Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 74
Filtrer
1.
J Virol ; 94(7)2020 03 17.
Article de Anglais | MEDLINE | ID: mdl-31941788

RÉSUMÉ

Protein kinases homologous to the US3 gene product (pUS3) of herpes simplex virus (HSV) are conserved throughout the alphaherpesviruses but are absent from betaherpesviruses and gammaherpesviruses. pUS3 homologs are multifunctional and are involved in many processes, including modification of the cytoskeleton, inhibition of apoptosis, and immune evasion. pUS3 also plays a role in efficient nuclear egress of alphaherpesvirus nucleocapsids. In the absence of pUS3, primary enveloped virions accumulate in the perinuclear space (PNS) in large invaginations of the inner nuclear membrane (INM), pointing to a modulatory function for pUS3 during deenvelopment. The HSV and pseudorabies virus (PrV) US3 genes are transcribed into two mRNAs encoding two pUS3 isoforms, which have different aminoterminal sequences and abundances. To test whether the two isoforms in PrV serve different functions, we constructed mutant viruses expressing exclusively either the larger minor or the smaller major isoform, a mutant virus with decreased expression of the smaller isoform, or a mutant with impaired kinase function. Respective virus mutants were investigated in several cell lines. Our results show that absence of the larger pUS3 isoform has no detectable effect on viral replication in cell culture, while full expression of the smaller isoform and intact kinase activity is required for efficient nuclear egress. Absence of pUS3 resulted in only minor titer reduction in most cell lines tested but disclosed a more severe defect in Madin-Darby bovine kidney cells. However, accumulations of primary virions in the PNS do not account for the observed titer reduction in PrV.IMPORTANCE A plethora of substrates and functions have been assigned to the alphaherpesviral pUS3 kinase, including a role in nuclear egress. In PrV, two different pUS3 isoforms are expressed, which differ in size, abundance, and intracellular localization. Their respective role in replication is unknown, however. Here, we show that efficient nuclear egress of PrV requires the smaller isoform and intact kinase activity, whereas absence of the larger isoform has no significant effect on viral replication. Thus, there is a clear distinction in function between the two US3 gene products of PrV.


Sujet(s)
Transport nucléaire actif , Noyau de la cellule/métabolisme , Herpèsvirus porcin de type 1/enzymologie , Protein-Serine-Threonine Kinases/composition chimique , Protéines virales/composition chimique , Animaux , Apoptose , Bovins , Chlorocebus aethiops , Cytosquelette/métabolisme , Génome viral , Herpèsvirus porcin de type 1/physiologie , Rein/cytologie , Mutation , Enveloppe nucléaire/métabolisme , Phénotype , Isoformes de protéines , Lapins , Cellules Vero , Assemblage viral
2.
J Virol ; 91(19)2017 10 01.
Article de Anglais | MEDLINE | ID: mdl-28724767

RÉSUMÉ

Herpesvirus capsids assemble in the nucleus, while final virion maturation proceeds in the cytoplasm. This requires that newly formed nucleocapsids cross the nuclear envelope (NE), which occurs by budding at the inner nuclear membrane (INM), release of the primary enveloped virion into the perinuclear space (PNS), and subsequent rapid fusion with the outer nuclear membrane (ONM). During this process, the NE remains intact, even at late stages of infection. In addition, the spacing between the INM and ONM is maintained, as is that between the primary virion envelope and nuclear membranes. The linker of nucleoskeleton and cytoskeleton (LINC) complex consists of INM proteins with a luminal SUN (Sad1/UNC-84 homology) domain connected to ONM proteins with a KASH (Klarsicht, ANC-1, SYNE homology) domain and is thought to be responsible for spacing the nuclear membranes. To investigate the role of the LINC complex during herpesvirus infection, we generated cell lines constitutively expressing dominant negative (dn) forms of SUN1 and SUN2. Ultrastructural analyses revealed a significant expansion of the PNS and the contiguous intracytoplasmic lumen, most likely representing endoplasmic reticulum (ER), especially in cells expressing dn-SUN2. After infection, primary virions accumulated in these expanded luminal regions, also very distant from the nucleus. The importance of the LINC complex was also confirmed by reduced progeny virus titers in cells expressing dn-SUN2. These data show that the intact LINC complex is required for efficient nuclear egress of herpesviruses, likely acting to promote fusion of primary enveloped virions with the ONM.IMPORTANCE While the viral factors for primary envelopment of nucleocapsids at the inner nuclear membrane are known to the point of high-resolution structures, the roles of cellular components and regulators remain enigmatic. Furthermore, the machinery responsible for fusion with the outer nuclear membrane is unsolved. We show here that dominant negative SUN2 interferes with efficient herpesvirus nuclear egress, apparently by interfering with fusion between the primary virion envelope and outer nuclear membrane. This identifies a new cellular component important for viral egress and implicates LINC complex integrity in nonconventional nuclear membrane trafficking.


Sujet(s)
Transport nucléaire actif/physiologie , Cytosquelette/métabolisme , Herpesviridae/croissance et développement , Enveloppe nucléaire/métabolisme , Matrice nucléaire/métabolisme , Transport nucléaire actif/génétique , Animaux , Lignée cellulaire , Herpesviridae/métabolisme , Protéines membranaires/génétique , Protéines associées aux microtubules/génétique , Protéines nucléaires/métabolisme , Lapins , Suidae , Assemblage viral/physiologie , Libération de particules virales/physiologie
3.
J Virol ; 89(21): 10959-69, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26311891

RÉSUMÉ

UNLABELLED: The incidence of human cowpox virus (CPXV) infections has increased significantly in recent years. Serological surveys have suggested wild rodents as the main CPXV reservoir. We characterized a CPXV isolated during a large-scale screening from a feral common vole. A comparison of the full-length DNA sequence of this CPXV strain with a highly virulent pet rat CPXV isolate showed a sequence identity of 96%, including a large additional open reading frame (ORF) of about 6,000 nucleotides which is absent in the reference CPXV strain Brighton Red. Electron microscopy analysis demonstrated that the vole isolate, in contrast to the rat strain, forms A-type inclusion (ATI) bodies with incorporated virions, consistent with the presence of complete ati and p4c genes. Experimental infections showed that the vole CPXV strain caused only mild clinical symptoms in its natural host, while all rats developed severe respiratory symptoms followed by a systemic rash. In contrast, common voles infected with a high dose of the rat CPXV showed severe signs of respiratory disease but no skin lesions, whereas infection with a low dose led to virus excretion with only mild clinical signs. We concluded that the common vole is susceptible to infection with different CPXV strains. The spectrum ranges from well-adapted viruses causing limited clinical symptoms to highly virulent strains causing severe respiratory symptoms. In addition, the low pathogenicity of the vole isolate in its eponymous host suggests a role of common voles as a major CPXV reservoir, and future research will focus on the correlation between viral genotype and phenotype/pathotype in accidental and reservoir species. IMPORTANCE: We report on the first detection and isolation of CPXV from a putative reservoir host, which enables comparative analyses to understand the infection cycle of these zoonotic orthopox viruses and the relevant genes involved. In vitro studies, including whole-genome sequencing as well as in vivo experiments using the Wistar rat model and the vole reservoir host allowed us to establish links between genomic sequences and the in vivo properties (virulence) of the novel vole isolate in comparison to those of a recent zoonotic CPXV isolated from pet rats in 2009. Furthermore, the role of genes present only in a reservoir isolate can now be further analyzed. These studies therefore allow unique insights and conclusions about the role of the rodent reservoir in CPXV epidemiology and transmission and about the zoonotic threat that these viruses represent.


Sujet(s)
Arvicolinae/virologie , Virus de la variole bovine/génétique , Virus de la variole bovine/physiologie , Réservoirs de maladies/virologie , Génotype , Phénotype , Animaux , Séquence nucléotidique , Analyse de regroupements , Microscopie électronique , Modèles génétiques , Annotation de séquence moléculaire , Données de séquences moléculaires , Rats , Rat Wistar , Analyse de séquence d'ADN , Similitude de séquences
4.
Virus Res ; 209: 76-86, 2015 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-25678269

RÉSUMÉ

Herpesvirus replication takes place in the nucleus and in the cytosol. After entering the cell, nucleocapsids are transported to nuclear pores where viral DNA is released into the nucleus. After gene expression and DNA replication new nucleocapsids are assembled which have to exit the nucleus for virion formation in the cytosol. Since nuclear pores are not wide enough to allow passage of the nucleocapsid, nuclear egress occurs by vesicle-mediated transport through the nuclear envelope. To this end, nucleocapsids bud at the inner nuclear membrane (INM) recruiting a primary envelope which then fuses with the outer nuclear membrane (ONM). In the absence of this regulated nuclear egress, mutants of the alphaherpesvirus pseudorabies virus have been described that escape from the nucleus after virus-induced nuclear envelope breakdown. Here we review these exit pathways and demonstrate that both can occur simultaneously under appropriate conditions.


Sujet(s)
Herpèsvirus porcin de type 1/physiologie , Enveloppe nucléaire/métabolisme , Assemblage viral , Libération de particules virales , Animaux , Modèles biologiques
5.
J Virol ; 89(3): 1825-37, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25410877

RÉSUMÉ

UNLABELLED: Since its emergence, Schmallenberg virus (SBV), a novel insect-transmitted orthobunyavirus which predominantly infects ruminants, has caused a large epidemic in European livestock. Newly developed inactivated vaccines are available, but highly efficacious and safe live vaccines are still not available. Here, the properties of novel recombinant SBV mutants lacking the nonstructural protein NSs (rSBVΔNSs) or NSm (rSBVΔNSm) or both of these proteins (rSBVΔNSs/ΔNSm) were tested in vitro and in vivo in type I interferon receptor knockout mice (IFNAR(-/-)) and in a vaccination/challenge trial in cattle. As for other bunyaviruses, both nonstructural proteins of SBV are not essential for viral growth in vitro. In interferon-defective BHK-21 cells, rSBVΔNSs and rSBVΔNSm replicated to levels comparable to that of the parental rSBV; the double mutant virus, however, showed a mild growth defect, resulting in lower final virus titers. Additionally, both mutants with an NSs deletion induced high levels of interferon and showed a marked growth defect in interferon-competent sheep SFT-R cells. Nevertheless, in IFNAR(-/-) mice, all mutants were virulent, with the highest mortality rate for rSBVΔNSs and a reduced virulence for the NSm-deleted virus. In cattle, SBV lacking NSm caused viremia and seroconversion comparable to those caused by the wild-type virus, while the NSs and the combined NSs/NSm deletion mutant induced no detectable virus replication or clinical disease after immunization. Furthermore, three out of four cattle immunized once with the NSs deletion mutant and all animals vaccinated with the virus lacking both nonstructural proteins were fully protected against a challenge infection. Therefore, the double deletion mutant will provide the basis for further developments of safe and efficacious modified live SBV vaccines which could be also a model for other viruses of the Simbu serogroup and related orthobunyaviruses. IMPORTANCE: SBV induces only mild clinical signs in adult ruminants but causes severe fetal malformation and, thereby, can have an important impact on animal welfare and production. As SBV is an insect-transmitted pathogen, vaccination will be one of the most important aspects of disease control. Here, mutant viruses lacking one or two proteins that essentially contribute to viral pathogenicity were tested as modified live vaccines in cattle. It could be demonstrated that a novel recombinant double deletion mutant is a safe and efficacious vaccine candidate. This is the first description of a putative modified live vaccine for the complete genus Orthobunyavirus, and in addition, such a vaccine type has never been tested in cattle for any virus of the entire family Bunyaviridae. Therefore, the described vaccine also represents the first model for a broad range of related viruses and is of high importance to the field.


Sujet(s)
Infections à Bunyaviridae/médecine vétérinaire , Maladies des bovins/prévention et contrôle , Orthobunyavirus/génétique , Orthobunyavirus/immunologie , Délétion de séquence , Protéines virales non structurales/génétique , Vaccins antiviraux/immunologie , Animaux , Infections à Bunyaviridae/immunologie , Infections à Bunyaviridae/prévention et contrôle , Bovins , Maladies des bovins/immunologie , Femelle , Mâle , Souris de lignée C57BL , Souris knockout , Vaccins atténués/administration et posologie , Vaccins atténués/génétique , Vaccins atténués/immunologie , Vaccins atténués/isolement et purification , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/génétique , Vaccins antiviraux/isolement et purification
6.
J Virol ; 89(4): 2002-12, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25505082

RÉSUMÉ

UNLABELLED: The herpesviral nuclear egress complex (NEC), consisting of pUL31 and pUL34 homologs, mediates efficient translocation of newly synthesized capsids from the nucleus to the cytosol. The tail-anchored membrane protein pUL34 is autonomously targeted to the nuclear envelope, while pUL31 is recruited to the inner nuclear membrane (INM) by interaction with pUL34. A nuclear localization signal (NLS) in several pUL31 homologs suggests importin-mediated translocation of the protein. Here we demonstrate that deletion or mutation of the NLS in pseudorabies virus (PrV) pUL31 resulted in exclusively cytosolic localization, indicating active nuclear export. Deletion or mutation of a predicted nuclear export signal (NES) in mutant constructs lacking a functional NLS resulted in diffuse nuclear and cytosolic localization, indicating that both signals are functional. pUL31 molecules lacking the complete NLS or NES were not recruited to the INM by pUL34, while site-specifically mutated proteins formed the NEC and partially complemented the defect of the UL31 deletion mutant. Our data demonstrate that the N terminus of pUL31, encompassing the NLS, is required for efficient nuclear targeting but not for pUL34 interaction, while the C terminus, containing the NES but not necessarily the NES itself, is required for complex formation and efficient budding of viral capsids at the INM. Moreover, pUL31-ΔNLS displayed a dominant negative effect on wild-type PrV replication, probably by diverting pUL34 to cytoplasmic membranes. IMPORTANCE: The molecular details of nuclear egress of herpesvirus capsids are still enigmatic. Although the key players, homologs of herpes simplex virus pUL34 and pUL31, which interact and form the heterodimeric nuclear egress complex, are well known, the molecular basis of this interaction and the successive budding, vesicle formation, and scission from the INM, as well as capsid release into the cytoplasm, remain largely obscure. Here we show that classical cellular targeting signals for nuclear import and export are important for proper localization and function of the NEC, thus regulating herpesvirus nuclear egress.


Sujet(s)
Herpèsvirus porcin de type 1/physiologie , Signaux d'export nucléaire , Signaux de localisation nucléaire , Protéines virales/génétique , Protéines virales/métabolisme , Animaux , Lignée cellulaire , Herpèsvirus porcin de type 1/génétique , Mutation , Liaison aux protéines , Transport des protéines , Lapins , Délétion de séquence
7.
PLoS One ; 9(12): e113344, 2014.
Article de Anglais | MEDLINE | ID: mdl-25437176

RÉSUMÉ

Newcastle disease virus (NDV), also designated as Avian paramyxovirus type 1 (APMV-1), is the causative agent of a notifiable disease of poultry but it exhibits different pathogenicity dependent on the virus strain. The molecular basis for this variability is not fully understood. The efficiency of activation of the fusion protein (F) is determined by presence or absence of a polybasic amino acid sequence at an internal proteolytic cleavage site which is a major determinant of NDV virulence. However, other determinants of pathogenicity must exist since APMV-1 of high (velogenic), intermediate (mesogenic) and low (lentogenic) virulence specify a polybasic F cleavage site. We aimed at elucidation of additional virulence determinants by constructing a recombinant virus that consists of a lentogenic NDV Clone 30 backbone and the F protein gene from a mesogenic pigeon paramyxovirus-1 (PPMV-1) isolate with an intracerebral pathogenicity index (ICPI) of 1.1 specifying the polybasic sequence R-R-K-K-R*F motif at the cleavage site. The resulting virus was characterized by an ICPI of 0.6, indicating a lentogenic pathotype. In contrast, alteration of the cleavage site G-R-Q-G-R*L of the lentogenic Clone 30 to R-R-K-K-R*F resulted in a recombinant virus with an ICPI of 1.36 which was higher than that of parental PPMV-1. Substitution of different regions of the F protein of Clone 30 by those of PPMV-1, while maintaining the polybasic amino acid sequence at the F cleavage site, resulted in recombinant viruses with ICPIs ranging from 0.59 to 1.36 suggesting that virulence is modulated by regions of the F protein other than the polybasic cleavage site.


Sujet(s)
Virus de la maladie de Newcastle/métabolisme , Virus de la maladie de Newcastle/pathogénicité , Protéines de fusion virale/composition chimique , Protéines de fusion virale/métabolisme , Motifs d'acides aminés , Animaux , Sites de fixation , Poulets/virologie , Cellules géantes/virologie , Virus de la maladie de Newcastle/physiologie , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion virale/génétique , Virion/métabolisme , Virion/pathogénicité , Virion/physiologie , Virulence , Réplication virale
8.
Arch Virol ; 159(12): 3257-68, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25150990

RÉSUMÉ

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.


Sujet(s)
Carpes (poisson)/virologie , Herpesviridae/composition chimique , Herpesviridae/génétique , Protéines virales structurales/analyse , Protéines virales structurales/génétique , Animaux , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Technique d'immunofluorescence indirecte
9.
Prev Vet Med ; 116(4): 423-34, 2014 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-24768435

RÉSUMÉ

In autumn 2011, a novel species of the genus Orthobunyavirus of the Simbu serogroup was discovered close to the German/Dutch border and named Schmallenberg virus (SBV). Since then, SBV has caused a large epidemic in European livestock. Like other viruses of the Simbu serogroup, SBV is transmitted by insect vectors. Adult ruminants may show a mild transient disease, while an infection during a critical period of pregnancy can lead to severe congenital malformation, premature birth or stillbirth. The current knowledge about the virus, its diagnosis, the spread of the epidemic, the impact and the possibilities for preventing infections with SBV is described and discussed.


Sujet(s)
Maladies de l'animal/épidémiologie , Maladies de l'animal/virologie , Infections à Bunyaviridae/médecine vétérinaire , Orthobunyavirus , Maladies de l'animal/diagnostic , Maladies de l'animal/prévention et contrôle , Maladies de l'animal/transmission , Animaux , Infections à Bunyaviridae/diagnostic , Infections à Bunyaviridae/épidémiologie , Infections à Bunyaviridae/prévention et contrôle , Infections à Bunyaviridae/transmission , Ceratopogonidae/virologie , Malformations/médecine vétérinaire , Malformations/virologie , Europe/épidémiologie , Vecteurs insectes/virologie , Orthobunyavirus/génétique , Orthobunyavirus/pathogénicité , Réaction de polymérisation en chaine en temps réel , Ruminants/virologie , Études séroépidémiologiques
10.
J Virol ; 88(11): 6003-11, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24623429

RÉSUMÉ

UNLABELLED: Herpesvirus capsid morphogenesis occurs in the nucleus, while final maturation takes place in the cytosol, requiring translocation of capsids through the nuclear envelope. The nuclear egress complex, consisting of homologs of herpes simplex virus pUL31 and pUL34, is required for efficient nuclear egress via primary envelopment and de-envelopment. Recently, we described an alternative mode of nuclear escape by fragmentation of the nuclear envelope induced by replication-competent pUL31 and pUL34 deletion mutants of the alphaherpesvirus pseudorabies virus (PrV), which had been selected by serial passaging in cell culture. Both passaged viruses carry congruent mutations in seven genes, including UL46, which encodes one of the major tegument proteins. Herpesvirus pUL46 homologs have recently been shown to activate the PI3K-Akt and ERK1/2 signaling pathways, which are involved in regulation of mitosis and apoptosis. Since in uninfected cells fragmentation of the nuclear envelope occurs during mitosis and apoptosis, we analyzed whether pUL46 of PrV is involved in signaling events impairing the integrity of the nuclear envelope. We show here that PrV pUL46 is able to induce phosphorylation of ERK1/2 and, thus, expression of ERK1/2 target genes but fails to activate the PI3K-Akt pathway. Deletion of UL46 from PrV-ΔUL34Pass and PrV-ΔUL31Pass or replacement by wild-type UL46 resulted in enhanced nuclear envelope breakdown, indicating that the mutations in pUL46 may limit the extent of NEBD. Thus, although pUL46 induces ERK1/2 phosphorylation, controlling the integrity of the nuclear envelope is independent of the ERK1/2 signaling pathway. IMPORTANCE: Herpesvirus nucleocapsids can leave the nucleus by regulated, vesicle-mediated transport through the nuclear envelope, designated nuclear egress, or by inducing nuclear envelope breakdown (NEBD). The viral proteins involved in NEBD are unknown. We show here that the pseudorabies virus tegument protein pUL46 induces the ERK1/2 signaling pathway and modulates NEBD. However, these two processes are independent and ERK1/2 signaling induced by pUL46 is not involved in herpesvirus-induced NEBD.


Sujet(s)
Transport nucléaire actif/physiologie , Antigènes viraux/métabolisme , Activation enzymatique/physiologie , Herpèsvirus porcin de type 1/physiologie , Système de signalisation des MAP kinases/physiologie , Enveloppe nucléaire/métabolisme , Protéines virales/métabolisme , Transport nucléaire actif/génétique , Analyse de variance , Technique de Western , Activation enzymatique/génétique , Cinétique , Luciferases , Microscopie électronique , Enveloppe nucléaire/virologie , Plasmides/génétique
11.
Vector Borne Zoonotic Dis ; 14(3): 220-2, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24575742

RÉSUMÉ

Sindbis virus (SINV) is an arbovirus that causes clinical symptoms, including arthritis, rash, and fever during acute human infections. In Europe, SINV outbreaks are largely restricted to northern Europe. Intrigued by the isolation of SINV from mosquitoes in southwestern Germany in 2009, we initiated a passive arbovirus-monitoring program in birds and analyzed a total of 685 samples. By this approach, we were able to detect a SINV in a Hooded Crow in Germany for the first time. It was possible to isolate SINV virus in cell cultures and even to visualize virus particles by electron microscopy. After the determination of the complete SINV genome sequence, the phylogenetic analysis revealed its close relationship to SINV genotype I sequences previously obtained from mosquitoes in Germany and Scandinavia. This first report on the isolation of viable SINV indicates the potential involvement of crows in an enzootic circulation of SINV in Germany and Central Europe.


Sujet(s)
Infections à alphavirus/médecine vétérinaire , Maladies des oiseaux/virologie , Corneilles/virologie , Culicidae/virologie , Virus Sindbis/isolement et purification , Infections à alphavirus/épidémiologie , Infections à alphavirus/transmission , Infections à alphavirus/virologie , Animaux , Séquence nucléotidique , Maladies des oiseaux/épidémiologie , Maladies des oiseaux/transmission , Réservoirs de maladies , Génotype , Allemagne/épidémiologie , Humains , Microscopie électronique , Données de séquences moléculaires , Phylogenèse , Analyse de séquence d'ADN , Virus Sindbis/classification , Virus Sindbis/ultrastructure
12.
J Virol ; 88(11): 6224-31, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24648464

RÉSUMÉ

UNLABELLED: Nuclear egress of herpesvirus capsids is mediated by a conserved heterodimeric complex of two viral proteins, designated pUL34 and pUL31 in herpes simplex virus and pseudorabies virus (PrV). pUL34, a tail-anchored membrane protein, is targeted to the nuclear envelope and recruits pUL31 to the inner nuclear membrane (INM) to provide the docking and envelopment machinery for the nascent capsid. While the less conserved C-terminal part of pUL34 is required for correct positioning of the nuclear egress complex (NEC) at the INM, the conserved N-terminal part functions as a docking site for pUL31. Since no crystal structure of NEC is available yet, structure-function studies depend on mutational analyses, with several approaches already being performed for different herpesvirus NECs. Here, we extended our studies on PrV pUL34 and identified two asparagine residues (N75, N103) and a dileucine motif (LL166/167), adjacent to an endoplasmic reticulum retention signal, which are absolutely required for NEC function. While the pUL34-N75A substitution mutant is unable to interact with pUL31, the pUL34-N103A mutant is nonfunctional, despite continuing complex formation. Surprisingly, mutant pUL34-G77A, which does not efficiently recruit pUL31 to the nuclear rim after cotransfection, nonetheless complements a UL34 deletion mutant, indicating that the NEC may be stabilized by additional viral factors during infection. IMPORTANCE: In the absence of a crystal structure of the nuclear egress complex (NEC) required for herpesvirus maturation, site-directed mutagenesis studies provide important information on critical amino acid residues. Here, we identify conserved amino acid residues in the membrane-bound component of the NEC which are relevant for its function.


Sujet(s)
Séquence conservée/génétique , Herpèsvirus porcin de type 1/génétique , Protéines virales/génétique , Libération de particules virales/génétique , Séquence d'acides aminés , Animaux , Technique de Western , Lignée cellulaire , Technique d'immunofluorescence , Test de complémentation , Microscopie confocale , Microscopie électronique , Données de séquences moléculaires , Mutagenèse dirigée , Lapins , Analyse de séquence d'ADN , Protéines virales/métabolisme
13.
J Gen Virol ; 95(Pt 1): 80-90, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24337965

RÉSUMÉ

Picornaviruses have been isolated from a variety of hosts, mainly mammals and birds. Here, we describe the sequence analysis of carp picornavirus 1 (CPV-1) F37/06 that was isolated from an organ pool (heart, brain, liver) of a common carp (Cyprinus carpio). This carp perished after an accidental discharge of liquid manure into a fish pond and presented without obvious clinical symptoms. Experimental intraperitoneal infection of young carp with CPV-1 revealed no clinical signs, but the virus was re-isolated from various organs. Sequence analysis of almost the complete genome (7632 nt excluding the poly-A tract) revealed a novel picornavirus clade. In phylogenetic trees, the polymerase sequence clusters with parechoviruses, duck hepatitis A virus, eel picornavirus and aquamavirus A. The ORF includes 6807 nt and encodes a polyprotein of 2269 amino acids. CPV-1 has a genome layout like that of picornaviruses except for the presence of two aphthovirus 2A-like NPGP sequence motifs: VPg+5'UTR[1AB-1C-1D-2A1(npgp)/2A2(npgp)-2B-2C(ATPase)/3A-3B(VPg)-3C(pro)-3D(pol)]3'UTR-poly-A. 2A1(npgp) and 2A2(npgp) are separated by 133 amino acids. The proteins 2A2(npgp), 2B, 3A and 3B(VPg) have no significant similarity to the corresponding proteins of other picornaviruses. Amino acid identities of the orthologous proteins P1, 2C, 3C(pro) and 3D(pol) range from 16.4 to 40.8 % in the eel picornavirus/CPV-1 comparison. 3D(pol) shows the closest similarity to eel picornavirus, with an amino acid identity of 40.8 %, followed by human parechovirus (36.5 %), duck hepatitis A virus (32.7 %) and swine pasivirus (29.3 %). Both the unique genome organization and low sequence similarity support the assignment of CPV-1 to a novel picornavirus species within a novel genus.


Sujet(s)
Aphthovirus/génétique , Carpes (poisson)/virologie , Maladies des poissons/virologie , Infections à Picornaviridae/médecine vétérinaire , Picornaviridae/génétique , Picornaviridae/isolement et purification , Régions 5' non traduites , Séquence d'acides aminés , Animaux , Aphthovirus/composition chimique , Aphthovirus/classification , Génome viral , Humains , Données de séquences moléculaires , Cadres ouverts de lecture , Phylogenèse , Picornaviridae/composition chimique , Picornaviridae/classification , Infections à Picornaviridae/virologie , Alignement de séquences , Protéines virales/composition chimique , Protéines virales/génétique
14.
PLoS One ; 8(9): e72530, 2013.
Article de Anglais | MEDLINE | ID: mdl-24023747

RÉSUMÉ

Newcastle disease virus (NDV), an avian paramyxovirus type 1, is a promising vector for expression of heterologous proteins from a variety of unrelated viruses including highly pathogenic avian influenza virus (HPAIV). However, pre-existing NDV antibodies may impair vector virus replication, resulting in an inefficient immune response against the foreign antigen. A chimeric NDV-based vector with functional surface glycoproteins unrelated to NDV could overcome this problem. Therefore, an NDV vector was constructed which carries the fusion (F) and hemagglutinin-neuraminidase (HN) proteins of avian paramyxovirus type 8 (APMV-8) instead of the corresponding NDV proteins in an NDV backbone derived from the lentogenic NDV Clone 30 and a gene expressing HPAIV H5 inserted between the F and HN genes. After successful virus rescue by reverse genetics, the resulting chNDVFHN PMV8H5 was characterized in vitro and in vivo. Expression and virion incorporation of the heterologous proteins was verified by Western blot and electron microscopy. Replication of the newly generated recombinant virus was comparable to parental NDV in embryonated chicken eggs. Immunization with chNDVFHN PMV8H5 stimulated full protection against lethal HPAIV infection in chickens without as well as with maternally derived NDV antibodies. Thus, tailored NDV vector vaccines can be provided for use in the presence or absence of routine NDV vaccination.


Sujet(s)
Immunité/immunologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/prévention et contrôle , Virus de la maladie de Newcastle/immunologie , Animaux , Poulets , Vaccins antiviraux/immunologie
15.
J Virol ; 87(17): 9733-41, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23824797

RÉSUMÉ

Herpesvirus nucleocapsids are assembled in the nucleus, whereas maturation into infectious virions takes place in the cytosol. Since, due to their size, nucleocapsids cannot pass the nuclear pores, they traverse the nuclear envelope by vesicle-mediated transport. Nucleocapsids bud at the inner nuclear membrane into the perinuclear space, forming primary enveloped particles and are released into the cytosol after fusion of the primary envelope with the outer nuclear membrane. The nuclear egress complex (NEC), consisting of the conserved herpesvirus proteins (p)UL31 and pUL34, is required for this process, whereas the viral glycoproteins gB and gH, which are essential for fusion during penetration, are not. We recently described herpesvirus-induced nuclear envelope breakdown (NEBD) as an alternative egress pathway used in the absence of the NEC. However, the molecular details of this pathway are still unknown. It has been speculated that glycoproteins involved in fusion during entry might play a role in NEBD. By deleting genes encoding glycoproteins gB and gH from the genome of NEBD-inducing pseudorabies viruses, we demonstrate that these glycoproteins are not required for NEBD but are still necessary for syncytium formation, again emphasizing fundamental differences in herpesvirus-induced alterations at the nuclear envelopes and plasma membranes of infected cells.


Sujet(s)
Herpèsvirus porcin de type 1/physiologie , Protéines de l'enveloppe virale/physiologie , Animaux , Lignée cellulaire , Délétion de gène , Gènes viraux , Cellules géantes/ultrastructure , Cellules géantes/virologie , Herpèsvirus porcin de type 1/génétique , Herpèsvirus porcin de type 1/ultrastructure , Microscopie électronique à transmission , Enveloppe nucléaire/virologie , Lapins , Protéines de l'enveloppe virale/génétique , Assemblage viral/physiologie , Libération de particules virales/physiologie
16.
J Virol ; 87(19): 10895-9, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23885066

RÉSUMÉ

A novel picornavirus was isolated from specimens of a diseased European eel (Anguilla anguilla). This virus induced a cytopathic effect in eel embryonic kidney cells and high mortality in a controlled transmission study using elvers. Eel picornavirus has a genome of 7,496 nucleotides that encodes a polyprotein of 2,259 amino acids. It has a typical picornavirus genome layout, but its low similarity to known viral proteins suggests a novel species in the family Picornaviridae.


Sujet(s)
Anguilla/virologie , Maladies des poissons/virologie , Génome viral , Picornaviridae/isolement et purification , Anguilla/génétique , Animaux , Maladies des poissons/génétique , Maladies des poissons/anatomopathologie , Phylogenèse , Picornaviridae/physiologie , Protéines virales/génétique
17.
Cell Microbiol ; 15(2): 200-12, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23046288

RÉSUMÉ

Lyssavirus matrix proteins (M) support virus budding and have accessory functions that may contribute to host cell manipulation and adaptation to specific hosts. Here, we show that rabies virus (RABV) and European Bat Lyssavirus Type 1 (EBLV-1) M proteins differ in targeting and accumulation at cellular membranes. In contrast to RABV M, EBLV-1 M expressed from authentic EBLV-1 or chimeric RABV accumulated at the Golgi apparatus. Chimeric M proteins revealed that Golgi association depends on the integrity of the entire EBLV-1 M protein. Since RABV and EBLV-1 M differ in the use of cellular membranes for particle formation, differential membrane targeting and transport of M might determine the site of virus production. Moreover, both RABV and EBLV-1 M were for the first time detected within the nucleus and in Negri body-like inclusions bodies. Whereas nuclear M may imply hitherto unknown functions of lyssavirus M in host cell manipulation, the presence of M in inclusion bodies may correlate with regulatory functions of M in virus RNA synthesis. The data strongly support a model in which targeting of lyssavirus M proteins to distinctintracellular sites is a key determinant of diverse features in lyssavirus replication, host adaptation and pathogenesis.


Sujet(s)
Membrane cellulaire/métabolisme , Corps d'inclusion/métabolisme , Lyssavirus/physiologie , Virus de la rage/physiologie , Infections à Rhabdoviridae/médecine vétérinaire , Protéines de la matrice virale/génétique , Virion/physiologie , Animaux , Lignée cellulaire , Membrane cellulaire/ultrastructure , Membrane cellulaire/virologie , Noyau de la cellule/métabolisme , Noyau de la cellule/ultrastructure , Noyau de la cellule/virologie , Chiroptera/virologie , Cricetinae , Appareil de Golgi/métabolisme , Appareil de Golgi/ultrastructure , Appareil de Golgi/virologie , Corps d'inclusion/ultrastructure , Corps d'inclusion/virologie , Microscopie électronique , Transport des protéines , Infections à Rhabdoviridae/virologie , Spécificité d'espèce , Transfection , Protéines de la matrice virale/métabolisme
18.
J Virol ; 87(4): 2011-22, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23236073

RÉSUMÉ

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.


Sujet(s)
ADN viral/génétique , Virus défectifs/génétique , Virus défectifs/immunologie , Herpèsvirus humain de type 4/génétique , Herpèsvirus humain de type 4/immunologie , Vaccins à pseudo-particules virales/immunologie , Lymphocytes T CD4+/immunologie , Cellules cultivées , Délétion de gène , Herpèsvirus humain de type 4/physiologie , Humains , Protéines membranaires/déficit , Vaccins à pseudo-particules virales/génétique , Protéines virales , Assemblage viral
19.
Cell Microbiol ; 15(2): 170-8, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23057731

RÉSUMÉ

Herpesvirus capsids are assembled in the nucleus of infected cells whereas final maturation occurs in the cytosol. To access the final maturation compartment, intranuclear capsids have to cross the nuclear envelope which represents a formidable barrier. They do so by budding at the inner nuclear membrane, thereby forming a primary enveloped particle residing in the perinuclear cleft. Formation of primary envelopes is driven by a heterodimeric complex of two conserved herpesviral proteins, designated in the herpes simplex virus nomenclature as pUL34, a tail-anchored transmembrane protein located in the nuclear envelope, and pUL31. This nuclear egress complex recruits viral and cellular kinases to soften the nuclear lamina and allowing access of capsids to the inner nuclear membrane. How capsids are recruited to the budding site and into the primary virus particle is still not completely understood, nor is the composition of the primary enveloped virion in the perinuclear cleft. Fusion of the primary envelope with the outer nuclear membrane then results in translocation of the capsid to the cytosol. This fusion event is clearly different from fusion during infectious entry of free virions into target cells in that it does not require the conserved essential core herpesvirus fusion machinery. Nuclear egress can thus be viewed as a vesicle (primary envelope)-mediated transport of cargo (capsids) through thenuclear envelope, a process which had been unique in cell biology. Only recently has a similar process been identified in Drosophila for nuclear egress of large ribonucleoprotein complexes. Thus, herpesviruses appear to subvert a hitherto cryptic cellular pathway for translocation of capsids from the nucleus to the cytosol.


Sujet(s)
Protéines de capside/physiologie , Vésicules cytoplasmiques/métabolisme , Cellules épithéliales/métabolisme , Herpesviridae/physiologie , Enveloppe nucléaire/métabolisme , Virion/physiologie , Libération de particules virales/physiologie , Transport biologique , Cytosol/métabolisme , Cytosol/virologie , Cellules épithéliales/ultrastructure , Cellules épithéliales/virologie , Herpesviridae/ultrastructure , Infections à Herpesviridae/virologie , Interactions hôte-pathogène , Humains , Fusion membranaire , Enveloppe nucléaire/virologie
20.
J Virol ; 86(12): 6512-21, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22491460

RÉSUMÉ

Herpesvirus nucleocapsids are translocated from their assembly site in the nucleus to the cytosol by acquisition of a primary envelope at the inner nuclear membrane which subsequently fuses with the outer nuclear membrane. This transport through the nuclear envelope requires homologs of the conserved herpesviral pUL31 and pUL34 proteins which form the nuclear egress complex (NEC). In its absence, 1,000-fold less virus progeny is produced. We isolated a UL34-negative mutant of the alphaherpesvirus pseudorabies virus (PrV), PrV-ΔUL34Pass, which regained replication competence after serial passages in cell culture by inducing nuclear envelope breakdown (NEBD) (B. G. Klupp, H. Granzow, and T. C. Mettenleiter, J. Virol. 85:8285-8292, 2011). To test whether this phenotype is unique, passaging experiments were repeated with a UL31 deletion mutant. After 60 passages, the resulting PrV-ΔUL31Pass replicated similarly to wild-type PrV. Ultrastructural analyses confirmed escape from the nucleus via NEBD, indicating an inherent genetic disposition in herpesviruses. To identify the mutated viral genes responsible for this phenotype, the genome of PrV-ΔUL34Pass was sequenced and compared to the genomes of parental PrV-Ka and PrV-ΔUL34. Targeted sequencing of PrV-ΔUL31Pass disclosed congruent mutations comprising genes encoding tegument proteins (pUL49, pUL46, pUL21, pUS2), envelope proteins (gI, pUS9), and protease pUL26. To investigate involvement of cellular pathways, different inhibitors of cellular kinases were tested. While induction of apoptosis or inhibition of caspases had no specific effect on the passaged mutants, roscovitine, a cyclin-dependent kinase inhibitor, and U0126, an inhibitor of MEK1/2, specifically impaired replication of the passaged mutants, indicating involvement of mitosis-related processes in herpesvirus-induced NEBD.


Sujet(s)
Herpèsvirus porcin de type 1/physiologie , Enveloppe nucléaire/métabolisme , Maladie d'Aujeszky/métabolisme , Protéines virales/métabolisme , Assemblage viral , Animaux , Lignée cellulaire , Herpèsvirus porcin de type 1/génétique , Données de séquences moléculaires , Enveloppe nucléaire/virologie , Maladie d'Aujeszky/virologie , Protéines virales/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...