Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(7): e1010187, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35816507

RESUMO

Nucleoli are membrane-less structures located within the nucleus and are known to be involved in many cellular functions, including stress response and cell cycle regulation. Besides, many viruses can employ the nucleolus or nucleolar proteins to promote different steps of their life cycle such as replication, transcription and assembly. While adeno-associated virus type 2 (AAV2) capsids have previously been reported to enter the host cell nucleus and accumulate in the nucleolus, both the role of the nucleolus in AAV2 infection, and the viral uncoating mechanism remain elusive. In all prior studies on AAV uncoating, viral capsids and viral genomes were not directly correlated on the single cell level, at least not in absence of a helper virus. To elucidate the properties of the nucleolus during AAV2 infection and to assess viral uncoating on a single cell level, we combined immunofluorescence analysis for detection of intact AAV2 capsids and capsid proteins with fluorescence in situ hybridization for detection of AAV2 genomes. The results of our experiments provide evidence that uncoating of AAV2 particles occurs in a stepwise process that is completed in the nucleolus and supported by alteration of the nucleolar structure.


Assuntos
Dependovirus , Desenvelopamento do Vírus , Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Células HeLa , Humanos , Hibridização in Situ Fluorescente
2.
J Virol ; 95(13): e0048621, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33853961

RESUMO

Wild-type adeno-associated virus (AAV) can only replicate in the presence of helper factors, which can be provided by coinfecting helper viruses such as adenoviruses and herpesviruses. The AAV genome consists of a linear, single-stranded DNA (ssDNA), which is converted into different molecular structures within the host cell. Using high-throughput sequencing, we found that herpes simplex virus 1 (HSV-1) coinfection leads to a shift in the type of AAV genome end recombination. In particular, open-end inverted terminal repeat (ITR) recombination was enhanced, whereas open-closed ITR recombination was reduced in the presence of HSV-1. We demonstrate that the HSV-1 protein ICP8 plays an essential role in HSV-1-mediated interference with AAV genome end recombination, indicating that the previously described ICP8-driven mechanism of HSV-1 genome recombination may be underlying the observed changes. We also provide evidence that additional factors, such as products of true late genes, are involved. Although HSV-1 coinfection significantly changed the type of AAV genome end recombination, no significant change in the amount of circular AAV genomes was identified. IMPORTANCE Adeno-associated virus (AAV)-mediated gene therapy represents one of the most promising approaches for the treatment of genetic diseases. Currently, various GMP-compatible production methods can be applied to manufacture clinical-grade vector, including methods that employ helper factors derived from herpes simplex virus 1 (HSV-1). Yet, to date, we do not fully understand how HSV-1 interacts with AAV. We observed that HSV-1 modulates AAV genome ends similarly to the genome recombination events observed during HSV-1 replication and postulate that further improvements of the HSV-1 production platform may enhance packaging of the recombinant AAV particles.


Assuntos
Dependovirus/crescimento & desenvolvimento , Dependovirus/genética , Genoma Viral/genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Recombinação Genética/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Coinfecção/patologia , Células HEK293 , Células HeLa , Herpes Simples/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Infecções por Parvoviridae/patologia , Sequências Repetidas Terminais/genética , Células Vero , Interferência Viral/genética , Replicação Viral/genética
3.
Int J Mol Sci ; 22(9)2021 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-33923223

RESUMO

Herpes Simplex Virus Type-1 (HSV-1) forms progeny in the nucleus within distinct membrane-less inclusions, the viral replication compartments (VRCs), where viral gene expression, DNA replication, and packaging occur. The way in which the VRCs maintain spatial integrity remains unresolved. Here, we demonstrate that the essential viral transcription factor ICP4 is an intrinsically disordered protein (IDP) capable of driving protein condensation and liquid-liquid phase separation (LLPS) in transfected cells. Particularly, ICP4 forms nuclear liquid-like condensates in a dose- and time-dependent manner. Fluorescence recovery after photobleaching (FRAP) assays revealed rapid exchange rates of EYFP-ICP4 between phase-separated condensates and the surroundings, akin to other viral IDPs that drive LLPS. Likewise, HSV-1 VRCs revealed by EYFP-tagged ICP4 retained their liquid-like nature, suggesting that they are phase-separated condensates. Individual VRCs homotypically fused when reaching close proximity and grew over the course of infection. Together, the results of this study demonstrate that the HSV-1 transcription factor ICP4 has characteristics of a viral IDP, forms condensates in the cell nucleus by LLPS, and can be used as a proxy for HSV-1 VRCs with characteristics of liquid-liquid phase-separated condensates.


Assuntos
Regulação Viral da Expressão Gênica , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Compartimentos de Replicação Viral , Animais , Núcleo Celular/metabolismo , Chlorocebus aethiops , Herpes Simples/genética , Herpes Simples/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Intrinsicamente Desordenadas/genética , Extração Líquido-Líquido , Transição de Fase , Células Vero
4.
PLoS One ; 12(6): e0179607, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28622358

RESUMO

In infected cells rotavirus (RV) replicates in viroplasms, cytosolic structures that require a stabilized microtubule (MT) network for their assembly, maintenance of the structure and perinuclear localization. Therefore, we hypothesized that RV could interfere with the MT-breakdown that takes place in mitosis during cell division. Using synchronized RV-permissive cells, we show that RV infection arrests the cell cycle in S/G2 phase, thus favoring replication by improving viroplasms formation, viral protein translation, and viral assembly. The arrest in S/G2 phase is independent of the host or viral strain and relies on active RV replication. RV infection causes cyclin B1 down-regulation, consistent with blocking entry into mitosis. With the aid of chemical inhibitors, the cytoskeleton network was linked to specific signaling pathways of the RV-induced cell cycle arrest. We found that upon RV infection Eg5 kinesin was delocalized from the pericentriolar region to the viroplasms. We used a MA104-Fucci system to identify three RV proteins (NSP3, NSP5, and VP2) involved in cell cycle arrest in the S-phase. Our data indicate that there is a strong correlation between the cell cycle arrest and RV replication.


Assuntos
Pontos de Checagem da Fase G2 do Ciclo Celular , Rotavirus/fisiologia , Pontos de Checagem da Fase S do Ciclo Celular , Transdução de Sinais , Replicação Viral/fisiologia , Animais , Ciclina B1/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/virologia , Cães , Células HEK293 , Humanos , Cinesinas/metabolismo , Macaca mulatta , Células Madin Darby de Rim Canino , Proteínas Virais/metabolismo
5.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515305

RESUMO

Adeno-associated virus 2 (AAV2) depends on the simultaneous presence of a helper virus such as herpes simplex virus 1 (HSV-1) for productive replication. At the same time, AAV2 efficiently blocks the replication of HSV-1, which would eventually limit its own replication by diminishing the helper virus reservoir. This discrepancy begs the question of how AAV2 and HSV-1 can coexist in a cell population. Here we show that in coinfected cultures, AAV2 DNA replication takes place almost exclusively in S/G2-phase cells, while HSV-1 DNA replication is restricted to G1 phase. Live microscopy revealed that not only wild-type AAV2 (wtAAV2) replication but also reporter gene expression from both single-stranded and double-stranded (self-complementary) recombinant AAV2 vectors preferentially occurs in S/G2-phase cells, suggesting that the preference for S/G2 phase is independent of the nature of the viral genome. Interestingly, however, a substantial proportion of S/G2-phase cells transduced by the double-stranded but not the single-stranded recombinant AAV2 vectors progressed through mitosis in the absence of the helper virus. We conclude that cell cycle-dependent AAV2 rep expression facilitates cell cycle-dependent AAV2 DNA replication and inhibits HSV-1 DNA replication. This may limit competition for cellular and viral helper factors and, hence, creates a biological niche for either virus to replicate.IMPORTANCE Adeno-associated virus 2 (AAV2) differs from most other viruses, as it requires not only a host cell for replication but also a helper virus such as an adenovirus or a herpesvirus. This situation inevitably leads to competition for cellular resources. AAV2 has been shown to efficiently inhibit the replication of helper viruses. Here we present a new facet of the interaction between AAV2 and one of its helper viruses, herpes simplex virus 1 (HSV-1). We observed that AAV2 rep gene expression is cell cycle dependent and gives rise to distinct time-controlled windows for HSV-1 replication. High Rep protein levels in S/G2 phase support AAV2 replication and inhibit HSV-1 replication. Conversely, low Rep protein levels in G1 phase permit HSV-1 replication but are insufficient for AAV2 replication. This allows both viruses to productively replicate in distinct sets of dividing cells.


Assuntos
Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Dependovirus/crescimento & desenvolvimento , Vírus Auxiliares/crescimento & desenvolvimento , Herpesvirus Humano 1/crescimento & desenvolvimento , Interferência Viral , Proteínas Virais/metabolismo , Replicação Viral , Linhagem Celular , Coinfecção , Expressão Gênica , Humanos , Microscopia , Cultura de Vírus
6.
PLoS One ; 12(1): e0170908, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28125695

RESUMO

As their names imply, parvoviruses of the genus Dependovirus rely for their efficient replication on the concurrent presence of a helpervirus, such as herpesvirus, adenovirus, or papilloma virus. Adeno-associated virus 2 (AAV2) is such an example, which in turn can efficiently inhibit the replication of each helpervirus by distinct mechanisms. In a previous study we have shown that expression of the AAV2 rep gene is not compatible with efficient replication of herpes simplex virus 1 (HSV-1). In particular, the combined DNA-binding and ATPase/helicase activities of the Rep68/78 proteins have been shown to exert opposite effects on the replication of AAV2 and HSV-1. While essential for AAV2 DNA replication these protein activities account for the Rep-mediated inhibition of HSV-1 replication. Here, we describe a novel Rep mutant (Rep-D371Y), which displayed an unexpected phenotype. Rep-D371Y did not block HSV-1 replication, but still supported efficient AAV2 replication, at least when a double-stranded AAV2 genome template was used. We also found that the capacity of Rep-D371Y to induce apoptosis and a Rep-specific DNA damage response was significantly reduced compared to wild-type Rep. These findings suggest that AAV2 Rep-helicase subdomains exert diverging activities, which contribute to distinct steps of the AAV2 life cycle. More important, the novel AAV2 mutant Rep-D371Y may allow deciphering yet unsolved activities of the AAV2 Rep proteins such as DNA second-strand synthesis, genomic integration or packaging, which all involve the Rep-helicase activity.


Assuntos
Replicação do DNA , Proteínas de Ligação a DNA/genética , Dependovirus/genética , Herpesvirus Humano 1/genética , Proteínas Virais/genética , Replicação Viral , Animais , Chlorocebus aethiops , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dependovirus/metabolismo , Herpesvirus Humano 1/metabolismo , Células Vero , Proteínas Virais/metabolismo
7.
J Virol ; 89(21): 11150-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292324

RESUMO

Adeno-associated virus type 2 is known to inhibit replication of herpes simplex virus 1 (HSV-1). This activity has been linked to the helicase- and DNA-binding domains of the Rep68/Rep78 proteins. Here, we show that Rep68 can bind to consensus Rep-binding sites on the HSV-1 genome and that the Rep helicase activity can inhibit replication of any DNA if binding is facilitated. Therefore, we hypothesize that inhibition of HSV-1 replication involves direct binding of Rep68/Rep78 to the HSV-1 genome.


Assuntos
DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dependovirus/genética , Genoma Viral/genética , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Sítios de Ligação/genética , Western Blotting , Dependovirus/metabolismo , Herpesvirus Humano 1/metabolismo , Humanos
8.
Eur J Immunol ; 41(9): 2544-55, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21748731

RESUMO

Cross-presentation is an important mechanism to elicit both immune defenses and tolerance. Although only a few DC subsets possess the machinery required for cross-presentation, little is known about differences in cross-presenting capabilities of DCs belonging to the same subpopulation but localized in different lymphoid organs. In this study, we demonstrate that steady-state thymic CD8(+) DCs can efficiently cross-prime naïve CD8(+) T cells in the absence of costimulation. Surprisingly, cross-priming by splenic CD8(+) DCs was dependent on licensing factors such as GM-CSF. In the absence of GM-CSF, antigen-MHC-class-I complexes were detected on thymic but not on splenic CD8(+) DCs, indicating that the cross-presentation capacity of the thymic subpopulation was higher. The observed cross-priming differences between thymic and splenic CD8(+) DCs did not correlate with differential antigen capture or costimulatory molecules found on the surface of DCs. Moreover, we did not detect overall impairment of antigen presentation, as peptide-loaded splenic CD8(+) DCs were able to induce CD8(+) T-cell proliferation. The observation that thymic CD8(+) DCs are more efficient than splenic CD8(+) DCs in T-cell cross-priming in the absence of licensing factors indicates that the requirements for efficient antigen presentation differ between these cells.


Assuntos
Apresentação Cruzada , Células Dendríticas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Linfócitos T/metabolismo , Animais , Antígenos CD8/biossíntese , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Tolerância Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Baço/citologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia
9.
Vet Microbiol ; 143(1): 37-44, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20211531

RESUMO

Bovine herpesvirus 1 (BoHV-1) and BoHV-5 are closely related (82% amino acid identity) but differ strongly in neuropathogenesis. The immediate-early gene for BICP0 is less conserved (70% amino acid identity) and may contribute to a dissimilar phenotype. A peculiar difference is a guanosine hexamer in the BICP0-1 gene which aligns with only five guanosines in the BICP0-5 gene and therefore results in a frameshift in the latter open reading frame. Thus, the C-terminal amino acid sequence (residues 643-676 of BICP0-1 vs. 655-720 of BICP0-5) is completely different. We introduced the BICP0-5 frameshift into the BoHV-1 genome cloned as a bacterial artificial chromosome (BoHV-1 BAC) using the Red recombination system with galK selection and counterselection. Transfection of MDBK cells with the resulting BAC produced recombinant virus that replicated like wild type BoHV-1 in vitro. Attempts to exchange the entire BICP0-1 gene by the BoHV-5 homolog using the same approach failed repeatedly. Therefore, we cotransfected purified BICP0(-)/galK(+)-BoHV-1 BAC DNA with a recombination plasmid coding for BICP0-5 with or without a HA tag into MDBK cells. BoHV-1 recombinants expressing the respective proteins were characterized. In vitro, all recombinants grew to similar titers as the parental viruses, which demonstrates that BICP0-5 compensates for the growth defect of BICP0(-)/galK(+)-BoHV-1 and functionally complements BICP0-1 of BoHV-1. We conclude that BICP0 may be suitable to positively select BoHV-1 recombinants with deletions or insertions of additional genes of interest.


Assuntos
Herpesvirus Bovino 1/metabolismo , Herpesvirus Bovino 5/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Transativadores/química , Transativadores/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Proteínas Virais/metabolismo , Animais , Bovinos , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , Mutação da Fase de Leitura , Teste de Complementação Genética , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/crescimento & desenvolvimento , Herpesvirus Bovino 5/genética , Humanos , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/genética , Recombinação Genética , Transativadores/metabolismo , Transfecção , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/química , Proteínas Virais/genética
10.
Vet Microbiol ; 113(3-4): 185-92, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16352405

RESUMO

The immediate-early (IE) protein BICP22 of bovine herpesvirus 1 (BHV-1) acts as transrepressor protein on viral promoters of different kinetic classes. In the present work, we looked for host cell targets of BICP22 using a yeast two-hybrid system and identified seven candidates: (1) JIK, a serine/threonine kinase of the sterile 20 protein (STE20) family that inhibits stress-related pathways; (2) cAMP response element binding protein-like 2 (CREBL2), which in its bZip domain shares homology with CREB, modulating transcription of cAMP responsive genes; (3) DNA-dependent ATPase and helicase (ATRX), a protein of the SNF2 family altering nucleosome structure; (4) scaffold attachment factor B (SAF-B), which helps to organize chromatin into topologically separated loops; (5) peptidylglycine alpha-amidating monooxygenase COOH-terminal interactor protein 1 (PAMCIP1), involved in regulation of the secretory pathway in the perinuclear area; (6) zinc finger protein (ZNF38) found in proliferating cells and possibly associated with meiosis in male and female gametogenesis; (7) FLJ22709, hypothetical protein conserved among various species, containing an occludin/ELL domain. To confirm some of the interactions by confocal fluorescence microscopy, BICP22 was tagged with red fluorescent protein in an amplicon, and selected target sequences were tagged with green fluorescent protein in plasmid expression vectors. Upon amplicon transduction of Vero cells and plasmid transfection, CREBL2 and ZNF38 both colocalized with BICP22 in distinct nuclear domains.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Bovino 1/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Regiões Promotoras Genéticas , Proteínas Repressoras/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Doenças dos Bovinos/virologia , Núcleo Celular/metabolismo , Chlorocebus aethiops , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Microscopia Confocal/veterinária , Microscopia de Fluorescência/veterinária , Dados de Sequência Molecular , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
J Virol ; 78(8): 3805-10, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15047795

RESUMO

The immediate-early protein, BICP0, of bovine herpesvirus 1 (BHV-1) transactivates a variety of viral and cellular genes. In a yeast two-hybrid cDNA library screening, we found that lipocalin-type prostaglandin D synthase, which catalyzes the production of prostaglandin D(2) (PGD(2)), is a cellular target of BICP0. We observed that, during wild-type BHV-1 infection, PGD(2) levels were increased intracellularly and decreased in the medium. These effects were absent upon infection with recombinant BHV-1 expressing beta-galactosidase instead of BICP0 (A2G2). Transient-expression assays showed that BICP0 alone caused a significant increase in PGD(2) levels in the cell. PGD(2) repressed BHV-1 replication in cultured cells. Antiviral activities of prostaglandins have been documented long ago, but their mode of action remains to be clarified. Here we provide evidence that PGD(2) impairs the transactivation ability of BICP0 that is necessary for efficient virus replication.


Assuntos
Herpesvirus Bovino 1/fisiologia , Prostaglandina D2/metabolismo , Transativadores/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Animais , Sequência de Bases , Bovinos , Linhagem Celular , DNA Viral/genética , Células HeLa , Herpesvirus Bovino 1/genética , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/metabolismo , Lipocalinas , Prostaglandina D2/farmacologia , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...