Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Vet Res ; 49(1): 31, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29587836

ABSTRACT

Marek's disease is a multi-faceted highly contagious disease affecting chickens caused by the Marek's disease alphaherpesvirus (MDV). MDV early infection induces a transient immunosuppression, which is associated with thymus and bursa of Fabricius atrophy. Little is known about the cellular processes involved in primary lymphoid organ atrophy. Here, by in situ TUNEL assay, we demonstrate that MDV infection results in a high level of apoptosis in the thymus and bursa of Fabricius, which is concomitant to the MDV lytic cycle. Interestingly, we observed that in the thymus most of the MDV infected cells at 6 days post-infection (dpi) were apoptotic, whereas in the bursa of Fabricius most of the apoptotic cells were uninfected suggesting that MDV triggers apoptosis by two different modes in these two primary lymphoid organs. In addition, a high decrease of cell proliferation was observed from 6 to 14 dpi in the bursa of Fabricius follicles, and not in the thymus. Finally, with an adapted absolute blood lymphocyte count, we demonstrate a major B-lymphopenia during the two 1st weeks of infection, and propose this method as a potent non-invasive tool to diagnose MDV bursa of Fabricius infection and atrophy. Our results demonstrate that the thymus and bursa of Fabricius atrophies are related to different cell mechanisms, with different temporalities, that affect infected and uninfected cells.


Subject(s)
Atrophy/veterinary , Chickens , Herpesvirus 2, Gallid/physiology , Lymphoid Tissue/pathology , Marek Disease/physiopathology , Poultry Diseases/physiopathology , Animals , Apoptosis , Atrophy/pathology , Atrophy/physiopathology , Atrophy/virology , Cell Proliferation , Lymphoid Tissue/physiopathology , Lymphopenia , Marek Disease/pathology , Marek Disease/virology , Poultry Diseases/pathology , Poultry Diseases/virology
2.
PLoS One ; 12(4): e0175259, 2017.
Article in English | MEDLINE | ID: mdl-28406989

ABSTRACT

Marek's disease virus is the etiological agent of a major lymphoproliferative disorder in poultry and the prototype of the Mardivirus genus. Primary avian somatic cells are currently used for virus replication and vaccine production, but they are largely refractory to any genetic modification compatible with the preservation of intact viral susceptibility. We explored the concept of induction of viral replication permissiveness in an established pluripotent chicken embryonic stem cell-line (cES) in order to derive a new fully susceptible cell-line. Chicken ES cells were not permissive for Mardivirus infection, but as soon as differentiation was triggered, replication of Marek's disease virus was detected. From a panel of cyto-differentiating agents, hexamethylene bis (acetamide) (HMBA) was found to be the most efficient regarding the induction of permissiveness. These initial findings prompted us to analyse the effect of HMBA on gene expression, to derive a new mesenchymal cell line, the so-called ESCDL-1, and monitor its susceptibility for Mardivirus replication. All Mardiviruses tested so far replicated equally well on primary embryonic skin cells and on ESCDL-1, and the latter showed no variation related to its passage number in its permissiveness for virus infection. Viral morphogenesis studies confirmed efficient multiplication with, as in other in vitro models, no extra-cellular virus production. We could show that ESCDL-1 can be transfected to express a transgene and subsequently cloned without any loss in permissiveness. Consequently, ESCDL-1 was genetically modified to complement viral gene deletions thus yielding stable trans-complementing cell lines. We herein claim that derivation of stable differentiated cell-lines from cES cell lines might be an alternative solution to the cultivation of primary cells for virology studies.


Subject(s)
Embryonic Stem Cells/virology , Mardivirus/physiology , Virus Replication/physiology , Acetamides/pharmacology , Animals , Cell Line , Chick Embryo , Chickens , Embryonic Stem Cells/metabolism , Marek Disease/metabolism , Virus Replication/drug effects
3.
Virol J ; 13: 7, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26742789

