Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Rev Med Brux ; 23 Suppl 2: 131-6, 2002.
Article in French | MEDLINE | ID: mdl-12584929

ABSTRACT

The clinic of medical oncology is mainly devoted to the development of new anticancer treatments based on molecular biology and immunology. The clinic was the first in Belgium to start a protocol of gene therapy. Scientific contributions deal with the role of various oncogens in cell transformation, the interaction between cancer and the immune system and, new tools for the molecular diagnosis of cancers. Focus was particularly put on the development of new vectors for gene therapy and antitumor cell vaccines for cell therapy.


Subject(s)
Oncology Service, Hospital , Belgium , Biomedical Research , Cell Transformation, Neoplastic , Hospitals, University , Humans , Neoplasms/therapy
2.
J Virol ; 75(3): 1284-93, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11152501

ABSTRACT

The production of wild-type-free stocks of recombinant parvovirus minute virus of mice [MVM(p)] is difficult due to the presence of homologous sequences in vector and helper genomes that cannot easily be eliminated from the overlapping coding sequences. We have therefore cloned and sequenced spontaneously occurring defective particles of MVM(p) with very small genomes to identify the minimal cis-acting sequences required for DNA amplification and virus production. One of them has lost all capsid-coding sequences but is still able to replicate in permissive cells when nonstructural proteins are provided in trans by a helper plasmid. Vectors derived from this particle produce stocks with no detectable wild-type MVM after cotransfection with new, matched, helper plasmids that present no homology downstream from the transgene.


Subject(s)
Defective Viruses/genetics , Genetic Vectors , Minute Virus of Mice/genetics , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Polymerase Chain Reaction , Virus Replication
3.
Hum Gene Ther ; 10(7): 1229-38, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10340554

ABSTRACT

We have derived a vector from the autonomous parvovirus MVM(p), which expresses human IL-2 specifically in transformed cells (Russell et al., J. Virol 1992;66:2821-2828). Testing the therapeutic potential of these vectors in vivo requires high-titer stocks. Stocks with a titer of 10(9) can be obtained after concentration and purification (Avalosse et al., J. Virol. Methods 1996;62:179-183), but this method requires large culture volumes and cannot easily be scaled up. We wanted to increase the production of recombinant virus at the initial transfection step. Poor vector titers could be due to inadequate genome amplification or to inefficient packaging. Here we show that intracellular amplification of MVM vector genomes is not the limiting factor for vector production. Several vector genomes of different size and/or structure were amplified to an equal extent. Their amplification was also equivalent to that of a cotransfected wild-type genome. We did not observe any interference between vector and wild-type genomes at the level of DNA amplification. Despite equivalent genome amplification, vector titers varied greatly between the different genomes, presumably owing to differences in packaging efficiency. Genomes with a size close to 100% that of wild type were packaged most efficiently with loss of efficiency at lower and higher sizes. However, certain genomes of identical size showed different packaging efficiencies, illustrating the importance of the DNA sequence, and probably its structure.


Subject(s)
DNA, Viral/physiology , Genetic Vectors , Minute Virus of Mice/genetics , Minute Virus of Mice/physiology , Virus Assembly , Bacteriophage lambda/genetics , Cell Line , DNA, Viral/chemistry , DNA, Viral/metabolism , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Particle Size , Plasmids/genetics , Transfection
4.
Eur J Cancer ; 33(10): 1648-53, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9389929

ABSTRACT

We have previously demonstrated lysis of non-established cultures of human mammary carcinoma cells by parvovirus H-1, which has little effect on the proliferation of corresponding normal cultures. In the present study, we examined this effect in a number of breast-tumour specimens and found them to differ as to the amplitude of their response to parvoviral attack. We first investigated whether the differences in cell sensitivity to parvovirus infection reflected the differentiation level of the initial tumour. Among the biochemical and anatomopathological indicators of original tumour differentiation, the presence of oestrogenic receptors (ER) was found to have a predictive value as to the sensitivity of derived cultures to the cytopathic effect of H-1 virus. The ER+ tumour-derived cultures showed an increased sensitivity to the lytic effect of H-1 virus compared with the ER-tumour-derived cultures, in spite of similar average proliferation rates for the two types of cultures. The proliferation rate was more heterogeneous among ER+ tumour-derived cultures and, in this group, the faster growing cultures were also the most sensitive. This observation was corroborated by the study of established cell lines retaining ER expression under in vitro culture conditions. Oestradiol was found to increase the sensitivity of these cells to the parvovirus in parallel with induction of proliferation. This effect appeared to be mediated by ER activation, since it was not observed in the ER-negative cell line MDA-MB-231. These data point to the importance of hormonal influences and cellular parameters, notably differentiation and proliferation, in determining the extent to which human cancer cells can be targets for the cytopathic effect of parvoviruses.


Subject(s)
Breast Neoplasms/complications , Breast Neoplasms/pathology , Parvoviridae Infections/complications , Parvovirus/physiology , Breast Neoplasms/chemistry , Cell Differentiation/physiology , Cell Division/physiology , Cell Survival/physiology , Female , Humans , Receptors, Estrogen/analysis , Tumor Cells, Cultured
5.
Gene Ther ; 3(10): 927-31, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8908507

ABSTRACT

MVM-based autonomous parvoviral vectors have been shown to target the expression of heterologous genes in neoplastic cells and are therefore of interest for cancer gene therapy. The traditional method for production of parvoviral vectors requires the cotransfection of vector and helper plasmids into MVM-permissive cell lines, but recombination between the cotransfected plasmids invariably gives rise to vector stocks that are heavily contaminated with wild-type MVM. Therefore, to minimise recombination between the vector and helper genomes we have utilised a cell line in which the MVM helper functions are expressed inducibly from a modified MVM genome that is stably integrated into the host cell chromosome. Using this MVM packaging cell line, we could reproducibly generate MVM vector stocks that contained no detectable helper virus.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Interleukin-2/genetics , Minute Virus of Mice/genetics , Animals , Cell Line , Mice
6.
J Virol ; 66(5): 2821-8, 1992 May.
Article in English | MEDLINE | ID: mdl-1560527

ABSTRACT

The prototype strain of minute virus of mice [MVM(p)] is an autonomous parvovirus with a tropism for cells expressing a neoplastically transformed phenotype. To generate gene transfer vectors for tumor-specific gene expression, human interleukin-2 (IL-2) and murine interleukin-4 (IL-4) genes were cloned under the control of the p38 late promoter of MVM(p). Upon transfection into permissive cells, the recombinant MVMIL2 or MVMIL4 DNA was excised, amplified, and, in the presence of a helper plasmid, packaged into recombinant viral particles. The recombinant viruses were able to transfer fully functional IL-2 and IL-4 genes to permissive target cells and retained the oncotropic host range properties of the parental virus. Following infection with MVMIL2, nontransformed fibroblasts of rodent (FR3T3) or human (MRC-5) origin produced minimal IL-2 compared with the high levels of IL-2 production observed in their transformed derivatives (FREJ4 and MRC-5V1).


Subject(s)
Cell Transformation, Viral , Genetic Vectors/genetics , Interleukins/biosynthesis , Minute Virus of Mice/genetics , Cell Line, Transformed/metabolism , DNA, Recombinant/genetics , DNA, Viral/genetics , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukins/genetics , Minute Virus of Mice/growth & development , Plasmids/genetics , Serial Passage , Virion/growth & development
7.
EMBO J ; 9(9): 2989-95, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2167840

ABSTRACT

Although its dependence on the target cell type is well established, the cytopathogenicity of parvoviruses has remained elusive to date as far as its mechanism is concerned. However, indirect evidence suggested that parvoviral non-structural (NS) proteins may be the cytotoxic effectors. In order to test this hypothesis, a molecular clone of parvovirus MVMp was modified, by replacing the P4 promoter of the NS transcription unit by the glucocorticoid-inducible promoter of the mouse mammary tumour virus. Clones of neoplastic human cells that had incorporated this construct and that were induced to produce NS proteins by dexamethasone, showed a cytopathic effect and eventually died. Our data strongly suggest that the intracellular accumulation of parvoviral NS products jeopardizes the survival of the cells, which cannot be detected unless a threshold protein concentration is reached. Interestingly, a cell variant could be isolated which resisted dexamethasone-induced killing, although it was fully inducible for the production of NS proteins. This variant was also unusually resistant to infection with MVMp virions, thus confirming the essential role played by the NS proteins in the parvoviral cytotoxicity and indicating that the cytocidal activity of the parvoviral NS products is modulated by cellular factors that may vary from one cell to another.


Subject(s)
Capsid/genetics , Cell Survival , Genes, Viral , Parvoviridae/genetics , Viral Core Proteins/genetics , Viral Structural Proteins/genetics , Capsid/metabolism , Cell Division , Cell Line , Cell Transformation, Viral , Clone Cells , Humans , Plasmids , Promoter Regions, Genetic , Restriction Mapping , Simian virus 40/genetics , Transfection , Viral Core Proteins/metabolism , Viral Nonstructural Proteins
8.
Virology ; 174(2): 576-84, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2137660

ABSTRACT

The interaction of parvovirus minute virus of mice (prototype strain, MVMp) with simian virus 40 (SV40)-transformed human cells (NB-E) was investigated by means of transfection with MVMp molecular clones derived from the infectious recombinant plasmid (pMM984). pMM984 inhibits stable transformation of NB-E cells to geneticin resistance (G418R) upon cotransfection with the selectable pSV2neo plasmid. We show here that this inhibition is not merely caused by a repression of marker gene expression from the SV40 early region promoter in pSV2neo and rather is likely to reflect the cytotoxic action of the parvovirus. Starting from plasmid pMM984, defined mutations were introduced into the genome of MVMp and more particularly into sequences coding for the NS-1 and/or NS-2 nonstructural proteins. In this way we could show that the NS-1 protein is necessary for the inhibition of transformation to G418R and that the NS-2 protein acts synergistically to enhance this effect. Moreover, results obtained with different viral mutants indicate that the inhibitory action of NS-1 on stable transformation can be dissociated from the ability of this protein both to transactivate the parvoviral p39 promoter of the capsid protein-encoding region and to drive parvoviral DNA amplification. Altogether these data point to a probable direct toxicity of MVMp nonstructural proteins for permissive host cells.


Subject(s)
Capsid/physiology , Cell Transformation, Viral , Minute Virus of Mice/pathogenicity , Parvoviridae/pathogenicity , Viral Core Proteins/physiology , Genetic Markers , Humans , Minute Virus of Mice/genetics , Promoter Regions, Genetic , Transcription, Genetic , Transfection , Viral Nonstructural Proteins
9.
J Mol Biol ; 179(3): 367-90, 1984 Nov 05.
Article in English | MEDLINE | ID: mdl-6096551

ABSTRACT

Indirect prophage induction is produced by transfer to recipients of u.v.-damaged F plasmid (95 kb). We tested whether the SOS signal can be produced by miniF, a 9.3 kb restriction fragment, coding for the replication and segregation functions of plasmid F. We used lambda miniF, a hybrid phage-plasmid. u.v.-irradiated lambda miniF induced prophages phi 80 or lambda and sfiA, a chromosomal SOS gene, in more than 50% of the infected cells. The maximal inducing dose produced about 0.5 pyrimidine dimers per kb and left 1% of lambda miniF survivors. Thus, the SOS signal produced by u.v.-damaged lambda miniF was almost as potent as that resulting from direct u.v.-irradiation of the lysogens. The u.v.-damaged vector lambda, devoid of miniF, failed to promote SOS induction. In contrast, efficient induction was observed when u.v.-damaged lambda miniF infected a lambda immune host, in which replication and expression of the phage genome were repressed. When replication and expression of the miniF genome was repressed by Hfr incompatibility, SOS induction was largely prevented. All these facts indicate that, in the hybrid lambda-miniF, it is the u.v.-damaged miniF that generates an SOS signal. To locate on the miniF genome the loci that are involved in the production of the SOS signal, we isolated deletions spanning all the miniF restriction fragments. We characterized six mutant phenotypes (Par+, Rep-, Fid-, Par-2, Par-1 and SOS-) related to four functions; partition, copy number, replication and SOS induction. A locus, we call lynA, 800 bp long, located by deletion mapping between the two origins of replication oriP and oriS is required for the production of an inducing signal. We postulate that indirect SOS induction by u.v.-damaged miniF results from the disturbance of the lynA function that may be involved in the co-segregation of F plasmid with the host chromosome.


Subject(s)
DNA Repair , F Factor , Virus Activation , Bacteriophage lambda/genetics , Bacteriophage lambda/growth & development , Chromosome Deletion , Chromosome Mapping , DNA Restriction Enzymes , F Factor/radiation effects , Mutation , Phenotype , Rec A Recombinases , Ultraviolet Rays , Virus Activation/radiation effects
10.
J Mol Biol ; 179(3): 571-6, 1984 Nov 05.
Article in English | MEDLINE | ID: mdl-6239927

ABSTRACT

We describe a novel mode of SOS induction, called gratuitous indirect induction, which is elicited when the maintenance of an intact lambda miniF introduced into a recipient was inhibited by a resident plasmid or by mutations in miniF that impaired partition or replication. Gratuitous induction required the presence of the lynA locus on miniF and was dependent on the host recA and lexA alleles. To account for gratuitous induction, we postulate that impairment of the normal co-regulation between partition and replication of miniF affects lynA functions whose disturbance leads to the production of an SOS signal.


Subject(s)
Virus Activation , Bacteriophage lambda/genetics , Bacteriophage lambda/growth & development , Chromosome Deletion , DNA Repair , F Factor , Gene Expression Regulation , Lysogeny , Mutation , Plasmids , Replicon , Ultraviolet Rays , Virus Activation/radiation effects
13.
Mol Gen Genet ; 163(3): 293-9, 1978 Jul 25.
Article in English | MEDLINE | ID: mdl-355855

ABSTRACT

The inactivation of lambda repressor was followed by the specific DNA binding assay during the course of lysogenic induction provoked by incubation at 42 degrees C of an E. coli tif-1 lysogenic strain. The presence of up to 400 microgram/ml chloramphenicol during the inducing treatment did not impair the loss of repressor binding activity, whilst concentrations of 200 microgram/ml neomycin and 100 microgram/ml rifampicin effectively inhibited the inactivation of lambda repressor. Residual protein synthesis in the presence of chloramphenicol, neomycin and rifampicin was 5%, 5% and 27% respectively of that observed in the drug-free control. This residual synthesis did not appear to involve amplification of the X-protein. These results suggest that tif-mediated inactivation of the lambda repressor requires the activation of some specific gene(s), the translation of which appears to be resistant to chloramphenicol.


Subject(s)
Chloramphenicol/pharmacology , Coliphages/genetics , Escherichia coli/genetics , Lysogeny/drug effects , Neomycin/pharmacology , Rifampin/pharmacology , Autoradiography , Bacterial Proteins/biosynthesis , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Genes, Regulator , Hot Temperature , RNA, Bacterial/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...