Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 6(39)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32978151

RESUMO

Cell entry of enveloped viruses requires specialized viral proteins that mediate fusion with the host membrane by substantial structural rearrangements from a metastable pre- to a stable postfusion conformation. This metastability renders the herpes simplex virus 1 (HSV-1) fusion glycoprotein B (gB) highly unstable such that it readily converts into the postfusion form, thereby precluding structural elucidation of the pharmacologically relevant prefusion conformation. By identification of conserved sequence signatures and molecular dynamics simulations, we devised a mutation that stabilized this form. Functionally locking gB allowed the structural determination of its membrane-embedded prefusion conformation at sub-nanometer resolution and enabled the unambiguous fit of all ectodomains. The resulting pseudo-atomic model reveals a notable conservation of conformational domain rearrangements during fusion between HSV-1 gB and the vesicular stomatitis virus glycoprotein G, despite their very distant phylogeny. In combination with our comparative sequence-structure analysis, these findings suggest common fusogenic domain rearrangements in all class III viral fusion proteins.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Herpesvirus Humano 1/genética , Humanos , Modelos Moleculares , Conformação Proteica , Internalização do Vírus
2.
Arch Virol ; 156(10): 1691-705, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21837416

RESUMO

Suid herpesvirus 1 (SuHV1, syn. Aujeszky's disease virus [ADV] or pseudorabies virus [PrV]), which belongs to the family Herpesviridae, subfamily Alphaherpesvirinae, genus Varicellovirus is the causative agent of Aujeszky's disease (AD, pseudorabies), a notifiable disease, that causes substantial economic losses to the swine industry in countries, where AD is present. Members of the family Suidae (true pigs) are the only natural hosts for PrV, although the virus can infect numerous other mammals including ruminants, carnivores and rodents. Despite the tremendous progress that has been made in controlling and eliminating PrV in domestic pigs, there is mounting evidence that PrV infections are more widespread in wild swine across the world than originally thought. Unfortunately, our understanding of the extent of PrV infections in these wild populations and of the threat to domestic swine is still fragmentary. This review aims at giving a global perspective on PrV infections in wild swine by scrutinizing the current state of knowledge concerning (i) the global occurrence of PrV infections in free-living populations of wild swine, e.g., wild boar and feral swine, (ii) the molecular characterization of wild swine PrV, (iii) infection characteristics of PrV in populations of wild swine, (iv) the risk of spillover infections to domestic pigs, (v) potential risk-mitigating measures, focusing on further research needs.


Assuntos
Herpesvirus Suídeo 1/isolamento & purificação , Pseudorraiva/epidemiologia , Doenças dos Suínos/epidemiologia , Animais , Animais Selvagens/virologia , Herpesvirus Suídeo 1/classificação , Herpesvirus Suídeo 1/genética , Filogenia , Pseudorraiva/virologia , Suínos , Doenças dos Suínos/virologia
3.
Epidemiol Infect ; 138(11): 1590-600, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20223047

RESUMO

Pseudorabies virus (PrV) infections appear to be more widely distributed in the European wild boar (Sus scrofa) population than assumed. In Europe, attempts to isolate and characterize the causative agents have been limited so far. We therefore collected and examined a total of 35 PrV isolates obtained from wild boar or hunting dogs in Germany, France, Spain, Italy, Slovakia and Hungary between 1993 and 2008. Restriction enzyme analysis of genomic DNA using BamHI showed that all isolates, except one, belonged to genogroup I but different subtypes were evident. For further investigations of the phylogenetic relationships, a 732-bp fragment of the glycoprotein C (gC) gene was amplified by PCR. Sequence analysis revealed about 40 variant positions within this fragment. Comparison of the nucleotide sequences supported the separation into a clade containing isolates from North-Rhine Westphalia, Rhineland-Palatinate (Germany), France and Spain (clade B) and an apparently more variable clade comprising isolates from Brandenburg, Baden-Wurttemberg, Saxony, Saxony-Anhalt (Germany), Slovakia, Hungary, Italy and France (clade A).


Assuntos
Doenças do Cão/virologia , Herpesvirus Suídeo 1/classificação , Pseudorraiva/virologia , Sus scrofa , Doenças dos Suínos/virologia , Sequência de Aminoácidos , Animais , Doenças do Cão/epidemiologia , Cães , Europa (Continente)/epidemiologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/isolamento & purificação , Dados de Sequência Molecular , Filogenia , Polimorfismo de Fragmento de Restrição , Pseudorraiva/epidemiologia , Suínos , Doenças dos Suínos/epidemiologia , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
J Virol ; 75(23): 11526-33, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689634

RESUMO

Glycoprotein B (gB) of pseudorabies virus (PrV) is essential for virus entry into target cells and direct viral cell-to-cell spread. Recently, we described a carboxy-terminally truncated derivative of PrV gB, gB-007, which was inefficiently incorporated into virions, was unable to complement infectivity, but was fully capable of restoring direct viral cell-to-cell spread of gB-negative PrV (R. Nixdorf, B. G. Klupp, and T. C. Mettenleiter, J. Virol. 74:7137-7145, 2000). Since recombinant PrV-007, which expresses gB-007 instead of wild-type gB, was able to spread directly from cell to cell, we attempted to obtain compensatory mutations leading to restoration of the entry defect by performing serial passages in cell culture. This procedure has previously been used to successfully restore entry defects in gD- or gL-deficient PrV mutants. From an initial titer of 100 PFU per ml in the supernatant, titers increased, reaching wild-type levels of up to 10(7) PFU after ca. 20 passages. One single-plaque isolate of the passaged mutant, designated PrV-007Pass, was further characterized. PrV-007Pass gB was efficiently incorporated into the viral envelope and restored infectivity to a gB-negative PrV mutant, PrV-gB(-). Interestingly, localization of PrV-007Pass gB in the plasma membrane was similar to that of PrV-007. In contrast, wild-type gB is mainly found in intracellular vesicles. Marker rescue experiments and trans-complementation assays demonstrated the presence of compensatory mutations within the gB gene of PrV-007Pass. DNA sequencing revealed two point mutations in the gB open reading frame of PrV-007Pass, resulting in amino acid substitutions at positions 305 and 744 of gB, both of which are required for compensation of the defect in PrV-007. Our data again demonstrate the power of reversion analysis of herpesviruses and suggest that cytosolic and ectodomains play a role in incorporation of gB into virions.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Mutação Puntual , Proteínas do Envelope Viral/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Genoma Viral , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Fenótipo , Testes de Precipitina , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral
5.
J Virol ; 75(21): 10054-64, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11581374

RESUMO

Infection of cells by herpesviruses is initiated by the interaction of viral envelope glycoproteins with cellular receptors. In the alphaherpesvirus pseudorabies virus (PrV), the causative agent of Aujeszky's disease in pigs, the essential glycoprotein D (gD) mediates secondary attachment of virions to target cells by binding to newly identified cellular receptors (R. J. Geraghty, C. Krummenacher, G. H. Cohen, R. J. Eisenberg, and P. G. Spear, Science 280:1618-1620, 1998). However, in the presence of compensatory mutations, infection can also occur in the absence of gD, as evidenced by the isolation in cell culture of an infectious gD-negative PrV mutant (PrV-gD(-) Pass) (J. Schmidt, B. G. Klupp, A. Karger, and T. C. Mettenleiter, J. Virol. 71:17-24, 1997). PrV-gD(-) Pass is replication competent with an only moderate reduction in specific infectivity but appears to bind to receptors different from those recognized by wild-type PrV (A. Karger, J. Schmidt, and T. C. Mettenleiter, J. Virol. 72:7341-7348, 1998). To analyze whether this alteration in receptor usage in vitro influences infection in vivo, the model host mouse and the natural host pig were intranasally infected with PrV-gD(-) Pass and were compared to animals infected by wild-type PrV. For mice, a comparable progress of disease was observed, and all animals infected with mutant virus died, although they exhibited a slight delay in the onset of symptoms and, correspondingly, a longer time to death. In contrast, whereas wild-type PrV-infected pigs showed clinical signs and histological and histopathological findings typical of PrV infection, no signs of disease were observed after infection with PrV-gD(-) Pass. Moreover, in these animals, virus-infected cells were not detectable by immunohistochemical staining of different organ samples and no virus could be isolated from nasal swabs. Mutations in glycoproteins B and H were found to correlate with, and probably contribute to, gD-independent infectivity. In conclusion, although PrV-gD(-) Pass is virulent in mice, it is apparently unable to infect the natural host, the pig. This altered host range in vivo correlates with a difference of receptor usage in vitro and demonstrates for the first time the importance of gD receptors in alphaherpesvirus infection of an animal host.


Assuntos
Herpesvirus Suídeo 1/patogenicidade , Mutação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/fisiologia , Suínos , Virulência
6.
J Virol ; 75(22): 11137-45, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602753

RESUMO

Penetration and propagation of herpesviruses in the nervous system require the action of several glycoproteins. To assay for a function of glycoproteins gC, gK, and gL in the neuroinvasiveness of pseudorabies virus (PrV), deletion mutants lacking one of these glycoproteins and corresponding rescuants were inoculated in the nasal cavity of adult mice. We demonstrate that the lack of gL almost prevented the virus from penetrating and propagating in trigeminal, sympathetic, and parasympathetic tracks innervating the nasal cavity, while the lack of gC and gK only slowed the invasion of the nervous system. The conclusion of this and previous studies is that only gB, gD, gH, and gL are indispensable for penetration into neurons, while gB, gH, and gL (and, in some categories of neurons, also gE and gI) are necessary for transneuronal transfer in the mouse model. The deletion of other glycoprotein genes has little effect on PrV neuroinvasiveness although it may affect the dissemination of the virus.


Assuntos
Herpesvirus Suídeo 1/patogenicidade , Neurônios/virologia , Proteínas do Envelope Viral/fisiologia , Administração Intranasal , Animais , Genótipo , Herpesvirus Suídeo 1/genética , Camundongos , Mutação , Proteínas do Envelope Viral/genética
7.
J Virol ; 75(19): 8927-36, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11533156

RESUMO

Herpesvirus envelopment is a two-step process which includes acquisition of a primary envelope resulting from budding of intranuclear capsids through the inner nuclear membrane. Fusion with the outer leaflet of the nuclear membrane releases nucleocapsids into the cytoplasm, which then gain their final envelope by budding into trans-Golgi vesicles. It has been shown that the UL34 gene product is required for primary envelopment of the alphaherpesvirus pseudorabies virus (PrV) (B. G. Klupp, H. Granzow, and T. C. Mettenleiter, J. Virol. 74:10063-10073, 2000). For secondary envelopment, several virus-encoded PrV proteins are necessary, including glycoproteins E, I, and M (A. R. Brack, J. M. Dijkstra, H. Granzow, B. G. Klupp, and T. C. Mettenleiter, J. Virol. 73:5364-5372, 1999). We show here that the product of the UL37 gene of PrV, which is a constituent of mature virions, is involved in secondary envelopment. Replication of a UL37 deletion mutant, PrV-DeltaUL37, was impaired in normal cells; this defect could be complemented on cells stably expressing UL37. Ultrastructural analysis demonstrated that intranuclear capsid maturation and budding of capsids into and release from the perinuclear space were unimpaired. However, secondary envelopment was drastically reduced. Instead, apparently DNA-filled capsids accumulated in the cytoplasm in large aggregates similar to those observed in the absence of glycoproteins E/I and M but lacking the surrounding electron-dense tegument material. Although displaying an ordered structure, capsids did not contact each other directly. We postulate that the UL37 protein is necessary for correct addition of other tegument proteins, which are required for secondary envelopment. In the absence of the UL37 protein, capsids interact with each other through unknown components but do not acquire the electron-dense tegument which is normally found around wild-type capsids during and after secondary envelopment. Thus, apposition of the UL37 protein to cytoplasmic capsids may be crucial for the addition of other tegument proteins, which in turn are able to interact with viral glycoproteins to mediate secondary envelopment.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas Estruturais Virais/fisiologia , Montagem de Vírus/fisiologia , Animais , Linhagem Celular , Coelhos , Vírion/fisiologia
8.
J Virol ; 75(8): 3675-84, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11264357

RESUMO

Egress of four important alphaherpesviruses, equine herpesvirus 1 (EHV-1), herpes simplex virus type 1 (HSV-1), infectious laryngotracheitis virus (ILTV), and pseudorabies virus (PrV), was investigated by electron microscopy of infected cell lines of different origins. In all virus-cell systems analyzed, similar observations were made concerning the different stages of virion morphogenesis. After intranuclear assembly, nucleocapsids bud at the inner leaflet of the nuclear membrane, resulting in enveloped particles in the perinuclear space that contain a sharply bordered rim of tegument and a smooth envelope surface. Egress from the perinuclear cisterna primarily occurs by fusion of the primary envelope with the outer leaflet of the nuclear membrane, which has been visualized for HSV-1 and EHV-1 for the first time. The resulting intracytoplasmic naked nucleocapsids are enveloped at membranes of the trans-Golgi network (TGN), as shown by immunogold labeling with a TGN-specific antiserum. Virions containing their final envelope differ in morphology from particles within the perinuclear cisterna by visible surface projections and a diffuse tegument. Particularly striking was the addition of a large amount of tegument material to ILTV capsids in the cytoplasm. Extracellular virions were morphologically identical to virions within Golgi-derived vesicles, but distinct from virions in the perinuclear space. Studies with gB- and gH-deleted PrV mutants indicated that these two glycoproteins, which are essential for virus entry and direct cell-to-cell spread, are dispensable for egress. Taken together, our studies indicate that the deenvelopment-reenvelopment process of herpesvirus maturation also occurs in EHV-1, HSV-1, and ILTV and that membrane fusion processes occurring during egress are substantially different from those during entry and direct viral cell-to-cell spread.


Assuntos
Alphaherpesvirinae/crescimento & desenvolvimento , Alphaherpesvirinae/ultraestrutura , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Deleção de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/ultraestrutura , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/crescimento & desenvolvimento , Herpesvirus Equídeo 1/metabolismo , Herpesvirus Equídeo 1/ultraestrutura , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/metabolismo , Herpesvirus Suídeo 1/ultraestrutura , Microscopia Eletrônica , Nucleocapsídeo/metabolismo , Nucleocapsídeo/ultraestrutura , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/ultraestrutura , Montagem de Vírus , Rede trans-Golgi/metabolismo , Rede trans-Golgi/ultraestrutura , Rede trans-Golgi/virologia
9.
J Virol ; 74(21): 10063-73, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11024135

RESUMO

Primary envelopment of several herpesviruses has been shown to occur by budding of intranuclear capsids through the inner nuclear membrane. By subsequent fusion of the primary envelope with the outer nuclear membrane, capsids are released into the cytoplasm and gain their final envelope by budding into vesicles in the trans-Golgi area. We show here that the product of the UL34 gene of pseudorabies virus, an alphaherpesvirus of swine, is localized in transfected and infected cells in the nuclear membrane. It is also detected in the envelope of virions in the perinuclear space but is undetectable in intracytoplasmic and extracellular enveloped virus particles. Conversely, the tegument protein UL49 is present in mature virus particles and absent from perinuclear virions. In the absence of the UL34 protein, acquisition of the primary envelope is blocked and neither virus particles in the perinuclear space nor intracytoplasmic capsids or virions are observed. However, light particles which label with the anti-UL49 serum are formed in the cytoplasm. We conclude that the UL34 protein is required for primary envelopment, that the primary envelope is biochemically different from the final envelope in that it contains the UL34 protein, and that perinuclear virions lack the tegument protein UL49, which is present in mature virions. Thus, we provide additional evidence for a two-step envelopment process in herpesviruses.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Membrana Nuclear/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Herpesvirus Suídeo 1/genética , Dados de Sequência Molecular , Mutação , Membrana Nuclear/virologia , Coelhos , Transfecção , Proteínas Virais/química , Proteínas Virais/genética , Vírion/metabolismo , Replicação Viral
10.
J Virol ; 74(15): 6760-8, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10888614

RESUMO

A transient transfection-fusion assay was established to investigate membrane fusion mediated by pseudorabies virus (PrV) glycoproteins. Plasmids expressing PrV glycoproteins under control of the immediate-early 1 promoter-enhancer of human cytomegalovirus were transfected into rabbit kidney cells, and the extent of cell fusion was quantitated 27 to 42 h after transfection. Cotransfection of plasmids encoding PrV glycoproteins B (gB), gD, gH, and gL resulted in formation of polykaryocytes, as has been shown for homologous proteins of herpes simplex virus type 1 (HSV-1) (A. Turner, B. Bruun, T. Minson, and H. Browne, J. Virol. 72:873-875, 1998). However, in contrast to HSV-1, fusion was also observed when the gD-encoding plasmid was omitted, which indicates that PrV gB, gH, and gL are sufficient to mediate fusion. Fusogenic activity was enhanced when a carboxy-terminally truncated version of gB (gB-008) lacking the C-terminal 29 amino acids was used instead of wild-type gB. With gB-008, only gH was required in addition for fusion. A very rapid and extended fusion was observed after cotransfection of plasmids encoding gB-008 and gDH, a hybrid protein consisting of the N-terminal 271 amino acids of gD fused to the 590 C-terminal amino acids of gH. This protein has been shown to substitute for gH, gD, and gL function in the respective viral mutants (B. G. Klupp and T. C. Mettenleiter, J. Virol. 73:3014-3022, 1999). Cotransfection of plasmids encoding PrV gC, gE, gI, gK, and UL20 with gB-008 and gDH had no effect on fusion. However, inclusion of a gM-expressing plasmid strongly reduced the extent of fusion. An inhibitory effect was also observed after inclusion of plasmids encoding gM homologs of equine herpesvirus 1 or infectious laryngotracheitis virus but only in conjunction with expression of the gM complex partner, the gN homolog. Inhibition by PrV gM was not limited to PrV glycoprotein-mediated fusion but also affected fusion induced by the F protein of bovine respiratory syncytial virus, indicating a general mechanism of fusion inhibition by gM.


Assuntos
Herpesvirus Suídeo 1/metabolismo , Fusão de Membrana/efeitos dos fármacos , Proteínas do Envelope Viral/fisiologia , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Animais , Bovinos , Células Cultivadas , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/metabolismo , Herpesvirus Suídeo 1/genética , Humanos , Plasmídeos/genética , Coelhos , Transfecção , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
11.
J Virol ; 74(15): 7137-45, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10888654

RESUMO

Glycoproteins homologous to the type I membrane glycoprotein B (gB) of herpes simplex virus 1 (HSV-1) are the most highly conserved glycoproteins within the family Herpesviridae and are present in members of each herpesvirus subfamily. In the alphaherpesvirus pseudorabies virus (PrV), gB is required for entry into target cells and for direct viral cell-to-cell spread. These processes, though related, appear to be distinct, and thus it was interesting to analyze whether they require different functions of gB. To this end, we established cell lines stably expressing different carboxy-terminally truncated versions of PrV gB by deleting either (i) one predicted intracytoplasmic alpha-helical domain encompassing putative YQRL and dileucine internalization signals, (ii) two predicted intracytoplasmic alpha-helical domains, (iii) the complete intracytoplasmic domain, or (iv) the intracytoplasmic domain and the transmembrane anchor region. Confocal laser scanning microscopy showed that gB derivatives lacking at least the last 29 amino acids (aa) localize close to the plasma membrane, while the full-length protein accumulates in intracellular aggregations. Trans-complementation studies with a gB-deleted PrV (PrV-gB(-)) demonstrated that the 29-aa truncated form lacking the putative internalization signals and the C-terminal alpha-helical domain (gB-008) was efficiently incorporated into PrV-gB(-) virions and efficiently complemented infectivity and cell-to-cell spread. Moreover, gB-008 exhibited an enhanced fusogenic activity. In contrast, gB proteins lacking both alpha-helical domains (gB-007), the complete intracytoplasmic domain, or the intracytoplasmic domain and transmembrane anchor were only inefficiently or not at all incorporated into PrV-gB(-) virions and did not complement infectivity. However, gB-007 was able to mediate cell-to-cell spread of PrV-gB(-). Similar phenotypes were observed when virus recombinants expressing gB-008 or gB-007, respectively, instead of wild-type gB were isolated and analyzed. Thus, our data show that internalization of gB is not required for gB incorporation into virions nor for its function in either entry or cell-to-cell spread. Moreover, they indicate different requirements for gB in these membrane fusion processes.


Assuntos
Herpesvirus Suídeo 1/patogenicidade , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Teste de Complementação Genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Coelhos , Recombinação Genética , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Vírion/metabolismo
12.
J Virol ; 74(11): 5083-90, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10799582

RESUMO

Glycoprotein K (gK) of pseudorabies virus (PrV) has recently been identified as a virion component which is dispensable for viral entry but required for direct cell-to-cell spread. Electron microscopic data suggested a possible function of gK in virus egress by preventing immediate fusion of released virus particles with the plasma membrane (B. G. Klupp, J. Baumeister, P. Dietz, H. Granzow, and T. C. Mettenleiter, J. Virol. 72:1949-1958, 1998). For more detailed analysis, a PrV mutant with a deletion of the UL53 (gK) open reading frame (ORF) from codons 48 to 275 was constructed, and the protein was analyzed with two monoclonal antibodies directed against PrV gK. The salient findings of this report are as follows. (i) From the PrV UL53 ORF, a functional gK is translated only from the first in-frame methionine. From the second in-frame methionine, a nonfunctional product is expressed which is not incorporated into virions. (ii) When constitutively expressed in a stable cell line without other viral proteins, gK is only incompletely processed. After superinfection with gK-deletion mutants, proper processing is restored and mature gK is incorporated into virions. (iii) The UL20 gene product is specifically required for processing of gK. gK is not correctly processed in a UL20 deletion mutant of PrV, and superinfection of gK-expressing cells with PrV-UL20(-) does not restore processing. However, all other known structural viral glycoproteins appear to be processed normally in PrV-UL20(-)-infected cells. (iv) Coexpression of gK and UL20 restored gK processing at least partially. Thus, our data show that the UL20 gene product is required for proper processing of PrV gK.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Cinética , Mutagênese , Fenótipo , Coelhos , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Vírion
13.
J Gen Virol ; 81(Pt 6): 1539-43, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10811937

RESUMO

The DNA sequence of a 2.4 kbp fragment located in the internal and terminal inverted repeat sequences of the pseudorabies virus genome determined in this study closes a gap between the previously described genes for the ICP4 and ICP22 homologues. The novel sequence contains no conserved herpesvirus open reading frames. Northern blot and cDNA analyses revealed a viral immediate-early transcript of 1.8 kb, which is spliced by the removal of two small introns close to its 5' end and which presumably represents the mRNA of the downstream open reading frame encoding the ICP22 homologue. Upstream of the transcribed region, an imperfect set of three directly repeated sequences was identified. Each of them contains a complementary pair of the alphaherpesvirus origin-binding protein recognition motif GTTCGCAC, spaced by AT-rich sequences. In vitro studies confirmed that the DNA fragment analysed includes a functional origin of viral DNA replication.


Assuntos
Genes Precoces , Herpesvirus Suídeo 1/genética , Sequências Repetitivas de Ácido Nucleico , Origem de Replicação , Sequências Repetidas Terminais , Animais , Sequência de Bases , Linhagem Celular , DNA Viral , Genoma Viral , Dados de Sequência Molecular , Suínos , Transcrição Gênica
14.
J Virol ; 74(9): 4004-16, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10756012

RESUMO

Glycoproteins M (gM), E (gE), and I (gI) of pseudorabies virus (PrV) are required for efficient formation of mature virions. The simultaneous absence of gM and the gE/gI complex results in severe deficiencies in virion morphogenesis and cell-to-cell spread, leading to drastically decreased virus titers and a small-plaque phenotype (A. Brack, J. Dijkstra, H. Granzow, B. G. Klupp, and T. C. Mettenleiter, J. Virol. 73:5364-5372, 1999). Serial passaging in noncomplementing cells of a virus mutant unable to express gM, gE, and gI resulted in a reversion of the small-plaque phenotype and restoration of infectious virus formation to the level of a gM(-) mutant. Genetic analyses showed that reversion of the phenotype was accompanied by a genomic rearrangement which led to the fusion of a portion of the gE gene encoding the cytoplasmic domain to the 3' end of the glycoprotein D gene, resulting in expression of a chimeric gD-gE protein. Since this indicated that the intracytoplasmic domain of gE was responsible for the observed phenotypic alterations, the UL10 (gM) gene was deleted in a PrV mutant, PrV-107, which specifically lacked the cytoplasmic tail of gE. Regarding one-step growth, plaque size, and virion formation as observed under the electron microscope, the mutant lacking gM and the gE cytoplasmic tail proved to be very similar to the gE/I/M triple mutant. Thus, our data indicate that it is the cytoplasmic tail of gE which is responsible for the observed phenotypic effects in conjunction with deletion of gM. We hypothesize that the cytoplasmic domain of gE specifically interacts with components of the capsid and/or tegument, leading to efficient secondary envelopment of intracytoplasmic capsids.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/fisiologia , Montagem de Vírus/fisiologia , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Genoma Viral , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/ultraestrutura , Corpos de Inclusão Viral , Mutagênese , Fenótipo , Coelhos , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Vírion/fisiologia
15.
J Virol ; 73(7): 5364-72, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364283

RESUMO

Glycoprotein M (gM), the product of the UL10 gene of pseudorabies virus (PrV), is one of the few nonessential glycoproteins conserved throughout the Herpesviridae. In contrast to wild-type PrV strains, the UL10 gene product of the attenuated PrV vaccine strain Bartha (PrV-Ba) is not modified by N-glycans due to a mutation in the DNA sequence encoding the consensus N-glycosylation motif. To assay function of the UL10 protein in PrV-Ba, a UL10-deletion mutant (PrV-Ba-UL10(-)) was isolated. Surprisingly, in contrast to gM-deleted wild-type PrV, PrV-Ba-UL10(-) was severely impaired in plaque formation, inducing only foci of very few infected RK13, Vero, and PSEK cells and tiny plaques on MDBK cells. Since this effect was significantly more dramatic than in wild-type PrV, additional mutations known to be present in PrV-Ba were analyzed for their contribution to this phenotype. trans-complementation of the mutated PrV-Ba UL21 or gC protein by the wild-type version had no influence on the observed phenotype. In contrast, complementation of the gE/gI deletion rescued the phenotype. The synergistic effect of deletions in gE/gI and gM on plaque size was verified by construction of a gE/I/M triple mutant derived from wild-type PrV which exhibited the same phenotype. The dramatic effect of deletion of gM on plaque size in a gE/I- virus background was mainly attributable to a function of gM, and not of the gM/gN complex, as shown by analysis of a gE/I/N triple mutant. Interestingly, despite the strong effect on plaque size, penetration was not significantly impaired. In noncomplementing cells infected with the gE/I/M triple mutant, electron microscopy showed absence of secondary envelopment in the cytoplasm but occurrence of intracytoplasmic accumulations of nucleocapsids in association with electron dense material, presumably tegument proteins. These structures were not observed after infection of cells expressing either gE/I or gM. We suggest that gE/I and gM are required for late stages in virion morphogenesis prior to final envelopment in the cytoplasm.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/fisiologia , Montagem de Vírus , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Deleção de Genes , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/ultraestrutura , Microscopia Eletrônica , Coelhos , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Vírion/fisiologia , Vírion/ultraestrutura
16.
J Virol ; 73(4): 3014-22, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10074151

RESUMO

Envelope glycoproteins gH and gL, which form a complex, are conserved throughout the family Herpesviridae. The gH-gL complex is essential for the fusion between the virion envelope and the cellular cytoplasmic membrane during penetration and is also required for direct viral cell-to-cell spread from infected to adjacent noninfected cells. It has been proposed for several herpesviruses that gL is required for proper folding, intracellular transport, and virion localization of gH. In pseudorabies virus (PrV), glycoprotein gL is necessary for infectivity but is dispensable for virion localization of gH. A virus mutant lacking gL, PrV-DeltagLbeta, is defective in entry into target cells, and direct cell-to-cell spread is drastically reduced, resulting in only single or small foci of infected cells (B. G. Klupp, W. Fuchs, E. Weiland, and T. C. Mettenleiter, J. Virol. 71:7687-7695, 1997). We used this limited cell-to-cell spreading ability of PrV-DeltagLbeta for serial passaging of cells infected with transcomplemented virus by coseeding with noninfected cells. After repeated passaging, plaque formation was restored and infectivity in the supernatant was observed. One single-plaque isolate, designated PrV-DeltagLPass, was further characterized. To identify the mutation leading to this gL-independent infectious phenotype, Southern and Western blot analyses, radioimmunoprecipitations, and DNA sequencing were performed. The results showed that rearrangement of a genomic region comprising part of the gH gene into a duplicated copy of part of the unique short region resulted in a fusion fragment predicted to encode a protein consisting of the N-terminal 271 amino acids of gD fused to the C-terminal 590 residues of gH. Western blotting and radioimmunoprecipitation with gD- and gH-specific antibodies verified the presence of a gDH fusion protein. To prove that this fusion protein mediates infectivity of PrV-DeltagLPass, cotransfection of PrV-DeltagLbeta DNA with the cloned fusion fragment was performed, and a cell line, Nde-67, carrying the fusion gene was established. After cotransfection, infectious gL-negative PrV was recovered, and propagation of PrV-DeltagLbeta on Nde-67 cells produced infectious virions. Thus, a gDH fusion polypeptide can compensate for function of the essential gL in entry and cell-to-cell spread of PrV.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas Virais de Fusão/fisiologia , Proteínas Virais/fisiologia , Animais , Chlorocebus aethiops , Regulação Viral da Expressão Gênica , Herpesvirus Suídeo 1/patogenicidade , Células Vero , Virulência/genética , Replicação Viral
17.
J Gen Virol ; 79 ( Pt 4): 851-4, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9568981

RESUMO

Glycoprotein M (gM) constitutes one of the rare examples of a nonessential glycoprotein conserved throughout all herpesvirus subfamilies. Whereas gM in wild-type pseudorabies virus (PrV) strains carries an N-glycan, gM of the attenuated strain Bartha is not glycosylated due to a point mutation in the N-glycosylation motif. Since PrV Bartha lacks glycoproteins E and I and carries a mutated gC, we analysed glycosylation of gM in isogenic PrV glycoprotein deletion mutants. Whereas gM was glycosylated normally in most mutants, two independent gC deletion mutants and a gI mutant expressed a nonglycosylated form of gM. DNA sequence analyses revealed the presence of point mutations in the N-glycosylation consensus motif. Surprisingly, mutations in strain Bartha, the two gC-deletion mutants and the gI mutant proved to be different, although all affected the N-glycosylation motif. Thus, our data show that different, apparently independent point mutations cause expression of nonglycosylated gM.


Assuntos
Herpesvirus Suídeo 1/genética , Mutação Puntual , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Sequência Conservada , DNA Viral/genética , Expressão Gênica , Genes Virais , Glicosilação , Dados de Sequência Molecular , Coelhos
18.
J Virol ; 72(3): 1949-58, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9499048

RESUMO

The pseudorabies virus (PrV) gene homologous to herpes simplex virus type 1 (HSV-1) UL53, which encodes HSV-1 glycoprotein K (gK), has recently been sequenced (J. Baumeister, B. G. Klupp, and T. C. Mettenleiter, J. Virol. 69:5560-5567, 1995). To identify the corresponding protein, a rabbit antiserum was raised against a 40-kDa glutathione S-transferase-gK fusion protein expressed in Escherichia coli. In Western blot analysis, this serum detected a 32-kDa polypeptide in PrV-infected cell lysates as well as a 36-kDa protein in purified virion preparations, demonstrating that PrV gK is a structural component of virions. After treatment of purified virions with endoglycosidase H, a 34-kDa protein was detected, while after incubation with N-glycosidase F, a 32-kDa protein was specifically recognized. This finding indicates that virion gK is modified by N-linked glycans of complex as well as high-mannose type. For functional analysis, the UL53 open reading frame was interrupted after codon 164 by insertion of a gG-lacZ expression cassette into the wild-type PrV genome (PrV-gKbeta) or by insertion of the bovine herpesvirus 1 gB gene into a PrV gB- genome (PrV-gK(gB)). Infectious mutant virus progeny was obtained only on complementing gK-expressing cells, suggesting that gK has an important function in the replication cycle. After infection of Vero cells with either gK mutant, only single infected cells or small foci of infected cells were visible. In addition, virus yield was reduced approximately 30-fold, and penetration kinetics showed a delay in entry which could be compensated for by phenotypic gK complementation. Interestingly, the plating efficiency of PrV-gKbeta was similar to that of wild-type PrV on complementing and noncomplementing cells, pointing to an essential function of gK in virus egress but not entry. Ultrastructurally, virus assembly and morphogenesis of PrV gK mutants in noncomplementing cells were similar to wild-type virus. However, late in infection, numerous nucleocapsids were found directly underneath the plasma membrane in stages typical for the entry process, a phenomenon not observed after wild-type virus infection and also not visible after infection of gK-complementing cells. Thus, we postulate that presence of gK is important to inhibit immediate reinfection.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Glicosilação , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/ultraestrutura , Mutagênese , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética , Vírion
19.
Virology ; 237(1): 113-22, 1997 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-9344913

RESUMO

Genes encoding homologs of the herpes simplex virus type 1 UL10 product, glycoprotein M, are conserved in all herpesviruses investigated so far. Recently, we identified pseudorabies virus (PrV) gM as a 45-kDa structural component of purified virions. A gM-PrV mutant could be propagated in cell culture, albeit at lower titers and with delayed penetration kinetics. Thus, gM has a nonessential but modulatory function in PrV infection. PrV gM is modified by addition of an N-linked glycan at a consensus sequence located between the predicted first and second hydrophobic region of the protein. This N-glycosylation site is conserved in all gM homologs sequenced so far, indicating an important functional role. To analyze intracellular processing of PrV gM, Western blot analyses were performed. In PrV-infected cells, mature 45-kDa gM as well as 33- and 35-kDa precursor forms were detectable. Presumably dimeric 90- and 70-kDa proteins were also observed. The 33- and 35-kDa proteins represent nonglycosylated and glycosylated precursors as shown by endoglycosidase digestions. Investigation of several PrV strains revealed that the UL10 product of PrV strain Bartha, an attenuated virus used as vaccine, was not modified by N-glycosylation. Sequence analysis showed that the N-glycosylation consensus sequence was altered from NDT to NDA, which resulted in loss of the N-glycosylation signal. To our knowledge, this is the only gM homolog identified so far which is not N-glycosylated. To investigate whether this form of the protein is functionally competent, the UL10 gene of strain Bartha was inserted into PrV strain Kaplan by substitution of the wild-type UL10 gene. The resulting recombinant expressed a UL10 protein lacking N-glycans. In vitro replication analyses did not reveal any difference in virus production, but plaque size and penetration kinetics were slightly reduced. In summary, we show that wild-type gM is modified by N-glycosylation at one conserved site. However, although this site is highly conserved throughout the herpesviruses, loss of N-glycans due to mutation of the consensus sequence had only a minor effect on propagation of PrV in cell culture.


Assuntos
Glicoproteínas/metabolismo , Herpesvirus Suídeo 1/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo , Animais , Chlorocebus aethiops , Glicoproteínas/genética , Glicosilação , Células Vero , Proteínas Virais/genética
20.
J Virol ; 71(10): 7687-95, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9311852

RESUMO

Herpesviruses contain a number of envelope glycoproteins which play important roles in the interaction between virions and target cells. Although several glycoproteins are not present in all herpesviruses, others, including glycoproteins H and L (gH and gL), are conserved throughout the Herpesviridae. To elucidate common properties and differences in herpesvirus glycoprotein function, corresponding virus mutants must be constructed and analyzed in different herpesvirus backgrounds. Analysis of gH- mutants of herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PrV) showed that in both viruses gH is essential for penetration and cell-to-cell spread and that its presence is required for virion localization of gL. Since gH homologs are found complexed with gL, it was of interest to assess the phenotype of gL- mutant viruses. By using this approach, HSV-1 gL has been shown to be required for entry and for virion localization of gH (C. Roop, L. Hutchinson, and D. Johnson, J. Virol. 67:2285-2297, 1993). To examine whether a similar phenotype is associated with lack of gL in another alphaherpesvirus, PrV, we constructed two independent gL- PrV mutants by insertion and deletion-insertion mutagenesis. The salient findings are as follows: (i) PrV gL is required for penetration of virions and cell-to-cell spread; (ii) unlike HSV-1, PrV gH is incorporated into the virion in the absence of gL; (iii) virion localization of gH in the absence of gL is not sufficient for infectivity; (iv) in the absence of gL, N-glycans on PrV gH are processed to a greater extent than in the presence of gL, indicating masking of N-glycans by association with gL; and (v) an anti-gL polyclonal antiserum is able to neutralize virion infectivity but did not inhibit cell-to-cell spread. Thus, whereas PrV gL is essential for virus replication, as is HSV-1 gL, gL- PrV mutants exhibit properties strikingly different from those of HSV-1. In conclusion, our data show an important functional role for PrV gL in the viral entry process, which is not explained by a chaperone-type mechanism in gH maturation and processing.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Herpesvirus Suídeo 1/patogenicidade , Proteínas do Envelope Viral/fisiologia , Vírion/fisiologia , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Rim , Cinética , Fusão de Membrana , Mutagênese , Mapeamento por Restrição , Suínos , Células Vero , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Vírion/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...