Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Gen Virol ; 93(Pt 7): 1519-1529, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22442112

ABSTRACT

Marek's disease virus (GaHV-2) is an alphaherpesvirus that induces T-cell lymphoma in chickens. The infection includes both lytic and latent stages. GaHV-2 encodes three clusters of microRNAs (miRNAs) located in the internal (I)/terminal (T) repeat (R) regions. We characterized transcripts encompassing the mdv1-mir-M9-M4 and mir-M11-M1 clusters located in the IR(L)/TR(L) region, upstream and downstream from the meq oncogene, respectively. By 5'- and 3'-RACE-PCR and targeted RT-PCR, we showed that mdv1-mir-M9-M4 could be transcribed from an unspliced transcript or from at least 15 alternatively spliced transcripts covering the IR(L)/TR(L) region, encompassing the meq and vIL-8 genes and localizing the mdv1-mir-M9-M4 cluster to the first intron at the 5'-end. However, all these transcripts, whether spliced or unspliced, seemed to start at the same transcriptional start site, their transcription being driven by a single promoter, prmiRM9M4. We demonstrated alternative promoter usage for the meq and vIL-8 genes, depending on the phase of GaHV-2 infection. During the latent phase, the prmiRM9M4 promoter drove transcription of the meq and vIL-8 genes and the mdv1-mir-M9-M4 cluster in the first intron of the corresponding transcripts. By contrast, during the lytic phase, this promoter drove the transcription only of the mdv1-mir-M9-M4 cluster to generate unspliced mRNA, the meq and vIL-8 genes being transcribed principally from their own promoters. Despite the expression of meq and the mdv1-mir-M9-M4 cluster under two different transcriptional processes during the latent and lytic phases, our data provide an explanation for meq expression and mdv1-mir-M4-5P overexpression in miRNA libraries from GaHV-2-infected cells, regardless of the phase of infection.


Subject(s)
Gene Expression Regulation, Viral , Mardivirus/genetics , Marek Disease/virology , Oncogene Proteins, Viral/biosynthesis , Virus Latency , Animals , Chickens , Gene Expression Profiling , Kinetics , MicroRNAs/biosynthesis , MicroRNAs/genetics , Promoter Regions, Genetic , Transcription Initiation Site , Transcription, Genetic
2.
J Gen Virol ; 92(Pt 6): 1273-1278, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21325479

ABSTRACT

All herpesviruses have a post-transcriptional regulatory protein that prevents precursor mRNA splicing and leads to the shutting off of host protein synthesis. The ICP27 protein of herpes simplex virus 1 (HSV-1) is the prototype of these proteins. Marek's disease virus (MDV-1), an alphaherpesvirus that induces lymphoma in birds, also has an ICP27 protein that is produced in lytic MDV-1-infected cells. We characterized this protein. We demonstrated ICP27 production in latently infected MSB-1 cells, but only on MDV-1 reactivation. ICP27 was found predominantly in specific structures within the nucleus. The ICP27 of MDV-1 colocalized and interacted with SR proteins. We demonstrated inhibitory effects of MDV-1 ICP27 on the splicing of both the viral vIL8 and cellular chTERT (telomerase reverse transcriptase) genes. Thus, the ICP27 of MDV-1 plays a similar role to the ICP27 of HSV-1 and may be involved in MDV-1 replication and the development of Marek's disease.


Subject(s)
Down-Regulation , Herpesvirus 2, Gallid/metabolism , Interleukin-8/genetics , Marek Disease/enzymology , RNA Splicing , Spliceosomes/metabolism , Telomerase/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chickens , Herpesvirus 2, Gallid/genetics , Interleukin-8/metabolism , Marek Disease/genetics , Marek Disease/virology , Molecular Sequence Data , Protein Binding , Protein Transport , Spliceosomes/genetics , Telomerase/metabolism , Viral Proteins/genetics
3.
Arch Virol ; 151(12): 2431-46, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16906478

ABSTRACT

We collected paraffin-embedded myelocytomatoses induced by subgroup J avian leukosis virus (ALV-J) in French poultry from 1992 to 2000. We used nested PCR to obtain the U3 LTR and the E element sequences that encompass putative binding sites for transcription factors. We observed minor mutations in the U3 sequences that rarely affected transcription factor binding sites, thus preserving the transcriptional potential of the U3 LTR. However, we observed a large variability in the E element sequences from both field and experimental tumor samples. This variability involved genomic rearrangements and various deletions that most often occurred between two direct repeat sequences. Moreover, in seven DNA samples of the 22 field tumors analyzed, we observed two different sequences for the E element region, suggesting that proviral genomes of two different sizes may be simultaneously present in a tumor. Even though most of the E element sequences were mutated or rearranged, all myelocytomatosis samples always exhibited one E element sequence containing at least one putative C/EBP binding site that was unaffected and still potentially functional.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/virology , Animals , Avian Leukosis Virus/classification , Avian Leukosis Virus/isolation & purification , Base Sequence , Bone Marrow/virology , DNA Primers , DNA, Viral/genetics , France , Heart/virology , Liver/virology , Molecular Sequence Data , Polymerase Chain Reaction/methods , Poultry/virology , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment , Sequence Homology, Nucleic Acid , Spleen/virology
4.
Poult Sci ; 83(9): 1479-88, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15384897

ABSTRACT

White Leghorn chicks homozygous for B19 MHC haplotype were selected for 18 generations on tumor regression after inoculation in the wing web with an SR-D strain of Rous sarcoma virus (RSV) at 4 wk of age. Each chick was assigned a tumor profile index (TPI) based on age at death and size of the tumor. During 18 generations, 2,010 birds were divergently selected on TPI for either progression or regression of the tumor (P and R lines). A Brody growth curve was fitted for each bird. Brody function parameters included the asymptotic tumor volume (A), the factor for increased growth in progression phase (K1), the factor for decreased growth in regression phase (K2), age at maximum volume (Tmax), and maximum volume of the tumor (Vmax). Tumor growth curves were found to be different according to line, sex, and restriction fragment pattern Y complex Rfp-Y MHC haplotype (Yw*15, Yw*16, and Yw*17). Within the P line, birds from the Yw*16 haplotype reached Vmax at an earlier age than Yw*15 and Yw*17, but with a lower Vmax value. Within the R line, tumor growth curves of birds from Yw*16 and Yw*17 haplotypes were similar. Rank correlations between the different parameters and TPI were low (between -0.26 and 0.36). Heritability estimated by the sire component was high for Vmax (0.73). Heritabilities of Tmax and K2 were moderate (0.20 to 0.23 for Tmax and 0.18 to 0.21 for K2) allowing these traits to be used as selection criteria. Heritabilities of A and K1 were lower than 0.12. Modeling the growth curve should contribute to better distinction between progressors and regressors.


Subject(s)
Avian Sarcoma Viruses/pathogenicity , Chickens/growth & development , Chickens/genetics , Poultry Diseases/pathology , Sarcoma, Avian/pathology , Animals , Disease Progression , Female , Haplotypes/genetics , Major Histocompatibility Complex/genetics , Male , Poultry Diseases/virology , Regression, Psychology , Sarcoma, Avian/virology , Sex Factors , Tumor Burden/genetics
5.
Mol Pathol ; 55(4): 262-72, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12147717

ABSTRACT

AIMS: Lymphoblastoid cell lines derived from Marek's disease virus (MDV) induced tumours have served as models of MDV latency and transformation. They are stable and can be cultured with no detectable MDV genomic alterations upon repeated passaging. An MDV transformed lymphoblastoid T cell line (T9 cell line) has been reported to contain a disrupted MDV BamHI-H fragment and a Rous associated virus insertional activation of the c-myb protooncogene. In an attempt to define the respective participation of c-myb and MDV in the transformed phenotype of T9 cells, an analysis of MDV oncogenic sequences (BamHI-H, BamHI-A, and EcoQ fragments) was performed in these cells. METHODS: Using two different passages of the T9 cell line (late and early passages), the organisation of the MDV oncogenic regions and their expression in these cells were analysed. In vivo assessment of the oncogenicity of the virus contained within these cells was assessed by injecting them into 1 day old chickens. RESULTS: In T9 cells maintained in culture for up to six months (late T9), the MDV ICP4 gene was disrupted, whereas the meq gene was actively transcribed. The alterations of the MDV genome in these cells correlated with the inability of the virus to induce the classic signs of Marek's disease in 1 day old chickens. However, early T9 cells submitted to a limited number of passages induced classic MDV pathogenicity, as efficiently as the MDV control cell line (T5), and did not show gross structural changes in the oncogenic MDV sequences. CONCLUSIONS: Although the expression pattern of the MDV oncogenes in early T9 cells was identical to the one reported for other MDV transformed cells, longterm culture of an MDV transformed cell line containing a RAV insertional activation of the c-myb protooncogene led to the disruption of the MDV BamHI-H and BamHI-A oncogenic regions. In the late T9 cells MEQ was the only detected MDV oncoprotein. These results suggest that in the late T9 cells the truncated MYB protein compensates for the loss of MDV oncoproteins and reinforce the possibility that MEQ and MYB cooperate in the maintenance of the transformed state and the tumorigenic potential of these cells.


Subject(s)
Cell Transformation, Viral/genetics , Herpesvirus 2, Gallid/genetics , Lymphoma, T-Cell/virology , Marek Disease/virology , Viral Proteins , Animals , Chickens , Genes, myb , Genome, Viral , Herpesvirus 2, Gallid/pathogenicity , Nuclear Proteins/genetics , Oncogenes , Peptide Fragments/genetics , Polymerase Chain Reaction/methods , Protein Precursors/genetics , Trans-Activators/genetics , Transforming Growth Factor beta1 , Tumor Cells, Cultured , Virulence/genetics
6.
J Gen Virol ; 82(Pt 1): 233-240, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11125176

ABSTRACT

The avian herpesvirus Marek's disease virus (MDV) has a worldwide distribution and is responsible for T-lymphoma in chickens. The question as to whether MDV poses a public health hazard to humans was first raised when the virus was isolated in 1967. However, no irrefutable results have been obtained in immunological and virological studies. We used a nested-PCR to detect MDV DNA in human serum samples. A total of 202 serum samples from individuals exposed and not exposed to poultry was tested by nested-PCR for a target sequence located in the MDV gD gene. The assay system was specific and sensitive, making it possible to detect a single copy of the target sequence. Forty-one (20%) of the 202 serum samples tested positive for MDV DNA. The prevalence of MDV DNA was not significantly different in the group exposed to poultry and the group not exposed to poultry. There was also no difference due to age or sex. Alignment of the 41 gD sequences amplified from human sera with eight gD sequences amplified from MDV-infected chicken sera showed a maximum nucleotide divergence of 1.65%. However, four 'hot-spot' mutation sites were identified, defining four groups. Interestingly, two groups contained only human MDV-gD sequences. The status of the MDV genome detected in human blood is discussed.


Subject(s)
DNA, Viral/analysis , Herpesvirus 2, Gallid/isolation & purification , Marek Disease/virology , Occupational Diseases/virology , Adult , Amino Acid Sequence , Animal Husbandry , Animals , Base Sequence , Chickens , Female , Herpesvirus 2, Gallid/genetics , Humans , Male , Marek Disease/blood , Marek Disease/epidemiology , Molecular Sequence Data , Occupational Diseases/blood , Occupational Diseases/epidemiology , Polymerase Chain Reaction , Prevalence , Sequence Alignment
7.
Virology ; 277(1): 58-65, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11062036

ABSTRACT

The purpose of this study was to determine to what extent nitric oxide (NO) may play a role in the antiviral-mediated effect of chicken IFN-gamma against the Marek's disease virus (MDV) RB-1B. NO-generating compounds S-nitroso-N-acetylpenicillamine (SNAP) and 3-morpholino-sydononimine (SIN-1) strongly inhibited RB-1B replication in chicken embryo fibroblasts (85%) in a dose-dependent manner. The addition of superoxide dismutase (SOD) did not alter the inhibitory effect of SIN-1, which is also known to generate superoxide anions. IFN-gamma-stimulated embryo fibroblasts almost totally suppressed viral replication and were high NO producers. Nevertheless, addition of N(G)-monomethyl-l-arginine (l-NMMA), a competitive inhibitor of NO synthase, inhibited NO production without preventing the dramatic viral suppression. IFN-gamma-stimulated chicken bone-marrow macrophages were good NO producers and demonstrated a specific cell dose-related inhibiting effect on RB-1B replication in bystander fibroblasts (around 60% at 10(6) macrophages). Adding l-NMMA together with oxygen scavengers such as SOD or d-mannitol restored viral replication almost completely. In conclusion, NO alone is a powerful inhibitor of MDV replication in chicken fibroblasts. Nevertheless, NO is not responsible for the direct inhibitory effect of the IFN-gamma treatment of fibroblasts and is only partially involved in the inhibitory effect of IFN-gamma-stimulated macrophages, which is also mediated by reactive oxygen intermediates.


Subject(s)
Herpesvirus 2, Gallid/physiology , Interferon-gamma/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide/physiology , Virus Replication/drug effects , Animals , Bone Marrow Cells/cytology , COS Cells , Cells, Cultured , Chick Embryo , Chickens , Chlorocebus aethiops , Coculture Techniques , Fibroblasts , Herpesvirus 2, Gallid/drug effects , Macrophages/physiology , Macrophages/virology , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine , Superoxide Dismutase/pharmacology , Superoxides/metabolism , Virus Replication/physiology , omega-N-Methylarginine/pharmacology
8.
Avian Dis ; 44(4): 776-89, 2000.
Article in English | MEDLINE | ID: mdl-11195631

ABSTRACT

Histocompatible B13/B13 white specific-pathogen-free leghorn chickens were used to investigate the effect of coinfection with Cryptosporidium baileyi and the HPRS 16 strain of Marek's disease virus (MDV) in chickens and to assess the pathogenicity of C. baileyi when MDV is given before or after the parasite. Groups of chickens concurrently infected with C. baileyi orally inoculated at day (D)4 and MDV inoculated at hatching (C4M0 group) or at D8 (C4M8 group) were compared with relevant control groups inoculated with only C. baileyi at D4 (C4 group), only MDV at hatching (M0 group) or at D8 (M8 group), and an uninoculated control group (UC group). The chickens were kept in isolator units until the end of the experiment at D62. Our results showed a considerable synergistic effect in concurrently infected chickens and more severe consequences when chickens received MDV before C. baileyi infection. In fact, except for a slight transitory weakness, the chickens in C4 group remained free of overt clinical signs and there was no mortality. However, coinfection with both pathogens induced more lasting or permanent oocyst shedding. Severe clinical cryptosporidiosis with weakness, anorexia, depression, growth retardation, and chronic and severe respiratory disease causing death occurred in all chickens in the C4M0 group between D12 and D43 and in 67% of the chickens in the C4M8 group between D17 and D57. Eighty-two percent and 33%, respectively, died before the development of specific Marek's disease lesions. Mortality rates were 27% and 33% in the M0 and M8 groups, respectively. The presence of MDV enhanced the establishment of more lasting cryptosporidial infection in the respiratory tract, esophagus, crop, proventriculus, and kidneys (only in C4M0 group) as well as in bursa of Fabricius, ceca, and cloaca. Serologic analysis showed that chickens with chronic cryptosporidiosis in the C4M8 group had an increased level of C. baileyi-specific immunoglobulin A. Our results may explain some cases of mortality in chickens naturally infected with MDV and Cryptosporidium.


Subject(s)
Cryptosporidiosis/veterinary , Cryptosporidium/physiology , Herpesvirus 2, Gallid/pathogenicity , Marek Disease/complications , Poultry Diseases , Animals , Chickens , Cryptosporidiosis/complications , Cryptosporidiosis/mortality , Marek Disease/mortality , Marek Disease/virology , Parasite Egg Count , Poultry Diseases/mortality
9.
Avian Pathol ; 29(6): 623-34, 2000 Dec.
Article in English | MEDLINE | ID: mdl-19184860

ABSTRACT

This study was performed to examine the effect of Marek's disease virus (MDV) serotype 1 vaccine (CVI988/Rispens) on the pathogenicity of Cryptosporidium baileyi , and to determine whether C. baileyi infection could prevent the development of vaccinal Marek's disease (MD) immunity in specific pathogen free (SPF) chickens. Sixty-eight SPF homozygous B13 White Leghorn chickens were divided into seven groups. C. baileyi was orally administered at 5 days of age (day 4) in chickens infected with Rispens vaccine at day 0 or at day 8 and challenged with HPRS-16 strain of oncogenic MDV at day 15. Relevant control groups were constituted. The chickens were kept in isolators until the end of the experiment at day 62. The parameters evaluated were clinical signs, kinetics of oocyst shedding, mortality, macroscopic and microscopic lesions, cryptosporidia location in various organs and serum anti- C. baileyi antibodies at days 42 and 62. Our results show that C. baileyi , which is considered to be non-pathogenic when inoculated orally, may become highly pathogenic. It induced severe mortality and developed in organs other than classical target sites when chickens were vaccinated with Rispens vaccine and challenged with the HPRS-16 strain of MDV.However,parasite infection does not prevent the induction of vaccinal immunity for MD. Our results also show that vaccination of B13 chickens at hatching induces higher protection against challenge with HPRS-16 MDV at day 15 than vaccination at day 8.

10.
Avian Dis ; 43(4): 738-44, 1999.
Article in English | MEDLINE | ID: mdl-10611989

ABSTRACT

Renal Cryptosporidiosis was experimentally induced during a study to investigate the pathogenicity of Cryptosporidium baileyi in specific-pathogen-free (SPF) chickens coinfected with Marek's disease virus (MDV). Cryptosporidium baileyi was administered orally at 4 days of age to chickens previously infected at hatching (day 0) with the HPRS 16 strain of oncogenic MDV. Three control groups received MDV at hatching, C. baileyi on day 4, or placebo consisting of distilled water. Renal cryptosporidiosis lesions were induced in the group coinfected with MDV and C. baileyi. The kidneys were markedly swollen and pale, with visible urate crystals in the ureters and surface tubules. Oocysts of C. baileyi were demonstrated in six of seven cases tested by a scoring method with modified Sheather's sugar solution on renal tissue scrapings and were confirmed in three cases by histologic examination of paraffin-embedded kidney sections. Histologic study also revealed subacute interstitial nephritis, acute ureteritis, and attachment of cryptosporidia on the epithelial cell surface of the ureters and collecting ducts, collecting tubules, and distal convoluted tubules. Various developmental stages of the parasite were present in the kidney sections. To our knowledge, this is the first report of experimentally induced renal cryptosporidiosis in SPF chickens coinfected with MDV.


Subject(s)
Cryptosporidiosis/veterinary , Cryptosporidium/isolation & purification , Kidney Diseases/veterinary , Kidney/pathology , Marek Disease/complications , Poultry Diseases/physiopathology , Animals , Chickens , Cryptosporidiosis/complications , Cryptosporidiosis/pathology , Cryptosporidium/classification , Kidney/parasitology , Kidney Diseases/complications , Kidney Diseases/parasitology , Marek Disease/pathology , Poultry Diseases/pathology , Specific Pathogen-Free Organisms , Ureter/parasitology , Ureter/pathology
11.
Avian Dis ; 40(4): 841-52, 1996.
Article in English | MEDLINE | ID: mdl-8980816

ABSTRACT

In order to investigate the possibility of producing transgenic chickens by injection of avian leukosis virus-based vectors into testis, we have analyzed the infection rate of testicular cells following inoculation of Rous-associated virus type 1 (RAV-1) into the gonads of adult and 1-wk-old brown leghorn males. Viroproduction, neutralizing antibody production, and vital DNA presence in testis, blood, muscle, and semen were analyzed at various times after infection. Inoculation of RAV-1 into the gonads of adult males resulted in a low level of viroproduction in testis and blood, followed by the appearance of neutralizing antibody 2 or 3 wk later. Neither viroproduction in semen nor viral DNA presence in sperm were detected even though the infected chickens were found to produce RAV-1 in testis. One week after intratesticular inoculation of 1-wk-old males with RAV-1, a high level of viroproduction was found in blood and testis, and viral DNA was detected in gonadal cells. Further, by 6 wk after inoculation, the production of virus decreased in all tissues, viral DNA could not longer be detected in the testis, and neutralizing antibodies appeared in blood. All together these data show that it is possible to infect testicular cells by direct inoculation of RAV-1 in the testis, and that the immune response of both adult and young chickens seems to reduce this infection. Moreover, no evidence of spermatozoa infection was found; this result suggests that RAV-1 inoculation into testis may not induce genetic transmission of virus, and consequently would not be useful in the production of transgenic chickens.


Subject(s)
Antibodies, Viral/metabolism , Avian Leukosis Virus/immunology , Avian Leukosis Virus/isolation & purification , Avian Leukosis/immunology , Chickens , Poultry Diseases/immunology , Semen/virology , Testis/virology , Animals , Avian Leukosis/genetics , Avian Leukosis/metabolism , Base Sequence , DNA, Viral/analysis , DNA, Viral/chemistry , DNA, Viral/genetics , Disease Vectors , Male , Muscle, Skeletal/chemistry , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/veterinary , Poultry Diseases/metabolism , Poultry Diseases/pathology , Semen/chemistry , Testis/chemistry
12.
Br Poult Sci ; 37(3): 661-4, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8842472

ABSTRACT

1. After intramagnal insemination egg production decreased drastically during the first two days and was equivalent to egg production of hens inseminated intravaginally for the remaining period of collection. 2. After magnal insemination, the fertility of eggs collected during the first week was 36.2% and only 3.6% during the second week. 3. In the case of intramagnal insemination, egg fertility in the first week was 88.1%, in the second week 81.8% and the third week 52.3%. 4. The eggs laid during the first day after intramagnal insemination were 83.3% fertile, indicating that treated spermatozoa fertilised the newly ovulated egg within 20 minutes of ovulation.


Subject(s)
Fertilization , Insemination, Artificial/veterinary , Spermatozoa/physiology , Animals , Chickens , Female , Insemination, Artificial/methods , Male , Oviposition , Ovulation , Ovum , Vagina
13.
Occup Environ Med ; 53(6): 403-10, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8758036

ABSTRACT

OBJECTIVES: To compare the prevalence of antibodies against Marek's disease herpes virus (MDV) and against avian leukosis viruses type C (ALV) in groups of workers exposed to poultry and in unexposed groups. METHODS: Antibodies directed against avian viral proteins were detected by enzyme linked immunosorbent assay in 549 subjects. Exposure to chickens was high in two subgroups: farmers on intensive chicken farms and workers at chicken slaughterhouses. One subgroup, traditional farmers on dairy or pig farms with poultry, had moderate exposure to poultry. Another subgroup, farmers and slaughterhouse workers on quail farms, had high exposure to quails. Three subgroups were not exposed to chickens: farmers on dairy or pig farms without poultry, workers at cattle slaughterhouses, and white collar workers. Also, MDV antibodies were tested after serum sample adsorption with chicken antigens in 134 serum samples. RESULTS: The prevalence of antibodies against MDV was significantly higher in the exposed subgroups than in unexposed groups (odds ratio (OR) 6.17; 95% confidence interval (95% CI) 3.91-9.75). No association was found between seroprevalence and age. However, higher prevalence was found among women and was related to duration of exposure to chickens. The concentration of antibodies from a few subjects remained very high after adsorption. Significant differences between the men and women were found for the prevalence of antibodies for ALV but were not related to exposure to chickens. CONCLUSIONS: The prevalence of antibodies against MDV was significantly higher among workers exposed to chickens and was related to sex and duration of exposure. The higher prevalence of antibodies against avian oncogenic viruses found among women compared with men may be induced by differences in exposure or by genetic factors. The meaning of these high titres could be related to the presence of MDV in humans. Because the involvement of animal oncogenic viruses in human cancer is indicated by epidemiological and some experimental studies, the integration of viral DNA in human cells needs to be investigated.


Subject(s)
Alpharetrovirus/immunology , Animal Husbandry , Antibodies, Viral/blood , Herpesvirus 2, Gallid/immunology , Occupational Exposure , Oncogenic Viruses/immunology , Poultry , Animals , Antibody Specificity , Female , Humans , Male , Sex Factors
15.
Transgenic Res ; 4(6): 369-77, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7581517

ABSTRACT

We have used vectors derived from avian leukosis viruses to transduce exogenous genes into early somatic stem cells of chicken embryos. The ecotropic helper cell line, Isolde, was used to generate stocks of NL-B vector carrying the Neo(r) selectable marker and the Escherichia coli lacZ gene. Microinjection of the NL-B vector directly beneath unincubated chicken embryo blastoderms resulted in infection of germline stem cells. One of the 16 male birds hatched (6.25%) from the injected embryos contained vector DNA sequences in its semen. Vector sequences were transmitted to G1 progeny at a frequency of 2.7%. Neo(r) and lacZ genes were transcribed in vitro in chicken embryo fibroblast cultures from transgenic embryos of the G2 progeny.


Subject(s)
Animals, Genetically Modified/genetics , Avian Leukosis Virus/genetics , Chickens/genetics , Genetic Vectors , Stem Cells , Animals , Base Sequence , Breeding , Cells, Cultured , Chick Embryo , DNA, Recombinant/analysis , DNA, Recombinant/blood , DNA, Viral/analysis , DNA, Viral/blood , Female , Fibroblasts , Gene Transfer Techniques , Kanamycin Kinase , Male , Microinjections , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Semen/chemistry , Transgenes/genetics , beta-Galactosidase/genetics
16.
Poult Sci ; 73(12): 1897-905, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7877946

ABSTRACT

Stage X blastodermal cells were isolated from freshly laid unincubated Brown Leghorn chicken eggs. Five hundred cells from Stage X Brown Leghorn embryos were injected into the subgerminal cavity of White Leghorn unincubated embryos exposed to 550 rad of gamma irradiation from a cesium-137 source. Of 712 White Leghorn embryos that were irradiated and injected with Brown Leghorn blastodermal cells, 52 (7.3%) survived to hatching. Somatic chimerism was examined in the melanocyte population and erythroid lineage. The presence of brown feathers indicating donor cell contribution to melanocyte pigmentation was observed in 23 (44%) out of the 52 hatched chicks. Analysis of blood DNA was performed using a probe that revealed an endogenous retroviral gag fragment specific for the donor genome. Three out of these 23 chimeric chickens exhibited the gag-specific fragment. To test germline chimerism, chickens that reached sexual maturity were mated with Brown Leghorns. Three somatically chimeric hens produced Brown Leghorn progeny at a rate of 30.7, 9.2, and 2.9% respectively, thus proving donor cell contribution to the germline differentiation. Chimeric chickens obtained after injection of nonirradiated embryos exhibited a lower extent of chimerism at the feather level and did not show any chimerism in the erythroid lineage and the germline, thus demonstrating the value of the use of compromised recipient embryos to produce chimeras in chickens. Nevertheless, the extent of somatic chimerism could not be used to predict the germline chimerism.


Subject(s)
Animals, Genetically Modified/embryology , Blastoderm/cytology , Cell Transplantation/veterinary , Chickens/genetics , Chimera/genetics , Animals , Animals, Genetically Modified/genetics , Cell Transplantation/methods , Chick Embryo/radiation effects , DNA/analysis , Feathers/anatomy & histology , Gamma Rays , Radiation Chimera
17.
Virology ; 195(2): 812-9, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8393249

ABSTRACT

To identify the nucleotide sequences responsible for the tumorigenic specificity of myeloblastosis-associated virus (MAV) we have established the complete nucleotide sequences of three infectious clones inducing either both osteopetrosis and nephroblastoma [MAV2(O)/2 and MAV2(O)p9] or only nephroblastoma [MAV1(N)], and compared their biological properties in the same chicken host strain. The MAV2(O)p9 originally described as a type 2 strain was found to carry a hybrid env gene containing sequences of both the types 1 and 2, and it induced milder and less rapid osteopetrosis than the original MAV2(O) clone when injected into Brown Leghorn chickens. These results, together with sequence comparisons between the MAV strains examined, suggest that subtle changes in the primary structure of the TM env protein's extracellular domain are likely to affect the tumorigenic potential of MAV.


Subject(s)
Avian Myeloblastosis Virus/pathogenicity , Gene Products, env/physiology , Osteopetrosis/microbiology , Amino Acid Sequence , Animals , Base Sequence , Chickens , DNA, Viral , Gene Products, env/genetics , Gene Products, env/metabolism , Kidney Neoplasms/microbiology , Molecular Sequence Data , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Wilms Tumor/microbiology
18.
Mol Cell Biol ; 12(1): 10-21, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309586

ABSTRACT

Histological and anatomopathological studies performed on 152 independent myeloblastosis-associated virus type 1 (MAV1)-induced nephroblastomas allowed us to precisely define the chronology of tumor development in chickens. Three tumors representing increasing developmental stages were used to construct genomic libraries and to study both the state of proviral genomes and the sites of MAV1 integration in genomic DNA. We established that increasing levels of proviral rearrangement, eventually leading to the elimination of infectious MAV genomes, were associated with tumor progression and that 22 individual tumors, representative of different developmental stages, did not contain any common MAV1 integration site. Cloning of cellular fragments flanking the MAV1-related proviruses in tumor DNA showed that each one of eight nephroblastomas tested expressed a high level of an as yet unidentified cellular gene (nov) whose transcription is normally arrested in adult kidney cells. Cloning of the normal nov gene established that in one tumor, fused long terminal repeat-truncated nov mRNA species were expressed, indicating that at least in that case, the high level of nov expression was under the control of the MAV long terminal repeat promoter. The normal nov gene encodes a putative 32-kDa secreted polypeptide, which is a member of a new family of proteins likely to be involved in cell growth regulation. We also showed that the expression of an amino-terminal-truncated nov product in chicken embryo fibroblasts was sufficient to induce their transformation.


Subject(s)
Avian Myeloblastosis Virus/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Immediate-Early Proteins , Intercellular Signaling Peptides and Proteins , Oncogene Proteins, Viral/genetics , Recombination, Genetic , Wilms Tumor/microbiology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cells, Cultured , Chickens , Connective Tissue Growth Factor , DNA , Gene Expression Regulation, Viral , Molecular Sequence Data , Proviruses/genetics , Repetitive Sequences, Nucleic Acid , Restriction Mapping , Sequence Alignment , Wilms Tumor/genetics
20.
Poult Sci ; 70(10): 2161-75, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1659694

ABSTRACT

In a Brown Leghorn chicken strain, four endogenous proviral loci have been identified. The DNA mapping data show strong homology between their structures and that of the Rous-associated virus O (RAV-O) genome. Two of them seem similar to ev3 and ev6 loci previously described in White Leghorn chickens; the two others are unknown in White Leghorns. Using DNA amplification methods, envelope genes of these endogenous viral structures have been partially sequenced. The results demonstrate that subgroup-specific sequences of the endogenous loci were largely homologous with those of RAV-O.


Subject(s)
Avian Leukosis Virus/genetics , Chickens/genetics , DNA/chemistry , Proviruses/genetics , Animals , Base Sequence , Blotting, Southern , Breeding , Chickens/microbiology , DNA/analysis , DNA Probes , Female , Male , Molecular Sequence Data , Polymerase Chain Reaction , Restriction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...