Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Virol ; 96(6): e0217821, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35045266

RESUMO

The assembly and egress of alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), within neurons is poorly understood. A key unresolved question is the structure of the viral particle that moves by anterograde transport along the axon, and two alternative mechanisms have been described. In the "married" model, capsids acquire their envelopes in the cell body and then traffic along axons as enveloped virions within a bounding organelle. In the "separate" model, nonenveloped capsids travel from the cell body into and along the axon, eventually encountering their envelopment organelles at a distal site, such as the nerve cell terminal. Here, we describe an "envelopment trap" to test these models using the dominant negative terminal endosomal sorting complex required for transport (ESCRT) component VPS4-EQ. Green fluorescent protein (GFP)-tagged VPS4-EQ was used to arrest HSV-1 or PRV capsid envelopment, inhibit downstream trafficking, and GFP-label envelopment intermediates. We found that GFP-VPS4-EQ inhibited trafficking of HSV-1 capsids into and along the neurites and axons of mouse CAD cells and rat embryonic primary cortical neurons, consistent with egress via the married pathway. In contrast, transport of HSV-1 capsids was unaffected in the neurites of human SK-N-SH neuroblastoma cells, consistent with the separate mechanism. Unexpectedly, PRV (generally thought to utilize the married pathway) also appeared to employ the separate mechanism in SK-N-SH cells. We propose that apparent differences in the methods of HSV-1 and PRV egress are more likely a reflection of the host neuron in which transport is studied rather than true biological differences between the viruses themselves. IMPORTANCE Alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), are pathogens of the nervous system. They replicate in the nerve cell body and then travel great distances along axons to reach nerve termini and spread to adjacent epithelial cells; however, key aspects of how these viruses travel along axons remain controversial. Here, we test two alternative mechanisms for transport, the married and separate models, by blocking envelope assembly, a critical step in viral egress. When we arrest formation of the viral envelope using a mutated component of the cellular ESCRT apparatus, we find that entry of viral particles into axons is blocked in some types of neurons but not others. This approach allows us to determine whether envelope assembly occurs prior to entry of viruses into axons or afterwards and, thus, to distinguish between the alternative models for viral transport.


Assuntos
Alphaherpesvirinae , Complexos Endossomais de Distribuição Requeridos para Transporte , Herpesvirus Humano 1 , Herpesvirus Suídeo 1 , Neurônios , Alphaherpesvirinae/metabolismo , Animais , Axônios/virologia , Linhagem Celular Tumoral , Células Cultivadas , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Humanos , Camundongos , Neurônios/virologia , Ratos , Montagem de Vírus/fisiologia , Internalização do Vírus
2.
Nature ; 599(7886): 662-666, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34789877

RESUMO

Neurotropic alphaherpesviruses initiate infection in exposed mucosal tissues and, unlike most viruses, spread rapidly to sensory and autonomic nerves where life-long latency is established1. Recurrent infections arise sporadically from the peripheral nervous system throughout the life of the host, and invasion of the central nervous system may occur, with severe outcomes2. These viruses directly recruit cellular motors for transport along microtubules in nerve axons, but how the motors are manipulated to deliver the virus to neuronal nuclei is not understood. Here, using herpes simplex virus type I and pseudorabies virus as model alphaherpesviruses, we show that a cellular kinesin motor is captured by virions in epithelial cells, carried between cells, and subsequently used in neurons to traffic to nuclei. Viruses assembled in the absence of kinesin are not neuroinvasive. The findings explain a critical component of the alphaherpesvirus neuroinvasive mechanism and demonstrate that these viruses assimilate a cellular protein as an essential proviral structural component. This principle of viral assimilation may prove relevant to other virus families and offers new strategies to combat infection.


Assuntos
Herpesvirus Humano 1/metabolismo , Herpesvirus Suídeo 1/metabolismo , Cinesinas/metabolismo , Movimento , Vírion/metabolismo , Montagem de Vírus , Animais , Transporte Biológico , Capsídeo/metabolismo , Linhagem Celular , Núcleo Celular/virologia , Chlorocebus aethiops , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Neurônios/metabolismo , Neurônios/virologia , Coelhos , Suínos
3.
Viruses ; 13(8)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34452486

RESUMO

The alphaherpesviruses are pathogens of the mammalian nervous system. Initial infection is commonly at mucosal epithelia, followed by spread to, and establishment of latency in, the peripheral nervous system. During productive infection, viral gene expression, replication of the dsDNA genome, capsid assembly and genome packaging take place in the infected cell nucleus, after which mature nucleocapsids emerge into the cytoplasm. Capsids must then travel to their site of envelopment at cytoplasmic organelles, and enveloped virions need to reach the cell surface for release and spread. Transport at each of these steps requires movement of alphaherpesvirus particles through a crowded and viscous cytoplasm, and for distances ranging from several microns in epithelial cells, to millimeters or even meters during egress from neurons. To solve this challenging problem alphaherpesviruses, and their assembly intermediates, exploit microtubule- and actin-dependent cellular motors. This review focuses upon the mechanisms used by alphaherpesviruses to recruit kinesin, myosin and dynein motors during assembly and egress.


Assuntos
Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Dineínas/metabolismo , Cinesinas/metabolismo , Miosinas/metabolismo , Montagem de Vírus , Liberação de Vírus , Actinas/metabolismo , Alphaherpesvirinae/patogenicidade , Capsídeo/metabolismo , Dineínas/genética , Interações entre Hospedeiro e Microrganismos , Humanos , Cinesinas/genética , Microtúbulos/metabolismo , Miosinas/genética , Vírion/metabolismo
4.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32825127

RESUMO

Herpes simplex virus type 1 (HSV-1) is a structurally complex enveloped dsDNA virus that has evolved to replicate in human neurons and epithelia. Viral gene expression, DNA replication, capsid assembly, and genome packaging take place in the infected cell nucleus, which mature nucleocapsids exit by envelopment at the inner nuclear membrane then de-envelopment into the cytoplasm. Once in the cytoplasm, capsids travel along microtubules to reach, dock, and envelope at cytoplasmic organelles. This generates mature infectious HSV-1 particles that must then be sorted to the termini of sensory neurons, or to epithelial cell junctions, for spread to uninfected cells. The focus of this review is upon our current understanding of the viral and cellular molecular machinery that enables HSV-1 to travel within infected cells during egress and to manipulate cellular organelles to construct its envelope.


Assuntos
Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Envelope Viral/metabolismo , Liberação de Vírus , Animais , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Patógeno , Humanos
5.
PLoS Pathog ; 16(6): e1008597, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32511265

RESUMO

During infection of neurons by alphaherpesviruses including Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) viral nucleocapsids assemble in the cell nucleus, become enveloped in the cell body then traffic into and down axons to nerve termini for spread to adjacent epithelia. The viral membrane protein US9p and the membrane glycoprotein heterodimer gE/gI play critical roles in anterograde spread of both HSV-1 and PRV, and several models exist to explain their function. Biochemical studies suggest that PRV US9p associates with the kinesin-3 motor KIF1A in a gE/gI-stimulated manner, and the gE/gI-US9p complex has been proposed to recruit KIF1A to PRV for microtubule-mediated anterograde trafficking into or along the axon. However, as loss of gE/gI-US9p essentially abolishes delivery of alphaherpesviruses to the axon it is difficult to determine the microtubule-dependent trafficking properties and motor-composition of Δ(gE/gI-US9p) particles. Alternatively, studies in HSV-1 have suggested that gE/gI and US9p are required for the appearance of virions in the axon because they act upstream, to help assemble enveloped virions in the cell body. We prepared Δ(gE/gI-US9p) mutant, and control parental PRV particles from differentiated cultured neuronal or porcine kidney epithelial cells and quantitated the efficiency of virion assembly, the properties of microtubule-dependent transport and the ability of viral particles to recruit kinesin motors. We find that loss of gE/gI-US9p has no significant effect upon PRV particle assembly but leads to greatly diminished plus end-directed traffic, and enhanced minus end-directed and bidirectional movement along microtubules. PRV particles prepared from infected differentiated mouse CAD neurons were found to be associated with either kinesin KIF1A or kinesin KIF5C, but not both. Loss of gE/gI-US9p resulted in failure to recruit KIF1A and KF5C, but did not affect dynein binding. Unexpectedly, while KIF5C was expressed in undifferentiated and differentiated CAD neurons it was only found associated with PRV particles prepared from differentiated cells.


Assuntos
Herpesvirus Suídeo 1 , Peptídeos e Proteínas de Sinalização Intracelular , Cinesinas/metabolismo , Lipoproteínas , Microtúbulos/metabolismo , Pseudorraiva , Proteínas do Envelope Viral , Proteínas Virais , Liberação de Vírus , Animais , Transporte Biológico Ativo , Linhagem Celular , Deleção de Genes , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinesinas/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Microtúbulos/genética , Microtúbulos/virologia , Pseudorraiva/genética , Pseudorraiva/metabolismo , Pseudorraiva/patologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
6.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748394

RESUMO

Capsid envelopment during assembly of the neurotropic herpesviruses herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV) in the infected cell cytoplasm is thought to involve the late-acting cellular ESCRT (endosomal sorting complex required for transport) components ESCRT-III and VPS4 (vacuolar protein sorting 4). However, HSV-1, unlike members of many other families of enveloped viruses, does not appear to require the ESCRT-I subunit TSG101 or the Bro1 domain-containing protein ALIX (Alg-2-interacting protein X) to recruit and activate ESCRT-III. Alternative cellular factors that are known to be capable of regulating ESCRT-III function include the ESCRT-II complex and other members of the Bro1 family. We therefore used small interfering RNA (siRNA) to knock down the essential ESCRT-II subunit EAP20/VPS25 (ELL-associated protein 20/vacuolar protein sorting 25) and the Bro1 proteins HD-PTP (His domain-containing protein tyrosine phosphatase) and BROX (Bro1 domain and CAAX motif containing). We demonstrated reductions in levels of the targeted proteins by Western blotting and used quantitative microscopic assays to confirm loss of ESCRT-II and HD-PTP function. We found that in single-step replication experiments, the final yields of HSV-1 were unchanged following loss of EAP20, HD-PTP, or BROX.IMPORTANCE HSV-1 is a pathogen of the human nervous system that uses its own virus-encoded proteins and the normal cellular ESCRT machinery to drive the construction of its envelope. How HSV-1 structural proteins interact with ESCRT components and which subsets of cellular ESCRT proteins are utilized by the virus remain largely unknown. Here, we demonstrate that an essential component of the ESCRT-II complex and two ESCRT-associated Bro1 proteins are dispensable for HSV-1 replication.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Capsídeo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Endossomos/metabolismo , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Ligação Proteica/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/genética , Interferência de RNA , Proteínas de Transporte Vesicular/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
7.
Viruses ; 11(12)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31861082

RESUMO

The Alphaherpesvirinae include the neurotropic pathogens herpes simplex virus and varicella zoster virus of humans and pseudorabies virus of swine. These viruses establish lifelong latency in the nuclei of peripheral ganglia, but utilize the peripheral tissues those neurons innervate for productive replication, spread, and transmission. Delivery of virions from replicative pools to the sites of latency requires microtubule-directed retrograde axonal transport from the nerve terminus to the cell body of the sensory neuron. As a corollary, during reactivation newly assembled virions must travel along axonal microtubules in the anterograde direction to return to the nerve terminus and infect peripheral tissues, completing the cycle. Neurotropic alphaherpesviruses can therefore exploit neuronal microtubules and motors for long distance axonal transport, and alternate between periods of sustained plus end- and minus end-directed motion at different stages of their infectious cycle. This review summarizes our current understanding of the molecular details by which this is achieved.


Assuntos
Alphaherpesvirinae/fisiologia , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Interações Hospedeiro-Patógeno , Microtúbulos/metabolismo , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/virologia , Alphaherpesvirinae/ultraestrutura , Animais , Axônios/metabolismo , Biomarcadores , Capsídeo/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Suscetibilidade a Doenças , Exocitose , Humanos , Estágios do Ciclo de Vida , Neurônios/metabolismo , Neurônios/virologia , Transporte Proteico
8.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996099

RESUMO

The Herpesviridae are structurally complex DNA viruses whose capsids undergo primary envelopment at the inner nuclear membrane and secondary envelopment at organelles in the cytoplasm. In both locations, there is evidence that envelope formation and scission involve the participation of multiple viral proteins and also the cellular ESCRT apparatus. It nevertheless appears that the best-understood viral strategies for ESCRT recruitment, those adopted by the retroviruses and many other families of enveloped RNA viruses, are not utilized by the Herpesviridae, at least during envelopment in the cytoplasm. Thus, although a large number of herpesvirus proteins have been assigned roles in envelopment, there is a dearth of candidates for the acquisition of the ESCRT complex and the control of envelope scission. This review summarizes our current understanding of ESCRT association by enveloped viruses, examines what is known of herpesvirus ESCRT utilization in the nucleus and cytoplasm, and identifies candidate cellular and viral proteins that could link enveloping herpesviruses to cellular ESCRT components.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Herpesviridae/crescimento & desenvolvimento , Herpesviridae/metabolismo , Capsídeo/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Montagem de Vírus , Liberação de Vírus
9.
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
10.
J Virol ; 90(16): 7257-7267, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27252536

RESUMO

UNLABELLED: UL36p (VP1/2) is the largest protein encoded by herpes simplex virus 1 (HSV-1) and resides in the innermost layer of tegument, the complex protein layer between the capsid and envelope. UL36p performs multiple functions in the HSV life cycle, including a critical but unknown role in capsid cytoplasmic envelopment. We tested whether UL36p is essential for envelopment because it is required to engage capsids with the cellular ESCRT/Vps4 apparatus. A green fluorescent protein (GFP)-fused form of the dominant negative ATPase Vps4-EQ was used to irreversibly tag ESCRT envelopment sites during infection by UL36p-expressing and UL36-null HSV strains. Using fluorescence microscopy and scanning electron microscopy, we quantitated capsid/Vps4-EQ colocalization and examined the ultrastructure of the corresponding viral assembly intermediates. We found that loss of UL36p resulted in a two-thirds reduction in the efficiency of capsid/Vps4-EQ association but that the remaining UL36p-null capsids were still able to engage the ESCRT envelopment apparatus. It appears that although UL36p helps to couple HSV capsids to the ESCRT pathway, this is likely not the sole reason for its absolute requirement for envelopment. IMPORTANCE: Envelopment of the HSV capsid is essential for the assembly of an infectious virion and requires the complex interplay of a large number of viral and cellular proteins. Critical to envelope assembly is the virally encoded protein UL36p, whose function is unknown. Here we test the hypothesis that UL36p is essential for the recruitment of cellular ESCRT complexes, which are also known to be required for envelopment.


Assuntos
Capsídeo/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus/fisiologia , ATPases Associadas a Diversas Atividades Celulares , Animais , Western Blotting , Chlorocebus aethiops , Citoplasma , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Herpes Simples/genética , Herpes Simples/metabolismo , Humanos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ATPases Vacuolares Próton-Translocadoras/genética , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/genética , Vírion/fisiologia , Replicação Viral
11.
J Virol ; 89(22): 11372-82, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26339048

RESUMO

UNLABELLED: UL36p (VP1/2) is the largest protein encoded by herpes simplex virus 1 (HSV-1) and resides in the innermost layer of the viral tegument, lying between the capsid and the envelope. UL36p performs multiple functions in the HSV life cycle, including an essential role in cytoplasmic envelopment. We earlier described the isolation of a virion-associated cytoplasmic membrane fraction from HSV-infected cells. Biochemical and ultrastructural analyses showed that the organelles in this buoyant fraction contain enveloped infectious HSV particles in their lumens and naked capsids docked to their cytoplasmic surfaces. These organelles can also recruit molecular motors and transport their cargo virions along microtubules in vitro. Here we examine the properties of these HSV-associated organelles in the absence of UL36p. We find that while capsid envelopment is clearly defective, a subpopulation of capsids nevertheless still associate with the cytoplasmic faces of these organelles. The existence of these capsid-membrane structures was confirmed by subcellular fractionation, immunocytochemistry, lipophilic dye fluorescence microscopy, thin-section electron microscopy, and correlative light and electron microscopy. We conclude that capsid-membrane binding can occur in the absence of UL36p and propose that this association may precede the events of UL36p-driven envelopment. IMPORTANCE: Membrane association and envelopment of the HSV capsid are essential for the assembly of an infectious virion. Envelopment involves the complex interplay of a large number of viral and cellular proteins; however, the function of most of them is unknown. One example of this is the viral protein UL36p, which is clearly essential for envelopment but plays a poorly understood role. Here we demonstrate that organelles utilized for HSV capsid envelopment still accumulate surface-bound capsids in the absence of UL36p. We propose that UL36p-independent binding of capsids to organelles occurs prior to the function of UL36p in capsid envelopment.


Assuntos
Proteínas do Capsídeo/metabolismo , Herpesvirus Humano 1/fisiologia , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus/fisiologia , Animais , Capsídeo/metabolismo , Linhagem Celular , Chlorocebus aethiops , Herpesvirus Humano 1/patogenicidade , Humanos , Células Vero , Proteínas Virais/genética
12.
J Virol ; 89(13): 6673-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25878109

RESUMO

UNLABELLED: Dynasore, a small-molecule inhibitor of the GTPase activity of dynamin, inhibits the entry of several viruses, including herpes simplex virus (HSV), but its impact on other steps in the viral life cycle has not been delineated. The current study was designed to test the hypothesis that dynamin is required for viral protein trafficking and thus has pleiotropic inhibitory effects on HSV infection. Dynasore inhibited HSV-1 and HSV-2 infection of human epithelial and neuronal cells, including primary genital tract cells and human fetal neurons and astrocytes. Similar results were obtained when cells were transfected with a plasmid expressing dominant negative dynamin. Kinetic studies demonstrated that dynasore reduced the number of viral capsids reaching the nuclear pore if added at the time of viral entry and that, when added as late as 8 h postentry, dynasore blocked the transport of newly synthesized viral proteins from the nucleus to the cytosol. Proximity ligation assays demonstrated that treatment with dynasore prevented the colocalization of VP5 and dynamin. This resulted in a reduction in the number of viral capsids isolated from sucrose gradients. Fewer capsids were observed by electron microscopy in dynasore-treated cells than in control-treated cells. There were also reductions in infectious progeny released into culture supernatants and in cell-to-cell spread. Together, these findings suggest that targeting dynamin-HSV interactions may provide a new strategy for HSV treatment and prevention. IMPORTANCE: HSV infections remain a global health problem associated with significant morbidity, particularly in neonates and immunocompromised hosts, highlighting the need for novel approaches to treatment and prevention. The current studies indicate that dynamin plays a role in multiple steps in the viral life cycle and provides a new target for antiviral therapy. Dynasore, a small-molecule inhibitor of dynamin, has pleiotropic effects on HSV-1 and HSV-2 infection and impedes viral entry, trafficking of viral proteins, and capsid formation.


Assuntos
Antirretrovirais/metabolismo , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 2/fisiologia , Hidrazonas/metabolismo , Proteínas Virais/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Centrifugação com Gradiente de Concentração , Dinaminas/metabolismo , Células Epiteliais/virologia , Humanos , Microscopia Eletrônica de Transmissão , Neurônios/virologia , Transporte Proteico/efeitos dos fármacos
13.
J Virol ; 88(24): 14467-78, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25297998

RESUMO

UNLABELLED: Herpes simplex virus (HSV) and, as reported here, pseudorabies virus (PRV) utilize the ESCRT apparatus to drive cytoplasmic envelopment of their capsids. Here, we demonstrate that blocking ESCRT-mediated envelopment using the dominant-negative inhibitor Vps4A-EQ (Vps4A in which glutamate [E] at position 228 in the ATPase active site is replaced by a glutamine [Q]) reduced the ability of HSV and PRV particles to subsequently traffic along microtubules in vitro. HSV and PRV capsid-associated particles with bound green fluorescent protein (GFP)-labeled Vps4A-EQ were readily detected by fluorescence microscopy in cytoplasmic extracts of infected cells. These Vps4A-EQ-associated capsid-containing particles bound to microtubules in vitro but were unable to traffic along them. Using a PRV strain expressing a fluorescent capsid and a fluorescently tagged form of the envelope protein gD, we found that similar numbers of gD-positive and gD-negative capsid-associated particles accumulated in cytoplasmic extracts under our conditions. Both classes of PRV particle bound to microtubules in vitro with comparable efficiency, and similar results were obtained for HSV using anti-gD immunostaining. The gD-positive and gD-negative PRV capsids were both capable of trafficking along microtubules in vitro; however, motile gD-positive particles were less numerous and their trafficking was more sensitive to the inhibitory effects of Vps4A-EQ. We discuss our data in the context of microtubule-mediated trafficking of naked and enveloped alphaherpesvirus capsids. IMPORTANCE: The alphaherpesviruses include several important human pathogens. These viruses utilize microtubule-mediated transport to travel through the cell cytoplasm; however, the molecular mechanisms of trafficking are not well understood. In this study, we have used a cell-free system to examine the requirements for microtubule trafficking and have attempted to distinguish between the movement of so-called "naked" and membrane-associated cytoplasmic alphaherpesvirus capsids.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Herpesvirus Suídeo 1/fisiologia , Interações Hospedeiro-Patógeno , Microtúbulos/virologia , Simplexvirus/fisiologia , Montagem de Vírus , Animais , Transporte Biológico , Chlorocebus aethiops , Células Vero
14.
Intervirology ; 56(1): 50-4, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22854264

RESUMO

BACKGROUND AND OBJECTIVE: During herpesvirus envelopment capsids, tegument polypeptides and membrane proteins assemble at the site of budding, and a cellular lipid bilayer becomes refashioned into a spherical envelope. A web of interactions between tegument proteins and the cytoplasmic tails of viral glycoproteins play a critical role in this process. We have previously demonstrated that for herpes simplex virus (HSV)-1 the cytoplasmic tail of glycoprotein H (gH) binds the tegument protein VP16. The HSV and pseudorabies virus (PRV) genomes are essentially collinear, and individual gene products show significant sequence homology. However, the demarcation of function often differs between PRV and HSV proteins. The goal of this study was to determine whether PRV gH and VP16 interact in a manner similar to their homologs in HSV. METHODS: A fusion protein pull-down assay was performed in which a PRV gH cytoplasmic tail-glutathione S-transferase fusion protein, bound to glutathione-Sepharose beads, was incubated with PRV-infected cell cytosol, washed and subjected to Western blot analysis using anti-PRV VP16 antisera. RESULTS: Western blots indicate that PRV VP16 does not specifically bind to the PRV gH tail. CONCLUSION: Our results highlight that, despite the relatively close evolutionary relationship between HSV and PRV, there are significant differences in their protein interactions that drive envelopment.


Assuntos
Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/fisiologia , Proteínas Estruturais Virais/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Herpesvirus Suídeo 1/metabolismo , Herpesvirus Suídeo 1/ultraestrutura , Dados de Sequência Molecular , Mapeamento de Interação de Proteínas , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo
15.
J Virol ; 83(10): 4757-65, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19279117

RESUMO

Herpes simplex virus 1 (HSV-1) nucleocapsids exit the nucleus by budding into the inner nuclear membrane, where they exist briefly as primary enveloped virions. These virus particles subsequently fuse their envelopes with the outer nuclear membrane, permitting nucleocapsids to then enter the cytoplasm and complete assembly. We have developed a method to isolate primary enveloped virions from HSV-1-infected cells and subjected the primary enveloped virion preparation to MALDI-MS/MS (matrix-assisted laser desorption ionization-tandem mass spectrometry) analyses. We identified most capsid proteins, a tegument protein (VP22), a glycoprotein (gD), and a cellular protein (annexin A2) in the primary enveloped virion preparation. We determined that annexin A2 does not play an essential role in infection under our experimental conditions. Elucidating the structure and biochemical properties of this unique virus assembly intermediate will provide new insights into HSV-1 biology.


Assuntos
Proteínas do Capsídeo/isolamento & purificação , Herpesvirus Humano 1/isolamento & purificação , Proteômica , Proteínas do Envelope Viral/isolamento & purificação , Vírion/isolamento & purificação , Animais , Anexina A2/isolamento & purificação , Células COS , Células Cultivadas , Chlorocebus aethiops , DNA Viral/análise , Herpesvirus Humano 1/metabolismo , Membrana Nuclear/virologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Vírion/metabolismo
16.
J Virol ; 82(15): 7388-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18495763

RESUMO

Microtubule-mediated anterograde transport is essential for the transport of herpes simplex virus type 1 (HSV-1) along axons, yet little is known regarding the mechanism and the machinery required for this process. Previously, we were able to reconstitute anterograde transport of HSV-1 on microtubules in an in vitro microchamber assay. Here we report that the large tegument protein UL36p is essential for this trafficking. Using a fluorescently labeled UL36 null HSV-1 strain, KDeltaUL36GFP, we found that it is possible to isolate a membrane-associated population of this virus. Although these viral particles contained normal amounts of tegument proteins VP16, vhs, and VP22, they displayed a 3-log decrease in infectivity and showed a different morphology compared to UL36p-containing virions. Membrane-associated KDeltaUL36GFP also displayed a slightly decreased binding to microtubules in our microchamber assay and a two-thirds decrease in the frequency of motility. This decrease in binding and motility was restored when UL36p was supplied in trans by a complementing cell line. These findings suggest that UL36p is necessary for HSV-1 anterograde transport.


Assuntos
Herpesvirus Humano 1/fisiologia , Microtúbulos/virologia , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Deleção de Genes , Genes Reporter , Teste de Complementação Genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Herpesvirus Humano 1/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão , Coloração e Rotulagem/métodos , Proteínas Virais/genética , Vírion/ultraestrutura , Replicação Viral
17.
J Virol ; 80(20): 10117-27, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17005689

RESUMO

Assembly of herpes simplex viruses (HSV) is a poorly understood process involving multiple redundant interactions between large number of tegument and envelope proteins. We have previously shown (G. E. Lee, G. A. Church, and D. W. Wilson, J. Virol. 77:2038-2045, 2003) that the virion host shutoff (Vhs) tegument protein is largely insoluble in HSV-infected cells and is also stably associated with membranes. Here we demonstrate that both insolubility and stable membrane binding are stimulated during the course of an HSV infection. Furthermore, we have found that the amino-terminal 42 residues of Vhs are sufficient to mediate membrane association and tegument incorporation when fused to a green fluorescent protein (GFP) reporter. Particle incorporation correlates with sorting to cytoplasmic punctate structures that may correspond to sites of HSV assembly. We conclude that the amino terminus of Vhs mediates targeting to sites of HSV assembly and to the viral tegument.


Assuntos
Membrana Celular/metabolismo , Herpesvirus Humano 1/fisiologia , Proteínas Virais/química , Proteínas Virais/fisiologia , Montagem de Vírus , Animais , Células COS , Chlorocebus aethiops , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Herpesvirus Humano 1/genética , Microscopia Imunoeletrônica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/análise , Ribonucleases , Solubilidade , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo
18.
J Virol ; 80(9): 4264-75, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611885

RESUMO

Microtubule-mediated anterograde transport of herpes simplex virus (HSV) from the neuronal cell body to the axon terminal is crucial for the spread and transmission of the virus. It is therefore of central importance to identify the cellular and viral factors responsible for this trafficking event. In previous studies, we isolated HSV-containing cytoplasmic organelles from infected cells and showed that they represent the first and only destination for HSV capsids after they emerge from the nucleus. In the present study, we tested whether these cytoplasmic compartments were capable of microtubule-dependent traffic. Organelles containing green fluorescent protein-labeled HSV capsids were isolated and found to be able to bind rhodamine-labeled microtubules polymerized in vitro. Following the addition of ATP, the HSV-associated organelles trafficked along the microtubules, as visualized by time lapse microscopy in an imaging microchamber. The velocity and processivity of trafficking resembled those seen for neurotropic herpesvirus traffic in living axons. The use of motor-specific inhibitors indicated that traffic was predominantly kinesin mediated, consistent with the reconstitution of anterograde traffic. Immunocytochemical studies revealed that the majority of HSV-containing organelles attached to the microtubules contained the trans-Golgi network marker TGN46. This simple, minimal reconstitution of microtubule-mediated anterograde traffic should facilitate and complement molecular analysis of HSV egress in vivo.


Assuntos
Microtúbulos/metabolismo , Simplexvirus/metabolismo , Transporte Biológico , Biomarcadores , Capsídeo/metabolismo , Linhagem Celular Tumoral , Citoplasma/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Microtúbulos/ultraestrutura , Ligação Proteica , Simplexvirus/ultraestrutura , Vírion/metabolismo
19.
J Virol ; 79(10): 6134-41, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15857998

RESUMO

Critical events in the life cycle of herpes simplex virus (HSV) are the binding of cytoplasmic capsids to cellular organelles and subsequent envelopment. Work from several laboratories suggests that these events occur as a result of a network of partially redundant interactions among the capsid surface, tegument components, and cytoplasmic tails of virally encoded glycoproteins. Consistent with this model, we previously showed that tegument protein VP16 can specifically interact with the cytoplasmic tail of envelope protein gH in vitro and in vivo when fused to glutathione S-transferase and to green fluorescent protein, respectively. In both instances, this association was strikingly temperature dependent: binding occurred only at 37 degrees C and not at lower temperatures. Here we demonstrate that virally expressed full-length gH and VP16 can be coimmunoprecipitated from HSV-infected cells and that this association is also critically dependent upon the physiological temperature. To investigate the basis of this temperature requirement, we performed one- and two-dimensional nuclear magnetic resonance spectroscopy on peptides with the sequence of the gH tail. We found that the gH tail is disorganized at temperatures permissive for binding but becomes structured at lower temperatures. Furthermore, a mutated tail unable to adopt this rigid conformation binds VP16 even at 4 degrees C. We hypothesize that the gH tail is unstructured under physiological conditions in order to maximize the number of potential tegument partners with which it may associate. Being initially disordered, the gH tail may adopt one of several induced conformations as it associates with VP16 or alternative components of the tegument, maximizing redundancy during particle assembly.


Assuntos
Citoplasma/metabolismo , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Ligação Proteica , Proteínas do Envelope Viral/metabolismo , Animais , Células COS , Chlorocebus aethiops , Herpesvirus Humano 1/metabolismo , Modelos Químicos , Temperatura , Células Vero , Proteínas do Envelope Viral/química , Montagem de Vírus
20.
J Gen Virol ; 86(Pt 2): 253-261, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15659744

RESUMO

Herpes simplex virus (HSV) capsids assemble, mature and package their viral genome in the nucleoplasm. They then exit the nucleus into the cytoplasm, where they acquire their final tegument and envelope. The molecular mechanism of cytoplasmic envelopment is unclear, but evidence suggests that the viral glycoprotein tails play an important role in the recruitment of tegument and capsids at the final envelopment site. However, due to redundancy in protein-protein interactions among the viral glycoproteins, genetic analysis of the role of individual glycoproteins in assembly has been difficult. To overcome this problem, a glutathione S-transferase fusion protein-binding assay was used in this study to test the interaction between the cytoplasmic tail of one specific viral glycoprotein, gD, and tegument proteins. The study demonstrated that the 38 kDa tegument protein VP22 bound specifically to the gD tail. This association was dependent on arginine and lysine residues at positions 5 and 6 in the gD tail. In addition, HSV-1 capsids bound the gD tail and exhibited a similar sequence dependence. It is concluded that VP22 may serve as a linker protein, mediating the interaction of the HSV capsid with gD.


Assuntos
Capsídeo/metabolismo , Simplexvirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Arginina/genética , Células COS , Capsídeo/ultraestrutura , Chlorocebus aethiops , Citoplasma/virologia , Lisina/genética , Dados de Sequência Molecular , Peso Molecular , Mutação , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas do Envelope Viral/genética , Proteínas Estruturais Virais/química , Montagem de Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA