Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 112022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35635544

RESUMO

A restriction factor induced by interferons blocks the replication of herpesviruses by disassembling the capsid proteins surrounding their genome.


Assuntos
Proteínas do Capsídeo , Herpesviridae , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Herpesviridae/genética , Interferons
2.
J Virol ; 95(20): e0075521, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34346766

RESUMO

The maturation process that occurs in most viruses is evolutionarily driven, as it resolves several conflicting virion assembly requirements. During herpesvirus assembly in a host cell nucleus, micron-long double-stranded herpes DNA is packaged into a nanometer-sized procapsid. This leads to strong confinement of the viral genome, resulting in tens of atmospheres of intracapsid DNA pressure. Yet, the procapsid is unstable due to weak reversible interactions between its protein subunits, which ensures free energy minimization and reduces assembly errors. In this work, we show that herpesviruses resolve these contradictory capsid requirements through a mechanical capsid maturation process facilitated by multifunctional auxiliary protein UL25. Through mechanical interrogation of herpes simplex virus 1 (HSV-1) capsid with atomic force microscopy nano-indentation, we show that UL25 binding at capsid vertices post-assembly provides the critical capsid reinforcement required for stable DNA encapsidation; the absence of UL25 binding leads to capsid rupture. Furthermore, we demonstrate that gradual capsid reinforcement is a feasible maturation mechanism facilitated by progressive UL25 capsid binding, which is likely correlated with DNA packaging progression. This work provides insight into elegantly programmed viral assembly machinery, where targeting of capsid assembly mechanics presents a new antiviral strategy that is resilient to the development of drug resistance. IMPORTANCE Most viruses undergo a maturation process from a weakly assembled particle to a stable virion. Herpesvirus capsid undergoes mechanical maturation to withstand tens of atmospheres of DNA pressure. We demonstrate that this mechanical capsid maturation is mainly facilitated through binding of auxiliary protein UL25 in herpes simplex virus 1 (HSV-1) capsid vertices. We show that UL25 binding provides the critical capsid reinforcement required for stable DNA encapsidation. Our data also suggest that gradual capsid reinforcement by progressive UL25 binding is a feasible capsid maturation mechanism, correlated with DNA packaging progression.


Assuntos
Proteínas do Capsídeo/genética , Herpesvirus Humano 1/metabolismo , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Chlorocebus aethiops , DNA Viral/genética , Genoma Viral , Herpesviridae/genética , Herpesvirus Humano 1/genética , Células Vero , Proteínas Virais/metabolismo , Vírion/metabolismo , Montagem de Vírus
3.
J Virol ; 94(24)2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32967953

RESUMO

The packaging of DNA into preformed capsids is a critical step during herpesvirus infection. For herpes simplex virus, this process requires the products of seven viral genes: the terminase proteins pUL15, pUL28, and pUL33; the capsid vertex-specific component (CVSC) proteins pUL17 and pUL25; and the portal proteins pUL6 and pUL32. The pUL6 portal dodecamer is anchored at one vertex of the capsid by interactions with the adjacent triplexes as well as helical density attributed to the pUL17 and pUL25 subunits of the CVSC. To define the roles and structures of the CVSC proteins in virus assembly and DNA packaging, we isolated a number of recombinant viruses expressing pUL25, pUL17, and pUL36 fused with green or red fluorescent proteins as well as viruses with specific deletions in the CVSC genes. Biochemical and structural studies of these mutants demonstrated that (i) four of the helices in the CVSC helix bundle can be attributed to two copies each of pUL36 and pUL25, (ii) pUL17 and pUL6 are required for capsid binding of the terminase complex in the nucleus, (iii) pUL17 is important for determining the site of the first cleavage reaction generating replicated genomes with termini derived from the long-arm component of the herpes simplex virus 1 (HSV-1) genome, (iv) pUL36 serves no direct role in cleavage/packaging, (v) cleavage and stable packaging of the viral genome involve an ordered interaction of the terminase complex and pUL25 with pUL17 at the portal vertex, and (vi) packaging of the viral genome results in a dramatic displacement of the portal.IMPORTANCE Herpes simplex virus 1 (HSV-1) is the causative agent of several pathologies ranging in severity from the common cold sore to life-threatening encephalitic infection. A critical step during productive HSV-1 infection is the cleavage and packaging of replicated, concatemeric viral DNA into preformed capsids. A key knowledge gap is how the capsid engages the replicated viral genome and the subsequent packaging of a unit-length HSV genome. Here, biochemical and structural studies focused on the unique portal vertex of wild-type HSV and packaging mutants provide insights into the mechanism of HSV genome packaging. The significance of our research is in identifying the portal proteins pUL6 and pUL17 as key viral factors for engaging the terminase complex with the capsid and the subsequent cleavage, packaging, and stable incorporation of the viral genome in the HSV-1 capsid.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Herpesvirus Humano 1/metabolismo , Proteínas Virais/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Microscopia Crioeletrônica , Empacotamento do DNA , DNA Viral/metabolismo , Endodesoxirribonucleases , Genes Virais , Genoma Viral , Herpesvirus Humano 1/genética , Células Vero , Proteínas Virais/química , Proteínas Virais/genética , Montagem de Vírus , Replicação Viral
4.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490590

RESUMO

The herpes simplex virus (HSV) capsid is released into the cytoplasm after fusion of viral and host membranes, whereupon dynein-dependent trafficking along microtubules targets it to the nuclear envelope. Binding of the capsid to the nuclear pore complex (NPC) is mediated by the capsid protein pUL25 and the capsid-tethered tegument protein pUL36. Temperature-sensitive mutants in both pUL25 and pUL36 dock at the NPC but fail to release DNA. The uncoating reaction has been difficult to study due to the rapid release of the genome once the capsid interacts with the nuclear pore. In this study, we describe the isolation and characterization of a truncation mutant of pUL25. Live-cell imaging and immunofluorescence studies demonstrated that the mutant was not impaired in penetration of the host cell or in trafficking of the capsid to the nuclear membrane. However, expression of viral proteins was absent or significantly delayed in cells infected with the pUL25 mutant virus. Transmission electron microscopy revealed capsids accumulated at nuclear pores that retained the viral genome for at least 4 h postinfection. In addition, cryoelectron microscopy (cryo-EM) reconstructions of virion capsids did not detect any obvious differences in the location or structural organization for the pUL25 or pUL36 proteins on the pUL25 mutant capsids. Further, in contrast to wild-type virus, the antiviral response mediated by the viral DNA-sensing cyclic guanine adenine synthase (cGAS) was severely compromised for the pUL25 mutant. These results demonstrate that the pUL25 capsid protein has a critical role in releasing viral DNA from NPC-bound capsids.IMPORTANCE Herpes simplex virus 1 (HSV-1) is the causative agent of several pathologies ranging in severity from the common cold sore to life-threatening encephalitic infection. Early steps in infection include release of the capsid into the cytoplasm, docking of the capsid at a nuclear pore, and release of the viral genome into the nucleus. A key knowledge gap is how the capsid engages the NPC and what triggers release of the viral genome into the nucleus. Here we show that the C-terminal region of the HSV-1 pUL25 protein is required for releasing the viral genome from capsids docked at nuclear pores. The significance of our research is in identifying pUL25 as a key viral factor for genome uncoating. pUL25 is found at each of the capsid vertices as part of the capsid vertex-specific component and implicates the importance of this complex for NPC binding and genome release.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA Viral/metabolismo , Herpesvirus Humano 1/fisiologia , Poro Nuclear/metabolismo , Desenvelopamento do Vírus , Animais , Proteínas do Capsídeo/genética , Chlorocebus aethiops , Microscopia , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Deleção de Sequência , Células Vero
5.
Adv Anat Embryol Cell Biol ; 223: 119-142, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28528442

RESUMO

Herpes simplex virus type I (HSV-1) is the causative agent of several pathologies ranging in severity from the common cold sore to life-threatening encephalitic infection. During productive lytic infection, over 80 viral proteins are expressed in a highly regulated manner, resulting in the replication of viral genomes and assembly of progeny virions. The virion of all herpesviruses consists of an external membrane envelope, a proteinaceous layer called the tegument, and an icosahedral capsid containing the double-stranded linear DNA genome. The capsid shell of HSV-1 is built from four structural proteins: a major capsid protein, VP5, which forms the capsomers (hexons and pentons), the triplex consisting of VP19C and VP23 found between the capsomers, and VP26 which binds to VP5 on hexons but not pentons. In addition, the dodecameric pUL6 portal complex occupies 1 of the 12 capsid vertices, and the capsid vertex specific component (CVSC), a heterotrimer complex of pUL17, pUL25, and pUL36, binds specifically to the triplexes adjacent to each penton. The capsid is assembled in the nucleus where the viral genome is packaged into newly assembled closed capsid shells. Cleavage and packaging of replicated, concatemeric viral DNA requires the seven viral proteins encoded by the UL6, UL15, UL17, UL25, UL28, UL32, and UL33 genes. Considerable advances have been made in understanding the structure of the herpesvirus capsid and the function of several of the DNA packaging proteins by applying biochemical, genetic, and structural techniques. This review is a summary of recent advances with respect to the structure of the HSV-1 virion capsid and what is known about the function of the seven packaging proteins and their interactions with each other and with the capsid shell.


Assuntos
Capsídeo/fisiologia , Empacotamento do DNA , Herpesviridae/fisiologia , Montagem de Vírus/fisiologia , Animais , Humanos , Proteínas Virais/metabolismo , Vírion/metabolismo
6.
Nat Struct Mol Biol ; 23(6): 531-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27111889

RESUMO

The herpesvirus capsid is a complex protein assembly that includes hundreds of copies of four major subunits and lesser numbers of several minor proteins, all of which are essential for infectivity. Cryo-electron microscopy is uniquely suited for studying interactions that govern the assembly and function of such large functional complexes. Here we report two high-quality capsid structures, from human herpes simplex virus type 1 (HSV-1) and the animal pseudorabies virus (PRV), imaged inside intact virions at ~7-Å resolution. From these, we developed a complete model of subunit and domain organization and identified extensive networks of subunit contacts that underpin capsid stability and form a pathway that may signal the completion of DNA packaging from the capsid interior to outer surface, thereby initiating nuclear egress. Differences in the folding and orientation of subunit domains between herpesvirus capsids suggest that common elements have been modified for specific functions.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Herpesvirus Humano 1/química , Regulação Alostérica , Animais , Capsídeo/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Herpes Simples/virologia , Herpesvirus Humano 1/ultraestrutura , Herpesvirus Suídeo 1/química , Herpesvirus Suídeo 1/ultraestrutura , Humanos , Modelos Moleculares , Conformação Proteica , Estabilidade Proteica , Subunidades Proteicas/química , Pseudorraiva/virologia
7.
Nat Chem Biol ; 10(10): 861-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25195012

RESUMO

DNA in the human Herpes simplex virus type 1 (HSV-1) capsid is packaged to a tight density. This leads to tens of atmospheres of internal pressure responsible for the delivery of the herpes genome into the cell nucleus. In this study we show that, despite its liquid crystalline state inside the capsid, the DNA is fluid-like, which facilitates its ejection into the cell nucleus during infection. We found that the sliding friction between closely packaged DNA strands, caused by interstrand repulsive interactions, is reduced by the ionic environment of epithelial cells and neurons susceptible to herpes infection. However, variations in the ionic conditions corresponding to neuronal activity can restrict DNA mobility in the capsid, making it more solid-like. This can inhibit intranuclear DNA release and interfere with viral replication. In addition, the temperature of the human host (37 °C) induces a disordering transition of the encapsidated herpes genome, which reduces interstrand interactions and provides genome mobility required for infection.


Assuntos
Capsídeo/química , Núcleo Celular/virologia , DNA Viral/química , Genoma Viral , Herpesvirus Humano 1/fisiologia , Animais , Chlorocebus aethiops , Herpesvirus Humano 1/química , Humanos , Cinética , Conformação de Ácido Nucleico , Transição de Fase , Temperatura , Células Vero , Replicação Viral
8.
Nucleic Acids Res ; 42(14): 9096-107, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25053840

RESUMO

Herpes simplex type 1 virus (HSV-1) and bacteriophage λ capsids undergo considerable structural changes during self-assembly and DNA packaging. The initial steps of viral capsid self-assembly require weak, non-covalent interactions between the capsid subunits to ensure free energy minimization and error-free assembly. In the final stages of DNA packaging, however, the internal genome pressure dramatically increases, requiring significant capsid strength to withstand high internal genome pressures of tens of atmospheres. Our data reveal that the loosely formed capsid structure is reinforced post-assembly by the minor capsid protein UL25 in HSV-1 and gpD in bacteriophage λ. Using atomic force microscopy nano-indentation analysis, we show that the capsid becomes stiffer upon binding of UL25 and gpD due to increased structural stability. At the same time the force required to break the capsid increases by ∼70% for both herpes and phage. This demonstrates a universal and evolutionarily conserved function of the minor capsid protein: facilitating the retention of the pressurized viral genome in the capsid. Since all eight human herpesviruses have UL25 orthologs, this discovery offers new opportunities to interfere with herpes replication by disrupting the precise force balance between the encapsidated DNA and the capsid proteins crucial for viral replication.


Assuntos
Bacteriófago lambda/fisiologia , Proteínas do Capsídeo/metabolismo , Capsídeo/ultraestrutura , Glicoproteínas/metabolismo , Herpesvirus Humano 1/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus , Animais , Bacteriófago lambda/metabolismo , Bacteriófago lambda/ultraestrutura , Capsídeo/metabolismo , Chlorocebus aethiops , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Células Vero
9.
J Virol ; 88(1): 225-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155374

RESUMO

During herpes simplex virus 1 (HSV-1) infection, empty procapsids are assembled and subsequently filled with the viral genome by means of a protein complex called the terminase, which is comprised of the HSV-1 UL15, UL28, and UL33 proteins. Biochemical studies of the terminase proteins have been hampered by the inability to purify the intact terminase complex. In this study, terminase complexes were isolated by tandem-affinity purification (TAP) using recombinant viruses expressing either a full-length NTAP-UL28 fusion protein (vFH476) or a C-terminally truncated NTAP-UL28 fusion protein (vFH499). TAP of the UL28 protein from vFH476-infected cells, followed by silver staining, Western blotting, and mass spectrometry, identified the UL15, UL28, and UL33 subunits, while TAP of vFH499-infected cells confirmed previous findings that the C terminus of UL28 is required for UL28 interaction with UL33 and UL15. Analysis of the oligomeric state of the purified complexes by sucrose density gradient ultracentrifugation revealed that the three proteins formed a complex with a molecular mass that is consistent with the formation of a UL15-UL28-UL33 heterotrimer. In order to assess the importance of conserved regions of the UL15 and UL28 proteins, recombinant NTAP-UL28 viruses with mutations of the putative UL28 metal-binding domain or within the UL15 nuclease domain were generated. TAP of UL28 complexes from cells infected with each domain mutant demonstrated that the conserved cysteine residues of the putative UL28 metal-binding domain and conserved amino acids within the UL15 nuclease domain are required for the cleavage and packaging functions of the viral terminase, but not for terminase complex assembly.


Assuntos
Endodesoxirribonucleases/isolamento & purificação , Herpesvirus Humano 1/enzimologia , Animais , Sequência de Bases , Southern Blotting , Western Blotting , Chlorocebus aethiops , Cromatografia de Afinidade , DNA Viral/química , Endodesoxirribonucleases/química , Espectrometria de Massas , Mutação , Células Vero
10.
J Am Chem Soc ; 135(30): 11216-21, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23829592

RESUMO

Herpes simplex virus type 1 (HSV-1) packages its micrometers-long double-stranded DNA genome into a nanometer-scale protein shell, termed the capsid. Upon confinement within the capsid, neighboring DNA strands experience repulsive electrostatic and hydration forces as well as bending stress associated with the tight curvature required of packaged DNA. By osmotically suppressing DNA release from HSV-1 capsids, we provide the first experimental evidence of a high internal pressure of tens of atmospheres within a eukaryotic human virus, resulting from the confined genome. Furthermore, the ejection is progressively suppressed by increasing external osmotic pressures, which reveals that internal pressure is capable of powering ejection of the entire genome from the viral capsid. Despite billions of years of evolution separating eukaryotic viruses and bacteriophages, pressure-driven DNA ejection has been conserved. This suggests it is a key mechanism for viral infection and thus presents a new target for antiviral therapies.


Assuntos
Genoma Viral/genética , Herpesvirus Humano 1/genética , Pressão Osmótica , Animais , Capsídeo/metabolismo , Chlorocebus aethiops , Empacotamento do DNA , DNA Viral/genética , Humanos , Células Vero
11.
J Virol ; 85(15): 7513-22, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21632758

RESUMO

The herpes simplex virus (HSV) UL17 and UL25 minor capsid proteins are essential for DNA packaging. They are thought to comprise a molecule arrayed in five copies around each of the capsid vertices. This molecule was initially termed the "C-capsid-specific component" (CCSC) (B. L. Trus et al., Mol. Cell 26:479-489, 2007), but as we have subsequently observed this feature on reconstructions of A, B, and C capsids, we now refer to it more generally as the "capsid vertex-specific component" (CVSC) (S. K. Cockrell et al., J. Virol. 85:4875-4887, 2011). We previously confirmed that UL25 occupies the vertex-distal region of the CVSC density by visualizing a large UL25-specific tag in reconstructions calculated from cryo-electron microscopy (cryo-EM) images. We have pursued the same strategy to determine the capsid location of the UL17 protein. Recombinant viruses were generated that contained either a small tandem affinity purification (TAP) tag or the green fluorescent protein (GFP) attached to the C terminus of UL17. Purification of the TAP-tagged UL17 or a similarly TAP-tagged UL25 protein clearly demonstrated that the two proteins interact. A cryo-EM reconstruction of capsids containing the UL17-GFP protein reveals that UL17 is the second component of the CVSC and suggests that UL17 interfaces with the other CVSC component, UL25, through its C terminus. The portion of UL17 nearest the vertex appears to be poorly constrained, which may provide flexibility in interacting with tegument proteins or the DNA-packaging machinery at the portal vertex. The exposed locations of the UL17 and UL25 proteins on the HSV-1 capsid exterior suggest that they may be attractive targets for highly specific antivirals.


Assuntos
Capsídeo , Empacotamento do DNA , Proteínas Virais/metabolismo , Animais , Sequência de Bases , Chlorocebus aethiops , Cromatografia de Afinidade , Microscopia Crioeletrônica , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Proteínas de Fluorescência Verde/genética , Células Vero , Proteínas Virais/genética
12.
J Virol ; 85(10): 4875-87, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21411517

RESUMO

The herpes simplex virus 1 (HSV-1) UL25 gene product is a minor capsid component that is required for encapsidation, but not cleavage, of replicated viral DNA. UL25 is located on the capsid surface in a proposed heterodimer with UL17, where five copies of the heterodimer are found at each of the capsid vertices. Previously, we demonstrated that amino acids 1 to 50 of UL25 are essential for its stable interaction with capsids. To further define the UL25 capsid binding domain, we generated recombinant viruses with either small truncations or amino acid substitutions in the UL25 N terminus. Studies of these mutants demonstrated that there are two important regions within the capsid binding domain. The first 27 amino acids are essential for capsid binding of UL25, while residues 26 to 39, which are highly conserved in the UL25 homologues of other alphaherpesviruses, were found to be critical for stable capsid binding. Cryo-electron microscopy reconstructions of capsids containing either a small tag on the N terminus of UL25 or the green fluorescent protein (GFP) fused between amino acids 50 and 51 of UL25 demonstrate that residues 1 to 27 of UL25 contact the hexon adjacent to the penton. A second region, most likely centered on amino acids 26 to 39, contacts the triplex that is one removed from the penton. Importantly, both of these UL25 capsid binding regions are essential for the stable packaging of full-length viral genomes.


Assuntos
Proteínas do Capsídeo/metabolismo , Herpesvirus Humano 1/fisiologia , Mapeamento de Interação de Proteínas , Substituição de Aminoácidos/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Herpesvirus Humano 1/ultraestrutura , Modelos Moleculares , Estrutura Quaternária de Proteína , Deleção de Sequência , Vírion/ultraestrutura , Montagem de Vírus
13.
J Mol Biol ; 397(2): 575-86, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20109467

RESUMO

The herpes simplex virus type 1 UL25 protein is one of seven viral proteins that are required for DNA cleavage and packaging. Together with UL17, UL25 forms part of an elongated molecule referred to as the C-capsid-specific component (CCSC). Five copies of the CCSC are located at each of the capsid vertices on DNA-containing capsids. To study the conformation of UL25 as it is folded on the capsid surface, we identified the sequence recognized by a UL25-specific monoclonal antibody and localized the epitope on the capsid surface by immunogold electron microscopy. The epitope mapped to amino acids 99-111 adjacent to the region of the protein (amino acids 1-50) that is required for capsid binding. In addition, cryo-EM reconstructions of C-capsids in which the green fluorescent protein (GFP) was fused within the N-terminus of UL25 localized the point of contact between UL25 and GFP. The result confirmed the modeled location of the UL25 protein in the CCSC density as the region that is distal to the penton with the N-terminus of UL25 making contact with the triplex one removed from the penton. Immunofluorescence experiments at early times during infection demonstrated that UL25-GFP was present on capsids located within the cytoplasm and adjacent to the nucleus. These results support the view that UL25 is present on incoming capsids with the capsid-binding domain of UL25 located on the surface of the mature DNA-containing capsid.


Assuntos
Proteínas do Capsídeo/análise , Proteínas do Capsídeo/química , Capsídeo/química , Herpesvirus Humano 1/química , Dobramento de Proteína , Anticorpos Monoclonais/metabolismo , Anticorpos Antivirais/metabolismo , Microscopia Crioeletrônica , Mapeamento de Epitopos , Processamento de Imagem Assistida por Computador , Microscopia Imunoeletrônica , Modelos Moleculares
14.
J Mol Biol ; 392(4): 885-94, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19631662

RESUMO

Ejection of DNA from the capsid is an early step in infection by all herpesviruses. Ejection or DNA uncoating occurs after a parental capsid has entered the host cell cytoplasm, migrated to the nucleus, and bound to a nuclear pore. DNA exits the capsid through the portal vertex and proceeds by way of the nuclear pore complex into the nucleoplasm where it is transcribed and replicated. Here, we describe use of an in vitro uncoating system to determine which genome end exits first from the herpes simplex virus 1 capsid. Purified DNA-containing capsids were bound to a solid surface and warmed under conditions in which some, but not all, of the DNA was ejected. Restriction endonuclease digestion was then used to identify the genomic origin of the ejected DNA. The results support the view that the S segment end exits the capsid first. Preferential release at the S end demonstrates that herpesvirus DNA uncoating conforms to the paradigm in double-stranded DNA bacteriophage where the last end packaged is the first to be ejected. Release of herpes simplex virus 1 DNA beginning at the S end causes the first gene to enter the host cell nucleus to be alpha4, a transcription factor required for expression of early genes.


Assuntos
Capsídeo/metabolismo , DNA Viral/metabolismo , Herpesvirus Humano 1/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Replicação do DNA/fisiologia , DNA Viral/efeitos dos fármacos , Endonucleases/metabolismo , Formaldeído/farmacologia , Genoma Viral/efeitos dos fármacos , Genoma Viral/fisiologia , Herpesvirus Humano 1/efeitos dos fármacos
15.
J Virol ; 83(1): 47-57, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945788

RESUMO

The herpes simplex virus protein UL25 attaches to the external vertices of herpes simplex virus type 1 capsids and is required for the stable packaging of viral DNA. To define regions of the protein important for viral replication and capsid attachment, the 580-amino-acid UL25 open reading frame was disrupted by transposon mutagenesis. The UL25 mutants were assayed for complementation of a UL25 deletion virus, and in vitro-synthesized protein was tested for binding to UL25-deficient capsids. Of the 11 mutants analyzed, 4 did not complement growth of the UL25 deletion mutant, and analysis of these and additional mutants in the capsid-binding assay demonstrated that UL25 amino acids 1 to 50 were sufficient for capsid binding. Several UL25 mutations were transferred into recombinant viruses to analyze the effect of the mutations on UL25 capsid binding and on DNA cleavage and packaging. Studies of these mutants demonstrated that amino acids 1 to 50 of UL25 are essential for its stable interaction with capsids and that the C terminus is essential for DNA packaging and the production of infectious virus through its interactions with other viral packaging or tegument proteins. Analysis of viral DNA cleavage demonstrated that in the absence of a functional UL25 protein, aberrant cleavage takes place at the unique short end of the viral genome, resulting in truncated viral genomes that are not retained in capsids. Based on these observations, we propose a model where UL25 is required for the formation of DNA-containing capsids by acting to stabilize capsids that contain full-length viral genomes.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus , Animais , Sítios de Ligação , Chlorocebus aethiops , Elementos de DNA Transponíveis , DNA Viral/metabolismo , Deleção de Genes , Teste de Complementação Genética , Mutagênese Insercional , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mapeamento de Interação de Proteínas , Células Vero , Proteínas Virais/genética
16.
J Virol ; 82(13): 6778-81, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18417585

RESUMO

The herpes simplex virus type 1 (HSV-1) portal is composed of a dodecamer of UL6 protein molecules whose incorporation into the capsid is mediated by interaction with the HSV-1 UL26.5 scaffold protein. Previous results with an in vitro capsid assembly assay demonstrated that nine amino acids (amino acids 143 to 151) of the UL26.5 protein are required for its interaction with UL6 and for incorporation of the portal complex into capsids. In the present study an HSV-1 mutant, bvFH411, was isolated and contained a deletion that removed the codons for UL26.5 amino acids 143 to 150. The mutant virus failed to produce infectious virus in noncomplementing cells, and only B capsids that contained only minor amounts of portal protein were made. These data corroborate our previous in vitro studies and demonstrate that amino acids 143 to 150 of UL26.5 are required for the formation of portal-containing HSV-1 capsids.


Assuntos
Capsídeo/metabolismo , Genoma Viral/genética , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Mapeamento Cromossômico , Cromossomos Artificiais Bacterianos/genética , Primers do DNA/genética , Herpesvirus Humano 1/ultraestrutura , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Mutação/genética , Células Vero , Proteínas Virais/genética
17.
Mol Cell ; 26(4): 479-89, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17531807

RESUMO

UL25 and UL17 are two essential minor capsid proteins of HSV-1, implicated in DNA packaging and capsid maturation. We used cryo-electron microscopy to examine their binding to capsids, whose architecture observes T = 16 icosahedral geometry. C-capsids (mature DNA-filled capsids) have an elongated two-domain molecule present at a unique, vertex-adjacent site that is not seen at other quasiequivalent sites or on unfilled capsids. Using SDS-PAGE and mass spectrometry to analyze wild-type capsids, UL25 null capsids, and denaturant-extracted capsids, we conclude that (1) the C-capsid-specific component is a heterodimer of UL25 and UL17, and (2) capsids have additional populations of UL25 and UL17 that are invisible in reconstructions because of sparsity and/or disorder. We infer that binding of the ordered population reflects structural changes induced on the outer surface as pressure builds up inside the capsid during DNA packaging. Its binding may signal that the C-capsid is ready to exit the nucleus.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Núcleo Celular/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Regulação Alostérica , Microscopia Crioeletrônica , Dimerização , Processamento de Imagem Assistida por Computador , Espectrometria de Massas , Ligação Proteica , Transporte Proteico , Proteínas Virais/metabolismo
18.
J Virol ; 80(24): 12312-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17035316

RESUMO

The UL28 protein of herpes simplex virus type 1 (HSV-1) is one of seven viral proteins required for the cleavage and packaging of viral DNA. Previous results indicated that UL28 interacts with UL15 and UL33 to form a protein complex (terminase) that is presumed to cleave concatemeric DNA into genome lengths. In order to define the functional domains of UL28 that are important for DNA cleavage/packaging, we constructed a series of HSV-1 mutants with linker insertion and nonsense mutations in UL28. Insertions that blocked DNA cleavage and packaging were found to be located in two regions of UL28: the first between amino acids 200 to 400 and the second between amino acids 600 to 740. Insertions located in the N terminus or in a region located between amino acids 400 and 600 did not affect virus replication. Insertions in the carboxyl terminus of the UL28 protein were found to interfere with the interaction of UL28 with UL33. In contrast, all of the UL28 insertion mutants were found to interact with UL15 but the interaction was reduced with mutants that failed to react with UL33. Together, these observations were consistent with previous conclusions that UL15 and UL33 interact directly with UL28 but interact only indirectly with each other. Revertant viruses that formed plaques on Vero cells were detected for one of the lethal UL28 insertion mutants. DNA sequence analysis, in combination with genetic complementation assays, demonstrated that a second-site mutation in the UL15 gene restored the ability of the revertant to cleave and package viral DNA. The isolation of an intergenic suppressor mutant provides direct genetic evidence of an association between the UL28 and UL15 proteins and demonstrates that this association is essential for DNA cleavage and packaging.


Assuntos
Herpesvirus Humano 1/genética , Mutação/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Southern Blotting , Chlorocebus aethiops , Empacotamento do DNA/genética , Teste de Complementação Genética , Immunoblotting , Imunoprecipitação , Dados de Sequência Molecular , Mutagênese Insercional , Análise de Sequência de DNA , Células Vero
19.
J Virol ; 80(13): 6286-94, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775316

RESUMO

UL25 is one of seven herpes simplex virus-encoded proteins involved specifically in DNA encapsidation. Its role appears to be to stabilize the capsid so that DNA is prevented from escaping once it has entered. To clarify the function of UL25, we have examined capsids with the goal of defining where it is located. Analysis of trypsin-treated capsids showed that UL25 is sensitive to cleavage like other proteins such as the major capsid and portal proteins that are exposed on the capsid surface. Internal proteins such as the scaffolding protein and protease were not affected under the same experimental conditions. Capsids were also examined by electron microscopy after staining with gold-labeled antibody specific for UL25. Images of stained capsids demonstrated that most labeled sites (71% in C capsids) were at capsid vertices, and most stained C capsids had label at more than one vertex. A quantitative immunoblotting method showed that the capsid contents of UL25 were 56, 20, and 75 copies per capsid in A, B, and C capsids, respectively. Finally, soluble UL25 protein was found to bind in vitro to purified capsids lacking it. The amount of bound UL25 corresponded to the amount present in B capsids, and bound UL25 was found by immunoelectron microscopy to be located predominantly at the capsid vertices. The results are interpreted to suggest that five UL25 molecules are found at or near each of the capsid vertices, where they are exposed on the capsid surface. Exposure on the surface is consistent with the view that UL25 is added to the capsid as DNA is packaged or during late stages of the packaging process.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Capsídeo/ultraestrutura , DNA Viral/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Montagem de Vírus/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Capsídeo/imunologia , Proteínas do Capsídeo/imunologia , Chlorocebus aethiops , Herpesvirus Humano 1/imunologia , Humanos , Microscopia Eletrônica de Transmissão , Ligação Proteica/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Células Vero
20.
J Biol Chem ; 281(26): 18193-200, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16638752

RESUMO

Herpesviruses are the second leading cause of human viral diseases. Herpes Simplex Virus types 1 and 2 and Varicella-zoster virus produce neurotropic infections such as cutaneous and genital herpes, chickenpox, and shingles. Infections of a lymphotropic nature are caused by cytomegalovirus, HSV-6, HSV-7, and Epstein-Barr virus producing lymphoma, carcinoma, and congenital abnormalities. Yet another series of serious health problems are posed by infections in immunocompromised individuals. Common therapies for herpes viral infections employ nucleoside analogs, such as Acyclovir, and target the viral DNA polymerase, essential for viral DNA replication. Although clinically useful, this class of drugs exhibits a narrow antiviral spectrum, and resistance to these agents is an emerging problem for disease management. A better understanding of herpes virus replication will help the development of new safe and effective broad spectrum anti-herpetic drugs that fill an unmet need. Here, we present the first crystal structure of a herpesvirus polymerase, the Herpes Simplex Virus type 1 DNA polymerase, at 2.7 A resolution. The structural similarity of this polymerase to other alpha polymerases has allowed us to construct high confidence models of a replication complex of the polymerase and of Acyclovir as a DNA chain terminator. We propose a novel inhibition mechanism in which a representative of a series of non-nucleosidic viral polymerase inhibitors, the 4-oxo-dihydroquinolines, binds at the polymerase active site interacting non-covalently with both the polymerase and the DNA duplex.


Assuntos
DNA Polimerase Dirigida por DNA/química , Exodesoxirribonucleases/química , Herpesvirus Humano 1/enzimologia , Proteínas Virais/química , Aciclovir/química , Antivirais/química , Sítios de Ligação , Cristalografia , Desenho de Fármacos , Farmacorresistência Viral , Herpesvirus Humano 1/efeitos dos fármacos , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Quinolinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...