Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(6): e1011204, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37289833

RESUMO

Efficient transmission of herpesviruses is essential for dissemination in host populations; however, little is known about the viral genes that mediate transmission, mostly due to a lack of natural virus-host model systems. Marek's disease is a devastating herpesviral disease of chickens caused by Marek's disease virus (MDV) and an excellent natural model to study skin-tropic herpesviruses and transmission. Like varicella zoster virus that causes chicken pox in humans, the only site where infectious cell-free MD virions are efficiently produced is in epithelial skin cells, a requirement for host-to-host transmission. Here, we enriched for heavily infected feather follicle epithelial skin cells of live chickens to measure both viral transcription and protein expression using combined short- and long-read RNA sequencing and LC/MS-MS bottom-up proteomics. Enrichment produced a previously unseen breadth and depth of viral peptide sequencing. We confirmed protein translation for 84 viral genes at high confidence (1% FDR) and correlated relative protein abundance with RNA expression levels. Using a proteogenomic approach, we confirmed translation of most well-characterized spliced viral transcripts and identified a novel, abundant isoform of the 14 kDa transcript family via IsoSeq transcripts, short-read intron-spanning sequencing reads, and a high-quality junction-spanning peptide identification. We identified peptides representing alternative start codon usage in several genes and putative novel microORFs at the 5' ends of two core herpesviral genes, pUL47 and ICP4, along with strong evidence of independent transcription and translation of the capsid scaffold protein pUL26.5. Using a natural animal host model system to examine viral gene expression provides a robust, efficient, and meaningful way of validating results gathered from cell culture systems.


Assuntos
Herpesviridae , Herpesvirus Galináceo 2 , Doença de Marek , Proteogenômica , Humanos , Animais , Galinhas , Herpesviridae/metabolismo , Herpesvirus Galináceo 2/genética
2.
PLoS Pathog ; 19(2): e1010959, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36749787

RESUMO

Conserved Herpesviridae protein kinases (CHPK) are conserved among all members of the Herpesviridae. Herpesviruses lacking CHPK propagate in cell culture at varying degrees, depending on the virus and cell culture system. CHPK is dispensable for Marek's disease herpesvirus (MDV) replication in cell culture and experimental infection in chickens; however, CHPK-particularly its kinase activity-is essential for horizontal transmission in chickens, also known as natural infection. To address the importance of CHPK during natural infection in chickens, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) based proteomics of samples collected from live chickens. Comparing modification of viral proteins in feather follicle epithelial (FFE) cells infected with wildtype or a CHPK-null virus, we identified the US10 protein (pUS10) as a potential target for CHPK in vivo. When expression of pUS10 was evaluated in cell culture and in FFE skin cells during in vivo infection, pUS10 was severely reduced or abrogated in cells infected with CHPK mutant or CHPK-null viruses, respectively, indicating a potential role for pUS10 in transmission. To test this hypothesis, US10 was deleted from the MDV genome, and the reconstituted virus was tested for replication, horizontal transmission, and disease induction. Our results showed that removal of US10 had no effect on the ability of MDV to transmit in experimentally infected chickens, but disease induction in naturally infected chickens was significantly reduced. These results show CHPK is necessary for pUS10 expression both in cell culture and in the host, and pUS10 is important for disease induction during natural infection.


Assuntos
Alphaherpesvirinae , Herpesviridae , Doença de Marek , Animais , Proteínas Quinases/metabolismo , Cromatografia Líquida , Galinhas , Espectrometria de Massas em Tandem , Herpesviridae/metabolismo , Alphaherpesvirinae/metabolismo , Proteínas Virais/metabolismo , Vírus Oncogênicos
3.
Sci Rep ; 13(1): 2044, 2023 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-36739336

RESUMO

Purinergic receptors (PRs) have been reported as potential therapeutic targets for many viral infections including herpesviruses, which urges the investigation into their role in Marek's disease (MD), a herpesvirus induced cancer in chickens that is an important pathogen for the poultry industry. MD is caused by MD virus (MDV) that has a similar viral life cycle as human varicella zoster virus in that it is shed from infected epithelial skin cells and enters the host through the respiratory route. In this report, PR responses during natural MDV infection and disease progression was examined in MD-resistant white Leghorns (WL) and MD-susceptible Pure Columbian (PC) chickens during natural infection. Whole lung lavage cells (WLLC) and liver tissue samples were collected from chickens infected but showing no clinical signs of MD (Infected) or presenting with clinical disease (Diseased). RNA was extracted followed by RT-qPCR analysis with gene specific primers against members of the P1, P2X, and P2Y PR families. Differential expression (p < 0.05) was observed in breed and disease conditions. Some PRs showed tissue specific expression (P1A1, P2X1, and P2X6 in WLLC) whereas others responded to MDV infection only in MD-susceptible (PC) chickens (P1A2A, P2X1, P2X5, P2X7). P2Y PRs had differential expression in both chicken lines in response to MDV infection and MD progression. This study is the first to our knowledge to examine PR responses during MDV infection and disease progression. These results suggest PR signaling may an important area of research for MDV replication and MD.


Assuntos
Herpesviridae , Herpesvirus Galináceo 2 , Doença de Marek , Animais , Humanos , Galinhas/genética , Herpesviridae/metabolismo , Herpesvirus Galináceo 2/genética , Suscetibilidade a Doenças , Progressão da Doença
4.
Virulence ; 13(1): 980-989, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35658809

RESUMO

One of the greatest achievements of the last century is the development of vaccines against viral diseases. Vaccines are essential for battling infectious diseases and many different formulations are available, including live attenuated vaccines. However, the use of live attenuated vaccines has the potential for adverse effects, including reversion of pathogenicity, recombination, and functional complementation in the host. Marek's disease is a serious disease in poultry controlled by live attenuated vaccines that has resulted in increased virulence over the decades. Recombination between circulating field viruses or vaccines is a proposed mechanism for the increase in virulence, however, complementation between vaccines and field strains has not been demonstrated in chickens. Here, we describe functional complementation of vaccines with virulent virus to functionally complement transmission and spread in the host. Using the natural virus-host model of Marek's disease in chickens, our results show dual infection of target cells in chickens with vaccine and virulent virus providing the opportunity for recombination or complementation to transpire. Interestingly, our controlled results showed no evidence of recombination between vaccine and virulent virus, but functional complementation occurred in two independent experiments providing proof for complementation during natural infection in vaccinated individuals. These results suggest complementation as a potential mechanism for vaccine-mediated viral evolution and the potential for complementation should be taken into consideration when developing novel vaccines.


Assuntos
Coinfecção , Doença de Marek , Doenças das Aves Domésticas , Vacinas Virais , Vírus , Animais , Galinhas , Doença de Marek/prevenção & controle , Vacinas Atenuadas/genética , Vacinas Virais/genética
5.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34704922

RESUMO

Members of the family Herpesviridae have enveloped, spherical virions with characteristic complex structures consisting of symmetrical and non-symmetrical components. The linear, double-stranded DNA genomes of 125-241 kbp contain 70-170 genes, of which 43 have been inherited from an ancestral herpesvirus. In general, herpesviruses have coevolved with and are highly adapted to their hosts, which comprise many mammalian, avian and reptilian species. Following primary infection, they are able to establish lifelong latent infection, during which there is limited viral gene expression. Severe disease is usually observed only in the foetus, the very young, the immunocompromised or following infection of an alternative host. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Herpesviridae, which is available at ictv.global/report/herpesviridae.


Assuntos
Genoma Viral , Herpesviridae , Animais , Evolução Molecular , Herpesviridae/classificação , Herpesviridae/genética , Herpesviridae/fisiologia , Herpesviridae/ultraestrutura , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Adaptação ao Hospedeiro , Vírion/química , Vírion/ultraestrutura , Latência Viral , Replicação Viral
6.
Viruses ; 13(8)2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34452285

RESUMO

Marek's disease (MD) in chickens is caused by Gallid alphaherpesvirus 2, better known as MD herpesvirus (MDV). Current vaccines do not block interindividual spread from chicken-to-chicken, therefore, understanding MDV interindividual spread provides important information for the development of potential therapies to protect against MD, while also providing a natural host to study herpesvirus dissemination. It has long been thought that glycoprotein C (gC) of alphaherpesviruses evolved with their host based on their ability to bind and inhibit complement in a species-selective manner. Here, we tested the functional importance of gC during interindividual spread and host specificity using the natural model system of MDV in chickens through classical compensation experiments. By exchanging MDV gC with another chicken alphaherpesvirus (Gallid alphaherpesvirus 1 or infectious laryngotracheitis virus; ILTV) gC, we determined that ILTV gC could not compensate for MDV gC during interindividual spread. In contrast, exchanging turkey herpesvirus (Meleagrid alphaherpesvirus 1 or HVT) gC could compensate for chicken MDV gC. Both ILTV and MDV are Gallid alphaherpesviruses; however, ILTV is a member of the Iltovirus genus, while MDV is classified as a Mardivirus along with HVT. These results suggest that gC is functionally conserved based on the virus genera (Mardivirus vs. Iltovirus) and not the host (Gallid vs. Meleagrid).


Assuntos
Antígenos Virais/metabolismo , Galinhas/virologia , Herpesvirus Galináceo 2/fisiologia , Doença de Marek/transmissão , Doença de Marek/virologia , Proteínas do Envelope Viral/metabolismo , Animais , Antígenos Virais/genética , Células Cultivadas , Herpesvirus Galináceo 1/classificação , Herpesvirus Galináceo 1/genética , Herpesvirus Meleagrídeo 1/classificação , Herpesvirus Meleagrídeo 1/genética , Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/genética , Proteínas Recombinantes/metabolismo , Perus/virologia , Proteínas do Envelope Viral/genética , Replicação Viral
7.
Pathogens ; 9(6)2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-32599844

RESUMO

Canine distemper virus (CDV) is a cause of significant disease in canids and increasingly recognized as a multi-host pathogen, particularly of non-canid families within Carnivora. CDV outbreaks in sympatric mesocarnivores are routinely diagnosed in the Forest Preserve District of Cook County, Illinois. CDV is diagnosed more commonly and the disease more severe in raccoons and striped skunks than in coyotes. Research in other species suggests host cell receptors may play a role in variable disease outcome, particularly, the signaling lymphocyte activation molecule (SLAM) located on lymphoid cells. To evaluate receptor differences, partial SLAM genes were sequenced, and predicted amino acid (AA) sequences and structural models of the proposed viral interface assessed. Of 263 aligned nucleotide base pairs, 36 differed between species with 24/36 differences between canid and non-canids. Raccoon and skunk predicted AA sequences had higher homology than coyote and raccoon/skunk sequences and 8/11 residue differences were between coyote and raccoons/skunks. Though protein structure was similar, few residue differences were associated with charge and electrostatic potential surface alterations between canids and non-canids. RNAScope®(Advanced Cell Diagnostics, Silicon Valley, USA) ISH revealed low levels of expression that did not differ significantly between species or tissue type. Results suggest that differences in host receptors may impact species-specific disease manifestation.

8.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32493818

RESUMO

The literature on the egress of different herpesviruses after secondary envelopment is contradictory. In this report, we investigated varicella-zoster virus (VZV) egress in a cell line from a child with Pompe disease, a glycogen storage disease caused by a defect in the enzyme required for glycogen digestion. In Pompe cells, both the late autophagy pathway and the mannose-6-phosphate receptor (M6PR) pathway are interrupted. We have postulated that intact autophagic flux is required for higher recoveries of VZV infectivity. To test that hypothesis, we infected Pompe cells and then assessed the VZV infectious cycle. We discovered that the infectious cycle in Pompe cells was remarkably different from that of either fibroblasts or melanoma cells. No large late endosomes filled with VZV particles were observed in Pompe cells; only individual viral particles in small vacuoles were seen. The distribution of the M6PR pathway (trans-Golgi network to late endosomes) was constrained in infected Pompe cells. When cells were analyzed with two different anti-M6PR antibodies, extensive colocalization of the major VZV glycoprotein gE (known to contain M6P residues) and the M6P receptor (M6PR) was documented in the viral highways at the surfaces of non-Pompe cells after maximum-intensity projection of confocal z-stacks, but neither gE nor the M6PR was seen in abundance at the surfaces of infected Pompe cells. Taken together, our results suggested that (i) Pompe cells lack a VZV trafficking pathway within M6PR-positive large endosomes and (ii) most infectious VZV particles in conventional cell substrates are transported via large M6PR-positive vacuoles without degradative xenophagy to the plasma membrane.IMPORTANCE The long-term goal of this research has been to determine why VZV, when grown in cultured cells, invariably is more cell associated and has a lower titer than other alphaherpesviruses, such as herpes simplex virus 1 (HSV1) or pseudorabies virus (PRV). Data from both HSV1 and PRV laboratories have identified a Rab6 secretory pathway for the transport of single enveloped viral particles from the trans-Golgi network within small vacuoles to the plasma membrane. In contrast, after secondary envelopment in fibroblasts or melanoma cells, multiple infectious VZV particles accumulated within large M6PR-positive late endosomes that were not degraded en route to the plasma membrane. We propose that this M6PR pathway is most utilized in VZV infection and least utilized in HSV1 infection, with PRV's usage being closer to HSV1's usage. Supportive data from other VZV, PRV, and HSV1 laboratories about evidence for two egress pathways are included.


Assuntos
Doença de Depósito de Glicogênio Tipo II/metabolismo , Herpesvirus Humano 3/metabolismo , Infecção pelo Vírus da Varicela-Zoster/fisiopatologia , Autofagia/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Varicela/virologia , Endossomos , Exocitose/fisiologia , Herpes Zoster/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 3/patogenicidade , Humanos , Macroautofagia/fisiologia , Receptor IGF Tipo 2/metabolismo , Vacúolos , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion , Rede trans-Golgi/metabolismo
9.
J Virol ; 94(5)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31801854

RESUMO

The Herpesviridae encode many conserved genes, including the conserved herpesvirus protein kinase (CHPK) that has multifunctional properties. In most cases, herpesviruses lacking CHPK can propagate in cell culture to various degrees, depending on the virus and cell culture system. However, in the natural animal model system of Marek's disease alphaherpesvirus (MDV) in chickens, CHPK is absolutely required for interindividual spread from chicken to chicken. The lack of biological reagents for chicken and MDV has limited our understanding of this important gene during interindividual spread. Here, we engineered epitope-tagged proteins in the context of virus infection in order to detect CHPK in the host. Using immunofluorescence assays and Western blotting during infection in cell culture and in chickens, we determined that the invariant lysine 170 (K170) of MDV CHPK is required for interindividual spread and autophosphorylation of CHPK and that mutation to methionine (M170) results in instability of the CHPK protein. Using these newly generated viruses allowed us to examine the expression of CHPK in infected chickens, and these results showed that mutant CHPK localization and late viral protein expression were severely affected in feather follicles wherein MDV is shed, providing important information on the requirement of CHPK for interindividual spread.IMPORTANCE Marek's disease in chickens is caused by Gallid alphaherpesvirus 2, better known as Marek's disease alphaherpesvirus (MDV). Current vaccines only reduce tumor formation but do not block interindividual spread from chicken to chicken. Understanding MDV interindividual spread provides important information for the development of potential therapies to protect against Marek's disease while also providing a reliable natural host in order to study herpesvirus replication and pathogenesis in animals. Here, we studied the conserved Herpesviridae protein kinase (CHPK) in cell culture and during infection in chickens. We determined that MDV CHPK is not required for cell-to-cell spread, for disease induction, and for oncogenicity. However, it is required for interindividual spread, and mutation of the invariant lysine (K170) results in stability issues and aberrant expression in chickens. This study is important because it addresses the critical role CHPK orthologs play in the natural host.


Assuntos
Alphaherpesvirinae/metabolismo , Galinhas/virologia , Herpesviridae/metabolismo , Doença de Marek/virologia , Proteínas Quinases/metabolismo , Proteínas Virais/metabolismo , Animais , Epitopos , Herpesvirus Galináceo 2 , Doença de Marek/transmissão , Modelos Moleculares , Doenças das Aves Domésticas/virologia , Proteínas Quinases/química , Proteínas Quinases/genética , Pele/patologia , Pele/virologia , Proteínas Virais/química , Proteínas Virais/genética
10.
Virology ; 534: 108-113, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31226665

RESUMO

Marek's disease virus (MDV) is a lymphotropic alphaherpesvirus that causes Marek's disease (MD) in chickens. RLORF4 is a MDV-specific gene that is systematically deleted during attenuation of MDV in vitro. Concomitantly, the expression of glycoprotein C (gC) is diminished during attenuation, suggesting these two changes may be linked. Original studies in which RLORF4 was deleted utilized an infectious clone that lacked gC expression due to a frame-shift mutation within the gC open reading frame (UL44). Here, we utilized an infectious clone in which gC expression was restored to test our hypothesis that RLORF4 is important for expression of MDV gC, and subsequently, interindividual spread. Contrary to our hypothesis, gC expression was unaltered during both in vitro and in vivo replication of RLORF4-null MDV and was able to efficiently transmit from chicken to chicken, conclusively showing that RLORF4 does not regulate gC expression and is not required for horizontal transmission.


Assuntos
Antígenos Virais/genética , Herpesvirus Galináceo 2/metabolismo , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/genética , Proteínas Virais/metabolismo , Animais , Antígenos Virais/metabolismo , Galinhas , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética
11.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30518650

RESUMO

The Herpesviridae conserved infected-cell protein 27 (ICP27) is essential for cell culture-based replication of most herpesviruses studied. For members of the Alphaherpesvirinae, ICP27 regulates the expression of many viral genes, including expression of pUL44 (gC), pUL47 (VP13/14), and pUL48 (VP16). These three viral proteins are dysregulated during Marek's disease alphaherpesvirus (MDV) replication in cell culture. MDV replicates in a highly cell-associated manner in cell culture, producing little to no infectious virus. In contrast, infectious cell-free MDV is produced in specialized feather follicle epithelial (FFE) cells of infected chickens, in which these three genes are abundantly expressed. This led us to hypothesize that MDV ICP27, encoded by gene UL54, is a defining factor for the dysregulation of gC, pUL47, and pUL48 and, ultimately, ineffective virus production in cell culture. To address ICP27's role in MDV replication, we generated recombinant MDV with ICP27 deleted (vΔ54). Interestingly, vΔ54 replicated, but plaque sizes were significantly reduced compared to those of parental viruses. The reduced cell-to-cell spread was due to ICP27 since plaque sizes were restored in rescued viruses, as well as when vΔ54 was propagated in cells expressing ICP27 in trans In chickens, vΔ54 replicated, induced disease, and was oncogenic but was unable to transmit from chicken to chicken. To our knowledge, this is the first report showing that the Herpesviridae conserved ICP27 protein is dispensable for replication and disease induction in its natural host.IMPORTANCE Marek's disease (MD) is a devastating oncogenic disease that affects the poultry industry and is caused by MD alphaherpesvirus (MDV). Current vaccines block induction of disease but do not block chicken-to-chicken transmission. There is a knowledge gap in our understanding of how MDV spreads from chicken to chicken. We studied the Herpesviridae conserved ICP27 regulatory protein in cell culture and during MDV infection in chickens. We determined that MDV ICP27 is important but not required for replication in both cell culture and chickens. In addition, MDV ICP27 was not required for disease induction or oncogenicity but was required for chicken-to-chicken transmission. This study is important because it addresses the role of ICP27 during infection in the natural host and provides important information for the development of therapies to protect chickens against MD.


Assuntos
Herpesviridae/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Alphaherpesvirinae/genética , Animais , Galinhas/virologia , Genes Virais , Herpesviridae/genética , Herpesviridae/patogenicidade , Infecções por Herpesviridae/metabolismo , Proteínas Imediatamente Precoces/fisiologia , Doença de Marek/genética , Doença de Marek/virologia , Aves Domésticas/virologia , Proteínas Virais
12.
Open Forum Infect Dis ; 5(6): ofy118, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30014002

RESUMO

BACKGROUND: The infectious cycle of varicella-zoster virus (VZV) after reactivation from the dorsal root ganglia includes replication and assembly of complete enveloped virions in the human skin to cause the characteristic herpes zoster (shingles). METHODS: To pursue studies of innate immunity to VZV infection, we have adapted a fetal skin organ culture model to a human neonatal foreskin explant model. RESULTS: Abundant expression of VZV IE62, gE, and gC was visualized by confocal microscopy while numerous enveloped virions were observed by electron microscopy in infected skin organ cultures. Microarray experiments demonstrated that the patterns of upregulated transcripts differed between VZV-infected cells and VZV-infected skin explants. One result stood out, namely a >30-fold elevated interleukin (IL)-6 level in the infected skin explant that was not present in the infected monolayer culture. The IL-6 results in the polyermase chain reaction (PCR) assay were reproduced by quantitative PCR testing with newly designed primers. To determine if increased transcription was accompanied by increased IL-6 expression, we quantitated the levels of IL-6 protein in the explant media at increasing intervals after infection. We found a statistically significant increase in IL-6 protein levels secreted into the media from VZV-infected skin explants as compared with mock-infected explants. CONCLUSIONS: The cellular stress response to VZV infection in neonatal skin explants included highly elevated levels of IL-6 transcription and expression. This skin organ model could be adapted to other viruses with a skin tropism, such as herpes simplex virus.

13.
Adv Anat Embryol Cell Biol ; 223: 195-224, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28528445

RESUMO

Interindividual spread of herpesviruses is essential for the virus life cycle and maintenance in host populations. For most herpesviruses, the virus-host relationship is close, having coevolved over millions of years resulting in comparatively high species specificity. The mechanisms governing interindividual spread or horizontal transmission are very complex, involving conserved herpesviral and cellular proteins during the attachment, entry, replication, and egress processes of infection. Also likely, specific herpesviruses have evolved unique viral and cellular interactions during cospeciation that are dependent on their relationship. Multiple steps are required for interindividual spread including virus assembly in infected cells; release into the environment, followed by virus attachment; and entry into new hosts. Should any of these steps be compromised, transmission is rendered impossible. This review will focus mainly on the natural virus-host model of Marek's disease virus (MDV) in chickens in order to delineate important steps during interindividual spread.


Assuntos
Infecções por Herpesviridae/transmissão , Infecções por Herpesviridae/virologia , Herpesviridae/fisiologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Proteínas Virais/metabolismo , Internalização do Vírus , Replicação Viral
14.
J Virol ; 90(19): 8673-85, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440906

RESUMO

UNLABELLED: Varicella-zoster virus (VZV) is an extremely cell-associated herpesvirus with limited egress of viral particles. The induction of autophagy in VZV-infected monolayers is easily detectable; inhibition of autophagy leads to decreased VZV glycoprotein biosynthesis and diminished viral titers. To explain how autophagic flux could exert a proviral effect on the VZV infectious cycle, we postulated that the VZV exocytosis pathway following secondary envelopment may converge with the autophagy pathway. This hypothesis depended on known similarities between VZV gE and autophagy-related (Atg) Atg9/Atg16L1 trafficking pathways. Investigations were carried out with highly purified fractions of VZV virions. When the virion fraction was tested for the presence of autophagy and endosomal proteins, microtubule-associated protein 1 light chain (MAP1LC3B) and Ras-like GTPase 11 (Rab11) were detected. By two-dimensional (2D) and 3D imaging after immunolabeling, both proteins also colocalized with VZV gE in a proportion of cytoplasmic vesicles. When purified VZV virions were enumerated after immunoelectron microscopy, gold beads were detected on viruses following incubation with antibodies to VZV gE (∼100%), Rab11 (50%), and LC3B (30%). Examination of numerous electron micrographs demonstrated that enveloped virions were housed in single-membraned vesicles; viral particles were not observed in autophagosomes. Taken together, our data suggested that some viral particles after secondary envelopment accumulated in a heterogeneous population of single-membraned vesicular compartments, which were decorated with components from both the endocytic pathway (Rab11) and the autophagy pathway (LC3B). The latter cytoplasmic viral vesicles resembled an amphisome. IMPORTANCE: VZV infection leads to increased autophagic flux, while inhibition of autophagy leads to a marked reduction in virus spread. In this investigation of the proviral role of autophagy, we found evidence for an intersection of viral exocytosis and autophagy pathways. Specifically, both LC3-II and Rab11 proteins copurified with some infectious VZV particles. The results suggested that a subpopulation of VZV particles were carried to the cell surface in single-walled vesicles with attributes of an amphisome, an organelle formed from the fusion of an endosome and an autophagosome. Our results also addressed the interpretation of autophagy/xenophagy results with mutated herpes simplex virus lacking its ICP34.5 neurovirulence gene (HSVΔ34.5). The VZV genome lacks an ICP34.5 ortholog, yet we found no evidence of VZV particles housed in a double-membraned autophagosome. In other words, xenophagy, a degradative process documented after infection with HSVΔ34.5, was not observed in VZV-infected cells.


Assuntos
Autofagia , Endossomos/metabolismo , Exocitose , Herpesvirus Humano 3/fisiologia , Vírion/metabolismo , Liberação de Vírus , Linhagem Celular , Humanos , Microscopia Imunoeletrônica , Proteínas Associadas aos Microtúbulos/análise , Proteínas do Envelope Viral/análise , Vírion/química , Proteínas rab de Ligação ao GTP/análise
15.
Virology ; 484: 213-226, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26117307

RESUMO

Marek's disease virus (MDV) is a lymphotropic alphaherpesvirus that replicates in a highly cell-associated manner in vitro. Production of infectious cell-free virus only occurs in feather follicle epithelial (FFE) cells of infected chicken skins. Previously, we described differential expression for a core alphaherpesvirus protein, pUL47 that was found to be abundantly expressed in FFE cells of infected chickens, while barely detectable during in vitro propagation. Here, we further examined the dynamics of expression of four tegument proteins within the UL46-49 locus during in vitro and in situ replication. All four proteins examined were expressed abundantly in situ, whereas both pUL47 and pUL48 expression were barely detectable in vitro. Replacement of the putative UL47 and UL48 promoters with the minimal cytomegalovirus promoter enhanced mRNA and protein expression in vitro. Interestingly, enhanced expression of pUL47 resulted in increased UL46, UL48, and UL49 transcripts that resulted in increased pUL46 and pUL48 expression.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/genética , Proteínas Estruturais Virais/metabolismo , Animais , Células Cultivadas , Galinhas , Células Epiteliais/virologia , Herpesvirus Galináceo 2/metabolismo
16.
Virus Res ; 201: 50-60, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25725150

RESUMO

Marek's disease virus (MDV) is a lymphotropic alphaherpesvirus and causes Marek's disease (MD) in chickens. RLORF4 is an MDV-specific gene located in the repeat long (RL) regions of the genome and is directly involved in attenuation. In this report, we generated recombinant (r)MDVs in which eGFP or mRFP was inserted in-frame of the 3' end of the RLORF4 gene. In vitro growth was unaffected and infected cells could be identified by using fluorescent microscopy. Interestingly, though inserted in-frame with RLORF4, eGFP and mRFP were expressed alone, confirming mRNA expression and splicing within the RL of MDV is complex. In vivo, rMDVs expressing mRFP or eGFP caused tumors similar to wild-type MDV. Fluorescent protein expression could be seen in spleen, tumor, and feather follicle epithelial cells. These results show that expression of fluorescent proteins within the RL region results in fluorescent rMDVs that still maintains full pathogenicity in the chicken.


