Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Commun Biol ; 6(1): 134, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36725983

RESUMO

DDX3X is a mammalian RNA helicase that regulates RNA metabolism, cancers, innate immunity and several RNA viruses. We discovered that herpes simplex virus 1, a nuclear DNA replicating virus, redirects DDX3X to the nuclear envelope where it surprisingly modulates the exit of newly assembled viral particles. DDX3X depletion also leads to an accumulation of virions in intranuclear herniations. Mechanistically, we show that DDX3X physically and functionally interacts with the virally encoded nuclear egress complex at the inner nuclear membrane. DDX3X also binds to and stimulates the incorporation in mature particles of pUs3, a herpes kinase that promotes viral nuclear release across the outer nuclear membrane. Overall, the data highlights two unexpected roles for an RNA helicase during the passage of herpes simplex viral particles through the nuclear envelope. This reveals a highly complex interaction between DDX3X and viruses and provides new opportunities to target viral propagation.


Assuntos
Infecções por Herpesviridae , Herpesvirus Humano 1 , Animais , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Membrana Nuclear/metabolismo , Núcleo Celular/metabolismo , Mamíferos
2.
PeerJ ; 10: e13721, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35833016

RESUMO

Coronaviruses have been at the forefront of the news for the last 2 years. Unfortunately, SARS-CoV-2, the etiologic agent for the COVID-19 pandemic, must be manipulated in biosecurity level 3 settings, which significantly limits research. Meanwhile, several less pathogenic human coronaviruses (HCoV) exist and can be studied in much more common biosafety level 2 laboratories. Among them, HCoV-OC43 is a good surrogate candidate for SARS-CoV-2 since both are phylogenetically related human Betacoronaviruses. However, one issue has been the lack of standardized means among laboratories to propagate and titer this less virulent coronavirus. The present study probes the optimal parameters to propagate HCoV-OC43. First, testing of five different cell lines (MRC-5, Huh7.5, Vero, HCT-8, HRT-18) indicated that the physiologically relevant MRC-5 human lung cell line produced among the highest viral titers. HRT-18 may however be an interesting alternative as they are quick growing cells that also led to higher viral titers and a better tropism for various HCoV-OC43 variants. We also probed the impact of serum and temperature during viral expansion and confirmed that the normal temperature of the upper respiratory track (33 °C) improves viral yields over the typical 37 °C used to grow many other viruses. Meanwhile, we did not notice any evidence that serum concentrations significantly affected the virus but interestingly noted that the virus grew quite efficiently in a serum-free media formulation. Meanwhile sonication of viral stocks somewhat improved viral titers. Four titration methods (plaque assays, TCID50-CPE, TCID50-IFA and TCID50-IPA) were also probed using two cell lines (VeroE6 and HRT-18). In our hands, plaque assays proved unreliable and quantification of the virus by scoring CPE positive wells was significantly less sensitive than antibody-based assays (IFA and IPA). While the latter methods were equally sensitive, we favor the TCID50-IPA method since simpler, faster and cheaper than the IFA protocol. Moreover, the HRT-18 cells appeared more sensitive to quantify the virus. Perhaps most importantly, these optimized protocols routinely led to high titer viral stocks in the order of 108 TCID50/ml magnitude, which should fulfill the requirements of most experimental settings.


Assuntos
COVID-19 , Coronavirus Humano OC43 , Humanos , Pandemias , SARS-CoV-2 , Linhagem Celular
3.
J Virol ; 96(9): e0045322, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35435724

RESUMO

Herpesviruses assemble new viral particles in the nucleus. These nucleocapsids bud through the inner nuclear membrane to produce enveloped viral particles in the perinuclear space before fusing with the outer nuclear membrane to reach the cytoplasm. This unusual route is necessary since viral capsids are too large to pass through nuclear pores. However, the transient perinuclear nucleocapsids (250 nm in diameter) are also larger than the width of the perinuclear space (30 to 50 nm). Interestingly, linker of the nucleoskeleton and cytoskeleton (LINC) components SUN and KASH connect the inner and outer nuclear membranes and regulate their spacing. Previous work by others on the related pseudorabies virus and human cytomegalovirus showed that they functionally interact with SUN proteins. To clarify the role of SUN proteins, we explored their impact on herpes simplex virus 1 (HSV-1), another herpesvirus. Using dominant negative SUN mutants and RNA interference, we show that HSV-1 propagation is dependent on the LINC complex. In contrast to pseudorabies virus, SUN2 disruption by either approach led to increased HSV-1 extracellular viral yields. This SUN2 dependency may be linked to its greater impact on perinuclear spacing in infected cells compared to SUN1. Finally, the virus itself seems to modulate perinuclear spacing. IMPORTANCE The large size of herpesviruses prevents them from travelling across the nuclear pores, and they instead egress across the two nuclear membranes, generating short-lived enveloped perinuclear virions. This poses a challenge as the perinuclear space is smaller than the virions. This implies the separation (unzipping) of the two nuclear membranes to accommodate the viral particles. The LINC complex bridges the two nuclear membranes and is an important regulator of perinuclear spacing. Work by others hint at its functional implication during pseudorabies virus and cytomegalovirus propagation. The present study probes the importance for HSV-1 of the SUN proteins, the LINC components found in the inner nuclear membrane. Using dominant negative constructs and RNA interference (RNAi), the data reveal that SUN2 exhibits antiviral propriety toward HSV-1, as disrupting the protein leads to increased viral yields. This is in contrast with that reported for pseudorabies and suggests that differences among herpesviruses may, once again, prevail.


Assuntos
Herpesvirus Humano 1 , Herpesvirus Suídeo 1 , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Animais , Núcleo Celular/metabolismo , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Membrana Nuclear/metabolismo , Nucleocapsídeo/metabolismo , Vírion/metabolismo
4.
J Virol ; 95(4)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33239454

RESUMO

Herpes simplex virus replicates in the nucleus, where new capsids are assembled. It produces procapsids devoid of nucleic acid but containing the preVP22a scaffold protein. These thermo-unstable particles then mature into A-, B- or C-nuclear icosahedral capsids, depending on their ability to shed the proteolytically processed scaffold and incorporation of the viral genome. To study how these viral capsids differ, we performed proteomics studies of highly enriched HSV-1 A-, B- and C-nuclear capsids, relying in part on a novel and powerful flow virometry approach to purify C-capsids. We found that the viral particles contained the expected capsid components and identified several tegument proteins in the C-capsid fraction (pUL21, pUL36, pUL46, pUL48, pUL49, pUL50, pUL51 and pUS10). Moreover, numerous ribosomal, hnRNPs and other host proteins, absent from the uninfected controls, were detected on the capsids with some of them seemingly specific to C-capsids (glycogen synthase, four different keratin-related proteins, fibronectin 1 and PCBP1). A subsequent proteomics analysis was performed to rule out the presence of protein complexes that may share similar density as the viral capsids but do not otherwise interact with them. Using pUL25 or VP5 mutant viruses incapable of assembling C-nuclear or all nuclear capsids, respectively, we confirmed the bulk of our initial findings. Naturally, it will next be important to address the functional relevance of these proteins.IMPORTANCE Much is known about the biology of herpesviruses. This includes their unique ability to traverse the two nuclear envelopes by sequential budding and fusion steps. For HSV-1, this implies the pUL31/pUL34 and pUL17/pUL25 complexes that may favor C-capsid egress. However, this selection process is not clear, nor are all the differences that distinguish A-, B- and C-capsids. The present study probes what proteins compose these capsids, including host proteins. This should open up new research avenues to clarify the biology of this most interesting family of viruses. It also reiterates the use of flow virometry as an innovative tool to purify viral particles.

5.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32817212

RESUMO

The glycoprotein M of herpes simplex virus 1 (HSV-1) is dynamically relocated from nuclear membranes to the trans-Golgi network (TGN) during infection, but molecular partners that promote this relocalization are unknown. Furthermore, while the presence of the virus is essential for this phenomenon, it is not clear if this is facilitated by viral or host proteins. Past attempts to characterize glycoprotein M (gM) interacting partners identified the viral protein gN by coimmunoprecipitation and the host protein E-Syt1 through a proteomics approach. Interestingly, both proteins modulate the activity of gM on the viral fusion machinery. However, neither protein is targeted to the nuclear membrane and consequently unlikely explains the dynamic regulation of gM nuclear localization. We thus reasoned that gM may transiently interact with other molecules. To resolve this issue, we opted for a proximity-dependent biotin identification (BioID) proteomics approach by tagging gM with a BirA* biotinylation enzyme and purifying BirA substrates on a streptavidin column followed by mass spectrometry analysis. The data identified gM and 170 other proteins that specifically and reproducibly were labeled by tagged gM at 4 or 12 h postinfection. Surprisingly, 35% of these cellular proteins are implicated in protein transport. Upon testing select candidate proteins, we discovered that XPO6, an exportin, is required for gM to be released from the nucleus toward the TGN. This is the first indication of a host or viral protein that modulates the presence of HSV-1 gM on nuclear membranes.IMPORTANCE The mechanisms that enable integral proteins to be targeted to the inner nuclear membrane are poorly understood. Herpes simplex virus 1 (HSV-1) glycoprotein M (gM) is an interesting candidate, as it is dynamically relocalized from nuclear envelopes to the trans-Golgi network (TGN) in a virus- and time-dependent fashion. However, it was, until now, unclear how gM was directed to the nucleus or evaded that compartment later on. Through a proteomic study relying on a proximity-ligation assay, we identified several novel gM interacting partners, many of which are involved in vesicular transport. Analysis of select proteins revealed that XPO6 is required for gM to leave the nuclear membranes late in the infection. This was unexpected, as XPO6 is an exportin specifically associated with actin/profilin nuclear export. This raises some very interesting questions about the interaction of HSV-1 with the exportin machinery and the cargo specificity of XPO6.


Assuntos
Herpesvirus Humano 1/metabolismo , Carioferinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Rede trans-Golgi/metabolismo , Biotinilação , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Expressão Gênica , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Carioferinas/genética , Glicoproteínas de Membrana/genética , Membrana Nuclear/virologia , Ligação Proteica , Transporte Proteico , Proteômica/métodos , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Coloração e Rotulagem/métodos , Estreptavidina/química , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Proteína ran de Ligação ao GTP/genética , Rede trans-Golgi/virologia
6.
Virologie (Montrouge) ; 24(4): 210-230, 2020 Aug 01.
Artigo em Francês | MEDLINE | ID: mdl-32795979

RESUMO

The Alphaherpesvirinae sub-family includes viruses primarily associated with cold sores, genital herpes, chicken pox and shingles in humans, but are responsible for several other pathologies and additionally infect many animals. These viruses are large entities that travel through various cellular compartments during their life cycle. As for the transport of cellular cargoes, this involves several budding and fusion steps as well as transport of viral particles along the cytoskeleton. Though the entry of these viruses in cells is generally well understood at the molecular level, the egress of newly assembled viral particles is poorly characterized. Albeit several viral genes have been implicated, their mode of action and the contribution of the cell remain to be clarified. The present review updates our current knowledge of the transport of herpes viruses and pinpoints open questions about the mechanisms they exploit.


Assuntos
Alphaherpesvirinae , Herpes Labial , Herpesvirus Humano 1 , Animais , Transporte Biológico , Humanos , Vírion
7.
Methods Mol Biol ; 2060: 279-288, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617184

RESUMO

The analysis of HSV-1 mature extracellular virions by proteomics requires highly enriched samples to limit false-positives and favor the detection of true components. The protocol described below involves the removal of highly contaminating serum proteins and purification of the virions by a series of differential and density centrifugation steps. In addition, L-particles, which are viral particles devoid of a genome and capsid but present in the extracellular milieu, are depleted on Ficoll 400 gradients. As previously reported, the resulting viral particles are free of most contaminants and suitable for mass spectrometry.


Assuntos
Herpesvirus Humano 1 , Proteômica , Vírion , Células HeLa , Herpesvirus Humano 1/química , Herpesvirus Humano 1/isolamento & purificação , Herpesvirus Humano 1/metabolismo , Humanos , Vírion/química , Vírion/isolamento & purificação , Vírion/metabolismo
8.
Methods Mol Biol ; 2060: 289-303, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617185

RESUMO

Flow cytometry has been instrumental in characterizing normal and infected cells. However, until recently, it was not possible to use such an approach to analyze small entities such as bacteria, let alone viruses, owing to the 0.5 µm resolution of most instruments. To circumvent this limitation, some laboratories decorate pathogens with antibodies or nanoparticles. Our laboratory instead exploits an alternative approach that relies on the staining of internal viral constituents with permeable SYTO dyes or the fluorescent tagging of individual viral proteinaceous components, whether capsid, tegument or glycoproteins. This opens up a range of new research avenues and, for example, enabled us to characterize individual herpes simplex virus type 1 particles, discern their different subpopulations, measure the heterogeneity of mature virions in terms of protein content, sort these viral particles with >90% purity and, for the first time, directly address the impact of this heterogeneity on viral fitness. This approach, coined flow virometry or nanoscale flow cytometry, allows for the study of a wide variety of pathogens with high statistical significance and the potential discovery of novel virulence factors.


Assuntos
Citometria de Fluxo , Herpesvirus Humano 1/metabolismo , Vírion/metabolismo , Células HeLa , Herpesvirus Humano 1/ultraestrutura , Humanos , Vírion/ultraestrutura
9.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232182

RESUMO

The assembly of new herpes simplex virus 1 (HSV-1) particles takes place in the nucleus. These particles then travel across the two nuclear membranes and acquire a final envelope from a cellular compartment. The contribution of the cell to the release of the virus is, however, little known. We previously demonstrated, using a synchronized infection, that the host protein kinase D and diacylglycerol, a lipid that recruits the kinase to the trans-Golgi network (TGN), promote the release of the virus from that compartment. Given the role this cellular protein plays in the herpes simplex virus 1 life cycle and the many molecules that modulate its activity, we aimed to determine to what extent this virus utilizes the protein kinase D pathway during a nonsynchronized infection. Several molecular protein kinase D (PKD) regulators were targeted by RNA interference and viral production monitored. Surprisingly, many of these modulators negatively impacted the extracellular release of the virus. Overexpression studies, the use of pharmacological reagents, and assays to monitor intracellular lipids implicated in the biology of PKD suggested that these effects were oddly independent of total intracellular diacylglycerol levels. Instead, mapping of the viral intermediates by electron microscopy suggested that some of these modulators could regulate distinct steps along the viral egress pathway, notably nuclear egress. Altogether, this suggests a more complex contribution of PKD to HSV-1 egress than originally anticipated and new research avenues to explore.IMPORTANCE Viruses are obligatory parasites that highjack numerous cellular functions. This is certainly true when it comes to transporting viral particles within the cell. Herpesviruses share the unique property of traveling through the two nuclear membranes by subsequent budding and fusion and acquiring their final envelope from a cellular organelle. Albeit disputed, the overall evidence from many laboratories points to the trans-Golgi network (TGN) as the source of that membrane. Moreover, past findings revealed that the host protein kinase D (PKD) plays an important role at that stage, which is significant given the known implication of that protein in vesicular transport. The present findings suggest that the PKD machinery not only affects the late stages of herpes simplex virus I egress but also modulates earlier steps, such as nuclear egress. This opens up new means to control these viruses.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas do Olho/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas de Membrana/metabolismo , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Liberação de Vírus , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas do Olho/antagonistas & inibidores , Proteínas do Olho/genética , Herpes Simples/genética , Herpes Simples/metabolismo , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Células Tumorais Cultivadas , Células Vero , Rede trans-Golgi
10.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046455

RESUMO

Enveloped viruses typically encode their own fusion machinery to enter cells. Herpesviruses are unusual, as they fuse with a number of cellular compartments throughout their life cycles. As uncontrolled fusion of the host membranes should be avoided in these events, tight regulation of the viral fusion machinery is critical. While studying herpes simplex virus 1 (HSV-1) glycoprotein gM, we identified the cellular protein E-Syt1 (extended synaptotagmin 1) as an interaction partner. The interaction took place in both infected and transfected cells, suggesting other viral proteins were not required for the interaction. Most interestingly, E-Syt1 is a member of the synaptotagmin family of membrane fusion regulators. However, the protein is known to promote the tethering of the endoplasmic reticulum (ER) to the plasma membrane. We now show that E-Syt1, along with the related E-Syt3, negatively modulates viral release into the extracellular milieu, cell-to-cell viral spread, and viral entry, all processes that implicate membrane fusion events. Similarly, these E-Syt proteins impacted the formation of virus-induced syncytia. Altogether, these findings hint at the modulation of the viral fusion machinery by the E-Syt family of proteins.IMPORTANCE Viruses typically encode their own fusion apparatus to enable them to enter cells. For many viruses, this means a single fusogenic protein. However, herpesviruses are large entities that express several accessory viral proteins to regulate their fusogenic activity. The present study hints at the additional participation of cellular proteins in this process, suggesting the host can also modulate viral fusion to some extent. Hence E-Syt proteins 1 and 3 seem to negatively modulate the different viral fusion events that take place during the HSV-1 life cycle. This could represent yet another innate immunity response to the virus.


Assuntos
Herpesvirus Humano 1/fisiologia , Fusão de Membrana , Glicoproteínas de Membrana/metabolismo , Sinaptotagminas/metabolismo , Proteínas Virais/metabolismo , Células Gigantes/virologia , Células HeLa , Herpesvirus Humano 1/genética , Humanos , Imunoprecipitação , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , RNA Interferente Pequeno , Sinaptotagminas/deficiência , Sinaptotagminas/genética , Proteínas Virais/química , Proteínas Virais/genética , Liberação de Vírus
11.
J Virol ; 92(3)2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29167334

RESUMO

For several decades, flow cytometry has been a common approach to analyze cells and sort them to near-purity. It enables one to probe inner cellular molecules, surface receptors, or infected cells. However, the analysis of smaller entities such as viruses and exocytic vesicles has been more difficult but is becoming mainstream. This has in part been due to the development of new instrumentation with resolutions below that of conventional cytometers. It is also attributed to the several means employed to fluorescently label viruses, hence enabling them to stand out from similarly sized particles representing background noise. Thus far, more than a dozen different viruses ranging in size from 40 nm to giant viruses have been probed by this approach, which was recently dubbed "flow virometry." These studies have collectively highlighted the breadth of the applications of this method, which, for example, has elucidated the maturation of dengue virus, served as quality control for vaccinia vaccines, and enabled the sorting of herpes simplex virus discrete viral particles. The present review focuses on the means employed to characterize and sort viruses by this powerful technology and on the emerging uses of flow virometry. It similarly addresses some of its current challenges and limitations.


Assuntos
Biomarcadores/análise , Citometria de Fluxo/métodos , Vírion/isolamento & purificação , Vírus da Dengue/isolamento & purificação , Humanos , Simplexvirus/isolamento & purificação
12.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275191

RESUMO

Several virulence genes have been identified thus far in the herpes simplex virus 1 genome. It is also generally accepted that protein heterogeneity among virions further impacts viral fitness. However, linking this variability directly with infectivity has been challenging at the individual viral particle level. To address this issue, we resorted to flow cytometry (flow virometry), a powerful approach we recently employed to analyze individual viral particles, to identify which tegument proteins vary and directly address if such variability is biologically relevant. We found that the stoichiometry of the UL37, ICP0, and VP11/12 tegument proteins in virions is more stable than the VP16 and VP22 tegument proteins, which varied significantly among viral particles. Most interestingly, viruses sorted for their high VP16 or VP22 content yielded modest but reproducible increases in infectivity compared to their corresponding counterparts containing low VP16 or VP22 content. These findings were corroborated for VP16 in short interfering RNA experiments but proved intriguingly more complex for VP22. An analysis by quantitative Western blotting revealed substantial alterations of virion composition upon manipulation of individual tegument proteins and suggests that VP22 protein levels acted indirectly on viral fitness. These findings reaffirm the interdependence of the virion components and corroborate that viral fitness is influenced not only by the genome of viruses but also by the stoichiometry of proteins within each virion.IMPORTANCE The ability of viruses to spread in animals has been mapped to several viral genes, but other factors are clearly involved, including virion heterogeneity. To directly probe whether the latter influences viral fitness, we analyzed the protein content of individual herpes simplex virus 1 particles using an innovative flow cytometry approach. The data confirm that some viral proteins are incorporated in more controlled amounts, while others vary substantially. Interestingly, this correlates with the VP16 trans-activating viral protein and indirectly with VP22, a second virion component whose modulation profoundly alters virion composition. This reaffirms that not only the presence but also the amount of specific tegument proteins is an important determinant of viral fitness.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Proteínas Estruturais Virais/metabolismo , Animais , Western Blotting , Chlorocebus aethiops , Citometria de Fluxo , Genes Virais , Proteína Vmw65 do Vírus do Herpes Simples/análise , Proteína Vmw65 do Vírus do Herpes Simples/química , Herpesvirus Humano 1/patogenicidade , RNA Interferente Pequeno , Células Vero , Proteínas Estruturais Virais/análise , Proteínas Estruturais Virais/química , Vírion/genética , Vírion/fisiologia , Montagem de Vírus
13.
J Neurochem ; 141(4): 592-605, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28222213

RESUMO

The axonal microtubule-associated protein TAU, involved in Alzheimer's disease (AD), can be found in the extracellular space where it could be taken up by neurons, an event that is believed to contribute to the propagation of tau pathology in the brain. Since the small GTPase Rab7A is involved in the trafficking of endosomes, autophagosomes, and lysosomes, and RAB7A gene expression and protein levels are up-regulated in AD patients, we tested the hypothesis that Rab7A was involved in tau secretion. We previously reported that both primary cortical neurons and HeLa cells over-expressing human TAU can release tau. Using these two cellular systems, we demonstrated that Rab7A regulates tau secretion. Upon Rab7A deletion, tau secretion was decreased. Consistent with this, the over-expression of a dominant negative and a constitutively active form of Rab7A decreased and increased tau secretion, respectively. A partial co-localization of tau and Rab7-positive structures in both neurons and HeLa cells indicated that a late endosomal compartment could be involved in its secretion. Collectively, the present data indicate that Rab7A regulates tau secretion and therefore the up-regulation of RAB7A reported in AD, could contribute to the extracellular accumulation of pathological TAU species that could result in the propagation of tau pathology in the AD brain.


Assuntos
Proteínas rab de Ligação ao GTP/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Endossomos/metabolismo , Deleção de Genes , Células HeLa , Humanos , Neurônios/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno , Regulação para Cima , Proteínas rab de Ligação ao GTP/genética , proteínas de unión al GTP Rab7
14.
J Virol ; 91(8)2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28148788

RESUMO

The human protein DDX3X is a DEAD box ATP-dependent RNA helicase that regulates transcription, mRNA maturation, and mRNA export and translation. DDX3X concomitantly modulates the replication of several RNA viruses and promotes innate immunity. We previously showed that herpes simplex virus 1 (HSV-1), a human DNA virus, incorporates DDX3X into its mature particles and that DDX3X is required for optimal HSV-1 infectivity. Here, we show that viral gene expression, replication, and propagation depend on optimal DDX3X protein levels. Surprisingly, DDX3X from incoming viral particles was not required for the early stages of the HSV-1 infection, but, rather, the protein controlled the assembly of new viral particles. This was independent of the previously reported ability of DDX3X to stimulate interferon type I production. Instead, both the lack and overexpression of DDX3X disturbed viral gene transcription and thus subsequent genome replication. This suggests that in addition to its effect on RNA viruses, DDX3X impacts DNA viruses such as HSV-1 by an interferon-independent pathway.IMPORTANCE Viruses interact with a variety of cellular proteins to complete their life cycle. Among them is DDX3X, an RNA helicase that participates in most aspects of RNA biology, including transcription, splicing, nuclear export, and translation. Several RNA viruses and a limited number of DNA viruses are known to manipulate DDX3X for their own benefit. In contrast, DDX3X is also known to promote interferon production to limit viral propagation. Here, we show that DDX3X, which we previously identified in mature HSV-1 virions, stimulates HSV-1 gene expression and, consequently, virion assembly by a process that is independent of its ability to promote the interferon pathway.


Assuntos
RNA Helicases DEAD-box/metabolismo , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , Montagem de Vírus , Animais , Linhagem Celular , Herpesvirus Humano 1/genética , Humanos , Replicação Viral
15.
J Gen Virol ; 96(11): 3313-3325, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26265177

RESUMO

Herpes simplex virus type 1 (HSV-1) glycoprotein M (gM/UL10) is a 473 aa type III transmembrane protein that resides in various membrane compartments. HSV-1 gM contains several putative trafficking motifs, but their functional relevance remains to be elucidated. We show here that transiently expressed gM 19­343 was sufficient for transport to the trans-Golgi network (TGN), whilst gM 133­473, where the first two transmembrane domains were deleted, and gM 1­342, which lacked the final residue of the last transmembrane domain, were retained in the endoplasmic reticulum (ER), indicating that all transmembrane domains are required for proper folding and ER exit. A series of bacterial artificial chromosome mutants revealed that in addition to the authentic start codon, translation of gM can be initiated at methionine 19 and 133/135. Whilst a protein lacking the first 18 residues supported WT-like growth, gM 133/135­473 resulted in reduced plaque diameters resembling a UL10 deletion mutant. An HSV-1 mutant encoding gM 1­342 showed similar growth characteristics and accumulated non-enveloped cytoplasmic particles, whilst gM 1­343 resulted in a gain of function, indicating that all transmembrane domains of the protein are important for viral growth. A C-terminal extension further supported viral propagation; however, the C-terminal trafficking motifs (residues 423­473) were completely dispensable. We propose a functional core within gM 19­343 comprised of all transmembrane domains that is sufficient to target the protein to the TGN, a favoured site for envelopment, and to support viral functions.


Assuntos
Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Rede trans-Golgi/virologia , Motivos de Aminoácidos , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Glicoproteínas de Membrana/genética , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Virais/genética
16.
J Virol ; 89(4): 2313-23, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25505065

RESUMO

UNLABELLED: Herpes simplex virus 1 (HSV-1) capsids are assembled in the nucleus, where they incorporate the viral genome. They then transit through the two nuclear membranes and are wrapped by a host-derived envelope. In the process, several HSV-1 proteins are targeted to the nuclear membranes, but their roles in viral nuclear egress are unclear. Among them, glycoprotein M (gM), a known modulator of virus-induced membrane fusion, is distributed on both the inner and outer nuclear membranes at the early stages of the infection, when no other viral glycoproteins are yet present there. Later on, it is found on perinuclear virions and ultimately redirected to the trans-Golgi network (TGN), where it cycles with the cell surface. In contrast, transfected gM is found only at the TGN and cell surface, hinting at an interaction with other viral proteins. Interestingly, many herpesvirus gM analogs interact with their gN counterparts, which typically alters their intracellular localization. To better understand how HSV-1 gM localization is regulated, we evaluated its ability to bind gN and discovered it does so in both transfected and infected cells, an interaction strongly weakened by the deletion of the gM amino terminus. Functionally, while gN had no impact on gM localization, gM redirected gN from the endoplasmic reticulum (ER) to the TGN. Most interestingly, gN overexpression stimulated the formation of syncytia in the context of an infection by a nonsyncytial strain, indicating that gM and gN not only physically but also functionally interact and that gN modulates gM's activity on membrane fusion. IMPORTANCE: HSV-1 gM is an important modulator of virally induced cell-cell fusion and viral entry, a process that is likely finely modulated in time and space. Until now, little was known of the proteins that regulate gM's activity. In parallel, gM is found in various intracellular locations at different moments, ranging from nuclear membranes, perinuclear virions, the TGN, cell surface, and mature extracellular virions. In transfected cells, however, it is found only on the TGN and cell surface, hinting that its localization is modulated by other viral proteins. The present study identifies HSV-1 gN as a binding partner for gM, in agreement with their analogs in other herpesviruses, but most excitingly shows that gN modulates gM's impact on HSV-1-induced membrane fusion. These findings open up new research avenues on the viral fusion machinery.


Assuntos
Herpesvirus Humano 1/fisiologia , Glicoproteínas de Membrana/metabolismo , Multimerização Proteica , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Humanos , Mapeamento de Interação de Proteínas
17.
Methods Mol Biol ; 1144: 181-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24671684

RESUMO

The analysis of herpes simplex virus type 1 mature extracellular virions by proteomics requires highly enriched samples to limit false positives and favor the detection of true components. The protocol described below involves the removal of highly contaminating serum proteins and purification of the virions by a series of differential and density centrifugation steps. In addition, L-particles, which are viral particles devoid of genome and capsid but present in the extracellular milieu, are depleted on Ficoll 400 gradients. As previously reported, the resulting viral particles are free of most contaminants and suitable for mass spectrometry.


Assuntos
Herpesvirus Humano 1/genética , Biologia Molecular/métodos , Proteínas Virais/biossíntese , Vírion/genética , Herpesvirus Humano 1/metabolismo , Humanos , Espectrometria de Massas , Proteômica
18.
J Virol ; 87(7): 3990-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23365427

RESUMO

Macroautophagy is a cellular pathway that degrades intracellular pathogens and contributes to antigen presentation. Herpes simplex virus 1 (HSV-1) infection triggers both macroautophagy and an additional form of autophagy that uses the nuclear envelope as a source of membrane. The present study constitutes the first in-depth analysis of nuclear envelope-derived autophagy (NEDA). We established LC3a as a marker that allowed us to distinguish between NEDA and macroautophagy in both immunofluorescence and flow cytometry. NEDA was observed in many different cell types, indicating that it is a general response to HSV-1 infection. This autophagic pathway is known to depend on the viral protein γ34.5, which can inhibit macroautophagy via binding to beclin-1. Using mutant viruses, we were able to show that binding of beclin-1 by γ34.5 had no effect on NEDA, demonstrating that NEDA is regulated differently than macroautophagy. Instead, NEDA was triggered in response to γ34.5 binding to protein phosphatase 1α, an interaction used by the virus to prevent host cells from shutting off protein translation. NEDA was not triggered when late viral protein production was inhibited with acyclovir or hippuristanol, indicating that the accumulation of these proteins might stress infected cells. Interestingly, expression of the late viral protein gH was sufficient to rescue NEDA in the context of infection with a virus that otherwise does not support strong late viral protein expression. We argue that NEDA is a cellular stress response triggered late during HSV-1 infection and might compensate for the viral alteration of the macroautophagic response.


Assuntos
Autofagia/fisiologia , Herpes Simples/fisiopatologia , Herpesvirus Humano 1/fisiologia , Membrana Nuclear/fisiologia , Biossíntese de Proteínas/fisiologia , Animais , Biomarcadores/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Imunofluorescência , Herpesvirus Humano 1/ultraestrutura , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Virais/metabolismo
19.
J Virol ; 87(9): 4895-906, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23408623

RESUMO

Herpes simplex virus type 1 particles are multilayered structures with a DNA genome surrounded by a capsid, tegument, and envelope. While the protein content of mature virions is known, the sequence of addition of the tegument and the intracellular compartments where this occurs are intensely debated. To probe this process during the initial stages of egress, we used two approaches: an in vitro nuclear egress assay, which reconstitutes the exit of nuclear capsids to the cytoplasm, and a classical nuclear capsid sedimentation assay. As anticipated, in vitro cytoplasmic capsids did not harbor UL34, UL31, or viral glycoproteins but contained US3. In agreement with previous findings, both nuclear and in vitro capsids were positive for ICP0 and ICP4. Unexpectedly, nuclear C capsids and cytoplasmic capsids produced in vitro without any cytosolic viral proteins also scored positive for UL36 and UL37. Immunoelectron microscopy confirmed that these tegument proteins were closely associated with nuclear capsids. When cytosolic viral proteins were present in the in vitro assay, no additional tegument proteins were detected on the capsids. As previously reported, the tegument was sensitive to high-salt extraction but, surprisingly, was stabilized by exogenous proteins. Finally, some tegument proteins seemed partially lost during egress, while others possibly were added at multiple steps or modified along the way. Overall, an emerging picture hints at the early coating of capsids with up to 5 tegument proteins at the nuclear stage, the shedding of some viral proteins during nuclear egress, and the acquisition of others tegument proteins during reenvelopment.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Montagem de Vírus , Capsídeo/ultraestrutura , Proteínas do Capsídeo/genética , Linhagem Celular , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestrutura , Humanos , Microscopia Imunoeletrônica , Eliminação de Partículas Virais
20.
PLoS One ; 8(1): e53276, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23301054

RESUMO

Viruses are strictly dependent on cells to propagate and many incorporate host proteins in their viral particles, but the significance of this incorporation is poorly understood. Recently, we performed the first comprehensive characterization of the mature herpes simplex virus type 1 (HSV-1) in which up to 49 distinct cellular proteins were identified by mass spectrometry. In the present study, we sought to identify if these cellular factors are relevant for the HSV-1 life cycle. To this end, we performed a small interfering RNA functional screen and found that 15 of these host proteins altered HSV-1 proliferation in cell culture, without any significant effect on cell viability. Moreover, the siRNA used had no negative consequences for Adenovirus type 5 propagation (with one exception) indicating that the modulation was specific for HSV-1 and not merely due to unhealthy cells. The positive host proteins include several Rab GTPases and other intracellular transport components as well as proteins involved in signal transduction, gene regulation and immunity. Remarkably, in most cases when virions were depleted for one of the above proteins, they replicated more poorly in subsequent infections in wild type cells. This highlights for the first time that both the cellular and virion-associated pools of many of these proteins actively contribute to viral propagation. Altogether, these findings underscore the power and biological relevance of combining proteomics and RNA interference to identify novel host-pathogen interactions.


Assuntos
Herpesvirus Humano 1/fisiologia , Interferência de RNA , Vírion/metabolismo , Replicação Viral , Animais , Proliferação de Células , Sobrevivência Celular , Chlorocebus aethiops , Regulação Viral da Expressão Gênica , Espectrometria de Massas , RNA Interferente Pequeno/metabolismo , Células Vero , Proteínas Virais/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA