Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38260396

ABSTRACT

Simian virus 40 (SV40) is a monkey virus associated with several types of human cancers. SV40 is most frequently detected in mesotheliomas, brain and bone tumors and lymphomas, but the mechanism for SV40 tumorigenesis in humans is not clear. SV40 relative Merkel cell polyomavirus (MCPyV) causes Merkel cell carcinoma (MCC) in humans by expressing truncated large tumor antigen (LT) caused by APOBEC cytidine deaminase family enzymes induced mutations. AID (activation-induced cytidine deaminase), a member of the APOBEC family, is the initiator of the antibody diversification process known as somatic hypermutation (SHM) and its aberrant expression and targeting is a frequent source of lymphomagenesis. In this study, we investigated whether AID-induced mutations could cause truncation of SV40 LT. We demonstrate that the SV40 enhancer has strong SHM targeting activity in several cell types and that AID-induced mutations accumulate to SV40 LT in B cells and kidney cells and cause truncated LT expression in B cells. Our results argue that the ability of the SV40 enhancer to target SHM to LT is a potential source of LT truncation events in various cell types that could contribute to carcinogenesis.

2.
J Immunol ; 208(1): 143-154, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34862258

ABSTRACT

Somatic hypermutation (SHM) drives the genetic diversity of Ig genes in activated B cells and supports the generation of Abs with increased affinity for Ag. SHM is targeted to Ig genes by their enhancers (diversification activators [DIVACs]), but how the enhancers mediate this activity is unknown. We show using chicken DT40 B cells that highly active DIVACs increase the phosphorylation of RNA polymerase II (Pol II) and Pol II occupancy in the mutating gene with little or no accompanying increase in elongation-competent Pol II or production of full-length transcripts, indicating accumulation of stalled Pol II. DIVAC has similar effect also in human Ramos Burkitt lymphoma cells. The DIVAC-induced stalling is weakly associated with an increase in the detection of ssDNA bubbles in the mutating target gene. We did not find evidence for antisense transcription, or that DIVAC functions by altering levels of H3K27ac or the histone variant H3.3 in the mutating gene. These findings argue for a connection between Pol II stalling and cis-acting targeting elements in the context of SHM and thus define a mechanistic basis for locus-specific targeting of SHM in the genome. Our results suggest that DIVAC elements render the target gene a suitable platform for AID-mediated mutation without a requirement for increasing transcriptional output.


Subject(s)
Avian Proteins/metabolism , B-Lymphocyte Subsets/immunology , Burkitt Lymphoma/immunology , Enhancer Elements, Genetic/genetics , Immunoglobulins/metabolism , RNA Polymerase II/metabolism , Animals , Antibody Diversity , Avian Proteins/genetics , Burkitt Lymphoma/genetics , Chickens , Cytidine Deaminase/genetics , Humans , Immunoglobulins/genetics , Lymphocyte Activation , Mutagenesis, Site-Directed , Mutation/genetics , RNA Polymerase II/genetics , Somatic Hypermutation, Immunoglobulin , Transcription, Genetic
3.
Viruses ; 13(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34960774

ABSTRACT

The chicken Tva cell surface protein, a member of the low-density lipoprotein receptor family, has been identified as an entry receptor for avian leukosis virus of classic subgroup A and newly emerging subgroup K. Because both viruses represent an important concern for the poultry industry, we introduced a frame-shifting deletion into the chicken tva locus with the aim of knocking-out Tva expression and creating a virus-resistant chicken line. The tva knock-out was prepared by CRISPR/Cas9 gene editing in chicken primordial germ cells and orthotopic transplantation of edited cells into the testes of sterilized recipient roosters. The resulting tva -/- chickens tested fully resistant to avian leukosis virus subgroups A and K, both in in vitro and in vivo assays, in contrast to their susceptible tva +/+ and tva +/- siblings. We also found a specific disorder of the cobalamin/vitamin B12 metabolism in the tva knock-out chickens, which is in accordance with the recently recognized physiological function of Tva as a receptor for cobalamin in complex with transcobalamin transporter. Last but not least, we bring a new example of the de novo resistance created by CRISPR/Cas9 editing of pathogen dependence genes in farm animals and, furthermore, a new example of gene editing in chicken.


Subject(s)
Avian Leukosis Virus/physiology , Avian Proteins/physiology , Chickens/virology , Receptors, Virus/physiology , Vitamin B 12/metabolism , Animals , Avian Leukosis Virus/classification , Avian Proteins/genetics , Chick Embryo , Female , Frameshift Mutation , Gene Editing , Gene Knockout Techniques , Male , Methylmalonic Acid/blood , Receptors, Virus/genetics
4.
Eur J Immunol ; 50(3): 380-395, 2020 03.
Article in English | MEDLINE | ID: mdl-31821534

ABSTRACT

Secondary diversification of the Ig repertoire occurs through somatic hypermutation (SHM), gene conversion (GCV), and class switch recombination (CSR)-three processes that are initiated by activation-induced cytidine deaminase (AID). AID targets Ig genes at orders of magnitude higher than the rest of the genome, but the basis for this specificity is poorly understood. We have previously demonstrated that enhancers and enhancer-like sequences from Ig genes are capable of stimulating SHM of neighboring genes in a capacity distinct from their roles in increasing transcription. Here, we use an in vitro proteomics approach to identify E-box, MEF2, Ets, and Ikaros transcription factor family members as potential binders of these enhancers. ChIP assays in the hypermutating Ramos B cell line confirmed that many of these factors bound the endogenous Igλ enhancer and/or the IgH intronic enhancer (Eµ) in vivo. Further investigation using SHM reporter assays identified binding sites for E2A and MEF2B in Eµ and demonstrated an association between loss of factor binding and decreases in the SHM stimulating activity of Eµ mutants. Our results provide novel insights into trans-acting factors that dictate SHM targeting and link their activity to specific DNA binding sites within Ig enhancers.


Subject(s)
Somatic Hypermutation, Immunoglobulin/physiology , Animals , Chickens , Genes, Immunoglobulin , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell Rep ; 29(12): 3902-3915.e8, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851922

ABSTRACT

Somatic hypermutation (SHM) introduces point mutations into immunoglobulin (Ig) genes but also causes mutations in other parts of the genome. We have used lentiviral SHM reporter vectors to identify regions of the genome that are susceptible ("hot") and resistant ("cold") to SHM, revealing that SHM susceptibility and resistance are often properties of entire topologically associated domains (TADs). Comparison of hot and cold TADs reveals that while levels of transcription are equivalent, hot TADs are enriched for the cohesin loader NIPBL, super-enhancers, markers of paused/stalled RNA polymerase 2, and multiple important B cell transcription factors. We demonstrate that at least some hot TADs contain enhancers that possess SHM targeting activity and that insertion of a strong Ig SHM-targeting element into a cold TAD renders it hot. Our findings lead to a model for SHM susceptibility involving the cooperative action of cis-acting SHM targeting elements and the dynamic and architectural properties of TADs.


Subject(s)
Enhancer Elements, Genetic/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Cell Line, Tumor , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , HEK293 Cells , Humans , Lentivirus , Male , Mutation/genetics , Plasmids/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
6.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31217247

ABSTRACT

Avian leukosis virus subgroup K (ALV-K) is composed of newly emerging isolates, which, in sequence analyses, cluster separately from the well-characterized subgroups A, B, C, D, E, and J. However, it remains unclear whether ALV-K represents an independent ALV subgroup with regard to receptor usage, host range, and superinfection interference. In the present study, we examined the host range of the Chinese infectious isolate JS11C1, an ALV-K prototype, and we found substantial overlap of species that were either resistant or susceptible to ALV-A and JS11C1. Ectopic expression of the chicken tva gene in mammalian cells conferred susceptibility to JS11C1, while genetic ablation of the tva gene rendered chicken DF-1 cells resistant to infection by JS11C1. Thus, tva expression is both sufficient and necessary for JS11C1 entry. Receptor sharing was also manifested in superinfection interference, with preinfection of cells with ALV-A, but not ALV-B or ALV-J, blocking subsequent JS11C1 infection. Finally, direct binding of JS11C1 and Tva was demonstrated by preincubation of the virus with soluble Tva, which substantially decreased viral infectivity in susceptible chicken cells. Collectively, these findings indicate that JS11C1 represents a new and bona fide ALV subgroup that utilizes Tva for cell entry and binds to a site other than that for ALV-A.IMPORTANCE ALV consists of several subgroups that are particularly characterized by their receptor usage, which subsequently dictates the host range and tropism of the virus. A few newly emerging and highly pathogenic Chinese ALV strains have recently been suggested to be an independent subgroup, ALV-K, based solely on their genomic sequences. Here, we performed a series of experiments with the ALV-K strain JS11C1, which showed its dependence on the Tva cell surface receptor. Due to the sharing of this receptor with ALV-A, both subgroups were able to interfere with superinfection. Because ALV-K could become an important pathogen and a significant threat to the poultry industry in Asia, the identification of a specific receptor could help in the breeding of resistant chicken lines with receptor variants with decreased susceptibility to the virus.


Subject(s)
Avian Leukosis Virus/pathogenicity , Avian Leukosis/genetics , Avian Proteins/genetics , Avian Proteins/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Animals , Avian Leukosis/metabolism , Avian Leukosis/virology , Avian Leukosis Virus/classification , Avian Leukosis Virus/physiology , Cell Line , Chickens , Disease Susceptibility , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Mesocricetus , Species Specificity , Virus Internalization
7.
Viruses ; 10(3)2018 03 08.
Article in English | MEDLINE | ID: mdl-29517993

ABSTRACT

Individual groups of retroviruses and retroviral vectors differ in their integration site preference and interaction with the host genome. Hence, immediately after infection genome-wide distribution of integrated proviruses is non-random. During long-term in vitro or persistent in vivo infection, the genomic position and chromatin environment of the provirus affects its transcriptional activity. Thus, a selection of long-term stably expressed proviruses and elimination of proviruses, which have been gradually silenced by epigenetic mechanisms, helps in the identification of genomic compartments permissive for proviral transcription. We compare here the extent and time course of provirus silencing in single cell clones of the K562 human myeloid lymphoblastoma cell line that have been infected with retroviral reporter vectors derived from avian sarcoma/leukosis virus (ASLV), human immunodeficiency virus type 1 (HIV) and murine leukaemia virus (MLV). While MLV proviruses remain transcriptionally active, ASLV proviruses are prone to rapid silencing. The HIV provirus displays gradual silencing only after an extended time period in culture. The analysis of integration sites of long-term stably expressed proviruses shows a strong bias for some genomic features-especially integration close to the transcription start sites of active transcription units. Furthermore, complex analysis of histone modifications enriched at the site of integration points to the accumulation of proviruses of all three groups in gene regulatory segments, particularly close to the enhancer loci. We conclude that the proximity to active regulatory chromatin segments correlates with stable provirus expression in various retroviral species.


Subject(s)
Alpharetrovirus/genetics , Chromatin/genetics , Genetic Vectors/genetics , HIV-1/genetics , Leukemia Virus, Murine/genetics , Proviruses/genetics , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Animals , Cell Line , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation, Viral , Gene Silencing , Gene Targeting , Humans , Mice , Plasmids/genetics , RNA Stability , Transcription Initiation Site , Virus Integration
8.
Nucleic Acids Res ; 45(22): 12752-12765, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29244184

ABSTRACT

Most retroviruses preferentially integrate into certain genomic locations and, as a result, their genome-wide integration patterns are non-random. We investigate the epigenetic landscape of integrated retroviral vectors and correlate it with the long-term stability of proviral transcription. Retroviral vectors derived from the avian sarcoma/leukosis virus expressing the GFP reporter were used to transduce the human myeloid lymphoblastoma cell line K562. Because of efficient silencing of avian retrovirus in mammalian cells, only ∼3% of established clones displayed stable proviral expression. We analyzed the vector integration sites in non-selected cells and in clones selected for the GFP expression. This selection led to overrepresentation of proviruses integrated in active transcription units, with particular accumulation in promoter-proximal areas. In parallel, we investigated the integration of vectors equipped with an anti-silencing CpG island core sequence. Such modification increased the frequency of stably expressing proviruses by one order. The modified vectors are also overrepresented in active transcription units, but stably expressed in distal parts of transcriptional units further away from promoters with marked accumulation in enhancers. These results suggest that integrated retroviruses subject to gradual epigenetic silencing during long-term cultivation. Among most genomic compartments, however, active promoters and enhancers protect the adjacent retroviruses from transcriptional silencing.


Subject(s)
Alpharetrovirus/genetics , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Animals , Cell Line , CpG Islands/genetics , Epigenesis, Genetic , Gene Silencing , Genetic Vectors/genetics , Humans , K562 Cells , Proviruses/genetics , Virus Integration/genetics
9.
J Virol ; 91(3)2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27881654

ABSTRACT

The J subgroup of avian leukosis virus (ALV-J) infects domestic chickens, jungle fowl, and turkeys. This virus enters the host cell through a receptor encoded by the tvj locus and identified as Na+/H+ exchanger 1. The resistance to avian leukosis virus subgroup J in a great majority of galliform species has been explained by deletions or substitutions of the critical tryptophan 38 in the first extracellular loop of Na+/H+ exchanger 1. Because there are concerns of transspecies virus transmission, we studied natural polymorphisms and susceptibility/resistance in wild galliforms and found the presence of tryptophan 38 in four species of New World quails. The embryo fibroblasts of New World quails are susceptible to infection with avian leukosis virus subgroup J, and the cloned Na+/H+ exchanger 1 confers susceptibility on the otherwise resistant host. New World quails are also susceptible to new avian leukosis virus subgroup J variants but resistant to subgroups A and B and weakly susceptible to subgroups C and D of avian sarcoma/leukosis virus due to obvious defects of the respective receptors. Our results suggest that the avian leukosis virus subgroup J could be transmitted to New World quails and establish a natural reservoir of circulating virus with a potential for further evolution. IMPORTANCE: Since its spread in broiler chickens in China and Southeast Asia in 2000, ALV-J remains a major enzootic challenge for the poultry industry. Although the virus diversifies rapidly in the poultry, its spillover and circulation in wild bird species has been prevented by the resistance of most species to ALV-J. It is, nevertheless, important to understand the evolution of the virus and its potential host range in wild birds. Because resistance to avian retroviruses is due particularly to receptor incompatibility, we studied Na+/H+ exchanger 1, the receptor for ALV-J. In New World quails, we found a receptor compatible with virus entry, and we confirmed the susceptibilities of four New World quail species in vitro We propose that a prospective molecular epidemiology study be conducted to identify species with the potential to become reservoirs for ALV-J.


Subject(s)
Avian Leukosis Virus/physiology , Avian Leukosis/genetics , Avian Leukosis/virology , Disease Susceptibility , Quail , Amino Acid Sequence , Amino Acids , Animals , Avian Leukosis/metabolism , Avian Leukosis Virus/classification , Cells, Cultured , Disease Resistance/genetics , Evolution, Molecular , Gene Expression , Genetic Loci , Host Specificity , Host-Pathogen Interactions , Phylogeny , Polymorphism, Genetic , Protein Interaction Domains and Motifs , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Virus Replication
10.
PLoS One ; 11(3): e0150589, 2016.
Article in English | MEDLINE | ID: mdl-26978658

ABSTRACT

J subgroup avian leukosis virus (ALV-J) infects domestic chicken, jungle fowl, and turkey and enters the host cell through a receptor encoded by tvj locus and identified as Na+/H+ exchanger 1 (NHE1). The resistance to ALV-J in a great majority of examined galliform species was explained by deletions or substitutions of the critical tryptophan 38 in the first extracellular loop of NHE1, and genetic polymorphisms around this site predict the susceptibility or resistance of a given species or individual. In this study, we examined the NHE1 polymorphism in domestic chicken breeds and documented quantitative differences in their susceptibility to ALV-J in vitro. In a panel of chicken breeds assembled with the aim to cover the maximum variability encountered in domestic chickens, we found a completely uniform sequence of NHE1 extracellular loop 1 (ECL1) without any source of genetic variation for the selection of ALV-J-resistant poultry. In parallel, we studied the natural polymorphisms of NHE1 in wild ducks and geese because of recent reports on ALV-J positivity in feral Asian species. In anseriform species, we demonstrate a specific and highly conserved critical ECL1 sequence without any homologue of tryptophan 38 in accordance with the resistance of duck cells to prototype ALV-J. Last, we demonstrated that the new Asian strains of ALV-J have not evolved their envelope glycoprotein to the entry the duck cells. Our results contribute substantially to the current discussion of possible heterotransmission of ALV-J and its spill-over into the wild ducks and geese.


Subject(s)
Animals, Domestic , Animals, Wild , Genetic Variation , Sodium-Hydrogen Exchangers/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , Chickens , Molecular Sequence Data , Sequence Homology, Amino Acid , Sodium-Hydrogen Exchangers/chemistry
11.
Oncotarget ; 5(16): 6923-35, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-25071011

ABSTRACT

Downregulation of MHC class I expression on tumour cells, a common mechanism by which tumour cells can escape from specific immune responses, can be associated with coordinated silencing of antigen-presenting machinery genes. The expression of these genes can be restored by IFNγ. In this study we documented association of DNA demethylation of selected antigen-presenting machinery genes located in the MHC genomic locus (TAP-1, TAP-2, LMP-2, LMP-7) upon IFNγ treatment with MHC class I upregulation on tumour cells in several MHC class I-deficient murine tumour cell lines (TC-1/A9, TRAMP-C2, MK16 and MC15). Our data also documented higher methylation levels in these genes in TC-1/A9 cells, as compared to their parental MHC class I-positive TC-1 cells. IFNγ-mediated DNA demethylation was relatively fast in comparison with demethylation induced by DNA methyltransferase inhibitor 5-azacytidine, and associated with increased histone H3 acetylation in the promoter regions of APM genes. Comparative transcriptome analysis in distinct MHC class I-deficient cell lines upon their treatment with either IFNγ or epigenetic agents revealed that a set of genes, significantly enriched for the antigen presentation pathway, was regulated in the same manner. Our data demonstrate that IFNγ acts as an epigenetic modifier when upregulating the expression of antigen-presenting machinery genes.


Subject(s)
Antigen Presentation/genetics , DNA Methylation , Fibrosarcoma/genetics , Genes, MHC Class I , Interferon-gamma/genetics , Interferon-gamma/metabolism , Animals , Down-Regulation , Epigenesis, Genetic , Fibrosarcoma/immunology , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Signal Transduction , Transfection , Tumor Cells, Cultured , Up-Regulation
12.
J Virol ; 88(6): 3505-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24403579

ABSTRACT

UNLABELLED: Transformation of rodent cells with avian Rous sarcoma virus (RSV) opened new ways to studying virus integration and expression in nonpermissive cells. We were interested in (i) the molecular changes accompanying fusion of RSV-transformed mammalian cells with avian cells leading to virus rescue and (ii) enhancement of this process by retroviral gene products. The RSV-transformed hamster RSCh cell line was characterized as producing only a marginal amount of env mRNA, no envelope glycoprotein, and a small amount of unprocessed Gag protein. Egress of viral unspliced genomic RNA from the nucleus was hampered, and its stability decreased. Cell fusion of the chicken DF-1 cell line with RSCh cells led to production of env mRNA, envelope glycoprotein, and processed Gag and virus-like particle formation. Proteosynthesis inhibition in DF-1 cells suppressed steps leading to virus rescue. Furthermore, new aberrantly spliced env mRNA species were found in the RSCh cells. Finally, we demonstrated that virus rescue efficiency can be significantly increased by complementation with the env gene and the highly expressed gag gene and can be increased the most by a helper virus infection. In summary, Env and Gag synthesis is increased after RSV-transformed hamster cell fusion with chicken fibroblasts, and both proteins provided in trans enhance RSV rescue. We conclude that the chicken fibroblast yields some factor(s) needed for RSV replication, particularly Env and Gag synthesis, in nonpermissive rodent cells. IMPORTANCE: One of the important issues in retrovirus heterotransmission is related to cellular factors that prevent virus replication. Rous sarcoma virus (RSV), a member of the avian sarcoma and leukosis family of retroviruses, is able to infect and transform mammalian cells; however, such transformed cells do not produce infectious virus particles. Using the well-defined model of RSV-transformed rodent cells, we established that the lack of virus replication is due to the absence of chicken factor(s), which can be supplemented by cell fusion. Cell fusion with permissive chicken cells led to an increase in RNA splicing and nuclear export of specific viral mRNAs, as well as synthesis of respective viral proteins and production of virus-like particles. RSV rescue by cell fusion can be potentiated by in trans expression of viral genes in chicken cells. We conclude that rodent cells lack some chicken factor(s) required for proper viral RNA processing and viral protein synthesis.


Subject(s)
Poultry Diseases/virology , Rous sarcoma virus/genetics , Sarcoma, Avian/virology , Animals , Cell Fusion , Cell Line, Transformed , Cell Transformation, Viral , Chickens , Cricetinae , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genetic Complementation Test , Rous sarcoma virus/physiology
13.
J Virol ; 87(15): 8399-407, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698309

ABSTRACT

Subgroup J avian leukosis virus (ALV-J) is unique among the avian sarcoma and leukosis viruses in using the multimembrane-spanning cell surface protein Na(+)/H(+) exchanger type 1 (NHE1) as a receptor. The precise localization of amino acids critical for NHE1 receptor activity is key in understanding the virus-receptor interaction and potential interference with virus entry. Because no resistant chicken lines have been described until now, we compared the NHE1 amino acid sequences from permissive and resistant galliform species. In all resistant species, the deletion or substitution of W38 within the first extracellular loop was observed either alone or in the presence of other incidental amino acid changes. Using the ectopic expression of wild-type or mutated chicken NHE1 in resistant cells and infection with a reporter recombinant retrovirus of subgroup J specificity, we studied the effect of individual mutations on the NHE1 receptor capacity. We suggest that the absence of W38 abrogates binding of the subgroup J envelope glycoprotein to ALV-J-resistant cells. Altogether, we describe the functional importance of W38 for virus entry and conclude that natural polymorphisms in NHE1 can be a source of host resistance to ALV-J.


Subject(s)
Avian Leukosis Virus/physiology , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Viral Tropism , Virus Internalization , Animals , Birds , DNA Mutational Analysis , Tryptophan/genetics , Tryptophan/metabolism
14.
Nucleic Acids Res ; 40(12): 5298-312, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22379139

ABSTRACT

The autonomous transcription of integrated retroviruses strongly depends on genetic and epigenetic effects of the chromatin at the site of integration. These effects are mostly suppressive and proviral activity can be finally silenced by mechanisms, such as DNA methylation and histone modifications. To address the role of the integration site at the whole-genome-scale, we performed clonal analysis of provirus silencing with an avian leucosis/sarcoma virus-based reporter vector and correlated the transcriptional silencing with the epigenomic landscape of respective integrations. We demonstrate efficient provirus silencing in human HCT116 cell line, which is strongly but not absolutely dependent on the de novo DNA methyltransferase activity, particularly of Dnmt3b. Proviruses integrated close to the transcription start sites of active genes into the regions enriched in H3K4 trimethylation display long-term stability of expression and are resistant to the transcriptional silencing after over-expression of Dnmt3a or Dnmt3b. In contrast, proviruses in the intergenic regions tend to spontaneous transcriptional silencing even in Dnmt3a(-/-) Dnmt3b(-/-) cells. The silencing of proviruses within genes is accompanied with DNA methylation of long terminal repeats, whereas silencing in intergenic regions is DNA methylation-independent. These findings indicate that the epigenomic features of integration sites are crucial for their permissivity to the proviral expression.


Subject(s)
Alpharetrovirus/genetics , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Proviruses/genetics , Virus Integration , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Humans , Transcription, Genetic , DNA Methyltransferase 3B
15.
J Virol ; 86(4): 2021-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22171251

ABSTRACT

The group of closely related avian sarcoma and leukosis viruses (ASLVs) evolved from a common ancestor into multiple subgroups, A to J, with differential host range among galliform species and chicken lines. These subgroups differ in variable parts of their envelope glycoproteins, the major determinants of virus interaction with specific receptor molecules. Three genetic loci, tva, tvb, and tvc, code for single membrane-spanning receptors from diverse protein families that confer susceptibility to the ASLV subgroups. The host range expansion of the ancestral virus might have been driven by gradual evolution of resistance in host cells, and the resistance alleles in all three receptor loci have been identified. Here, we characterized two alleles of the tva receptor gene with similar intronic deletions comprising the deduced branch-point signal within the first intron and leading to inefficient splicing of tva mRNA. As a result, we observed decreased susceptibility to subgroup A ASLV in vitro and in vivo. These alleles were independently found in a close-bred line of domestic chicken and Indian red jungle fowl (Gallus gallus murghi), suggesting that their prevalence might be much wider in outbred chicken breeds. We identified defective splicing to be a mechanism of resistance to ASLV and conclude that such a type of mutation could play an important role in virus-host coevolution.


Subject(s)
Alpharetrovirus/physiology , Avian Proteins/genetics , Chickens/genetics , Genetic Predisposition to Disease , Poultry Diseases/genetics , RNA Splicing , Receptors, Virus/genetics , Sarcoma, Avian/genetics , Sequence Deletion , Alpharetrovirus/genetics , Amino Acid Sequence , Animals , Avian Proteins/metabolism , Base Sequence , Chickens/metabolism , Chickens/virology , Introns , Molecular Sequence Data , Poultry Diseases/metabolism , Poultry Diseases/virology , Receptors, Virus/metabolism , Sarcoma, Avian/metabolism , Sarcoma, Avian/virology
16.
J Virol ; 84(9): 4204-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20147411

ABSTRACT

Retroviruses and retrovirus-derived vectors integrate nonrandomly into the genomes of host cells with specific preferences for transcribed genes, gene-rich regions, and CpG islands. However, the genomic features that influence the transcriptional activities of integrated retroviruses or retroviral vectors are poorly understood. We report here the cloning and characterization of avian sarcoma virus integration sites from chicken tumors. Growing progressively, dependent on high and stable expression of the transduced v-src oncogene, these tumors represent clonal expansions of cells bearing transcriptionally active replication-defective proviruses. Therefore, integration sites in our study distinguished genomic loci favorable for the expression of integrated retroviruses and gene transfer vectors. Analysis of integration sites from avian sarcoma virus-induced tumors showed strikingly nonrandom distribution, with proviruses found prevalently within or close to transcription units, particularly in genes broadly expressed in multiple tissues but not in tissue-specifically expressed genes. We infer that proviruses integrated in these genomic areas efficiently avoid transcriptional silencing and remain active for a long time during the growth of tumors. Defining the differences between unselected retroviral integration sites and sites selected for long-terminal-repeat-driven gene expression is relevant for retrovirus-mediated gene transfer and has ramifications for gene therapy.


Subject(s)
Avian Sarcoma Viruses/physiology , Chromosomes/virology , Proviruses/physiology , Sarcoma, Avian/virology , Virus Integration , Animals , Avian Sarcoma Viruses/genetics , Chickens , Gene Expression , Genetic Therapy/methods , Genetic Vectors , Proviruses/genetics
17.
J Virol ; 82(16): 7818-27, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550662

ABSTRACT

Unmethylated CpG islands are known to keep adjacent promoters transcriptionally active. In the CpG island adjacent to the adenosine phosphoribosyltransferase gene, the protection against transcriptional silencing can be attributed to the short CpG-rich core element containing Sp1 binding sites. We report here the insertion of this CpG island core element, IE, into the long terminal repeat of a retroviral vector derived from Rous sarcoma virus, which normally suffers from progressive transcriptional silencing in mammalian cells. IE insertion into a specific position between enhancer and promoter sequences led to efficient protection of the integrated vector from silencing and gradual CpG methylation in rodent and human cells. Individual cell clones with IE-modified reporter vectors display high levels of reporter expression for a sustained period and without substantial variegation in the cell culture. The presence of Sp1 binding sites is important for the protective effect of IE, but at least some part of the entire antisilencing capacity is maintained in IE with mutated Sp1 sites. We suggest that this strategy of antisilencing protection by the CpG island core element may prove generally useful in retroviral vectors.


Subject(s)
Avian Leukosis Virus/metabolism , CpG Islands , Gene Silencing , Sarcoma, Avian/genetics , Sarcoma, Avian/virology , Transcription, Genetic , Animals , Binding Sites , Birds , Flow Cytometry , Genes, Reporter , Humans , Models, Biological , Mutation , Rous sarcoma virus/metabolism , Sp1 Transcription Factor/metabolism , Terminal Repeat Sequences
18.
J Virol ; 82(5): 2097-105, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094190

ABSTRACT

The avian sarcoma and leukosis virus (ASLV) family of retroviruses contains five highly related envelope subgroups (A to E) thought to have evolved from a common viral ancestor in the chicken population. Three genetic loci in chickens determine the susceptibility or resistance of cells to infection by the subgroup A to E ASLVs. Some inbred lines of chickens display phenotypes that are somewhere in between either efficiently susceptible or resistant to infection by specific subgroups of ASLV. The tvb gene encodes the receptor for subgroups B, D, and E ASLVs. The wild-type Tvb(S1) receptor confers susceptibility to subgroups B, D, and E ASLVs. In this study, the genetic defect that accounts for the altered susceptibility of an inbred chicken line, line M, to infection by ASLV(B), ASLV(D), and ASLV(E) was identified. The tvb gene in line M, tvb(r2), encodes a mutant Tvb(S1) receptor protein with a substitution of a serine for a cysteine at position 125 (C125S). Here, we show that the C125S substitution in Tvb(S1) significantly reduces the susceptibility of line M cells to infection by ASLV(B) and ASLV(D) and virtually eliminates susceptibility to ASLV(E) infection both in cultured cells and in the incidence and growth of avian sarcoma virus-induced sarcomas in chickens. The C125S substitution significantly reduces the binding affinity of the Tvb(S1) receptor for the subgroup B, D, and E ASLV envelope glycoproteins. These are the first results that demonstrate a possible role of the cysteine-rich domain 3 in the function of the Tvb receptors.


Subject(s)
Alpharetrovirus/pathogenicity , Amino Acid Substitution , Genetic Predisposition to Disease , Receptors, Virus/physiology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Alleles , Alpharetrovirus/classification , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chick Embryo , DNA Primers , Flow Cytometry , Membrane Fusion , Molecular Sequence Data , Receptors, Virus/chemistry , Receptors, Virus/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Species Specificity
19.
Reproduction ; 134(3): 445-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17709563

ABSTRACT

Chicken testicular cells, including spermatogonia, transplanted into the testes of recipient cockerels sterilized by repeated gamma-irradiation repopulate the seminiferous epithelium and resume the exogenous spermatogenesis. This procedure could be used to introduce genetic modifications into the male germ line and generate transgenic chickens. In this study, we present a successful retroviral infection of chicken testicular cells and consequent transduction of the retroviral vector into the sperm of recipient cockerels. A vesicular stomatitis virus glycoprotein G-pseudotyped recombinant retroviral vector, carrying the enhanced green fluorescent protein reporter gene was applied to the short-term culture of dispersed testicular cells. The efficiency of infection and the viability of infected cells were analyzed by flow cytometry. No significant CpG methylation was detected in the infected testicular cells, suggesting that epigenetic silencing events do not play a role at this stage of germ line development. After transplantation into sterilized recipient cockerels, these retrovirus-infected testicular cells restored exogenous spermatogenesis within 9 weeks with approximately the same efficiency as non-infected cells. Transduction of the reporter gene encoding the green fluorescent protein was detected in the sperms of recipient cockerels with restored spermatogenesis. Our data demonstrate that, similarly as in mouse and rat, the transplantation of retrovirus-infected spermatogonia provides an efficient system to introduce genes into the chicken male germ line.


Subject(s)
Chickens , Genetic Vectors/administration & dosage , Spermatozoa/virology , Testis/virology , Transduction, Genetic/methods , Vesicular stomatitis Indiana virus/genetics , Animals , Animals, Genetically Modified , Cells, Cultured , Flow Cytometry , Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Infertility, Male/therapy , Infertility, Male/veterinary , Male , Microscopy, Fluorescence , Spermatozoa/physiology , Spermatozoa/transplantation , Testis/cytology , Testis/transplantation
20.
Arch Biochem Biophys ; 455(2): 136-43, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17056000

ABSTRACT

Active, wild-type v-Src and its kinase-dead double Y416F-K295N mutant were expressed in hamster fibroblasts. Expression of the active v-Src induced activation of endogenous c-Src and increased general protein-tyrosine phosphorylation in the infected cells. Expression of the kinase-dead mutant induced hypophosphorylation of Tyr416 of the endogenous c-Src. The inactivation of c-Src was reversible, as confirmed by in vitro kinase activity of c-Src immunoprecipitated from the kinase-dead v-Src-expressing cells. Both activation and inactivation of c-Src may be explained by direct interaction of the v-Src and c-Src that may either facilitate transphosphorylation of the regulatory Tyr416 in the activation loop, or prevent it by formation of transient dead-end complexes of the Y416F-K295N mutant with c-Src. The interaction was also indicated by co-localization of v- and c-Src proteins in immunofluorescent images of the infected cells. These results suggest that dimerization of Src plays an important role in the regulation of Src tyrosine kinase activity.


Subject(s)
Fibroblasts/metabolism , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Protein-Tyrosine Kinases/metabolism , CSK Tyrosine-Protein Kinase , Cell Line , Enzyme Activation , Gene Expression Regulation, Enzymologic/physiology , Humans , Mutagenesis, Site-Directed , Mutation , Structure-Activity Relationship , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL