Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Biochem Soc Trans ; 50(1): 95-105, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35076655

RESUMO

Viruses can be enveloped or non-enveloped, and require a host cell to replicate and package their genomes into new virions to infect new cells. To accomplish this task, viruses hijack the host-cell machinery to facilitate their replication by subverting and manipulating normal host cell function. Enveloped viruses can have severe consequences for human health, causing various diseases such as acquired immunodeficiency syndrome (AIDS), seasonal influenza, COVID-19, and Ebola virus disease. The complex arrangement and pleomorphic architecture of many enveloped viruses pose a challenge for the more widely used structural biology techniques, such as X-ray crystallography. Cryo-electron tomography (cryo-ET), however, is a particularly well-suited tool for overcoming the limitations associated with visualizing the irregular shapes and morphology enveloped viruses possess at macromolecular resolution. The purpose of this review is to explore the latest structural insights that cryo-ET has revealed about enveloped viruses, with particular attention given to their architectures, mechanisms of entry, replication, assembly, maturation and egress during infection. Cryo-ET is unique in its ability to visualize cellular landscapes at 3-5 nanometer resolution. Therefore, it is the most suited technique to study asymmetric elements and structural rearrangements of enveloped viruses during infection in their native cellular context.


Assuntos
Vírus/ultraestrutura , Microscopia Crioeletrônica , Ebolavirus/metabolismo , Ebolavirus/ultraestrutura , Tomografia com Microscopia Eletrônica , HIV-1/metabolismo , HIV-1/ultraestrutura , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Humanos , SARS-CoV-2/metabolismo , SARS-CoV-2/ultraestrutura , Vírus/metabolismo
2.
mBio ; 12(2)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33727359

RESUMO

Herpes simplex virus 1 (HSV-1) requires seven proteins to package its genome through a vertex in its capsid, one of which is the portal protein, pUL6. The portal protein is also thought to facilitate assembly of the procapsid. While the portal has been visualized in mature capsids, we aimed to elucidate its role in the assembly and maturation of procapsids using cryo-electron tomography (cryoET). We identified the portal vertex in individual procapsids, calculated a subtomogram average, and compared that with the portal vertex in empty mature capsids (A-capsids). The resulting maps show the portal on the interior surface with its narrower end facing outwards, while maintaining close contact with the capsid shell. In the procapsid, the portal is embedded in the underlying scaffold, suggesting that assembly involves a portal-scaffold complex. During maturation, the capsid shell angularizes with a corresponding outward movement of the vertices. We found that in A-capsids, the portal translocates outward further than the adjacent capsomers and strengthens its contacts with the capsid shell. Our methodology also allowed us to determine the number of portal vertices in each capsid, with most having one per capsid, but some none or two, and rarely three. The predominance of a single portal per capsid supports facilitation of the assembly of the procapsid.IMPORTANCE Herpes simplex virus 1 (HSV-1) infects a majority of humans, causing mostly mild disease but in some cases progressing toward life-threatening encephalitis. Understanding the life cycle of the virus is important to devise countermeasures. Production of the virion starts with the assembly of an icosahedral procapsid, which includes DNA packaging proteins at a vertex, one of which is the dodecameric portal protein. The procapsid then undergoes maturation and DNA packaging through the portal, driven by a terminase complex. We used cryo-electron tomography to visualize the portal in procapsids and compare them to mature empty capsids. We found the portal located inside one vertex interacting with the scaffold protein in the procapsid. On maturation, the scaffold is cleaved and dissociates, the capsid angularizes, and the portal moves outward, interacting closely with the capsid shell. These transformations may provide a basis for the development of drugs to prevent HSV-1 infections.


Assuntos
Capsídeo/metabolismo , Capsídeo/ultraestrutura , Microscopia Crioeletrônica/métodos , Tomografia com Microscopia Eletrônica/métodos , Herpesvirus Humano 1/ultraestrutura , Proteínas Virais/metabolismo , Montagem de Vírus , Proteínas do Capsídeo/genética , Herpesvirus Humano 1/metabolismo
3.
J Biol Chem ; 296: 100236, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33380421

RESUMO

Herpesviruses are large and complex viruses that have a long history of coevolution with their host species. One important factor in the virus-host interaction is the alteration of intracellular morphology during viral replication with critical implications for viral assembly. However, the details of this remodeling event are not well understood, in part because insufficient tools are available to deconstruct this highly heterogeneous process. To provide an accurate and reliable method of investigating the spatiotemporal dynamics of virus-induced changes to cellular architecture, we constructed a dual-fluorescent reporter virus that enabled us to classify four distinct stages in the infection cycle of herpes simplex virus-1 at the single cell level. This timestamping method can accurately track the infection cycle across a wide range of multiplicities of infection. We used high-resolution fluorescence microscopy analysis of cellular structures in live and fixed cells in concert with our reporter virus to generate a detailed and chronological overview of the spatial and temporal reorganization during viral replication. The highly orchestrated and striking relocation of many organelles around the compartments of secondary envelopment during transition from early to late gene expression suggests that the reshaping of these compartments is essential for virus assembly. We furthermore find that accumulation of HSV-1 capsids in the cytoplasm is accompanied by fragmentation of the Golgi apparatus with potential impact on the late steps of viral assembly. We anticipate that in the future similar tools can be systematically applied for the systems-level analysis of intracellular morphology during replication of other viruses.


Assuntos
Complexo de Golgi/genética , Herpesvirus Humano 1/genética , Microscopia de Fluorescência , Replicação Viral/genética , Animais , Capsídeo/ultraestrutura , Chlorocebus aethiops , Citoplasma/genética , Citoplasma/ultraestrutura , Citoplasma/virologia , Genes Reporter/genética , Complexo de Golgi/ultraestrutura , Complexo de Golgi/virologia , Herpesvirus Humano 1/ultraestrutura , Humanos , Análise de Célula Única , Análise Espaço-Temporal , Células Vero , Montagem de Vírus/genética
4.
Sci Rep ; 10(1): 17596, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-33077791

RESUMO

Cryo electron microscopy (cryo-EM), a key method for structure determination involves imaging purified material embedded in vitreous ice. Images are then computationally processed to obtain three-dimensional structures approaching atomic resolution. There is increasing interest in extending structural studies by cryo-EM into the cell, where biological structures and processes may be imaged in context. The limited penetrating power of electrons prevents imaging of thick specimens (> 500 nm) however. Cryo-sectioning methods employed to overcome this are technically challenging, subject to artefacts or involve specialised and costly equipment. Here we describe the first structure of herpesvirus capsids determined by sub-tomogram averaging from nuclei of eukaryotic cells, achieved by cryo-electron tomography (cryo-ET) of re-vitrified cell sections prepared using the Tokuyasu method. Our reconstructions confirm that the capsid associated tegument complex is present on capsids prior to nuclear egress. We demonstrate that this method is suited to both 3D structure determination and correlative light/electron microscopy, thus expanding the scope of cryogenic cellular imaging.


Assuntos
Capsídeo/ultraestrutura , Núcleo Celular/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Vírion/ultraestrutura , Animais , Linhagem Celular , Cricetinae , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica
5.
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
6.
Methods Mol Biol ; 2060: 343-354, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617189

RESUMO

Transmission electron microscopy (TEM) provides the resolution necessary to identify both viruses and subcellular components of cells infected with many types of viruses, including herpes simplex virus. Recognized as a powerful tool in both diagnostic and research-based virology laboratories, TEM has made possible the identification of new viruses and has contributed to the elucidation of virus life cycle and virus-host cell interaction.While there are many sample preparation techniques for TEM, conventional processing using chemical fixation and resin embedding remains a useful technique, available in virtually all EM laboratories, for studying virus/cell ultrastructure. In this chapter, we describe the preparation of herpes simplex virus infected primary neurons, grown on plastic coverslips, to allow for sectioning of neurons and axons in their growth plane. This technique allows for TEM examination of cell bodies, axons, growth cones and varicosities, providing powerful insights into virus-cell interaction.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Microscopia Eletrônica de Transmissão , Neurônios , Herpes Simples/metabolismo , Herpes Simples/patologia , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Humanos , Neurônios/metabolismo , Neurônios/ultraestrutura , Neurônios/virologia
7.
Methods Mol Biol ; 2060: 355-364, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617190

RESUMO

Transmission immunoelectron microscopy allows for the ultrastructural detection and localization of herpes simplex virus-1 (HSV-1) particles and viral proteins within the infected cell and their relation to the cell cytoskeleton, cellular proteins, vesicles, membranes, and organelles. For the successful application of immunoelectron microscopy, preservation of cell ultrastructure and of epitope antigenicity is essential during sample preparation. This chapter describes the use of chemical fixation followed by rapid cooling of HSV-1 infected sensory neurons in the presence of sucrose as a cryoprotectant to achieve optimal preservation of cell morphology and the use of freeze substitution and resin polymerization at low temperatures for preservation of protein antigenicity. In order to examine HSV-1 infection in the specialized compartments of the neurons (cell body, axons, and growth cones), neurons cultured on plastic coverslips are flat embedded prior to resin polymerization. Overall, this method allows for the ultrathin sectioning and immunogold labeling of the neurons and their axons in growth plane.


Assuntos
Gânglios Espinais , Herpes Simples , Herpesvirus Humano 1 , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Neurônios , Animais , Galinhas , Gânglios Espinais/metabolismo , Gânglios Espinais/ultraestrutura , Gânglios Espinais/virologia , Herpes Simples/metabolismo , Herpes Simples/patologia , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/ultraestrutura , Neurônios/virologia , Ratos
8.
PLoS Pathog ; 15(12): e1008209, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790506

RESUMO

The processes of cell attachment and membrane fusion of Herpes Simplex Virus 1 involve many different envelope glycoproteins. Viral proteins gC and gD bind to cellular receptors. Upon binding, gD activates the gH/gL complex which in turn activates gB to trigger membrane fusion. Thus, these proteins must be located at the point of contact between cellular and viral envelopes to interact and allow fusion. Using super-resolution microscopy, we show that gB, gH/gL and most of gC are distributed evenly round purified virions. In contrast, gD localizes essentially as clusters which are distinct from gB and gH/gL. Upon cell binding, we observe that all glycoproteins, including gD, have a similar ring-like pattern, but the diameter of these rings was significantly smaller than those observed on cell-free viruses. We also observe that contrary to cell-free particles, gD mostly colocalizes with other glycoproteins on cell-bound particles. The differing patterns of localization of gD between cell-free and cell-bound viruses indicates that gD can be reorganized on the viral envelope following either a possible maturation of the viral particle or its adsorption to the cell. This redistribution of glycoproteins upon cell attachment could contribute to initiate the cascade of activations leading to membrane fusion.


Assuntos
Herpesvirus Humano 1/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Linhagem Celular , Glicoproteínas/metabolismo , Glicoproteínas/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Humanos , Microscopia/métodos , Proteínas do Envelope Viral/ultraestrutura , Vírion/ultraestrutura , Ligação Viral , Internalização do Vírus
9.
Viruses ; 11(10)2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31614678

RESUMO

During lytic herpes simplex virus 1 (HSV-1) infection, the expansion of the viral replication compartments leads to an enrichment of the host chromatin in the peripheral nucleoplasm. We have shown previously that HSV-1 infection induces the formation of channels through the compacted peripheral chromatin. Here, we used three-dimensional confocal and expansion microscopy, soft X-ray tomography, electron microscopy, and random walk simulations to analyze the kinetics of host chromatin redistribution and capsid localization relative to their egress site at the nuclear envelope. Our data demonstrated a gradual increase in chromatin marginalization, and the kinetics of chromatin smoothening around the viral replication compartments correlated with their expansion. We also observed a gradual transfer of capsids to the nuclear envelope. Later in the infection, random walk modeling indicated a gradually faster transport of capsids to the nuclear envelope that correlated with an increase in the interchromatin channels in the nuclear periphery. Our study reveals a stepwise and time-dependent mechanism of herpesvirus nuclear egress, in which progeny viral capsids approach the egress sites at the nuclear envelope via interchromatin spaces.


Assuntos
Cromatina/virologia , Infecções por Herpesviridae/patologia , Herpesvirus Humano 1 , Liberação de Vírus , Animais , Linhagem Celular , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Chlorocebus aethiops , Cromatina/ultraestrutura , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/ultraestrutura , Humanos , Microscopia Eletrônica , Microscopia de Fluorescência , Tomografia por Raios X , Células Vero , Replicação Viral
10.
PLoS Biol ; 17(6): e3000316, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31199794

RESUMO

Infections with human herpesviruses are ubiquitous and a public health concern worldwide. Current treatments reduce the severity of some symptoms associated to herpetic infections but neither remove the viral reservoir from the infected host nor protect from the recurrent symptom outbreaks that characterise herpetic infections. The difficulty in therapeutically tackling these viral systems stems in part from their remarkably large proteomes and the complex networks of physical and functional associations that they tailor. This study presents our efforts to unravel the complexity of the interactome of herpes simplex virus type 1 (HSV1), the prototypical herpesvirus species. Inspired by our previous work, we present an improved and more integrative computational pipeline for the protein-protein interaction (PPI) network reconstruction in HSV1, together with a newly developed consensus clustering framework, which allowed us to extend the analysis beyond binary physical interactions and revealed a system-level layout of higher-order functional associations in the virion proteome. Additionally, the analysis provided new functional annotation for the currently undercharacterised protein pUS10. In-depth bioinformatics sequence analysis unravelled structural features in pUS10 reminiscent of those observed in some capsid-associated proteins in tailed bacteriophages, with which herpesviruses are believed to share a common ancestry. Using immunoaffinity purification (IP)-mass spectrometry (MS), we obtained additional support for our bioinformatically predicted interaction between pUS10 and the inner tegument protein pUL37, which binds cytosolic capsids, contributing to initial tegumentation and eventually virion maturation. In summary, this study unveils new, to our knowledge, insights at both the system and molecular levels that can help us better understand the complexity behind herpesvirus infections.


Assuntos
Biologia Computacional/métodos , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Animais , Capsídeo/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Bases de Dados Factuais , Herpes Simples/metabolismo , Humanos , Hidroliases/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas , Relação Estrutura-Atividade , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Vírion/metabolismo , Montagem de Vírus
11.
Nature ; 570(7760): 257-261, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31142842

RESUMO

Herpesviruses are enveloped viruses that are prevalent in the human population and are responsible for diverse pathologies, including cold sores, birth defects and cancers. They are characterized by a highly pressurized pseudo-icosahedral capsid-with triangulation number (T) equal to 16-encapsidating a tightly packed double-stranded DNA (dsDNA) genome1-3. A key process in the herpesvirus life cycle involves the recruitment of an ATP-driven terminase to a unique portal vertex to recognize, package and cleave concatemeric dsDNA, ultimately giving rise to a pressurized, genome-containing virion4,5. Although this process has been studied in dsDNA phages6-9-with which herpesviruses bear some similarities-a lack of high-resolution in situ structures of genome-packaging machinery has prevented the elucidation of how these multi-step reactions, which require close coordination among multiple actors, occur in an integrated environment. To better define the structural basis of genome packaging and organization in herpes simplex virus type 1 (HSV-1), we developed sequential localized classification and symmetry relaxation methods to process cryo-electron microscopy (cryo-EM) images of HSV-1 virions, which enabled us to decouple and reconstruct hetero-symmetric and asymmetric elements within the pseudo-icosahedral capsid. Here we present in situ structures of the unique portal vertex, genomic termini and ordered dsDNA coils in the capsid spooled around a disordered dsDNA core. We identify tentacle-like helices and a globular complex capping the portal vertex that is not observed in phages, indicative of herpesvirus-specific adaptations in the DNA-packaging process. Finally, our atomic models of portal vertex elements reveal how the fivefold-related capsid accommodates symmetry mismatch imparted by the dodecameric portal-a longstanding mystery in icosahedral viruses-and inform possible DNA-sequence recognition and headful-sensing pathways involved in genome packaging. This work showcases how to resolve symmetry-mismatched elements in a large eukaryotic virus and provides insights into the mechanisms of herpesvirus genome packaging.


Assuntos
Microscopia Crioeletrônica , Empacotamento do DNA , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestrutura , Conformação de Ácido Nucleico , Capsídeo/química , Capsídeo/ultraestrutura , DNA Viral/química , DNA Viral/ultraestrutura , Herpesvirus Humano 1/química , Modelos Moleculares , Vírion/química , Vírion/genética , Vírion/ultraestrutura
12.
Cells ; 8(2)2019 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-30717447

RESUMO

The primary envelopment/de-envelopment of Herpes viruses during nuclear exit is poorly understood. In Herpes simplex virus type-1 (HSV-1), proteins pUL31 and pUL34 are critical, while pUS3 and some others contribute; however, efficient membrane fusion may require additional host proteins. We postulated that vesicle fusion proteins present in the nuclear envelope might facilitate primary envelopment and/or de-envelopment fusion with the outer nuclear membrane. Indeed, a subpopulation of vesicle-associated membrane protein-associated protein B (VAPB), a known vesicle trafficking protein, was present in the nuclear membrane co-locating with pUL34. VAPB knockdown significantly reduced both cell-associated and supernatant virus titers. Moreover, VAPB depletion reduced cytoplasmic accumulation of virus particles and increased levels of nuclear encapsidated viral DNA. These results suggest that VAPB is an important player in the exit of primary enveloped HSV-1 virions from the nucleus. Importantly, VAPB knockdown did not alter pUL34, calnexin or GM-130 localization during infection, arguing against an indirect effect of VAPB on cellular vesicles and trafficking. Immunogold-labelling electron microscopy confirmed VAPB presence in nuclear membranes and moreover associated with primary enveloped HSV-1 particles. These data suggest that VAPB could be a cellular component of a complex that facilitates UL31/UL34/US3-mediated HSV-1 nuclear egress.


Assuntos
Núcleo Celular/metabolismo , Herpesvirus Humano 1/fisiologia , Fusão de Membrana , Proteínas de Transporte Vesicular/metabolismo , Liberação de Vírus/fisiologia , Replicação Viral/fisiologia , Animais , Núcleo Celular/ultraestrutura , Chlorocebus aethiops , Células HeLa , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/ultraestrutura , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Microssomos/metabolismo , Microssomos/ultraestrutura , Membrana Nuclear/metabolismo , Células Vero , Proteínas Virais/metabolismo , Vírion/metabolismo , Vírion/ultraestrutura
13.
J Biochem ; 164(6): 397-406, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30107464

RESUMO

Heat shock protein 90 (Hsp90) has been identified as an essential host factor for the infection and replication of several viruses, including HSV-1. Recent works have clearly shown that Hsp90 plays a role in the early stages of HSV-1 infection, including nuclear import and DNA replication. However, the role of Hsp90 in the late stages of HSV-1 infection remains unclear. In this study, we found that Hsp90 was up-regulated during late viral infection. Treatment with the Hsp90 inhibitor AT-533 significantly decreased the intracellular and extracellular virus titers, and strongly inhibited nucleocapsid egress from the nucleus. More detailed studies revealed that AT-533 inhibited the nuclear egress of the viral nucleocapsid by suppressing the expression and translocation of nuclear-associated proteins pUL31 and pUL34. In addition, we found that AT-533 hindered the assembly of virus particles possibly though affecting the localization of glycoproteins in the endoplasmic reticulum and Golgi apparatus. These results thus invoke a new role for Hsp90 in the nucleocapsid egress and viral maturation of HSV-1, and further promote the development of Hsp90 inhibitors as potential anti-HSV-1 drugs.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Núcleo Celular/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Herpesvirus Humano 1/efeitos dos fármacos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Montagem de Vírus/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Benzamidas/efeitos adversos , Benzoquinonas/efeitos adversos , Benzoquinonas/farmacologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/ultraestrutura , Cinética , Lactamas Macrocíclicas/efeitos adversos , Lactamas Macrocíclicas/farmacologia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Células Vero , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
14.
PLoS Biol ; 16(6): e2006191, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29924793

RESUMO

Herpesviruses include many important human pathogens such as herpes simplex virus, cytomegalovirus, varicella-zoster virus, and the oncogenic Epstein-Barr virus and Kaposi sarcoma-associated herpesvirus. Herpes virions contain a large icosahedral capsid that has a portal at a unique 5-fold vertex, similar to that seen in the tailed bacteriophages. The portal is a molecular motor through which the viral genome enters the capsid during virion morphogenesis. The genome also exits the capsid through the portal-vertex when it is injected through the nuclear pore into the nucleus of a new host cell to initiate infection. Structural investigations of the herpesvirus portal-vertex have proven challenging, owing to the small size of the tail-like portal-vertex-associated tegument (PVAT) and the presence of the tegument layer that lays between the nucleocapsid and the viral envelope, obscuring the view of the portal-vertex. Here, we show the structure of the herpes simplex virus portal-vertex at subnanometer resolution, solved by electron cryomicroscopy (cryoEM) and single-particle 3D reconstruction. This led to a number of new discoveries, including the presence of two previously unknown portal-associated structures that occupy the sites normally taken by the penton and the Ta triplex. Our data revealed that the PVAT is composed of 10 copies of the C-terminal domain of pUL25, which are uniquely arranged as two tiers of star-shaped density. Our 3D reconstruction of the portal-vertex also shows that one end of the viral genome extends outside the portal in the manner described for some bacteriophages but not previously seen in any eukaryote viruses. Finally, we show that the viral genome is consistently packed in a highly ordered left-handed spool to form concentric shells of DNA. Our data provide new insights into the structure of a molecular machine critical to the biology of an important class of human pathogens.


Assuntos
Capsídeo/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Capsídeo/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , DNA Viral/química , Genoma Viral , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Imageamento Tridimensional , Modelos Biológicos , Modelos Moleculares , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/ultraestrutura , Montagem de Vírus
15.
Nat Struct Mol Biol ; 25(5): 416-424, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29728654

RESUMO

Viral fusogens merge viral and cell membranes during cell penetration. Their ectodomains drive fusion by undergoing large-scale refolding, but little is known about the functionally important regions located within or near the membrane. Here we report the crystal structure of full-length glycoprotein B (gB), the fusogen from herpes simplex virus, complemented by electron spin resonance measurements. The membrane-proximal (MPR), transmembrane (TMD), and cytoplasmic (CTD) domains form a uniquely folded trimeric pedestal beneath the ectodomain, which balances dynamic flexibility with extensive, stabilizing membrane interactions. The postfusion conformation of the ectodomain suggests that the CTD likewise adopted the postfusion form. However, hyperfusogenic mutations, which destabilize the prefusion state of gB, target key interfaces and structural motifs that reinforce the observed CTD structure. Thus, a similar CTD structure must stabilize gB in its prefusion state. Our data suggest a model for how this dynamic, membrane-dependent 'clamp' controls the fusogenic refolding of gB.


Assuntos
Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Fusão de Membrana/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Ligação Viral , Animais , Células CHO , Cricetulus , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Herpesvirus Humano 1/genética , Conformação Proteica , Células Sf9 , Proteínas do Envelope Viral/genética , Internalização do Vírus
16.
Science ; 360(6384)2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622628

RESUMO

Herpes simplex viruses (HSVs) rely on capsid-associated tegument complex (CATC) for long-range axonal transport of their genome-containing capsids between sites of infection and neuronal cell bodies. Here we report cryo-electron microscopy structures of the HSV-1 capsid with CATC up to 3.5-angstrom resolution and atomic models of multiple conformers of capsid proteins VP5, VP19c, VP23, and VP26 and tegument proteins pUL17, pUL25, and pUL36. Crowning every capsid vertex are five copies of heteropentameric CATC, each containing a pUL17 monomer supporting the coiled-coil helix bundle of a pUL25 dimer and a pUL36 dimer, thus positioning their flexible domains for potential involvement in nuclear capsid egress and axonal capsid transport. Notwithstanding newly discovered fold conservation between triplex proteins and bacteriophage λ protein gpD and the previously recognized bacteriophage HK97 gp5-like fold in VP5, HSV-1 capsid proteins exhibit extraordinary diversity in forms of domain insertion and conformational polymorphism, not only for interactions with tegument proteins but also for encapsulation of large genomes.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Herpesvirus Humano 1/química , Animais , Capsídeo/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Chlorocebus aethiops , Microscopia Crioeletrônica , Glicoproteínas/química , Glicoproteínas/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Humanos , Conformação Proteica em alfa-Hélice , Células Vero
17.
J Biol Chem ; 292(38): 15611-15621, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28743747

RESUMO

During lytic infection, herpes simplex virus (HSV) DNA is replicated by a mechanism involving DNA recombination. For instance, replication of the HSV-1 genome produces X- and Y-branched structures, reminiscent of recombination intermediates. HSV-1's replication machinery includes a trimeric helicase-primase composed of helicase (UL5) and primase (UL52) subunits and a third subunit, UL8. UL8 has been reported to stimulate the helicase and primase activities of the complex in the presence of ICP8, an HSV-1 protein that functions as an annealase, a protein that binds complementary single-stranded DNA (ssDNA) and facilitates its annealing to duplex DNA. UL8 also influences the intracellular localization of the UL5/UL52 subunits, but UL8's catalytic activities are not known. In this study we used a combination of biochemical techniques and transmission electron microscopy. First, we report that UL8 alone forms protein filaments in solution. Moreover, we also found that UL8 binds to ssDNAs >50-nucletides long and promotes the annealing of complementary ssDNA to generate highly branched duplex DNA structures. Finally, UL8 has a very high affinity for replication fork structures containing a gap in the lagging strand as short as 15 nucleotides, suggesting that UL8 may aid in directing or loading the trimeric complex onto a replication fork. The properties of UL8 uncovered here suggest that UL8 may be involved in the generation of the X- and Y-branched structures that are the hallmarks of HSV replication.


Assuntos
DNA Helicases/metabolismo , DNA Primase/metabolismo , Replicação do DNA , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Sequência de Bases , DNA de Cadeia Simples/biossíntese , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , Herpesvirus Humano 1/ultraestrutura , Peso Molecular
18.
Planta Med ; 83(6): 509-518, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27706530

RESUMO

Herpes simplex virus infections persist throughout the lifetime of the host and affect more than 80 % of the humans worldwide. The intensive use of available therapeutic drugs has led to undesirable effects, such as drug-resistant strains, prompting the search for new antiherpetic agents. Although diverse bioactivities have been identified in Schinus terebinthifolia, its antiviral activity has not attracted much attention. The present study evaluated the antiherpetic effects of a crude hydroethanolic extract from the stem bark of S. terebinthifolia against Herpes simplex virus type 1 in vitro and in vivo as well as its genotoxicity in bone marrow in mammals and established the chemical composition of the crude hydroethanolic extract based on liquid chromatography-diode array detector-mass spectrometry and MS/MS. The crude hydroethanolic extract inhibited all of the tested Herpes simplex virus type 1 strains in vitro and was effective in the attachment and penetration stages, and showed virucidal activity, which was confirmed by transmission electron microscopy. The micronucleus test showed that the crude hydroethanolic extract had no genotoxic effect at the concentrations tested. The crude hydroethanolic extract afforded protection against lesions that were caused by Herpes simplex virus type 1 in vivo. Liquid chromatography-diode array detector-mass spectrometry and MS/MS identified 25 substances, which are condensed tannins mainly produced by a B-type linkage and prodelphinidin and procyanidin units.


Assuntos
Anacardiaceae/química , Antivirais/farmacocinética , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/efeitos dos fármacos , Extratos Vegetais/farmacologia , Animais , Antivirais/química , Antivirais/isolamento & purificação , Antivirais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Cromatografia Líquida , Feminino , Herpes Simples/virologia , Herpesvirus Humano 1/ultraestrutura , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Casca de Planta/química , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Espectrometria de Massas em Tandem , Taninos/análise , Taninos/química , Células Vero
19.
J Virol ; 91(2)2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27852850

RESUMO

The tegument of herpesviruses is a highly complex structural layer between the nucleocapsid and the envelope of virions. Tegument proteins play both structural and regulatory functions during replication and spread, but the interactions and functions of many of these proteins are poorly understood. Here we focus on two tegument proteins from herpes simplex virus 1 (HSV-1), pUL7 and pUL51, which have homologues in all other herpesviruses. We have now identified that HSV-1 pUL7 and pUL51 form a stable and direct protein-protein interaction, their expression levels rely on the presence of each other, and they function as a complex in infected cells. We demonstrate that expression of the pUL7-pUL51 complex is important for efficient HSV-1 assembly and plaque formation. Furthermore, we also discovered that the pUL7-pUL51 complex localizes to focal adhesions at the plasma membrane in both infected cells and in the absence of other viral proteins. The expression of pUL7-pUL51 is important to stabilize focal adhesions and maintain cell morphology in infected cells and cells infected with viruses lacking pUL7 and/or pUL51 round up more rapidly than cells infected with wild-type HSV-1. Our data suggest that, in addition to the previously reported functions in virus assembly and spread for pUL51, the pUL7-pUL51 complex is important for maintaining the attachment of infected cells to their surroundings through modulating the activity of focal adhesion complexes. IMPORTANCE: Herpesviridae is a large family of highly successful human and animal pathogens. Virions of these viruses are composed of many different proteins, most of which are contained within the tegument, a complex structural layer between the nucleocapsid and the envelope within virus particles. Tegument proteins have important roles in assembling virus particles as well as modifying host cells to promote virus replication and spread. However, little is known about the function of many tegument proteins during virus replication. Our study focuses on two tegument proteins from herpes simplex virus 1 that are conserved in all herpesviruses: pUL7 and pUL51. We demonstrate that these proteins directly interact and form a functional complex that is important for both virus assembly and modulation of host cell morphology. Further, we identify for the first time that these conserved herpesvirus tegument proteins localize to focal adhesions in addition to cytoplasmic juxtanuclear membranes within infected cells.


Assuntos
DNA Helicases/metabolismo , DNA Primase/metabolismo , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Complexos Multiproteicos/metabolismo , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo , Animais , Chlorocebus aethiops , DNA Helicases/genética , DNA Primase/genética , Regulação Viral da Expressão Gênica , Células HEK293 , Herpesvirus Humano 1/ultraestrutura , Humanos , Ligação Proteica , Transporte Proteico , Células Vero , Proteínas da Matriz Viral/genética , Proteínas Virais/genética , Montagem de Vírus
20.
J Virol Methods ; 241: 46-51, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28012897

RESUMO

Our laboratory was one of the first to engineer a live fluorescent tag, enhanced green fluorescent protein (eGFP), that marked the capsid of herpes simplex virus type 1 (HSV-1) and subsequently maturing virus as the particle made its way to the cell surface. In the present study we sought to increase the repertoire of colors available as fusion to the small capsid protein, VP26, so that they can be used alone or in conjunction with other fluorescent tags (fused to other HSV proteins) to follow the virus as it enters and replicates within the cell. We have now generated viruses expressing VP26 fusions with Cerulean, Venus, mOrange, tdTomato, mCherry, and Dronpa3 fluorescent proteins. These fusions were made in a repaired UL35 gene (VP26) background. These fusions do not affect the replication properties of the virus expressing the fusion polypeptide and the fusion tag was stably associated with intranuclear capsids and mature virions. Of note we could not isolate viruses expressing fusions with fluorescent proteins that have a tendency to dimerize.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Herpesvirus Humano 1/ultraestrutura , Animais , Linhagem Celular , Membrana Celular/genética , Chlorocebus aethiops , Cor , Corantes Fluorescentes , Proteínas de Fluorescência Verde , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/química , Células Vero , Replicação Viral , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA