Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Neoplasma ; 63(3): 402-10, 2016.
Article in English | MEDLINE | ID: mdl-26925785

ABSTRACT

Tubulocystic renal cell carcinoma (TRCC) represents a rare tumor with incidence lower than 1 % of all renal carcinomas. This study was undertaken to contribute to characterization of molecular signatures associated with TRCC and to compare them with the features of papillary renal cell carcinoma (PRCC) at the level of genome wide methylation analysis.We performed methylated DNA immunoprecipitation (MeDIP) coupled with microarray analysis (Roche NimbleGen). Using the CHARM package, we compared the levels of gene methylation between paired samples of tumors and control renal tissues of each examined individual. We found significant global demethylation in all tumor samples in comparison with adjacent kidney tissues of normal histological appearance but no significant differences in gene methylation between the both compared tumor entities. Therefore we focused on characterization of differentially methylated regions between both tumors and control tissues. We found 42 differentially methylated genes.Hypermethylated genes for protocadherins (PCDHG) and genes coding for products associated with functions of plasma membrane were evaluated as significantly overrepresented among hypermethylated genes detected in both types of renal cell carcinomas.In our pilot study, we provide the first evidence that identical features in the process of carcinogenesis leading to TRCC and/or to PRCC may be found at the gene methylation level.


Subject(s)
Carcinoma, Papillary/genetics , Carcinoma, Renal Cell/genetics , DNA Methylation , Kidney Neoplasms/genetics , Adult , Aged , Carcinogenesis/genetics , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Female , Genome-Wide Association Study , Humans , Immunoprecipitation , Kidney Neoplasms/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pilot Projects
2.
Clin Genet ; 90(3): 230-7, 2016 09.
Article in English | MEDLINE | ID: mdl-26841933

ABSTRACT

Kabuki syndrome (KS) is a dominantly inherited disorder mainly due to de novo pathogenic variation in KMT2D or KDM6A genes. Initially, a representative cohort of 14 Czech cases with clinical features suggestive of KS was analyzed by experienced clinical geneticists in collaboration with other specialties, and observed disease features were evaluated according to the 'MLL2-Kabuki score' defined by Makrythanasis et al. Subsequently, the aforementioned genes were Sanger sequenced and copy number variation analysis was performed by MLPA, followed by genome-wide array CGH testing. Pathogenic variants in KMT2D resulting in protein truncation in 43% (6/14; of which 3 are novel) of all cases were detected, while analysis of KDM6A was negative. MLPA analysis was negative in all instances. One female patient bears a 6.6 Mb duplication of the Xp21.2-Xp21.3 region that is probably disease causing. Subjective KS phenotyping identified predictive clinical features associated with the presence of a pathogenic variant in KMT2D. We provide additional evidence that this scoring approach fosters prioritization of patients prior to KMT2D sequencing. We conclude that KMT2D sequencing followed by array CGH is a diagnostic strategy with the highest diagnostic yield.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Face/abnormalities , Hematologic Diseases/diagnosis , Hematologic Diseases/genetics , Histone Demethylases/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Vestibular Diseases/diagnosis , Vestibular Diseases/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Child , Child, Preschool , Comparative Genomic Hybridization , Czech Republic , Face/physiopathology , Female , Genome, Human , Hematologic Diseases/physiopathology , Humans , Infant , Male , Phenotype , Vestibular Diseases/physiopathology
4.
Anim Genet ; 35(3): 176-81, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147387

ABSTRACT

Avian sarcoma and leucosis viruses (ASLV) are classified into six major subgroups (A to E and J) according to the properties of the viral envelope proteins and the usage of cellular receptors for virus entry. Subgroup A and B receptors are identified molecularly and their genomic positions TVA and TVB are mapped. The subgroup C receptor is unknown, its genomic locus TVC is reported to be genetically linked to TVA, which resides on chicken chromosome 28. In this study, we used two chicken inbred lines that carry different alleles coding for resistance (TVC(R) and sensitivity (TVC(S)) to infection by subgroup C viruses. A backross population of these lines was tested for susceptibility to subgroup C infection and genotyped for markers from chicken chromosome 28. We confirmed the close linkage between TVA and TVC loci. Further, we have described the position of TVC on chromosome 28 relative to markers from the consensus map of the chicken genome.


Subject(s)
Avian Sarcoma Viruses/genetics , Chickens/genetics , Chromosome Mapping , Phenotype , Receptors, Virus/genetics , Viral Envelope Proteins/genetics , Alleles , Animals , Avian Sarcoma Viruses/classification , Chickens/virology , Crosses, Genetic , DNA Primers , Microsatellite Repeats/genetics
5.
Folia Biol (Praha) ; 49(3): 100-9, 2003.
Article in English | MEDLINE | ID: mdl-12859018

ABSTRACT

We have studied the pathogenic changes in Khaki Campbell ducks injected in mid embryogenesis with ALV subgroup C virus td daPR-C derived from a molecular clone. The employed duck flock was shown to be highly genetically homogeneous and was controlled for the absence of current infections. Clear symptoms of wasting disease, which appeared since one week post hatching, represented the early consequence of the virus infection. They were manifested by decreased body weight, including clear involution of thymic tissue and pronounced anaemia. Microscopically, thymuses of infected animals displayed lymphatic depletion, clearly visible in the lobular cortex. Similarly, in the bursa Fabricii follicles, a marked reduction of the cortical layer and a decrease in folicullar centres was revealed. A decrease in the antibody response correlated with bursa Fabricii atrophy. The clear signs of anaemia were confirmed by haematological measurements, red blood cell count, haematocrit value and haemoglobin included. On the basis of these and additional observations we propose that inoculation of duck embryos provides a suitable model for analysis of the wasting disease produced by ALV-C.


Subject(s)
Avian Leukosis Virus/metabolism , Avian Leukosis/virology , Wasting Syndrome/virology , Animals , Animals, Newborn , Antibodies, Viral , Apoptosis , Brucella abortus/metabolism , Bursa of Fabricius/pathology , Ducks/embryology , Ducks/virology , Kinetics , Thymus Gland/pathology , Time Factors , Vaccination
6.
Folia Biol (Praha) ; 49(2): 51-62, 2003.
Article in English | MEDLINE | ID: mdl-12779013

ABSTRACT

Tight regulation of the Src tyrosine kinase activity is essential for a variety of cellular processes, namely transitions of the cell cycle. The peaks of Src activity are dependent on its posttranslational modifications as well as on the regulation of gene expression. The 3'UTRs of mRNAs are often crucial for rapid changes of the protein level. The chicken c-src 3'UTR effects on gene expression have been explored. The c-src 3'UTR decreased the in vivo tumorigenic potential of the src-activated mutants in chickens. This corresponds with the finding that the c-src 3'UTR reduced the Src protein and src mRNA levels and luciferase activity in vitro. Our results suggest that the chicken c-src 3'UTR plays a role in the negative control of gene expression, either transcriptionally or posttranscriptionally.


Subject(s)
3' Untranslated Regions/genetics , Cell Cycle Proteins/genetics , Cell Cycle/genetics , Gene Expression Regulation/genetics , Genes, src/genetics , Animals , CSK Tyrosine-Protein Kinase , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Chick Embryo , Down-Regulation/genetics , Genes, Regulator/genetics , Luciferases/genetics , Mutation/genetics , Neurons/cytology , Neurons/metabolism , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , src-Family Kinases
7.
Avian Pathol ; 30(1): 33-42, 2001 Feb.
Article in English | MEDLINE | ID: mdl-19184871

ABSTRACT

The breeding history of the first inbred strain of Khaki Campbell ducks is presented. The genetic homogeneity of this strain was tested on the basis of serum amyloid A (SAA) polymorphism and it was established that it harbours only the SAA allele A, which is expressed in liver, lung and bursa of Fabricius tissues. Pathogenic changes in control and avian leukosis virus-C (ALV-C) persistently infected ducks were evaluated during the period spanning 1 to 10 months after hatching. In both groups, AA amyloidosis was revealed and characterized. In spite of the inbred nature of animals, the incidence of amyloid A deposition varied among experiments, suggesting that additional non-genetic factors are involved. Similar variation was found in ALV-C persistently infected ducks, where only in one out of three experiments was the incidence ofAA amyloidosis significantly higher than in controls.

8.
Gene ; 261(1): 181-8, 2000 Dec 30.
Article in English | MEDLINE | ID: mdl-11164049

ABSTRACT

Retroviruses are known to integrate in the host cell genome as proviruses, and therefore they are prone to cell-mediated control at the transcriptional and posttranscriptional levels. This plays an important role especially after retrovirus heterotransmission to foreign species, but also to differentiated cells. In addition to host cell-mediated blocks in provirus expression, also so far undefined host specificities, deciding upon the pathogenic manifestation of retrovirus heterotransmission, are in play. In this respect, we discuss especially the occurrence of wasting disease and immunodeficiency syndrome, which we established also in avian species using avian leukosis virus subgroup C (ALV-C) inoculated in mid-embryogenesis in duck or chicken embryos. The problem of provirus downregulation in foreign species or in differentiated cells has been in the recent years approached experimentally. From a series of observations it became apparent that provirus downregulation is mediated by its methylation, especially in the region of proviral enhancer-promoter located in long terminal repeats (LTR). Several strategies have been devised in order to protect the provirus from methylation using LTR modification and/or introducing in the LTR sequence motifs acting as antimethylation tags. In such a way the expression of retroviruses and vectors in foreign species, as well as in differentiated cells, has been significantly improved. The complexity of the mechanisms involved in provirus downregulation and further possibilities to modulate it are discussed.


Subject(s)
Gene Silencing , Proviruses/genetics , Retroviridae/genetics , Animals , Humans , Species Specificity
9.
Arch Virol ; 144(9): 1779-807, 1999.
Article in English | MEDLINE | ID: mdl-10542026

ABSTRACT

Proviral DNA load and expression of avian leukosis viruses of subgroup C (ALV-C) in ducks infected in mid embryogenesis were studied using quantitative PCR, RT-PCR, in situ hybridization employing ALV-specific riboprobe, and immunohistochemistry. A group of long-term surviving, non-reviremic ducks was selected for the study and compared to control reviremic animals in order to obtain information about persisting retroviruses in different duck tissues. A widespread distribution of proviruses in the tested tissues was found, but the proviral load was significantly lower in non-reviremic in comparison to reviremic animals. The only exception were brain and blood cells, in which no significant difference in the quantity of integrated proviruses was found between both categories of ducks, thus indicating an exceptional position of the brain and blood cells among all tested tissues. Contrary to reviremic, the proviruses were not transcribed in non-reviremic ducks, with the exception of brain and thymus. In the majority of non-reviremic ducks viral RNA was revealed in the brain, but no infectious virus could be recovered from this tissue. The opposite situation was observed in the thymus, where infectious virus was recovered but viral RNA remained below the detection limit of the assay. As revealed by in situ analysis, infected cells were either disseminated or focally distributed in tissues. From the long-term follow up of ALV-C in intraembryonally infected ducks we conclude that this model is suitable for the study of retrovirus persistence accompained both by the presence and absence of reviremias. The possible consequences of transmission and long-term persistence of retroviruses in the heterologous host for retroviral evolution are discussed.


Subject(s)
Avian Leukosis Virus/isolation & purification , Avian Leukosis/virology , Ducks/virology , Proviruses/isolation & purification , Animals , Antibodies, Viral/blood , Avian Leukosis/immunology , Avian Leukosis Virus/genetics , Avian Leukosis Virus/immunology , Avian Leukosis Virus/physiology , DNA, Viral/blood , DNA, Viral/isolation & purification , Ducks/embryology , Immunohistochemistry , In Situ Hybridization , Neutralization Tests , Polymerase Chain Reaction , Proviruses/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Viral Load , Viremia/virology
10.
Virology ; 255(1): 171-81, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10049832

ABSTRACT

Rous sarcoma virus (RSV) enhancer sequences in the long terminal repeat (LTR) have previously been shown to be sensitive to CpG methylation. We report further that the high density methylation of the RSV LTR-driven chloramphenicol acetyltransferase reporter is needed for full transcriptional inhibition in chicken embryo fibroblasts and for suppression of tumorigenicity of the RSV proviral DNA in chickens. In nonpermissive mammalian cells, however, the low density methylation is sufficient for full inhibition. The time course of inhibition differs strikingly in avian and mammalian cells: although immediately inhibited in mammalian cells, the methylated RSV LTR-driven reporter is fully inhibited with a significant delay after transfection in avian cells. Moreover, transcriptional inhibition can be overridden by transfection with a high dose of the methylated reporter plasmid in chicken cells but not in hamster cells. The LTR, v-src, LTR proviral DNA is easily capable of inducing sarcomas in chickens but not in hamsters. In contrast, Moloney murine leukemia virus LTR-driven v-src induces sarcomas in hamsters with high incidence. Therefore, the repression of integrated RSV proviruses in rodent cells is directed against the LTR.


Subject(s)
Avian Sarcoma Viruses/genetics , Gene Expression Regulation, Viral , Terminal Repeat Sequences , 3T3 Cells , Animals , Cell Line , Chick Embryo , Chickens , Cricetinae , DNA Methylation , DNA, Viral , Mice , Moloney murine leukemia virus/genetics , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Proviruses/genetics , Sarcoma, Experimental , Time Factors , Transcription, Genetic , Transfection
11.
Gene ; 208(1): 73-82, 1998 Feb 16.
Article in English | MEDLINE | ID: mdl-9479051

ABSTRACT

Although the Rous sarcoma virus (RSV) long terminal repeat (LTR) is an efficient promoter of transcription, most RSV proviruses are down-regulated upon retroviral integration in non-permissive mammalian cells. Among other mechanisms, DNA methylation has been shown to be involved in proviral silencing. The presence of Sp1 binding sites has been demonstrated to be essential for protection of a CpG island and also non-island DNA regions from de novo methylation. Also, the presence of these sites in the LTRs correlates with the transcriptional activity of certain proviral structures. Using transient and stable transfection assays, we demonstrate that insertion of Sp1 binding sites into the RSV LTR remarkably increases expression of the LTR-driven genes in permissive and non-permissive cells, despite the reported negative effect of insertion of the non-specific DNA into the LTR promoter/enhancer sequences. Particular arrangement of inserted Sp1 sites was effective even in stably transfected reporter gene constructs into non-permissive mammalian cells, where additional factors exert negative effects on expression.


Subject(s)
Avian Sarcoma Viruses/genetics , Gene Expression Regulation, Viral , Repetitive Sequences, Nucleic Acid , Sp1 Transcription Factor/metabolism , Animals , Binding Sites , Cell Line , DNA Methylation , Genes, Reporter , Plasmids , Transfection , Tumor Cells, Cultured
12.
Oncogene ; 13(12): 2717-25, 1996 Dec 19.
Article in English | MEDLINE | ID: mdl-9000146

ABSTRACT

We previously reported the isolation of PR2257, a novel avian sarcoma retrovirus which transduced the c-src protooncogene. The v-src gene of PR2257 differs from the c-src gene by a sequence change after amino acid 525, resulting in the replacement of tyrosine 527 by a valine, and an extension of the open reading frame into the non coding region of c-src. We investigated the respective roles of Tyr527 mutation and of the C-terminal extension in activating the oncogenic properties of c-src. Therefore we overexpressed the wild type c-src gene and c-src variants, carrying either a substitution of tyrosine 527 or an extension of the C-terminus or both modifications in combination, in chicken embryo fibroblasts and post mitotic neuroretina (NR) cells, using replication defective retroviruses. We also used in vivo inoculation of plasmid DNA to assess the tumorigenicity of the various c-src genes. We report that, in contrast to previous results, overexpression of c-src is sufficient to induce NR cell division. While mutation of tyrosine 527 alone significantly activates c-src transforming and tumorigenic properties, its combination with the C-terminal extension of PR2257 confers to this gene full oncogenic properties and increased metastatic potential as compared to the v-src of Rous sarcoma virus strains.


Subject(s)
Cell Differentiation/genetics , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Genes, src/genetics , src-Family Kinases/genetics , Animals , Chickens , Genetic Vectors , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Phenotype , Retroviridae/genetics , Transfection , src-Family Kinases/metabolism
13.
Gene ; 174(1): 9-17, 1996 Sep 26.
Article in English | MEDLINE | ID: mdl-8863723

ABSTRACT

The tumor hamster cell line H-19 harbors a single copy LTR, v-src, LTR provirus that becomes permanently transcriptionally suppressed in morphological revertants segregating at high rate from this cell line. Our previous data document that the provirus suppression is mediated by epigenetic cell-regulatory mechanisms. In this report, we concentrate on cellular sequences neighboring the integration site. The locus is unique for Syrian hamster and is not detectable in DNA of several animal species. No restriction sites that usually hint at the presence of CpG islands were found in the significantly close vicinity of the provirus. Nevertheless, the chromosomal DNA flanking the provirus is rich in GC content (57.8%). We localized a 0.5-kb region downstream from the provirus that remarkably inhibits transcription in the transient expression assay and is effective both on the homologous RSV LTR promoter/enhancer and heterologous SV40 promoter. We propose that a cellular trans-acting factor is involved in the silencing of the reporter gene. Since this activity is comparable both in transformed and revertant cells, we speculate that this down-regulatory region makes the permissive integration locus prone to provirus silencing initiated by other fluctuating stimuli.


Subject(s)
Avian Sarcoma Viruses/physiology , DNA, Viral/genetics , Genes, src , Proviruses/genetics , Repetitive Sequences, Nucleic Acid/genetics , Transcriptional Activation , Virus Integration/genetics , Animals , Cricetinae , DNA/genetics , Mesocricetus , Molecular Sequence Data , Sequence Analysis, DNA , Tumor Cells, Cultured
14.
J Autoimmun ; 9(2): 129-38, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8738956

ABSTRACT

The early, predictable, onset of spontaneous autoimmune thyroiditis (SAT) in Obese strain (OS) chickens provides a unique opportunity to analyse the mechanisms initiating autoimmunity which is virtually impossible to obtain in humans. In this study we focused on the respective roles of viruses and macrophages in the initiation of SAT. To analyse viruses, leukosis virus-free OS chickens were bred over three generations and reared under gnotobiotic conditions. By 2 weeks of age there were no differences in the levels of thyroid mononuclear cell infiltration between these and control animals. The role of mononuclear phagocytes in SAT was determined by their depletion via injecting newborn OS chicks with silica or carrageenan and dichloro-methylene diphosphonate encapsulated into liposomes. Although this treatment did not substantially change the amount of macrophages in primary lymphoid organs or blood, there was destruction of splenic architecture and, most importantly, mononuclear cell infiltration of the thyroids was significantly lower compared to controls. The role of activated macrophages in SAT is discussed.


Subject(s)
Avian Leukosis/complications , Macrophages/immunology , Thyroiditis, Autoimmune/etiology , Animals , Chick Embryo , Chickens , Disease Models, Animal , Germ-Free Life , Obesity/complications , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/virology
16.
Folia Biol (Praha) ; 42(5): 245-55, 1996.
Article in English | MEDLINE | ID: mdl-8997643

ABSTRACT

Ducks intraembryonally infected with avian leukosis viruses of subgroup C (ALV-C) were followed for a long period (up to 6.8 years), and the viraemia and production of virus-neutralizing antibodies were measured. In three independent experiments comprising ducks inoculated with uncloned and/or molecularly cloned ALV-C, we found that after the elimination of primary post-hatching viraemia, reviraemia could be detected in 60-70% of infected animals. Based on the course of viraemia, the individual ducks were assigned to four different groups: Group I (no reviraemia), Group II (one transient reviraemic period), Group III (one persistent reviraemic period), Group IV (fluctuating reviraemia). In comparison to sera from ducks included in Group I and/or II, a significant decrease in neutralizing activity of sera from animals comprised in Group III and/or IV was observed. Two out of four reviraemic viruses were not neutralized by antiserum against ALV-C, instead their infectivity was enhanced. Long-term follow-up of the cell-associated virus revealed that its rescuability by cocultivation with chicken embryo fibroblasts fluctuated in about 50% of animals. In the reviraemic phase of infection, integrated proviruses could be detected by Southern blotting in a majority of tissues examined. Our data document that many features recognized in lentiviruses are valid also for oncoviruses transmitted to heterologous hosts and substantiate further the suitability of ALV-C-infected ducks as a model for studying persistent retroviral infection.


Subject(s)
Avian Leukosis Virus/isolation & purification , Avian Leukosis/virology , Viremia/virology , Animals , Antibodies, Viral/blood , Avian Leukosis/blood , Avian Leukosis/immunology , Avian Leukosis Virus/genetics , Avian Leukosis Virus/immunology , DNA, Viral/analysis , Ducks/embryology , Kinetics , Neutralization Tests , Proviruses/genetics , Time Factors , Tissue Distribution
17.
Exp Clin Immunogenet ; 12(4): 272-82, 1995.
Article in English | MEDLINE | ID: mdl-8919361

ABSTRACT

The oncogenic potential of different strains of Rous sarcoma virus (RSV) varies significantly in two Mhc(B) congenic chicken lines CB and CC and in their F1 hybrids with the unrelated inbred line IA. The Bryan high-titer pseudotype of RSV of antigenic subgroups D and B (BH-D, and BH-B) was highly oncogenic, eliciting mostly fatal, progressively growing tumors. Visceral tumor formation and erythroblastosis were seen after challenge with BH-B in CB and CC chickens, but not in their F1 hybrids with IA. Tumor regression induced by a low-oncogenic RSV strain PR-C elicited a protective immunity capable of preventing not only the progressive growth of challenge tumors at the site of inoculation, but also visceral tumor formation and erythroblastosis.


Subject(s)
Avian Sarcoma Viruses/immunology , Cell Transformation, Neoplastic , Cell Transformation, Viral , Sarcoma, Avian/etiology , Sarcoma, Avian/prevention & control , Viral Vaccines/immunology , Viscera , Animals , Antibodies, Viral/pharmacology , Cell Division/immunology , Chickens , Immunity, Cellular , Sarcoma, Avian/mortality
18.
J Gen Virol ; 75 ( Pt 10): 2777-81, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7931166

ABSTRACT

Avian sarcoma virus PR2257 transduced de novo the c-src gene and about 900 bp of 3' non-coding sequences belonging to the src locus. This virus contains only one mutation in the c-src coding sequence causing a reading frame shift after Pro-525. The molecular clone studied was derived from a cell line of transformed quail fibroblasts, C7. It contains endogenous virus (ev) derived sequences in the U5 and 3' non-coding regions, indicating that multiple recombination occurred with endogenous virus. Here we investigated the possible evolution of PR2257 when the original tumour was repeatedly passaged in vivo. After 16 passages a new virus, designated PR2257/16, appeared with a tenfold higher titre. The sequence of PR2257/16 was determined and showed that PR2257/16 resulted from recombination of PR2257 with the env gene of the helper virus (td daPR-C). This recombination expanded the env gene content in PR2257/16 and, in addition, five point mutations occurred in its genome. Because we thought that an endogenous virus might be involved in the mechanism of c-src transduction, we also reinvestigated the presence of ev sequences in PR2257 proviruses from several early passages of the original tumour. We found that in contrast with the first isolate from the C7 cell line, the provirus in these tumours did not contain such sequences. These results do not support the hypothesis that endogenous sequences were involved in the transduction process.


Subject(s)
Avian Sarcoma Viruses/genetics , Biological Evolution , Genes, src , Animals , Base Sequence , Cell Line , Cloning, Molecular , Genes, env , Helper Viruses/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Proviruses/genetics , Quail , Recombination, Genetic , Sequence Homology, Nucleic Acid
19.
Cell Growth Differ ; 5(3): 277-85, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8018560

ABSTRACT

The highly malignant line of morphologically transformed H-19 hamster tumor cells that harbor a single LTR, v-src, LTR provirus segregates morphologically flat revertants at the rate of 1.4 to 2.4 x 10(-3)/cell/cycle. Revertants behave like almost nonmalignant cells; they keep the provirus within an unaltered junction DNA fragment. However, the provirus is methylated, permanently transcriptionally silent, and not rescuable. Using the polymerase chain reaction, we have synthesized the whole proviral structure from two revertants and established that the left-hand long terminal repeats assuring transcription remained structurally intact. Moreover, the cloned proviral DNAs from three revertants were shown to produce tumors in chickens. The unusually high reversion rate together with the finding of structural integrity of proviral transcriptional signals in revertants indicate strongly that the reversion has been mediated by cell-regulatory mechanisms.


Subject(s)
Cell Line, Transformed/cytology , Cell Line, Transformed/microbiology , Genes, src , Proviruses/genetics , Suppression, Genetic , Transcription, Genetic , Animals , Base Sequence , Chick Embryo , Chickens , Cricetinae , Mesocricetus , Molecular Sequence Data , Mutation , Repetitive Sequences, Nucleic Acid , Time Factors , Virus Integration
20.
Immunogenetics ; 40(4): 257-65, 1994.
Article in English | MEDLINE | ID: mdl-8082891

ABSTRACT

The growth pattern (progression/regression) of v-src DNA- and Rous sarcoma virus (RSV)-induced tumors was analogous on a panel of inbred chicken lines. The decisive role of the major histocompatibility complex [Mhc(B)] alleles in resistance to the progression of these tumors was formally proved in segregating backcross populations. The immune mechanism of tumor regression was demonstrated by both in vivo and in vitro assays. A protective effect of v-src-specific immunity against RSV challenge was shown in Rous sarcoma regressor, line CB (B12/B12). Immune cells from regressors of v-src DNA-induced tumors can protect syngeneic hosts from the development of tumor after challenge with both v-src DNA and RSV. Suppression of RSV-induced tumor cell growth in vitro was also achieved by the use of cocultivation with spleen cells from chickens in which v-src DNA-induced tumors had regressed. This in vitro sarcoma-specific response was Mhc(B)-restricted. Chickens of the congenic Rous sarcoma progressor line CC (B4/B4) are sometimes able to regress v-src DNA-induced tumors, but immune cells can only slow the growth of v-src DNA-induced tumors in syngeneic hosts. This suggests that the primary reason for the susceptibility of CC chickens is a weak v-src-specific immune response. Furthermore, some of the v-src DNA-induced tumors were transplantable across the Mhc(B) barrier. The growth of tumor allografts was able to be facilitated when immunological tolerance to the B-F/L region antigens (class I and class II) had been established. This demonstrated that a high tumorigenicity of the transplantable tumor was not due to the lack of Mhc(B) antigens on tumor cells.


Subject(s)
Avian Sarcoma Viruses/genetics , Chickens/immunology , Genes, src , Major Histocompatibility Complex , Neoplasms, Experimental/immunology , Oncogene Protein pp60(v-src)/immunology , Animals , Cell Transformation, Viral , DNA, Viral/genetics , Female , Inbreeding , Male , Repetitive Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...