Assuntos
Expressão Gênica , Genes Reporter , Genoma Viral , Proteínas Luminescentes/análise , Mardivirus/fisiologia , Doença de Marek/virologia , Coloração e Rotulagem/métodos , Animais , Galinhas , Plumas/patologia , Plumas/virologia , Proteínas Luminescentes/genética , Mardivirus/genética , Mardivirus/crescimento & desenvolvimento , Mardivirus/patogenicidade , Doença de Marek/patologia , Microscopia de Fluorescência , Mutagênese Insercional , Neoplasias/patologia , Neoplasias/virologia , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Baço/virologia , Replicação Viral
17.
Avian Dis ; 57(2 Suppl): 503-8, 2013 06.
Artigo em Inglês | MEDLINE | ID: mdl-23901768

RESUMO

The role of pp38 in the pathogenesis of Marek's disease (MD) has not been fully elucidated. Previously, we reported the presence of two splice variants (Spl A and Spl B) for pp38. We also reported that the wild-type pp38 (WT), as well as the mutated pp38 (MUT), altered the oxidative phosphorylation pathway in infected cells. To determine whether the different forms of pp38 are important for the pathogenesis of MD, we generated RB-1B-based bacterial artificial chromosome (BAC) clones expressing pp38MUT, pp38Sp1 A, and pp38Spl B. Infectious viruses were recovered from these BAC clones in chick kidney cells (CKC). The Spl A and Spl B viruses had significantly smaller plaque sizes and replicated to a lesser degree in CKC than the WT and MUT viruses. Two in vivo experiments were conducted by inoculating 7-day-old P2a chicks with 1000 plaque-forming units of each virus. In the first experiment, chicks were sacrificed at 4, 8, 11, and 15 days postinfection (PI). WT and MUT viruses had similar viremia levels using virus isolation and quantitative real-time PCR (qPCR) assays, whereas Spl A and Spl B viruses had significantly lower viremia levels than WT and MUT viruses. In the second experiment, we showed that tumor development and MD mortality were similar in the WT- and MUT-infected chickens, with all birds MD positive at 5 wk PI. In contrast, chickens infected with Spl B and Spl A had a significantly lower MD incidence at 11 wk PI, when the experiment was terminated.


Assuntos
Transformação Celular Neoplásica , Galinhas , Mardivirus/genética , Mardivirus/patogenicidade , Doença de Marek/imunologia , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Animais , Transformação Celular Neoplásica/imunologia , Células Cultivadas , Embrião de Galinha , Cromossomos Artificiais Bacterianos/genética , Mardivirus/metabolismo , Doença de Marek/virologia , Fosfoproteínas/genética , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Recombinação Genética , Organismos Livres de Patógenos Específicos , Proteínas Virais/genética
18.
Avian Dis ; 57(2 Suppl): 509-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23901769

RESUMO

Marek's disease (MD) is a highly transmissible, herpesvirus-associated malignancy of chickens and turkeys caused by Marek's disease virus (MDV). MD is currently controlled through the use of nonsterilizing vaccines composed of antigenically related, apathogenic herpesviruses Mardivirus 2 (MDV-2), Meleagrid herpesvirus 1 (herpesvirus of turkeys, HVT), or attenuated MDV-1 strain CVI988 (Rispens). Since the mid-1960s, field strains of MDV have increased in virulence, due, in part, to the widespread use of vaccines since the early 1970s. One mutation that we have identified common to very virulent field strains (vv and vv+MDVs) since the 1990s has been a mutation in the UL1 gene, encoding glycoprotein L (gL). This mutation, a 12-nucleotide (nt) deletion in the signal peptide of gL, has been associated with increased virulence and decreased vaccine protection in the context of challenge with a vv+MDV, strain TK. To determine whether this mutation alone was sufficient to confer increased virulence, we introduced this mutation into the transmission-competent pRB-1B bacterial artificial chromosome (BAC) using two-step, Red-mediated recombination. The resulting mutant, pRB-1BgLdelta, was tested for changes in replication in cell culture using multistep growth curves, plaque size analysis, viral burst analysis, and the ability to compete with the parental virus when co-transfected at different ratios and sequentially passaged. In addition, we examined this mutant for changes in pathogenicity in inoculated and contact-exposed unvaccinated and vaccinated chickens. Our data show minor differences in plaque sizes in cell culture, but no discernible changes in the infection of specific-pathogen-free (SPF) leghorn chickens. We therefore conclude that although this mutation is indeed common to MDV field strains isolated in the eastern United States, it is insufficient to confer increased virulence or loss of vaccine protection previously observed for a vv+MDV strain having this mutation.


Assuntos
Galinhas , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/imunologia , Proteínas Oncogênicas Virais/genética , Doenças das Aves Domésticas/imunologia , Proteínas do Envelope Viral/genética , Animais , Células Cultivadas , Embrião de Galinha , Cromossomos Artificiais Bacterianos/genética , Vacinas contra Herpesvirus/genética , Vacinas contra Herpesvirus/imunologia , Doença de Marek/virologia , Mutação , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
19.
Virology ; 437(2): 110-7, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23399034

RESUMO

Marek's disease virus (MDV) encodes an ubiquitin-specific protease (USP) within its UL36 gene. USP is highly conserved among herpesviruses and was shown to be important for MDV replication and pathogenesis in MDV's natural host, the chicken. To further investigate the role of MDV USP, several recombinant (r) MDVs were generated and their in vitro phenotypes were evaluated using plaque size and growth kinetics assays. We discovered that the N-terminus of pUL36 is essential for MDV replication and could not be complemented by ectopic expression of MDV USP. In addition, we demonstrated that the region located between the conserved glutamine (Q85) and leucine (L106) residues comprising the active site cysteine (C98) is also essential for MDV replication. Based on the analyses of the rMDVs generated here, we concluded that MDV USP likely contributes to the structure and/or stability of pUL36 and affects replication and oncogenesis of MDV beyond its enzymatic activity.


Assuntos
Endopeptidases/metabolismo , Mardivirus/fisiologia , Replicação Viral , Sequência de Aminoácidos , Animais , Linhagem Celular , Galinhas , Análise Mutacional de DNA , Endopeptidases/genética , Mardivirus/genética , Mardivirus/crescimento & desenvolvimento , Mardivirus/patogenicidade , Doença de Marek/patologia , Doença de Marek/virologia , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Alinhamento de Sequência , Proteases Específicas de Ubiquitina , Ensaio de Placa Viral
20.
PLoS One ; 7(5): e37428, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22629393

RESUMO

Hosts can be infected with multiple herpesviruses, known as superinfection; however, superinfection of cells is rare due to the phenomenon known as superinfection inhibition. It is believed that dual infection of cells occurs in nature, based on studies examining genetic exchange between homologous alphaherpesviruses in the host, but to date, this has not been directly shown in a natural model. In this report, gallid herpesvirus 2 (GaHV-2), better known as Marek's disease virus (MDV), was used in its natural host, the chicken, to determine whether two homologous alphaherpesviruses can infect the same cells in vivo. MDV shares close similarities with the human alphaherpesvirus, varicella zoster virus (VZV), with respect to replication in the skin and exit from the host. Recombinant MDVs were generated that express either the enhanced GFP (eGFP) or monomeric RFP (mRFP) fused to the UL47 (VP13/14) herpesvirus tegument protein. These viruses exhibited no alteration in pathogenic potential and expressed abundant UL47-eGFP or -mRFP in feather follicle epithelial cells in vivo. Using laser scanning confocal microscopy, it was evident that these two similar, but distinguishable, viruses were able to replicate within the same cells of their natural host. Evidence of superinfection inhibition was also observed. These results have important implications for two reasons. First, these results show that during natural infection, both dual infection of cells and superinfection inhibition can co-occur at the cellular level. Secondly, vaccination against MDV with homologous alphaherpesvirus like attenuated GaHV-2, or non-oncogenic GaHV-3 or meleagrid herpesvirus (MeHV-1) has driven the virus to greater virulence and these results implicate the potential for genetic exchange between homologous avian alphaherpesviruses that could drive increased virulence. Because the live attenuated varicella vaccine is currently being administered to children, who in turn could be superinfected by wild-type VZV, this could potentiate recombination events of VZV as well.


Assuntos
Células Epiteliais/virologia , Herpes Zoster/virologia , Herpesvirus Galináceo 2/metabolismo , Herpesvirus Humano 3/metabolismo , Doença de Marek/virologia , Pele/virologia , Superinfecção/virologia , Animais , Galinhas , Coinfecção , Células Epiteliais/metabolismo , Herpes Zoster/metabolismo , Herpesvirus Galináceo 2/genética , Herpesvirus Humano 3/genética , Pele/metabolismo , Superinfecção/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...