ABSTRACT

BACKGROUND: Marek's disease is a virus disease with worldwide distribution that causes major losses to poultry production. Vaccines against Marek's disease virus, an oncogenic alphaherpesvirus, reduce tumour formation but have no effect on virus shedding. Successful horizontal virus transmission is linked to the active viral replication in feather follicle epithelial cells of infected chickens, from which infectious viral particles are shed into the environment. The feather follicle epithelium is the sole tissue in which those infectious particles are produced and no in vitro cell-systems can support this highly efficient morphogenesis. We previously characterized embryonic stem-cell-derived keratinocytes, showing they display a marker-gene profile similar to skin keratinocytes, and therefore we tested their susceptibility to Marek's disease virus infection. FINDINGS: We show herein that keratinocytes derived from chicken embryonic stem-cells are fully permissive to the replication of either non-pathogenic or pathogenic Marek's disease viruses. All viruses replicated on all three keratinocyte lines and kinetics of viral production as well as viral loads were similar to those obtained on primary cells. Morphogenesis studies were conducted on infected keratinocytes and on corneocytes, showing that all types of capsids/virions were present inside the cells, but extracellular viruses were absent. CONCLUSIONS: The keratinocyte lines are the first epithelial cell-line showing ectodermal specific markers supporting Marek's disease virus replication. In this in vitro model the replication lead to the production of cell-associated viral progeny. Further work will be devoted to the study of relationship between 3D differentiation of keratinocytes and Marek's disease virus replication.


Subject(s)
Embryonic Stem Cells/cytology , Keratinocytes/cytology , Keratinocytes/virology , Mardivirus/physiology , Virus Replication , Animals , Cells, Cultured , Chick Embryo , Mardivirus/ultrastructure , Marek Disease/virology
4.
J Gen Virol ; 97(2): 480-486, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26612074

ABSTRACT

T-lymphocytes are central targets of Marek's disease, a major chicken disease induced by the oncogenic alphaherpesvirus Marek's disease virus (MDV). T-lymphocyte infection is also associated with immunosuppression and virus latency. To decipher viral morphogenesis in T-lymphocytes, we used the recombinant vRB-1B 47EGFP marker virus to generate a new lymphoblastoid cell line, 3867K, that exhibited typical properties of other MDV-transformed chicken cell lines in term of cell markers, reactivation rate and infectivity. Examination of reactivating EGFP-positive 3867K cells by transmission electron microscopy revealed the presence of most types of herpesvirus particles inside the cells but no extracellular ones. Quantification of virion types indicated only 5% cytoplasmic particles, with 0.5% being mature. This study demonstrated that MDV morphogenesis is complete upon reactivation in T-lymphocytes, albeit with poor efficiency, with a defect in the exit of virions from the nucleus and secondary envelopment, as occurs in infected fibroblasts.


Subject(s)
Herpesvirus 2, Gallid/physiology , T-Lymphocytes/virology , Virion/ultrastructure , Virus Activation , Virus Assembly , Animals , Cell Line , Chickens , Fibroblasts/virology , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Herpesvirus 2, Gallid/genetics , Microscopy, Electron, Transmission , Molecular Biology/methods , Virology/methods
5.
Virology ; 484: 213-226, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26117307

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Gallid/genetics , Viral Structural Proteins/metabolism , Animals , Cells, Cultured , Chickens , Epithelial Cells/virology , Herpesvirus 2, Gallid/metabolism
6.
PLoS One ; 10(3): e0121751, 2015.
Article in English | MEDLINE | ID: mdl-25812141

ABSTRACT

Merkel cell polyomavirus (MCPyV) is the first polyomavirus clearly associated with a human cancer, i.e. the Merkel cell carcinoma (MCC). Polyomaviruses are small naked DNA viruses that induce a robust polyclonal antibody response against the major capsid protein (VP1). However, the polyomavirus VP1 capsid protein epitopes have not been identified to date. The aim of this study was to identify the neutralizing epitopes of the MCPyV capsid. For this goal, four VP1 mutants were generated by insertional mutagenesis in the BC, DE, EF and HI loops between amino acids 88-89, 150-151, 189-190, and 296-297, respectively. The reactivity of these mutants and wild-type VLPs was then investigated with anti-VP1 monoclonal antibodies and anti-MCPyV positive human sera. The findings together suggest that immunodominant conformational neutralizing epitopes are present at the surface of the MCPyV VLPs and are clustered within BC and EF loops.


Subject(s)
Capsid Proteins/immunology , Epitopes/immunology , Merkel cell polyomavirus/immunology , Protein Interaction Domains and Motifs/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Cross Reactions/immunology , Epitope Mapping , Female , Humans , Immunodominant Epitopes/immunology , Merkel cell polyomavirus/genetics , Mice , Models, Molecular , Mutation , Protein Conformation
7.
Stem Cell Res ; 14(2): 224-37, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25702531

ABSTRACT

A common challenge in avian cell biology is the generation of differentiated cell-lines, especially in the keratinocyte lineage. Only a few avian cell-lines are available and very few of them show an interesting differentiation profile. During the last decade, mammalian embryonic stem cell-lines were shown to differentiate into almost all lineages, including keratinocytes. Although chicken embryonic stem cells had been obtained in the 1990s, few differentiation studies toward the ectodermal lineage were reported. Consequently, we explored the differentiation of chicken embryonic stem cells toward the keratinocyte lineage by using a combination of stromal induction, ascorbic acid, BMP4 and chicken serum. During the induction period, we observed a downregulation of pluripotency markers and an upregulation of epidermal markers. Three homogenous cell populations were derived, which were morphologically similar to chicken primary keratinocytes, displaying intracellular lipid droplets in almost every pavimentous cell. These cells could be serially passaged without alteration of their morphology and showed gene and protein expression profiles of epidermal markers similar to chicken primary keratinocytes. These cells represent an alternative to the isolation of chicken primary keratinocytes, being less cumbersome to handle and reducing the number of experimental animals used for the preparation of primary cells.


Subject(s)
Embryonic Stem Cells/cytology , Keratinocytes/cytology , Animals , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Cells, Cultured , Chickens , Embryonic Stem Cells/metabolism , Humans , Keratinocytes/metabolism
8.
PLoS One ; 9(6): e100004, 2014.
Article in English | MEDLINE | ID: mdl-24945933

ABSTRACT

Marek's disease is one of the most common viral diseases of poultry affecting chicken flocks worldwide. The disease is caused by an alphaherpesvirus, the Marek's disease virus (MDV), and is characterized by the rapid onset of multifocal aggressive T-cell lymphoma in the chicken host. Although several viral oncogenes have been identified, the detailed mechanisms underlying MDV-induced lymphomagenesis are still poorly understood. Many viruses modulate cell cycle progression to enhance their replication and persistence in the host cell, in the case of some oncogenic viruses ultimately leading to cellular transformation and oncogenesis. In the present study, we found that MDV, like other viruses, is able to subvert the cell cycle progression by triggering the proliferation of low proliferating chicken cells and a subsequent delay of the cell cycle progression into S-phase. We further identified the tegument protein VP22 (pUL49) as a major MDV-encoded cell cycle regulator, as its vector-driven overexpression in cells lead to a dramatic cell cycle arrest in S-phase. This striking functional feature of VP22 appears to depend on its ability to associate with histones in the nucleus. Finally, we established that VP22 expression triggers the induction of massive and severe DNA damages in cells, which might cause the observed intra S-phase arrest. Taken together, our results provide the first evidence for a hitherto unknown function of the VP22 tegument protein in herpesviral reprogramming of the cell cycle of the host cell and its potential implication in the generation of DNA damages.


Subject(s)
Cell Cycle Checkpoints , DNA Damage , Mardivirus/metabolism , S Phase , Viral Proteins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Chickens , DNA Breaks, Double-Stranded , Histones/metabolism , Marek Disease/pathology , Protein Transport , Subcellular Fractions/metabolism
9.
Vet Res ; 44: 125, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24359464

ABSTRACT

Marek's disease virus (MDV) is an alpha-herpesvirus causing Marek's disease in chickens, mostly associated with T-cell lymphoma. VP22 is a tegument protein abundantly expressed in cells during the lytic cycle, which is essential for MDV spread in culture. Our aim was to generate a pathogenic MDV expressing a green fluorescent protein (EGFP) fused to the N-terminus of VP22 to better decipher the role of VP22 in vivo and monitor MDV morphogenesis in tumors cells. In culture, rRB-1B EGFP22 led to 1.6-fold smaller plaques than the parental virus. In chickens, the rRB-1B EGFP22 virus was impaired in its ability to induce lymphoma and to spread in contact birds. The MDV genome copy number in blood and feathers during the time course of infection indicated that rRB-1B EGFP22 reached its two major target cells, but had a growth defect in these two tissues. Therefore, the integrity of VP22 is critical for an efficient replication in vivo, for tumor formation and horizontal transmission. An examination of EGFP fluorescence in rRB-1B EGFP22-induced tumors showed that about 0.1% of the cells were in lytic phase. EGFP-positive tumor cells were selected by cytometry and analyzed for MDV morphogenesis by transmission electron microscopy. Only few particles were present per cell, and all types of virions (except mature enveloped virions) were detected unequivocally inside tumor lymphoid cells. These results indicate that MDV morphogenesis in tumor cells is more similar to the morphorgenesis in fibroblastic cells in culture, albeit poorly efficient, than in feather follicle epithelial cells.


Subject(s)
Chickens , Herpesvirus 2, Gallid/physiology , Herpesvirus 2, Gallid/pathogenicity , Marek Disease/virology , Poultry Diseases/virology , Viral Proteins/genetics , Animals , Carcinogenesis , Cells, Cultured , Green Fluorescent Proteins , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/metabolism , Marek Disease/pathology , Marek Disease/transmission , Poultry Diseases/pathology , Poultry Diseases/transmission , Viral Proteins/metabolism , Virulence , Virus Replication
10.
PLoS One ; 7(8): e44072, 2012.
Article in English | MEDLINE | ID: mdl-22952878

ABSTRACT

Marek's Disease Virus (MDV) is an avian alpha-herpesvirus that only spreads from cell-to-cell in cell culture. While its cell-to-cell spread has been shown to be dependent on actin filament dynamics, the mechanisms regulating this spread remain largely unknown. Using a recombinant BAC20 virus expressing an EGFPVP22 tegument protein, we found that the actin cytoskeleton arrangements and cell-cell contacts differ in the center and periphery of MDV infection plaques, with cells in the latter areas showing stress fibers and rare cellular projections. Using specific inhibitors and activators, we determined that Rho-ROCK pathway, known to regulate stress fiber formation, and Rac-PAK, known to promote lamellipodia formation and destabilize stress fibers, had strong contrasting effects on MDV cell-to-cell spread in primary chicken embryo skin cells (CESCs). Inhibition of Rho and its ROCKs effectors led to reduced plaque sizes whereas inhibition of Rac or its group I-PAKs effectors had the adverse effect. Importantly, we observed that the shape of MDV plaques is related to the semi-ordered arrangement of the elongated cells, at the monolayer level in the vicinity of the plaques. Inhibition of Rho-ROCK signaling also resulted in a perturbation of the cell arrangement and a rounding of plaques. These opposing effects of Rho and Rac pathways in MDV cell-to-cell spread were validated for two parental MDV recombinant viruses with different ex vivo spread efficiencies. Finally, we demonstrated that Rho/Rac pathways have opposing effects on the accumulation of N-cadherin at cell-cell contact regions between CESCs, and defined these contacts as adherens junctions. Considering the importance of adherens junctions in HSV-1 cell-to-cell spread in some cell types, this result makes of adherens junctions maintenance one potential and attractive hypothesis to explain the Rho/Rac effects on MDV cell-to-cell spread. Our study provides the first evidence that MDV cell-to-cell spread is regulated by Rho/Rac signaling.


Subject(s)
Herpesvirus 2, Gallid/physiology , Marek Disease/pathology , Signal Transduction , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Communication/drug effects , Cellular Microenvironment/drug effects , Chick Embryo , Herpesvirus 2, Gallid/drug effects , Lipopolysaccharides/pharmacology , Lysophospholipids/pharmacology , Marek Disease/virology , Movement/drug effects , Polymerization/drug effects , Signal Transduction/drug effects , Skin/drug effects , Skin/embryology , Skin/pathology , Skin/virology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stress Fibers/drug effects , Stress Fibers/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors
11.
Virus Res ; 159(1): 32-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21557972

ABSTRACT

Avian influenza virus (AIV) infections of the chicken occur via the respiratory route. Unlike ducks which are considered as a natural AIV reservoir, chickens are highly susceptible to AIV infections and do not possess the RIG-I pattern recognition receptor involved in triggering the antiviral interferon response. To study the chicken innate immune response to AIV in the respiratory tract, we established an epithelial cell line (CLEC213) from lung explants of white leghorn chickens. CLEC213 cells exhibited a polyhedral morphology and formed cohesive clusters bound through tight junctions as assessed by electron microscopy. Expression of E-cadherin but not vimentin could be detected as expected for cells of epithelial origin. In addition, CLEC213 cells showed characteristics similar to those of mammalian type II pneumocytes, including the presence of intracytoplasmic vacuoles filled with a mucopolysaccharide material, alkaline phosphatase activity, transcription of chicken lung collectins genes (cLL and SPA), and some intracytoplasmic lamellar-like bodies. CLEC213 cells showed a constitutive expression level of TLR3 and TLR4 and were responsive to stimulation with the respective agonists, poly (I:C) and LPS: between 4h and 24h after treatment, a strong increase in the expression of IFN-α, IFN-ß and IL-8 genes could be detected. Furthermore, CLEC213 cells supported efficient growth of the low pathogenicity avian influenza virus H6N2 (A/duck/France/05057a/2005) in the presence or the absence of trypsin in the culture media. At 4h post-infection, the H6N2 virus induced highly elevated levels of expression of IFN-α and IL-8, moderately elevated levels of LITAF, TGF-ß4 and CCL5. However, an increase of IFN-ß gene expression could not be detected in response to AIV infection. In conclusion, like mammalian type II pneumocytes, CLEC213 are able to mount a robust cytokine and chemokine immune response to microbial patterns and viral infection. We hypothesize that they could derive from lung atrial granular cells. The involvement of such type of lung epithelial cells in the respiratory tract defence of the chicken can thus be further studied.


Subject(s)
Cell Line , Epithelial Cells/physiology , Epithelial Cells/virology , Influenza A virus/growth & development , Animals , Chickens , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Influenza A virus/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , Lung , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism
12.
J Biol Chem ; 285(17): 13233-43, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20172856

ABSTRACT

The influenza A virus PB1-F2 protein, encoded by an alternative reading frame in the PB1 polymerase gene, displays a high sequence polymorphism and is reported to contribute to viral pathogenesis in a sequence-specific manner. To gain insights into the functions of PB1-F2, the molecular structure of several PB1-F2 variants produced in Escherichia coli was investigated in different environments. Circular dichroism spectroscopy shows that all variants have a random coil secondary structure in aqueous solution. When incubated in trifluoroethanol polar solvent, all PB1-F2 variants adopt an alpha-helix-rich structure, whereas incubated in acetonitrile, a solvent of medium polarity mimicking the membrane environment, they display beta-sheet secondary structures. Incubated with asolectin liposomes and SDS micelles, PB1-F2 variants also acquire a beta-sheet structure. Dynamic light scattering revealed that the presence of beta-sheets is correlated with an oligomerization/aggregation of PB1-F2. Electron microscopy showed that PB1-F2 forms amorphous aggregates in acetonitrile. In contrast, at low concentrations of SDS, PB1-F2 variants exhibited various abilities to form fibers that were evidenced as amyloid fibers in a thioflavin T assay. Using a recombinant virus and its PB1-F2 knock-out mutant, we show that PB1-F2 also forms amyloid structures in infected cells. Functional membrane permeabilization assays revealed that the PB1-F2 variants can perforate membranes at nanomolar concentrations but with activities found to be sequence-dependent and not obviously correlated with their differential ability to form amyloid fibers. All of these observations suggest that PB1-F2 could be involved in physiological processes through different pathways, permeabilization of cellular membranes, and amyloid fiber formation.


Subject(s)
Amyloid/chemistry , Cell Membrane/chemistry , Influenza A virus/chemistry , Viral Proteins/chemistry , Acetonitriles/chemistry , Amyloid/genetics , Amyloid/metabolism , Amyloid/ultrastructure , Animals , Benzothiazoles , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Dogs , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza A virus/pathogenicity , Mutation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thiazoles/chemistry , Trifluoroethanol/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Vet Res ; 40(4): 28, 2009.
Article in English | MEDLINE | ID: mdl-19284966

ABSTRACT

Marek's disease virus type 1 (MDV-1) shows a strict dependency on the direct cell-to-cell spread for its propagation in cell culture. As MDV-1 shows an impaired nuclear egress in cell culture, we wished to address the characterization of capsid/tegument genes which may intervene in the maturation of intranuclear capsids. Orthologs of UL17 are present in all herpesviruses and, in all reported case, were shown to be essential for viral growth, playing a role in capsid maturation and DNA packaging. As only HSV-1 and PrV UL17 proteins have been characterized so far, we wished to examine the role of MDV-1 pUL17 in virus replication. To analyze MDV-1 UL17 gene function, we created deletion mutants or point mutated the open reading frame (ORF) to interrupt its coding phase. We established that a functional ORF UL17 is indispensable for MDV-1 growth. We chose to characterize the virally encoded protein by tagging the 729 amino-acid long protein with a repeat of the HA peptide that was fused to its C-terminus. Protein pUL17 was identified in infected cell extracts as an 82 kDa protein which localized to the nucleus, colocalizing with VP5, the major capsid protein, and VP13/14, a major tegument protein. By using green fluorescent protein fusion and HA tagged proteins expressed under the cytomegalovirus IE gene enhancer/promoter (P(CMV IE)), we showed that MDV-1 pUL17 nuclear distribution in infected cells is not an intrinsic property. Although our results strongly suggest that another viral protein retains (or relocate) pUL17 to the nucleus, we report that none of the tegument protein tested so far were able to mediate pUL17 relocation to the nucleus.


Subject(s)
Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/physiology , Viral Proteins/physiology , Virus Replication/physiology , Animals , COS Cells , Chick Embryo , Chlorocebus aethiops , Epitopes , Gene Deletion , Skin/cytology
14.
Vet Res ; 38(3): 419-33, 2007.
Article in English | MEDLINE | ID: mdl-17506972

ABSTRACT

Marek's disease virus (MDV) is an oncogenic alphaherpesvirus that is highly contagious in poultry. Recombinant RB-1B (rRB-1B) reconstituted from an infectious genome cloned as a bacterial artificial chromosome (BAC) is unable to spread horizontally, quite in contrast to parental RB-1B. This finding suggests the presence of one or several mutations in cloned relative to parental viral DNA. Sequence analyses of the pRB-1B bacmid identified a one-nucleotide insertion in the UL13 orthologous gene that causes a frame-shift mutation and thereby results in a theoretical truncated UL13 protein (176 aa vs. 513 aa in parental RB-1B). UL13 genes are conserved among alphaherpesviruses and encode protein kinases. Using two-step "en passant" mutagenesis, we restored the UL13 ORF in pRB-1B. After transfection of UL13-positive pRB-1B DNA (pRB-1B*UL13), the resulting, repaired virus did not exhibit a difference in cell-to cell spread (measured by plaque sizes) and in UL13 transcripts in culture compared to parental rRB-1B virus. Although 89% of the chickens inoculated with rRB-1B*UL13 virus developed tumors in visceral organs, none of the contact birds did. MDV antigens were clearly expressed in the feather tips of rRB-1B infected chickens, suggesting that the UL13 gene mutation did not alter virus tropism of the feather follicle. The results indicate that the correction in UL13 gene alone is not sufficient to restore in vivo spreading capabilities of the rRB-1B virus, and that other region(s) of pRB-1B might be involved in the loss-of-function phenotype. This finding also shows for the first time that a full UL13 ORF is dispensable for MDV tumor formation and feather follicle tropism.


Subject(s)
Disease Transmission, Infectious/veterinary , Mardivirus , Marek Disease/transmission , Marek Disease/virology , Protein Kinases/genetics , Animals , Base Sequence , Chickens , DNA, Viral/chemistry , Feathers/physiology , Feathers/virology , Frameshift Mutation , Mardivirus/genetics , Mardivirus/isolation & purification , Mardivirus/pathogenicity , Molecular Sequence Data , Open Reading Frames , Point Mutation , Poultry Diseases/transmission , Poultry Diseases/virology , Protein Kinases/physiology , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Specific Pathogen-Free Organisms
15.
J Virol ; 79(10): 5889-99, 2005 May.
Article in English | MEDLINE | ID: mdl-15857974

ABSTRACT

Expression levels of Marek's disease virus (MDV) glycoprotein C (gC) are significantly reduced after serial virus passage in cell culture. Reduced gC expression coincides with enhanced MDV growth in vitro and attenuation. To analyze this phenomenon in detail, a full-length infectious MDV clone was modified by Red-based and shuttle mutagenesis in Escherichia coli. Besides a gC-negative deletion mutant harboring a kanamycin resistance gene, a markerless mutant with the U(L)44 gene deleted was constructed. On the basis of this deletion mutant, the original or a modified U(L)44 gene with a mutated start codon (AUG-->ACG) was reinserted into the authentic locus. Similarly, mutants expressing authentic gC or the start codon mutation under the control of a strong constitutive promoter were generated. In vitro studies demonstrated that gC deletion mutants induced twofold-larger plaques than the parental virus did, whereas constitutive overexpression of the glycoprotein resulted in a more than twofold reduction in plaque size. In addition, plaque sizes of the gC deletion mutant were reduced when virus was grown using supernatants from cells infected with parental virus, but supernatants obtained from cells infected with the gC deletion mutant had no measurable effect on plaque size. The results indicated that (i) expression of MDV gC, albeit at low levels in a highly passaged virus, had a significant negative impact on the cell-to-cell spread capabilities of the virus, which was alleviated in its absence and exacerbated by its overexpression, and that (ii) this activity was mediated by the secreted form of MDV gC.


Subject(s)
Antigens, Viral/metabolism , Mardivirus/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Cells, Cultured , Culture Media, Conditioned , Gene Expression Regulation, Viral , Mardivirus/growth & development , Molecular Sequence Data , Point Mutation , Sequence Alignment , Viral Envelope Proteins/genetics , Virus Replication
16.
J Virol ; 76(4): 1959-70, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11799190

ABSTRACT

Experiments were conducted to investigate the roles of Marek's disease virus serotype 1 (MDV-1) major tegument proteins VP11/12, VP13/14, VP16, and VP22 in viral growth in cultured cells. Based on a bacterial artificial chromosome clone of MDV-1 (BAC20), mutant viruses were constructed in which the MDV-1 homologs of UL46, UL47, UL48, or UL49 were deleted alone and in various combinations. It could be demonstrated that the UL46, UL47, and UL48 genes are dispensable for MDV-1 growth in chicken embryonic skin and quail muscle QM7 cells, although the generated virus mutants exhibited reduced plaque sizes in all cell types investigated. In contrast, a UL49-negative MDV-1 (20 Delta 49) and a UL48-UL49 (20 Delta 48-49) doubly negative mutant were not able to produce MDV-1-specific plaques on either cell type. It was confirmed that this growth restriction is dependent on the absence of VP22 expression, because growth of these mutant viruses could be partially restored on cells that were cotransfected with a UL49 expression plasmid. In addition, we were able to demonstrate that cell-to-cell spread of MDV-1 conferred by VP22 is dependent on the expression of amino acids 37 to 187 of MDV-1 VP22, because expression plasmids containing MDV-1 UL49 mutant genes with deletions of amino acids 1 to 37 or 188 to 250 were still able to restore partial growth of the 20 Delta 49 and 20 Delta 48-49 viruses. These results demonstrate for the first time that an alphaherpesvirus UL49-homologous gene is essential for virus growth in cell culture.


Subject(s)
Gene Deletion , Herpesvirus 2, Gallid/growth & development , Herpesvirus 2, Gallid/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Animals , Cells, Cultured , Chick Embryo , Chromosomes, Artificial, Bacterial/genetics , Fluorescent Antibody Technique , Marek Disease/virology , Muscles/cytology , Muscles/virology , Skin/cytology , Skin/virology , Virus Replication
17.
J Gen Virol ; 81(Pt 9): 2219-2230, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950980

ABSTRACT

Genes UL49 and UL48 of Marek's disease virus 1 (MDV-1) strain RB1B, encoding the respective homologues of herpes simplex virus type 1 (HSV-1) genes VP22 and VP16, were cloned into a baculovirus vector. Seven anti-VP22 MAbs and one anti-VP16 MAb were generated and used to identify the tegument proteins in cells infected lytically with MDV-1. The two genes are known to be transcribed as a single bicistronic transcript, and the detection of only one of the two proteins (VP22) in MSB-1 lymphoma and in chicken embryo skin cells infected with MDV-1 prompted the study of the transcription/translation of the UL49-48 sequence in an in vivo and in vitro expression system. VP16 was expressed in vitro at detectable levels, whereas it could only be detected at a lower level in a more controlled environment. It was demonstrated that VP22 is phosphorylated in insect cells and possesses the remarkable property of being imported into all cells in a monolayer. VP22 localized rapidly and efficiently to nuclei, like its HSV-1 counterpart. The DNA-binding property of VP22 is also reported and a part of the region responsible for this activity was identified between aa 16 and 37 in the N-terminal region of the protein.


Subject(s)
DNA, Viral/genetics , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Herpesvirus 2, Gallid/genetics , Viral Proteins/genetics , Viral Structural Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Chick Embryo , Cloning, Molecular , DNA/metabolism , Molecular Sequence Data , Protein Biosynthesis , Protein Processing, Post-Translational , Spodoptera , Structure-Activity Relationship , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...