Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(4): e0140622, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37022163

RESUMO

The genomes of numerous herpesviruses have been cloned as infectious bacterial artificial chromosomes. However, attempts to clone the complete genome of infectious laryngotracheitis virus (ILTV), formally known as Gallid alphaherpesvirus-1, have been met with limited success. In this study, we report the development of a cosmid/yeast centromeric plasmid (YCp) genetic system to reconstitute ILTV. Overlapping cosmid clones were generated that encompassed 90% of the 151-Kb ILTV genome. Viable virus was produced by cotransfecting leghorn male hepatoma (LMH) cells with these cosmids and a YCp recombinant containing the missing genomic sequences - spanning the TRS/UL junction. An expression cassette for green fluorescent protein (GFP) was inserted within the redundant inverted packaging site (ipac2), and the cosmid/YCp-based system was used to generate recombinant replication-competent ILTV. Viable virus was also reconstituted with a YCp clone containing a BamHI linker within the deleted ipac2 site, further demonstrating the nonessential nature of this site. Recombinants deleted in the ipac2 site formed plaques undistinguished from those viruses containing intact ipac2. The 3 reconstituted viruses replicated in chicken kidney cells with growth kinetics and titers similar to the USDA ILTV reference strain. Specific pathogen-free chickens inoculated with the reconstituted ILTV recombinants succumbed to levels of clinical disease similar to that observed in birds inoculated with wildtype viruses, demonstrating the reconstituted viruses were virulent. IMPORTANCE Infectious laryngotracheitis virus (ILTV) is an important pathogen of chicken with morbidity of 100% and mortality rates as high as 70%. Factoring in decreased production, mortality, vaccination, and medication, a single outbreak can cost producers over a million dollars. Current attenuated and vectored vaccines lack safety and efficacy, leaving a need for better vaccines. In addition, the lack of an infectious clone has also impeded understanding viral gene function. Since infectious bacterial artificial chromosome (BAC) clones of ILTV with intact replication origins are not feasible, we reconstituted ILTV from a collection of yeast centromeric plasmids and bacterial cosmids, and identified a nonessential insertion site within a redundant packaging site. These constructs and the methodology necessary to manipulate them will facilitate the development of improved live virus vaccines by modifying genes encoding virulence factors and establishing ILTV-based viral vectors for expressing immunogens of other avian pathogens.


Assuntos
Cosmídeos , Herpesvirus Galináceo 1 , Mutagênese , Plasmídeos , Animais , Masculino , Galinhas , Cosmídeos/genética , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/genética , Herpesvirus Galináceo 1/patogenicidade , Plasmídeos/genética , Doenças das Aves Domésticas/virologia , Saccharomyces cerevisiae/genética , Linhagem Celular , Genoma Viral/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
Viruses ; 13(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34835004

RESUMO

African swine fever virus (ASFV), causing an OIE-notifiable viral disease of swine, is spreading over the Eurasian continent and threatening the global pig industry. Here, we conducted the first proteome analysis of ASFV-infected primary porcine monocyte-derived macrophages (moMΦ). In parallel to moMΦ isolated from different pigs, the stable porcine cell line WSL-R was infected with a recombinant of ASFV genotype IX strain "Kenya1033". The outcome of the infections was compared via quantitative mass spectrometry (MS)-based proteome analysis. Major differences with respect to the expression of viral proteins or the host cell response were not observed. However, cell-specific expression of some individual viral proteins did occur. The observed modulations of the host proteome were mainly related to cell characteristics and function. Overall, we conclude that both infection models are suitable for use in the study of ASFV infection in vitro.


Assuntos
Vírus da Febre Suína Africana , Macrófagos/virologia , Proteoma/metabolismo , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Animais , Linhagem Celular , Suínos , Proteínas Virais , Replicação Viral
3.
mBio ; 12(3)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33947756

RESUMO

Herpesvirus entry and spread requires fusion of viral and host cell membranes, which is mediated by the conserved surface glycoprotein B (gB). Upon activation, gB undergoes a major conformational change and transits from a metastable prefusion to a stable postfusion conformation. Although gB is a structural homolog of low-pH-triggered class III fusogens, its fusion activity depends strictly on the presence of the conserved regulatory gH/gL complex and nonconserved receptor binding proteins, which ensure that fusion occurs at the right time and space. How gB maintains its prefusion conformation and how gB fusogenicity is controlled remain poorly understood. Here, we report the isolation and characterization of a naturally selected pseudorabies virus (PrV) gB able to mediate efficient gH/gL-independent virus-cell and cell-cell fusion. We found that the control exerted on gB by the accompanying viral proteins is mediated via its cytosolic domain (CTD). Whereas gB variants lacking the CTD are inactive, a single mutation of a conserved asparagine residue in an alpha-helical motif of the ectodomain recently shown to be at the core of the gB prefusion trimer compensated for CTD absence and uncoupled gB from regulatory viral proteins, resulting in a hyperfusion phenotype. This phenotype was transferred to gB homologs from different alphaherpesvirus genera. Overall, our data propose a model in which the central helix acts as a molecular switch for the gB pre-to-postfusion transition by conveying the structural status of the endo- to the ectodomain, thereby governing their cross talk for fusion activation, providing a new paradigm for herpesvirus fusion regulation.IMPORTANCE The class III fusion protein glycoprotein B (gB) drives membrane fusion during entry and spread of herpesviruses. To mediate fusion, gB requires activation by the conserved gH/gL complex by a poorly defined mechanism. A detailed molecular-level understanding of herpesvirus membrane fusion is of fundamental virological interest and has considerable potential for the development of new therapeutics blocking herpesvirus cell invasion and spread. Using in vitro evolution and targeted mutagenesis of three different animal alphaherpesviruses, we identified a single conserved amino acid in a regulatory helix in the center of the gB ectodomain that enables efficient gH/gL-independent entry and plays a crucial role in the pre-to-postfusion transition of gB. Our results propose that the central helix is a key regulatory element involved in the intrastructural signal transduction between the endo- and ectodomain for fusion activation. This study expands our understanding of herpesvirus membrane fusion and uncovers potential targets for therapeutic interventions.


Assuntos
Aminoácidos/genética , Evolução Molecular Direcionada , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Aminoácidos/química , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Conformação Proteica , Células Vero , Proteínas do Envelope Viral/química
4.
Virus Res ; 261: 21-30, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30543872

RESUMO

Cyprinid herpesvirus 3 (CyHV-3) or koi herpesvirus is a global pathogen causing mass mortality in koi and common carp, against which improved vaccines are urgently needed. In this study we investigated the role of four nonessential, but immunogenic envelope glycoproteins encoded by members of the ORF25 gene family (ORF25, ORF65, ORF148 and ORF149) during CyHV-3 replication. Single deletion of ORF65 did not affect in vitro replication, and deletion of ORF148 even slightly enhanced virus growth on common carp brain (CCB) cells. Deletions of ORF25 or ORF149 led to reduced plaque sizes and virus titers, which was due to delayed entry into host cells. An ORF148/ORF149 double deletion mutant exhibited wild-type like growth indicating opposing functions of the two proteins. Electron microscopy of CCB cells infected with either mutant did not indicate any effects on virion formation and maturation in nucleus or cytoplasm, nor on release of enveloped particles. The ORF148, ORF149 and double deletion mutants were also tested in animal experiments using juvenile carp, and proved to be insufficiently attenuated for use as live virus vaccines. However, surviving fish were protected against challenge with wild-type CyHV-3, demonstrating that these antibody inducing proteins are dispensable for an efficient immune response in vivo.


Assuntos
Doenças dos Peixes/prevenção & controle , Deleção de Genes , Glicoproteínas/metabolismo , Infecções por Herpesviridae/veterinária , Herpesviridae/fisiologia , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Carpas , Núcleo Celular/virologia , Células Cultivadas , Citoplasma/virologia , Doenças dos Peixes/patologia , Doenças dos Peixes/virologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Herpesviridae/genética , Herpesviridae/imunologia , Herpesviridae/ultraestrutura , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Microscopia Eletrônica , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Carga Viral , Ensaio de Placa Viral , Vírion/ultraestrutura , Virulência
5.
J Virol ; 92(12)2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29618646

RESUMO

Herpesvirus membrane fusion depends on the core fusion machinery, comprised of glycoproteins B (gB) and gH/gL. Although gB structurally resembles autonomous class III fusion proteins, it strictly depends on gH/gL to drive membrane fusion. Whether the gH/gL complex needs to be membrane anchored to fulfill its function and which role the gH cytoplasmic (CD) and transmembrane domains (TMD) play in fusion is unclear. While the gH CD and TMD play an important role during infection, soluble gH/gL of herpes simplex virus 1 (HSV-1) seems to be sufficient to mediate cell-cell fusion in transient assays, arguing against an essential contribution of the CD and TMD. To shed more light on this apparent discrepancy, we investigated the role of the CD and TMD of the related alphaherpesvirus pseudorabies virus (PrV) gH. For this purpose, we expressed C-terminally truncated and soluble gH and replaced the TMD with a glycosylphosphatidylinositol (gpi) anchor. We also generated chimeras containing the TMD and/or CD of PrV gD or HSV-1 gH. Proteins were characterized in cell-based fusion assays and during virus infection. Although truncation of the CD resulted in decreased membrane fusion activity, the mutant proteins still supported replication of gH-negative PrV, indicating that the PrV gH CD is dispensable for viral replication. In contrast, PrV gH lacking the TMD, membrane-anchored via a lipid linker, or comprising the PrV gD TMD were nonfunctional, highlighting the essential role of the gH TMD for function. Interestingly, despite low sequence identity, the HSV-1 gH TMD could substitute for the PrV gH TMD, pointing to functional conservation.IMPORTANCE Enveloped viruses depend on membrane fusion for virus entry. While this process can be mediated by only one or two proteins, herpesviruses depend on the concerted action of at least three different glycoproteins. Although gB has features of bona fide fusion proteins, it depends on gH and its complex partner, gL, for fusion. Whether gH/gL prevents premature fusion or actively triggers gB-mediated fusion is unclear, and there are contradictory results on whether gH/gL function requires stable membrane anchorage or whether the ectodomains alone are sufficient. Our results show that in pseudorabies virus gH, the transmembrane anchor plays an essential role for gB-mediated fusion while the cytoplasmic tail is not strictly required.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Fusão de Membrana/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Domínios Proteicos/genética , Coelhos , Proteínas do Envelope Viral/genética , Replicação Viral/genética
6.
J Virol ; 92(9)2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29437979

RESUMO

Many viral envelope proteins are modified by asparagine (N)-linked glycosylation, which can influence their structure, physicochemical properties, intracellular transport, and function. Here, we systematically analyzed the functional relevance of N-linked glycans in the alphaherpesvirus pseudorabies virus (PrV) glycoprotein H (gH), which is an essential component of the conserved core herpesvirus fusion machinery. Upon gD-mediated receptor binding, the heterodimeric complex of gH and gL activates gB to mediate fusion of the viral envelope with the host cell membrane for viral entry. gH contains five potential N-linked glycosylation sites at positions 77, 162, 542, 604, and 627, which were inactivated by conservative mutations (asparagine to glutamine) singly or in combination. The mutated proteins were tested for correct expression and fusion activity. Additionally, the mutated gH genes were inserted into the PrV genome for analysis of function during virus infection. Our results demonstrate that all five sites are glycosylated. Inactivation of the PrV-specific N77 or the conserved N627 resulted in significantly reduced in vitro fusion activity, delayed penetration kinetics, and smaller virus plaques. Moreover, substitution of N627 greatly affected transport of gH in transfected cells, resulting in endoplasmic reticulum (ER) retention and reduced surface expression. In contrast, mutation of N604, which is conserved in the Varicellovirus genus, resulted in enhanced in vitro fusion activity and viral cell-to-cell spread. These results demonstrate a role of the N-glycans in proper localization and function of PrV gH. However, even simultaneous inactivation of all five N-glycosylation sites of gH did not severely inhibit formation of infectious virus particles.IMPORTANCE Herpesvirus infection requires fusion of the viral envelope with cellular membranes, which involves the conserved fusion machinery consisting of gB and the heterodimeric gH/gL complex. The bona fide fusion protein gB depends on the presence of the gH/gL complex for activation. Viral envelope glycoproteins, such as gH, usually contain N-glycans, which can have a strong impact on their folding, transport, and functions. Here, we systematically analyzed the functional relevance of all five predicted N-linked glycosylation sites in the alphaherpesvirus pseudorabies virus (PrV) gH. Despite the fact that mutation of specific sites affected gH transport, in vitro fusion activity, and cell-to-cell spread and resulted in delayed penetration kinetics, even simultaneous inactivation of all five N-glycosylation sites of gH did not severely inhibit formation of infectious virus particles. Thus, our results demonstrate a modulatory but nonessential role of N-glycans for gH function.


Assuntos
Membrana Celular/metabolismo , Herpesvirus Suídeo 1/fisiologia , Fusão de Membrana/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Retículo Endoplasmático/virologia , Glicosilação , Coelhos , Replicação Viral
7.
J Virol ; 91(9)2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28228592

RESUMO

Several envelope glycoproteins are involved in herpesvirus entry into cells, direct cell-to-cell spread, and induction of cell fusion. The membrane fusion protein glycoprotein B (gB) and the presumably gB-activating heterodimer gH/gL are essential for these processes and conserved throughout the Herpesviridae However, after extended cell culture passage of gL-negative mutants of the alphaherpesvirus pseudorabies virus (PrV), phenotypic revertants could be isolated which had acquired spontaneous mutations affecting the gL-interacting N-terminal part of the gH ectodomain (gDH and gHB4.1) (B. G. Klupp and T. C. Mettenleiter, J Virol 73:3014-3022, 1999; C. Schröter, M. Vallbracht, J. Altenschmidt, S. Kargoll, W. Fuchs, B. G. Klupp, and T. C. Mettenleiter, J Virol 90:2264-2272, 2016). To investigate the functional relevance of this part of gH in more detail, we introduced an in-frame deletion of 66 codons at the 5' end of the plasmid-cloned gH gene (gH32/98). The N-terminal signal peptide was retained, and the deletion did not affect expression or processing of gH but abrogated its function in in vitro fusion assays. Insertion of the engineered gH gene into the PrV genome resulted in a defective mutant (pPrV-gH32/98K), which was incapable of entry and spread. Interestingly, in vitro activity of mutated gH32/98 was restored when it was coexpressed with hyperfusogenic gBB4.1, obtained from a passaged gL deletion mutant of PrV. Moreover, the entry and spread defects of pPrV-gH32/98K were compensated by the mutations in gBB4.1 in cis, as well as in trans, independent of gL. Thus, PrV gL and the gL-interacting domain of gH are not strictly required for function.IMPORTANCE Membrane fusion is crucial for infectious entry and spread of enveloped viruses. While many enveloped viruses require only one or two proteins for receptor binding and membrane fusion, herpesvirus infection depends on several envelope glycoproteins. Besides subfamily-specific receptor binding proteins, the core fusion machinery consists of the conserved fusion protein gB and the gH/gL complex. The role of the latter is unclear, but it is hypothesized to interact with gB for fusion activation. Using isogenic virus recombinants, we demonstrate here that gL and the gL-binding domain of PrV gH are not strictly required for membrane fusion during virus entry and spread when concomitantly mutations in gB are present which increase its fusogenicity. Thus, our results strongly support the notion of a functional gB-gH interaction during the fusion process.


Assuntos
Herpesvirus Suídeo 1/genética , Fusão de Membrana/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular , Herpesvirus Suídeo 1/metabolismo , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Coelhos , Deleção de Sequência/genética , Replicação Viral/genética
8.
J Virol ; 90(1): 421-32, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491153

RESUMO

UNLABELLED: Membrane fusion is indispensable for entry of enveloped viruses into host cells. The conserved core fusion machinery of the Herpesviridae consists of glycoprotein B (gB) and the gH/gL complex. Recently, crystal structures of gH/gL of herpes simplex virus 2 (HSV-2) and Epstein-Barr virus and of a core fragment of pseudorabies virus (PrV) gH identified four structurally conserved gH domains. To investigate functional conservation, chimeric genes encoding combinations of individual domains of PrV and herpes simplex virus 1 (HSV-1) gH were expressed in rabbit kidney cells, and their processing and transport to the cell surface, as well as activity in fusion assays including gB, gD, and gL of PrV or HSV-1, were analyzed. Chimeric gH containing domain I of HSV-1 and domains II to IV of PrV exhibited limited fusion activity in the presence of PrV gB and gD and HSV-1 gL, but not of PrV gL. More strikingly, chimeric gH consisting of PrV domains I to III and HSV-1 domain IV exhibited considerable fusion activity together with PrV gB, gD, and gL. Replacing PrV gB with the HSV-1 protein significantly enhanced this activity. A cell line stably expressing this chimeric gH supported replication of gH-deleted PrV. Our results confirm the specificity of domain I for gL binding, demonstrate functional conservation of domain IV in two alphaherpesviruses from different genera, and indicate species-specific interactions of this domain with gB. They also suggest that gH domains II and III might form a structural and functional unit which does not tolerate major substitutions. IMPORTANCE: Envelope glycoprotein H (gH) is essential for herpesvirus-induced membrane fusion, which is required for host cell entry and viral spread. Although gH is structurally conserved within the Herpesviridae, its precise role and its interactions with other components of the viral fusion machinery are not fully understood. Chimeric proteins containing domains of gH proteins from different herpesviruses can serve as tools to elucidate the molecular basis of gH function. The present study shows that the C-terminal part of human herpesvirus 1 (herpes simplex virus 1) gH can functionally substitute for the corresponding part of suid herpesvirus 1 (pseudorabies virus) gH, whereas other tested combinations proved to be nonfunctional. Interestingly, the exchangeable fragment included the membrane-proximal end of the gH ectodomain (domain IV), which is most conserved in sequence and structure and might be capable of transient membrane interaction during fusion.


Assuntos
Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Multimerização Proteica , Proteínas Recombinantes/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Herpesvirus Humano 1/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Coelhos , Proteínas Recombinantes/genética , Especificidade por Substrato , Proteínas do Envelope Viral/genética
9.
J Virol ; 90(5): 2285-93, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26656711

RESUMO

UNLABELLED: Herpesviruses infect cells using the conserved core fusion machinery composed of glycoprotein B (gB) and gH/gL. The gH/gL complex plays an essential but still poorly characterized role in membrane fusion and cell tropism. Our previous studies demonstrated that the conserved disulfide bond (DB) C278/C335 in domain II (D-II) of Epstein-Barr virus (EBV) gH has an epithelial cell-specific function, whereas the interface of D-II/D-III is involved in formation of the B cell entry complex by binding to gp42. To extend these studies, we compared gH of the alphaherpesvirus pseudorabies virus (PrV) with gH of the gammaherpesvirus EBV to identify functionally equivalent regions critical for gH function during entry. We identified several conserved amino acids surrounding the conserved DB that connects three central helices of D-III of PrV and EBV gH. The present study verified that the conserved DB and several contacting amino acids in D-III modulate cell surface expression and thereby contribute to gH function. In line with this finding, we found that DB C404/C439 and T401 are important for cell-to-cell spread and efficient entry of PrV. This parallel comparison between PrV and EBV gH function brings new insights into how gH structure impacts fusion function during herpesvirus entry. IMPORTANCE: The alphaherpesvirus PrV is known for its neuroinvasion, whereas the gammaherpesvirus EBV is associated with cancer of epithelial and B cell origin. Despite low amino acid conservation, PrV gH and EBV gH show strikingly similar structures. Interestingly, both PrV gH and EBV gH contain a structural motif composed of a DB and supporting amino acids which is highly conserved within the Herpesviridae. Our study verified that PrV gH uses a minimal motif with the DB as the core, whereas the DB of EBV gH forms extensive connections through hydrogen bonds to surrounding amino acids, ensuring the cell surface expression of gH/gL. Our study verifies that the comparative analysis of distantly related herpesviruses, such as PrV and EBV, allows the identification of common gH functions. In addition, we provide an understanding of how functional domains can evolve over time, resulting in subtle differences in domain structure and function.


Assuntos
Análise Mutacional de DNA , Herpesvirus Suídeo 1/fisiologia , Herpesvirus Humano 4/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Linhagem Celular , Sequência Conservada , Herpesvirus Suídeo 1/genética , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Análise de Sequência de DNA , Proteínas do Envelope Viral/genética
10.
J Virol ; 90(5): 2264-72, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26656712

RESUMO

UNLABELLED: Entry of herpesviruses depends on the combined action of viral glycoprotein B (gB) and the heterodimeric gH/gL complex, which are activated by binding of the virion to specific cellular receptors. While gB carries signatures of a bona fide fusion protein, efficient membrane fusion requires gH/gL. However, although gB and gH/gL are essential for entry, the alphaherpesvirus pseudorabies virus (PrV) is capable of limited cell-to-cell spread in the absence of gL. To understand gH/gL function in more detail, the limited spread of PrV-ΔgL was used for reversion analyses by serial cell culture passages. In a first experiment, an infectious gL-negative mutant in which gL function was replaced by generation of a gD-gH hybrid protein was isolated (B. G. Klupp and T. C. Mettenleiter, J Virol 73:3014-3022, 1999). In a second, independent experiment PrV-ΔgLPassB4.1, which also replicated productively without gL, was isolated. Sequence analysis revealed mutations in gH but also in gB and gD. In a transfection-based fusion assay, two amino acid substitutions in the N-terminal part of gH(B4.1) (L(70)P and W(103)R) were found to be sufficient to compensate for lack of gL, while mutations present in gB(B4.1) enhanced fusogenicity. Coexpression of gB(B4.1) with the homologous gH(B4.1) resulted in strongly increased syncytium formation, which was further augmented by truncation of the gB(B4.1) C-terminal 29 amino acids. Nevertheless, gH was still required for membrane fusion. Surprisingly, coexpression of gD(B4.1) blocked syncytium formation in the fusion assays, which could be attributed to a V(106)A substitution within the ectodomain of gD(B4.1). IMPORTANCE: In contrast to many other enveloped viruses, herpesviruses rely on the concerted action of four viral glycoproteins for membrane fusion during infectious entry. Although the highly conserved gB shows signatures of a fusion protein, for fusion induction it requires the gH/gL complex, whose role is still elusive. Here we demonstrated fusion activation by gH in the absence of gL after reversion analysis of gL-deleted pseudorabies virus. This gL-independent fusion activity depended on single amino acid exchanges affecting the gL-binding domain in gH, increasing fusogenicity in gB and allowing negative fusion regulation by gD. Thus, our results provide novel information on the interplay in the fusion machinery of herpesviruses.


Assuntos
Deleção de Genes , Herpesvirus Suídeo 1/fisiologia , Supressão Genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Análise Mutacional de DNA , DNA Viral/química , DNA Viral/genética , Herpesvirus Suídeo 1/genética , Coelhos , Seleção Genética , Análise de Sequência de DNA , Inoculações Seriadas
11.
J Gen Virol ; 96(8): 2349-2354, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25908778

RESUMO

Glycoprotein gH is essential for herpesvirus-induced membrane fusion during entry and cell-to-cell spread. Structural analyses of gH homologues revealed a conserved syntaxin-like bundle motif composed of three α-helices. Previous studies showed that targeted disruption of any of these helices strongly impaired maturation, cell surface expression and fusion activity of pseudorabies virus gH, as well as formation and spread of infectious virus. After passaging of one corresponding mutant (pPrV-gH-V275P) these replication defects were widely corrected by an adjacent spontaneous amino acid substitution (V271A). Although the doubly mutated gH was still non-functional in fusion assays, its targeted reinsertion into the cloned virus genome (pPrV-gH-V275P-V271A) led to a 200-fold increase in plaque sizes and 10,000-fold higher virus titres, compared with pPrV-gH-V275P. Thus, our results demonstrate that structural requirements for gH function in in vitro assays and virus replication are different, and that minor amounts of mature gH in virions are sufficient for productive replication.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Mutação de Sentido Incorreto , Pseudorraiva/virologia , Doenças dos Suínos/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Replicação Viral , Animais , Linhagem Celular , Herpesvirus Suídeo 1/química , Herpesvirus Suídeo 1/genética , Modelos Moleculares , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Suínos , Proteínas do Envelope Viral/metabolismo , Vírion/química , Vírion/genética , Vírion/fisiologia
12.
J Virol ; 89(2): 1364-76, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25392216

RESUMO

UNLABELLED: Enveloped viruses utilize membrane fusion for entry into, and release from, host cells. For entry, members of the Herpesviridae require at least three envelope glycoproteins: the homotrimeric gB and a heterodimer of gH and gL. The crystal structures of three gH homologues, including pseudorabies virus (PrV) gH, revealed four conserved domains. Domain II contains a planar ß-sheet ("fence") and a syntaxin-like bundle of three α-helices (SLB), similar to those found in eukaryotic fusion proteins, potentially executing an important role in gH function. To test this hypothesis, we introduced targeted mutations into the PrV gH gene, which either disrupt the helices of the SLB by introduction of proline residues or covalently join them by artificial intramolecular disulfide bonds between themselves, to the adjacent fence region, or to domain III. Disruption of either of the three α-helices of the SLB (A250P, V275P, V298P) severely affected gH function in in vitro fusion assays and replication of corresponding PrV mutants. Considerable defects in fusion activity of gH, as well as in penetration kinetics and cell-to-cell spread of PrV mutants, were also observed after disulfide linkage of two α-helices within the SLB (A284C-S291C) or between SLB and domain III (H251C-L432C), as well as by insertions of additional cysteine pairs linking fence, SLB, and domain III. In vitro fusion activity of mutated gH could be partly restored by reduction of the artificial disulfide bonds. Our results indicate that the structure and flexibility of the SLB are relevant for the function of PrV gH in membrane fusion. IMPORTANCE: Mutational analysis based on crystal structures of proteins is a powerful tool to understand protein function. Here, we continued our study of pseudorabies virus gH, a part of the core fusion machinery of herpesviruses. We previously showed that the "flap" region in domain IV of PrV gH is important for its function. We now demonstrate that mutations within domain II that interfere with integrity or flexibility of a syntaxin-like three-helix bundle also significantly impair gH function during fusion. These studies provide important insights into the structural requirements of gH for function in fusion.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Replicação Viral , Animais , Linhagem Celular , Análise Mutacional de DNA , Herpesvirus Suídeo 1/genética , Modelos Moleculares , Conformação Proteica , Estrutura Terciária de Proteína , Coelhos
13.
J Virol ; 88(22): 13064-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25187552

RESUMO

UNLABELLED: Membrane fusion in herpesviruses requires viral glycoproteins (g) gB and gH/gL. While gB is considered the actual fusion protein but is nonfusogenic per se, the function of gH/gL remains enigmatic. Crystal structures for different gH homologs are strikingly similar despite only moderate amino acid sequence conservation. A highly conserved sequence motif comprises the residues serine-proline-cysteine corresponding to positions 437 to 439 in pseudorabies virus (PrV) gH. The PrV-gH structure shows that proline(438) induces bending at the end of an alpha-helix, thereby placing cysteine(404) and cysteine(439) in juxtaposition to allow formation of a strictly conserved disulfide bond. However, PrV vaccine strain Bartha unexpectedly carries a serine at this conserved position. To test the influence of this substitution, we constructed different gH chimeras carrying proline or serine at position 438 in gH derived from either PrV strain Kaplan or strain Bartha. Mutants expressing gH with serine(438) showed reduced fusion activity in transient-fusion assays and during infection, with delayed penetration kinetics and a small-plaque phenotype which indicates that proline(438) is important for efficient fusion. A more drastic effect was observed when disulfide bond formation was completely blocked by mutation of cysteine(404) to serine. Although PrV expressing gHC(404)S was viable, plaque size and penetration kinetics were drastically reduced. Alteration of serine(438) to proline in gH of strain Bartha enhanced cell-to-cell spread and penetration kinetics, but restoration of full activity required additional alteration of aspartic acid to valine at position 59. IMPORTANCE: The role of the gH/gL complex in herpesvirus membrane fusion is still unclear. Structural studies predicted a critical role for proline(438) in PrV gH to allow the formation of a conserved disulfide bond and correct protein folding. Functional analyses within this study corroborated these structural predictions: mutation of this residue resulted in a drastic impairment of membrane fusion kinetics not only in vitro in transient transfection-fusion assays but also during virus infection. Elimination of formation of the disulfide bond yielded the same phenotype in transient assays but had a more drastic effect on virus replication. Thus, our studies add important information to structure-function analyses of herpesvirus gH.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Prolina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Análise Mutacional de DNA , Viabilidade Microbiana , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Prolina/genética , Conformação Proteica , Controle Social Formal , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral
14.
Arch Virol ; 159(12): 3257-68, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25150990

RESUMO

As a prerequisite for development of improved vaccines and diagnostic tools for control of the fish pathogen koi herpesvirus, or cyprinid herpesvirus 3 (CyHV-3), we have started to identify putative viral envelope and capsid proteins. The complete or partial CyHV-3 open reading frames ORF25, ORF65, ORF92, ORF99, ORF136, ORF138, ORF146, ORF148, and ORF149 were expressed as bacterial fusion proteins, which were then used for preparation of monospecific rabbit antisera. All of the sera that were obtained detected their target proteins in cells transfected with the corresponding eukaryotic expression plasmids. However, only the type I membrane proteins pORF25, pORF65, pORF99, pORF136 and pORF149 and the major capsid protein pORF92 were sufficiently abundant and immunogenic to permit unambiguous detection in CyHV-3-infected cells. In indirect immunofluorescence tests (IIFT), sera from naturally or experimentally CyHV-3-infected carp and koi predominantly reacted with cells transfected with expression plasmids encoding pORF25, pORF65, pORF148, and pORF149, which represent a family of related CyHV-3 membrane proteins. Moreover, several neutralizing monoclonal antibodies raised against CyHV-3 virions proved to be specific for pORF149 in IIFT of transfected cells and in immunoelectron microscopic analysis of CyHV-3 particles. Since pORF149 appears to be an immunorelevant envelope protein of CyHV-3, a recombinant baculovirus was generated for its expression in insect cells, and pORF149 was shown to be incorporated into pseudotyped baculovirus particles, which might be suitable as diagnostic tools or subunit vaccines.


Assuntos
Carpas/virologia , Herpesviridae/química , Herpesviridae/genética , Proteínas Estruturais Virais/análise , Proteínas Estruturais Virais/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Técnica Indireta de Fluorescência para Anticorpo
15.
J Virol ; 87(4): 2011-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23236073

RESUMO

Virus-like particles (VLPs) from hepatitis B and human papillomaviruses have been successfully used as preventative vaccines against these infectious agents. These VLPs consist of a self-associating capsid polymer formed from a single structure protein and are devoid of viral DNA. Since virions from herpesviruses consist of a large number of molecules of viral and cellular origin, generating VLPs from a subset of these would be a particularly arduous task. Therefore, we have adopted an alternative strategy that consists of producing DNA-free defective virus particles in a cell line infected by a herpesvirus mutant incapable of packaging DNA. We previously reported that an Epstein-Barr virus (EBV) mutant devoid of the terminal repeats (ΔTR) that act as packaging signals in herpesviruses produces substantial amounts of VLPs and of light particles (LPs). However, ΔTR virions retained some infectious genomes, and although these mutants had lost their transforming abilities, this poses potential concerns for clinical applications. Therefore, we have constructed a series of mutants that lack proteins involved in maturation and assessed their ability to produce viral DNA-free VLP/LPs. Some of the introduced mutations were deleterious for capsid maturation and virus production. However, deletion of BFLF1/BFRF1A or of BBRF1 resulted in the production of DNA-free VLPs/LPs. The ΔBFLF1/BFRF1A viruses elicited a potent CD4(+) T-cell response that was indistinguishable from the one obtained with wild-type controls. In summary, the defective particles produced by the ΔBFLF1/BFRF1A mutant fulfill the criteria of efficacy and safety expected from a preventative vaccine.


Assuntos
DNA Viral/genética , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Deleção de Genes , Herpesvirus Humano 4/fisiologia , Humanos , Proteínas de Membrana/deficiência , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas Virais , Montagem de Vírus
16.
J Virol ; 86(15): 8002-13, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623768

RESUMO

Glycoprotein H (gH) is an envelope protein conserved in the Herpesviridae. Together with glycoprotein B (gB), the heterodimeric complex of gH and glycoprotein L (gL) mediates penetration and direct viral cell-to-cell spread. In herpes simplex and pseudorabies virus (PrV), coexpression of gH/gL, gB, and gD induces membrane fusion to form polykaryocytes. The recently determined crystal structure of a core fragment of PrV gH revealed marked structural similarity to other gH proteins (M. Backovic et al., Proc. Natl. Acad. Sci. U. S. A. 107:22635-22640, 2010). Within the membrane-proximal part (domain IV), a conserved negatively charged surface loop (flap) is flanked by intramolecular disulfide bonds. Together with an N-linked carbohydrate moiety, this flap covers an underlying patch of hydrophobic residues. To investigate the functional relevance of these structures, nonconservative amino acid substitutions were introduced by site-directed mutagenesis. The mutated proteins were tested for correct expression, fusion activity, and functional complementation of gH-deleted PrV. Several single amino acid changes within the flap and the hydrophobic patch were tolerated, and deletion of the glycosylation site had only minor effects. However, multiple alanine substitutions within the flap or the hydrophobic patch led to significant defects. gH function was also severely affected by disruption of the disulfide bond at the C terminus of the flap and after introduction of cysteine pairs designed to bridge the central part of the flap with the hydrophobic patch. Interestingly, all mutated gH proteins were able to complement gH-deleted PrV, but fusion-deficient gH mutants resulted in a pronounced delay in virus entry.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Herpesvirus Suídeo 1/genética , Interações Hidrofóbicas e Hidrofílicas , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pseudorraiva/genética , Pseudorraiva/metabolismo , Proteínas do Envelope Viral/genética
17.
J Virol ; 84(16): 8153-62, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534865

RESUMO

Homologs of the pseudorabies virus (PrV) essential large tegument protein pUL36 are conserved throughout the Herpesviridae. pUL36 functions during transport of the nucleocapsid to and docking at the nuclear pore as well as during virion formation after nuclear egress in the cytoplasm. Deletion analyses revealed several nonessential regions within the 3,084-amino-acid PrV pUL36 (S. Böttcher, B. G. Klupp, H. Granzow, W. Fuchs, K. Michael, and T. C. Mettenleiter, J. Virol. 80:9910-9915, 2006; S. Böttcher, H. Granzow, C. Maresch, B. Möhl, B. G. Klupp, and T. C. Mettenleiter, J. Virol. 81:13403-13411, 2007), while the C-terminal 62 amino acids are essential for virus replication (K. Coller, J. Lee, A. Ueda, and G. Smith, J. Virol. 81:11790-11797, 2007). To identify additional functional domains, we performed random mutagenesis of PrV pUL36 by transposon-mediated insertion of a 15-bp linker. By this approach, 26 pUL36 insertion mutants were selected and tested in transient transfection assays for their ability to complement one-step growth and/or viral spread of a PrV UL36 null mutant. Ten insertion mutants in the N-terminal half and 10 in the C terminus complemented both, whereas six insertion mutants clustering in the center of the protein did not complement in either assay. Interestingly, several insertions within conserved parts yielded positive complementation, including those located within the essential C-terminal 62 amino acids. For 15 mutants that mediated productive replication, stable virus recombinants were isolated and further characterized by plaque assay, in vitro growth analysis, and electron microscopy. Except for three mutant viruses, most insertion mutants replicated like wild-type PrV. Two insertion mutants, at amino acids (aa) 597 and 689, were impaired in one-step growth and viral spread and exhibited a defect in virion maturation in the cytoplasm. In contrast, one functional insertion (aa 1800) in a region which otherwise yielded only nonfunctional insertion mutants was impaired in viral spread but not in one-step growth without a distinctive ultrastructural phenotype. In summary, these studies extend and refine previous analyses of PrV pUL36 and demonstrate the different sensitivities of different regions of the protein to functional loss by insertion.


Assuntos
Elementos de DNA Transponíveis , Herpesvirus Suídeo 1/fisiologia , Mutagênese Insercional , Proteínas Estruturais Virais/fisiologia , Replicação Viral , Animais , Linhagem Celular , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Citoplasma/ultraestrutura , Citoplasma/virologia , Deleção de Genes , Teste de Complementação Genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/ultraestrutura , Microscopia Eletrônica de Transmissão , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Coelhos , Transfecção , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética , Vírion/ultraestrutura
18.
J Gen Virol ; 91(Pt 4): 847-57, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19940061

RESUMO

Infectious laryngotracheitis is an important respiratory disease of chickens that is caused by an alphaherpesvirus [infectious laryngotracheitis virus (ILTV); Gallid herpesvirus 1]. As herpesvirus envelope glycoproteins are main targets of the humoral host immune response, they are of particular interest for development of vaccines, as well as of diagnostic tools. The conserved, N-glycosylated envelope protein gC has been identified as a major surface antigen of ILTV. To study the function of gC, we now isolated a gC-deleted ILTV recombinant as well as a gC rescuant after co-transfection of permissive chicken cells with virion DNA and transfer plasmids containing engineered subgenomic fragments. Like other alphaherpesvirus homologues, ILTV gC proved to be non-essential for replication. ILTV-DeltagC exhibited delayed penetration kinetics and slightly reduced plaque sizes in cultured chicken cells, whereas virus titres were not reduced significantly compared with wild-type or gC-rescued virus. In vivo studies revealed that ILTV-DeltagC is attenuated in chickens. However, infection with high doses of ILTV-DeltagC was still fatal for approximately 20 % of the animals, whereas wild-type or gC-rescued ILTV led to 50 % mortality. Interestingly, innate and specific immune responses against ILTV-DeltagC were not reduced but enhanced, and surviving chickens were protected completely against challenge infection. Furthermore, ILTV-DeltagC might serve as a basis for marker vaccines permitting differentiation between vaccinated and field-virus-infected animals, as gC-specific antibodies could be detected easily in sera of animals infected with wild-type ILTV.


Assuntos
Herpesvirus Galináceo 1/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Galinhas , Genoma Viral , Herpesvirus Galináceo 1/genética , Herpesvirus Galináceo 1/imunologia , Herpesvirus Galináceo 1/patogenicidade , Proteínas do Envelope Viral/imunologia , Virulência , Internalização do Vírus , Replicação Viral
19.
J Gen Virol ; 90(Pt 7): 1560-1568, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19297610

RESUMO

In the absence of the tegument protein pUL37, virion formation of pseudorabies virus (PrV) and herpes simplex virus type 1 (HSV-1) is severely impaired. Non-enveloped nucleocapsids accumulate in clusters in the cytoplasm, whereas only a few enveloped particles can be detected. Although a contribution of pUL37 to nuclear egress of HSV-1 has been suggested, the nuclear stages of morphogenesis are not impaired in PrV-DeltaUL37-infected cells. Moreover, HSV-1 pUL37 has been described as essential for replication, whereas PrV is able to replicate productively without pUL37, although to lower titres than wild-type virus. Thus, there may be functional differences between the respective pUL37 proteins. This study compared the phenotypes of UL37-deleted PrV and HSV-1 in parallel assays, using a novel pUL37 deletion mutant of HSV-1 strain KOS, HSV-1DeltaUL37[86-1035]. Aggregates of seemingly 'naked' nucleocapsids were present in the cytoplasm of African green monkey (Vero) or rabbit kidney (RK13) cells infected with HSV-1DeltaUL37[86-1035] or PrV-DeltaUL37. Nuclear retention of nucleocapsids was not observed in either virus. However, in contrast to PrV-DeltaUL37, HSV-1DeltaUL37[86-1035] was unable to replicate productively in, and to form plaques on, either Vero or RK13 cells. Trans-complementation of respective deletion mutants with the heterologous pUL37 did not ensue. These data demonstrate that the conserved pUL37 in HSV-1 and PrV have similar but distinct functions.


Assuntos
Deleção de Genes , Herpesvirus Humano 1/fisiologia , Herpesvirus Suídeo 1/fisiologia , Proteínas Estruturais Virais/genética , Montagem de Vírus , Animais , Linhagem Celular , Núcleo Celular/virologia , Chlorocebus aethiops , Citoplasma/virologia , Teste de Complementação Genética , Herpesvirus Humano 1/genética , Herpesvirus Suídeo 1/genética , Nucleocapsídeo/metabolismo , Fenótipo , Coelhos , Ensaio de Placa Viral , Replicação Viral
20.
J Virol ; 83(7): 3389-96, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19144717

RESUMO

After fusion of the envelope of herpesvirus particles with the host cell plasma membrane, incoming nucleocapsids are transported to nuclear pores. Inner tegument proteins pUL36, pUL37, and pUS3 remain attached to the nucleocapsid after entry and therefore might mediate interactions between the nucleocapsid and cellular microtubule-associated motor proteins during transport. To assay for the role of pUL37 in this process, we constructed a pUL37-deleted pseudorabies virus mutant, PrV-DeltaUL37/UL35GFP, which expresses a fusion protein of green fluorescent protein (GFP) and the nonessential small capsid protein pUL35, resulting in the formation of fluorescently labeled capsids. Confocal laser-scanning microscopy of rabbit kidney cells infected with PrV-DeltaUL37/UL35GFP revealed that, whereas penetration was not affected in the absence of pUL37, nuclear translocation of incoming particles was delayed by approximately 1 h compared to PrV-UL35GFP, but not abolished. In contrast, phenotypically complemented pUL37-containing virions of PrV-DeltaUL37/UL35GFP exhibited wild type-like entry kinetics. Thus, the presence of pUL37 is required for rapid nuclear translocation of incoming nucleocapsids.


Assuntos
Capsídeo/metabolismo , Núcleo Celular/virologia , Herpesvirus Suídeo 1/fisiologia , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Herpesvirus Suídeo 1/genética , Microscopia Confocal , Transporte Proteico , Coelhos , Coloração e Rotulagem/métodos , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA