESUMO

Knowledge of the subcellular localization of viral proteins in a host cell or virus-infected cell is very important because it is closely related to their destructive tendencies and consequences. Facing the avalanche of new protein sequences discovered in the post genomic era, we are challenged to develop automated methods for quickly and accurately predicting the location sites of viral proteins in a host cell; the information thus acquired is particularly important for medical science and antiviral drug design. In view of this, a new fusion classifier called "Virus-mPLoc" was established by hybridizing the gene ontology information, functional domain information, and sequential evolutionary information. The new predictor not only can more accurately predict the location sites of viral proteins in a host cell, but also have the capacity to identify the multiple-location virus proteins, which is beyond the reach of any existing predictors specialized for viral proteins. For reader's convenience, a user-friendly web-server for Virus-mPLoc was designed that is freely accessible at http://www.csbio.sjtu.edu.cn/bioinf/virus-multi/.


Assuntos
Estruturas da Membrana Celular/química , Biologia Computacional/métodos , Frações Subcelulares , Vírus/química , Bases de Dados de Proteínas , Humanos , Frações Subcelulares/química , Frações Subcelulares/virologia
13.
Virology ; 398(2): 187-93, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20042211

RESUMO

Poxviruses encode a redox system for intramolecular disulfide bond formation in cytoplasmic domains of viral proteins. Our objectives were to determine the kinetics and intracellular location of disulfide bond formation. The vaccinia virus L1 myristoylated membrane protein, used as an example, has three intramolecular disulfide bonds. Reduced and disulfide-bonded forms of L1 were distinguished by electrophoretic mobility and reactivity with monoclonal and polyclonal antibodies. Because disulfide bonds formed during 5 min pulse labeling with radioactive amino acids, a protocol was devised in which dithiothreitol was present at this step. Disulfide bond formation was detected by 2 min after removal of reducing agent and was nearly complete in 10 min. When the penultimate glycine residue was mutated to prevent myristoylation, L1 was mistargeted to the endoplasmic reticulum and disulfide bond formation failed to occur. These data suggested that viral membrane association was required for oxidation of L1, providing specificity for the process.


Assuntos
Dissulfetos/metabolismo , Frações Subcelulares/virologia , Vacínia/metabolismo , Western Blotting , Eletroforese em Gel de Poliacrilamida , Células HeLa , Humanos , Cinética , Microscopia Confocal , Oxirredução , Frações Subcelulares/metabolismo
14.
PLoS One ; 4(12): e8325, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20016834

RESUMO

BACKGROUND: Flavivirus infected cells produce infectious virions and subviral particles, both of which are formed by the assembly of prM and E envelope proteins and are believed to undergo the same maturation process. Dengue recombinant subviral particles have been produced in cell cultures with either modified or chimeric proteins but not using the native forms of prM and E. METHODOLOGY/PRINCIPAL FINDINGS: We have used a codon optimization strategy to obtain an efficient expression of native viral proteins and production of recombinant subviral particles (RSPs) for all four dengue virus (DV) serotypes. A stable HeLa cell line expressing DV1 prME was established (HeLa-prME) and RSPs were analyzed by immunofluorescence and transmission electron microscopy. We found that E protein is mainly present in the endoplasmic reticulum (ER) where assembly of RSPs could be observed. Biochemical characterization of DV1 RSPs secretion revealed both prM protein cleavage and homodimerization of E proteins before their release into the supernatant, indicating that RSPs undergo a similar maturation process as dengue virus. Pulse chase experiment showed that 8 hours are required for the secretion of DV1 RSPs. We have used HeLa-prME to develop a semi-quantitative assay and screened a human siRNA library targeting genes involved in membrane trafficking. Knockdown of 23 genes resulted in a significant reduction in DV RSP secretion, whereas for 22 others we observed an increase of RSP levels in cell supernatant. CONCLUSIONS/SIGNIFICANCE: Our data describe the efficient production of RSPs containing native prM and E envelope proteins for all dengue serotypes. Dengue RSPs and corresponding producing cell lines are safe and novel tools that can be used in the study of viral egress as well as in the development of vaccine and drugs against dengue virus.


Assuntos
Vírus da Dengue/classificação , Vírus da Dengue/fisiologia , Proteínas Virais/metabolismo , Vírion/metabolismo , Montagem de Vírus/fisiologia , Centrifugação com Gradiente de Concentração , Códon/genética , Vírus da Dengue/genética , Retículo Endoplasmático/virologia , Genes Virais/genética , Células HeLa , Humanos , Microscopia Eletrônica , Biossíntese de Proteínas , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Via Secretória , Sorotipagem , Frações Subcelulares/virologia , Proteínas Virais/genética
15.
J Virol ; 82(17): 8673-86, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18562514

RESUMO

Varicella-zoster virus (VZV) open reading frame (ORF) 63 protein (ORF63p) is one of six VZV ORFs shown to be transcribed and translated in latently infected human dorsal root ganglia. ORF63p accumulates exclusively in the cytoplasm of latently infected sensory neurons, whereas it is both nuclear and cytoplasmic during lytic infection and following reactivation from latency. Here, we demonstrate that infection of primary guinea pig enteric neurons (EN) with an adenovirus expressing ORF63p results in the exclusive cytoplasmic localization of the protein reminiscent of its distribution during latent VZV infection in humans. We show that the addition of the simian virus 40 large-T-antigen nuclear localization signal (NLS) results in the nuclear import of ORF63p in EN and that the ORF63p endogenous NLSs are functional in EN when fused to a heterologous protein. These data suggest that the cytoplasmic localization of ORF63p in EN results from the masking of the NLSs, thus blocking nuclear import. However, the coexpression of ORF61p, a strictly lytic VZV protein, and ORF63p in EN results in the nuclear import of ORF63p in a proteasome-dependent manner, and both ORF63p NLSs are required for this event. We propose that the cytoplasmic localization of ORF63p in neurons results from NLS masking and that the expression of ORF61p removes this block, allowing nuclear import to proceed.


Assuntos
Núcleo Celular/metabolismo , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Neurônios/virologia , Proteínas do Envelope Viral/metabolismo , Transporte Ativo do Núcleo Celular , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Linhagem Celular Transformada , Transformação Celular Viral , Células Cultivadas , Sistema Nervoso Entérico/citologia , Células Epiteliais/virologia , Técnica Indireta de Fluorescência para Anticorpo , Gânglios/citologia , Glutationa Transferase/metabolismo , Cobaias , Herpes Zoster/virologia , Herpesvirus Humano 3/patogenicidade , Humanos , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/genética , Luciferases de Renilla/metabolismo , Dados de Sequência Molecular , Plasmídeos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares/metabolismo , Frações Subcelulares/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral
17.
J Gen Virol ; 88(Pt 7): 1977-1985, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17554031

RESUMO

The replicase of Semliki Forest virus (SFV) consists of four non-structural proteins, designated nsP1-4, and is bound to cellular membranes via an amphipathic peptide and palmitoylated cysteine residues of nsP1. It was found that mutations preventing nsP1 palmitoylation also attenuated virus replication. The replacement of these cysteines by alanines, or their deletion, abolished virus viability, possibly due to disruption of interactions between nsP1 and nsP4, which is the catalytic subunit of the replicase. However, during a single infection cycle, the ability of the virus to replicate was restored due to accumulation of second-site mutations in nsP1. These mutations led to the restoration of nsP1-nsP4 interaction, but did not restore the palmitoylation of nsP1. The proteins with palmitoylation-site mutations, as well as those harbouring compensatory mutations in addition to palmitoylation-site mutations, were enzymically active and localized, at least in part, on the plasma membrane of transfected cells. Interestingly, deletion of 7 aa including the palmitoylation site of nsP1 had a relatively mild effect on virus viability and no significant impact on nsP1-nsP4 interaction. Similarly, the change of cysteine to alanine at the palmitoylation site of nsP1 of Sindbis virus had only a mild effect on virus replication. Taken together, these findings indicate that nsP1 palmitoylation as such is not the factor determining the ability to bind to cellular membranes and form a functional replicase complex. Instead, these abilities may be linked to the three-dimensional structure of nsP1 and the capability of nsP1 to interact with other components of the viral replicase complex.


Assuntos
Genes Virais , Mutação , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/fisiologia , Proteínas não Estruturais Virais/genética , Animais , Sítios de Ligação/genética , Células COS , Chlorocebus aethiops , Cricetinae , Células HeLa , Humanos , Ácidos Palmíticos/química , Replicon , Vírus da Floresta de Semliki/patogenicidade , Frações Subcelulares/virologia , Transfecção , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Virulência/genética , Replicação Viral/genética
18.
J Hepatol ; 46(1): 53-9, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17107733

RESUMO

BACKGROUND/AIMS: The establishment of HCV replicon systems strongly improved the research on the replication processes but poorly advanced our knowledge on the subcellular localization of the structural glycoproteins, mainly due to their low expression. We sought to verify whether reinforcing E1E2 expression in the context of both HCV genomic and subgenomic replicon from either homologous or heterologous strains leads to formation of supramolecular structures including structural and nonstructural proteins. METHODS: Robust expression of HCV glycoproteins was achieved by stable expression of E1E2p7 from genotype 1a and 1b. RESULTS: In these cells, E1 and E2 triggered the formation of dot-like structures in which they co-localized with core and the nonstructural proteins NS3 and NS5A. Confocal microscopy analyses suggested that accumulation of HCV proteins occurs in an ER-derived subcompartment. Moreover, by labeling de novo-synthesized HCV RNA, we showed that these structures constitute a site of viral RNA synthesis. CONCLUSIONS: Expression in trans of HCV glycoproteins in the context of replicative viral genome or subgenome generates accumulation of structural and nonstructural viral proteins in peculiar cytoplasmic structures. The simultaneous presence of viral RNA, structural and nonstructural protein suggests that these complexes represent not only sites of HCV replication but also potential places of viral pre-budding.


Assuntos
Hepacivirus/fisiologia , Proteínas Virais/metabolismo , Sequência de Bases , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , DNA Viral/genética , Retículo Endoplasmático/virologia , Expressão Gênica , Genoma Viral , Hepacivirus/genética , Humanos , Neoplasias Hepáticas/virologia , Complexos Multiproteicos , Replicon , Frações Subcelulares/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Replicação Viral
19.
J Virol ; 81(4): 1888-98, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17151128

RESUMO

We had previously identified active autonomous copies of the MusD long terminal repeat-retrotransposon family, which have retained transpositional activity. These elements are closely related to betaretroviruses but lack an envelope (env) gene. Here we show that these elements encode strictly intracellular virus-like particles that can unambiguously be identified by electron microscopy. We demonstrate intracellular maturation of the particles, with a significant proportion of densely packed cores for wild-type MusD but not for a protease mutant. We show that the molecular origin of this unexpected intracellular localization is solely dependent on the N-terminal part of the Gag protein, which lacks a functional sequence for myristoylation and plasma membrane targeting: replacement of the N-terminal domain of the MusD matrix protein by that of its closest relative-the Mason-Pfizer monkey virus-led to targeting of the MusD Gag to the plasma membrane, with viral particles budding and being released into the cell supernatant. These particles can further be pseudotyped with a heterologous envelope protein and become infectious, thus "reconstituting" a functional retrovirus prone to proviral insertions. Consistent with its retroviral origin, a sequence with a constitutive transport element-like activity can further be identified at the MusD 3' untranslated region. A molecular scenario is proposed that accounts for the transition, during evolution, from an ancestral infectious betaretrovirus to the strictly intracellular MusD retrotransposon, involving not only the loss of the env gene but also an inability to escape the cell--via altered targeting of the Gag protein--resulting de facto in the generation of a very successful "intracellularized" insertional mutagen.


Assuntos
Retrovirus Endógenos/genética , Evolução Molecular , Provírus/genética , Regiões 3' não Traduzidas/genética , Sequência de Aminoácidos , Animais , Gatos , Células Cultivadas , Retrovirus Endógenos/ultraestrutura , Feminino , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Humanos , Camundongos , Microscopia Eletrônica , Dados de Sequência Molecular , Estrutura Terciária de Proteína/genética , Provírus/ultraestrutura , Alinhamento de Sequência , Frações Subcelulares/ultraestrutura , Frações Subcelulares/virologia , Proteínas da Matriz Viral
20.
J Gen Virol ; 87(Pt 9): 2605-2614, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16894199

RESUMO

Dengue virus, a member of the family Flaviviridae of positive-strand RNA viruses, has seven non-structural proteins: NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5. Except for enzymic activities contained within NS3 and NS5, the roles of the other proteins in virus replication and pathogenesis are not well defined. In this study, a physical interaction between NS4B and the helicase domain of NS3 was identified by using a yeast two-hybrid assay. This interaction was further confirmed by biochemical pull-down and immunoprecipitation assays, both with purified proteins and with dengue virus-infected cell lysates. NS4B co-localized with NS3 in the perinuclear region of infected human cells. Furthermore, NS4B dissociated NS3 from single-stranded RNA and consequently enhanced the helicase activity of NS3 in an in vitro unwinding assay. These results suggest that NS4B modulates dengue virus replication via its interaction with NS3.


Assuntos
Vírus da Dengue/fisiologia , RNA Viral/metabolismo , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Cricetinae , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Humanos , Técnicas In Vitro , Substâncias Macromoleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , RNA Helicases/genética , RNA Helicases/fisiologia , RNA Viral/química , RNA Viral/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia , Frações Subcelulares/virologia , Técnicas do Sistema de Duplo-Híbrido , Proteínas não Estruturais Virais/genética , Virulência , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...