Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Klin Onkol ; 33(1): 32-38, 2020.
Article in English | MEDLINE | ID: mdl-32075387

ABSTRACT

BACKGROUND: Signal transducer and activator of transcription (STAT) proteins are cytoplasmic transcription factors that transmit the signal of cytokines, hormones and growth factors. STAT proteins control fundamental cellular processes including survival, proliferation and differentiation. Inappropriate activation of STATs might contribute to cellular transformation and leukaemogenesis. About 70% of all solid and haematological tumours exhibit aberrant STAT3 expression and/or activation, highlighting its essential role in tumourigenesis. Aberrant STAT3 activation has been found in several solid tumours and haematologic malignancies. Importantly, constitutive activation of STAT proteins has been found in several leukaemias including acute myeloid leukaemia, acute promyelocytic leukaemia, acute lymphoblastic leukaemia, chronic myeloid leukaemia and chronic lymphocytic leukaemia (CLL). Constitutively activated STAT3 plays an important role in CLL biology. CLL cells harbour constitutive phosphorylation on S727 and acetylation on K685 and transient phosphorylation on Y705 residues. Moreover, STAT3 messenger RNA expression is significantly higher in CLL cells compared to healthy B-lymphocytes. Interestingly, STAT3 inhibition was disclosed as an important by-product of ibrutinib treatment in CLL patients. PURPOSE: The purpose of this review is to describe the consequences of STAT3 dysregulation in CLL cells. Here, we discuss aberrantly modified processes by STAT3 activation in CLL cells such as proliferation, apoptosis, B cell receptor signalling, cytokine secretion, immune checkpoint regulation, microRNA regulation, free fatty acid metabolism and electron transport chain in the mitochondria.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , STAT3 Transcription Factor/metabolism , Adenine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use
2.
Cell Death Dis ; 6: e1852, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26247737

ABSTRACT

The ATM-p53 DNA-damage response (DDR) pathway has a crucial role in chemoresistance in CLL, as indicated by the adverse prognostic impact of genetic aberrations of TP53 and ATM. Identifying and distinguishing TP53 and ATM functional defects has become relevant as epigenetic and posttranscriptional dysregulation of the ATM/p53 axis is increasingly being recognized as the underlying cause of chemoresistance. Also, specific treatments sensitizing TP53- or ATM-deficient CLL cells are emerging. We therefore developed a new ATM-p53 functional assay with the aim to (i) identify and (ii) distinguish abnormalities of TP53 versus ATM and (iii) enable the identification of additional defects in the ATM-p53 pathway. Reversed transcriptase multiplex ligation-dependent probe amplification (RT-MLPA) was used to measure ATM and/or p53-dependent genes at the RNA level following DNA damage using irradiation. Here, we showed that this assay is able to identify and distinguish three subgroups of CLL tumors (i.e., TP53-defective, ATM-defective and WT) and is also able to detect additional samples with a defective DDR, without molecular aberrations in TP53 and/or ATM. These findings make the ATM-p53 RT-MLPA functional assay a promising prognostic tool for predicting treatment responses in CLL.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Gene Expression Regulation, Leukemic , Multiplex Polymerase Chain Reaction/methods , Mutation , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Biological Assay , DNA Damage , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Gamma Rays , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , RNA, Neoplasm/genetics , Sensitivity and Specificity , Tumor Suppressor Protein p53/metabolism , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
3.
Leukemia ; 29(5): 1133-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25371178

ABSTRACT

Mutations or deletions in TP53 or ATM are well-known determinants of poor prognosis in chronic lymphocytic leukemia (CLL), but only account for approximately 40% of chemo-resistant patients. Genome-wide sequencing has uncovered novel mutations in the splicing factor sf3b1, that were in part associated with ATM aberrations, suggesting functional synergy. We first performed detailed genetic analyses in a CLL cohort (n=110) containing ATM, SF3B1 and TP53 gene defects. Next, we applied a newly developed multiplex assay for p53/ATM target gene induction and measured apoptotic responses to DNA damage. Interestingly, SF3B1 mutated samples without concurrent ATM and TP53 aberrations (sole SF3B1) displayed partially defective ATM/p53 transcriptional and apoptotic responses to various DNA-damaging regimens. In contrast, NOTCH1 or K/N-RAS mutated CLL displayed normal responses in p53/ATM target gene induction and apoptosis. In sole SF3B1 mutated cases, ATM kinase function remained intact, and γH2AX formation, a marker for DNA damage, was increased at baseline and upon irradiation. Our data demonstrate that single mutations in sf3b1 are associated with increased DNA damage and/or an aberrant response to DNA damage. Together, our observations may offer an explanation for the poor prognosis associated with SF3B1 mutations.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Cohort Studies , DNA Damage , DNA Mutational Analysis , Doxorubicin/pharmacology , Flow Cytometry , Gene Deletion , Genome, Human , Histones/metabolism , Humans , Imidazoles/pharmacology , Piperazines/pharmacology , Prognosis , RNA Splicing Factors , Receptor, Notch1/genetics , Tumor Suppressor Protein p53/genetics , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
4.
Leukemia ; 29(4): 877-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25287991

ABSTRACT

In chronic lymphocytic leukemia (CLL), the worst prognosis is associated with TP53 defects with the affected patients being potentially directed to alternative treatment. Therapy administration was shown to drive the selection of new TP53 mutations in CLL. Using ultra-deep next-generation sequencing (NGS), we performed a detailed analysis of TP53 mutations' clonal evolution. We retrospectively analyzed samples that were assessed as TP53-wild-type (wt) by FASAY from 20 patients with a new TP53 mutation detected in relapse and 40 patients remaining TP53-wt in relapse. Minor TP53-mutated subclones were disclosed in 18/20 patients experiencing later mutation selection, while only one minor-clone mutation was observed in those patients remaining TP53-wt (n=40). We documented that (i) minor TP53 mutations may be present before therapy and may occur in any relapse; (ii) the majority of TP53-mutated minor clones expand to dominant clone under the selective pressure of chemotherapy, while persistence of minor-clone mutations is rare; (iii) multiple minor-clone TP53 mutations are common and may simultaneously expand. In conclusion, patients with minor-clone TP53 mutations carry a high risk of mutation selection by therapy. Deep sequencing can shift TP53 mutation identification to a period before therapy administration, which might be of particular importance for clinical trials.


Subject(s)
B-Lymphocytes/metabolism , Clonal Evolution/genetics , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Clonal Evolution/drug effects , Clone Cells , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Mutation , Recurrence , Retrospective Studies , Signal Transduction , Survival Analysis , Tumor Suppressor Protein p53/metabolism
5.
Leukemia ; 26(7): 1458-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22297721

ABSTRACT

Recent evidence suggests that - in addition to 17p deletion - TP53 mutation is an independent prognostic factor in chronic lymphocytic leukemia (CLL). Data from retrospective analyses and prospective clinical trials show that ∼5% of untreated CLL patients with treatment indication have a TP53 mutation in the absence of 17p deletion. These patients have a poor response and reduced progression-free survival and overall survival with standard treatment approaches. These data suggest that TP53 mutation testing warrants integration into current diagnostic work up of patients with CLL. There are a number of assays to detect TP53 mutations, which have respective advantages and shortcomings. Direct Sanger sequencing of exons 4-9 can be recommended as a suitable test to identify TP53 mutations for centers with limited experience with alternative screening methods. Recommendations are provided on standard operating procedures, quality control, reporting and interpretation. Patients with treatment indications should be investigated for TP53 mutations in addition to the work-up recommended by the International workshop on CLL guidelines. Patients with TP53 mutation may be considered for allogeneic stem cell transplantation in first remission. Alemtuzumab-based regimens can yield a substantial proportion of complete responses, although of short duration. Ideally, patients should be treated within clinical trials exploring new therapeutic agents.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/genetics , Practice Guidelines as Topic , Tumor Suppressor Protein p53/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Prognosis
6.
Leukemia ; 24(12): 2072-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861914

ABSTRACT

The TP53 mutation profile in chronic lymphocytic leukemia (CLL) and the correlation of TP53 mutations with allele status or associated molecular genetics are currently unknown. We performed a large mutation analysis of TP53 at four centers and characterized the pattern of TP53 mutations in CLL. We report on 268 mutations in 254 patients with CLL. Missense mutations appeared in 74% of cases compared with deletions and insertions (20%), nonsense (4%) and splice site (2%) mutations. The majority (243 of 268) of mutations were located in the DNA-binding domain. Transitions were found in 131 of 268 mutations, with only 41 occurring at methylated CpG sites (15%), suggesting that transitions at CpGs are uncommon. The codons most frequently mutated were at positions 175, 179, 248 and 273; in addition, we detected a common 2-nt deletion in the codon 209. Most mutations (199 of 259) were accompanied by deletion of the other allele (17p-). Interestingly, trisomy 12 (without 17p-) was only found in one of 60 cases with TP53 mutation (without 17p-) compared with 60 of 16 in the cohort without mutation (P=0.006). The mutational profile was not different in the cohorts with and without previous therapy, suggesting that the mechanism underlying the development of mutations may be similar, independent of treatment.


Subject(s)
Genes, p53 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Chromatography, High Pressure Liquid , CpG Islands , Humans
8.
Mol Immunol ; 45(5): 1525-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17920683

ABSTRACT

Abnormalities of the TP53 gene are associated with a particularly severe prognosis in patients with B-cell chronic lymphocytic leukemia (B-CLL). This tumor-suppressor is mostly inactivated by the deletion of one and point mutation of the other allele and has not been previously shown to be hypermutated in B-CLL. We identified two patients whose lymphocytes showed repeatedly an extensive proportion of TP53 mutated cells by FASAY analysis (the yeast functional assay) and harbored various TP53 mutations, mostly single-base substitutions, in individual cells. The mutation targeting exhibited characteristic traits of the somatic hypermutation process. In the first patient (harboring the unmutated IgVH locus) a significant bias to point mutations at CG pairs (21/25; P=0.009), their remarkable preference for the RGYW/WRCY motives (28%) and the highest expression of the activation-induced cytidine deaminase (AID) mRNA among the 34 tested B-CLL samples. In the second patient no CG bias was observed but the targeting of point mutations into the RGYW/WRCY motives was even more prominent here (7/16; 44%). Moreover, six out of eight point mutations affecting AT pairs were localized in the WA/TW motives, which are also characteristic for the somatic hypermutations. This patient, who was IgVH-mutated, already did not express any significant amount of the AID transcript. Our findings add a new aspect to the mosaic of the p53 mutability in B-CLL.


Subject(s)
Genes, p53 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Cytidine Deaminase/genetics , Humans , Lymphocytes/pathology , Point Mutation , Somatic Hypermutation, Immunoglobulin
9.
Cas Lek Cesk ; 145(11): 855-8; discussion 859-60, 2006.
Article in Czech | MEDLINE | ID: mdl-17168419

ABSTRACT

BACKGROUND: Chronic lymphocytic leukemia is a heterogeneous disease manifesting with a variable clinical course. It is evident from many studies, that the division into two main prognostic categories is possible on the basis of mutation status of the immunoglobulin heavy-chain gene. The objective of our work was to identify a presence or absence of IgVH gene mutations in B-CLL patients which are monitored or treated on hematological clinics and to determine the presence of individual D and J, subgenes in malignant population of B-cells. METHODS AND RESULTS: A nucleotide sequence of IgVH gene of neoplastic cells was analyzed by appropriate molecular-genetic methods. RNA/cDNA was collected from 358 patients and a spectrum of individual subgenes translocations was identified. Our results show that 56.3% of patients manifested an unmutated variable (VH) segment. It is expected from the published data that this group of patients will suffer from aggressive course of the disease and will exhibit a substantially shorter survival in comparison to patients possessing somatic hypermutations. An expanded population of leukemic B-cells showed increased occurrence of clones whose variable segments belong to three different families. VH3 alleles are the ones most frequently used. A frequency of unmutated alleles is prominently shifted into families with V I homology. The preferred "diversity and joining" segments are D3, D2 and JH 4 and JH 6. CONCLUSIONS: The analysis of heavy chain immunoglobulin gene after recombinant VH-D-J11 segments translocation belongs to a standard hematooncological investigation. The results are an important prognostic criterion for prediction of expected disease aggressivity and for a minimal residual disease monitoring.


Subject(s)
Base Sequence , Genes, Immunoglobulin Heavy Chain/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Humans , Immunoglobulin Variable Region , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Prognosis , Translocation, Genetic
11.
J Inherit Metab Dis ; 27(4): 523-8, 2004.
Article in English | MEDLINE | ID: mdl-15303010

ABSTRACT

Low-density lipoprotein receptor (LDLR) is a cell-surface glycoprotein that mediates specific uptake and catabolism of plasma LDL. Mutations located in the coding region of the LDLR gene affect the structure and function of the protein and cause familial hypercholesterolaemia (FH). Mutations in the regulatory regions of the gene are rare, but in some cases have been shown to alter the transcriptional activity of the gene and cause the FH phenotype as well. Adult heterozygous FH individuals have a markedly raised plasma cholesterol that is associated with accelerated atherosclerosis and premature coronary heart disease. The aim of this study was the functional characterization of a promoter mutation in the LDLR gene in one family from the register of Czech FH subjects. Molecular screening revealed that three members of this family carried a -27C > T nucleotide transition in the promoter sequence (calculated from the start of transcription). All three manifested a heterozygous FH phenotype. This new mutation is located between the TATA box and sterol-dependent regulatory element repeat 3. Using a luciferase reporter assay system, we analysed the transcriptional efficiency of the normal and mutant alleles. The mutation reduced promoter activity to background level. Another new promoter mutation -60C > T was identified in an unrelated patient in the conserved nucleotide sequence of the sterol-dependent regulation element repeat 2 which virtually abolished the promoter activity. We assume a causal effect of this -60C > T transition on the basis of its position in the promoter sequence.


Subject(s)
Hyperlipoproteinemia Type II/genetics , Mutation , Promoter Regions, Genetic , Receptors, LDL/genetics , Adolescent , Base Sequence , Cell Line , Child , Czech Republic , Female , Heterozygote , Humans , Liver , Luciferases/genetics , Male , Middle Aged , Phenotype , TATA Box , Transfection
12.
Cas Lek Cesk ; 143(2): 84-9, 2004.
Article in Czech | MEDLINE | ID: mdl-15077568

ABSTRACT

In spite of the fact that many papers dealing with the chronic lymphocytic leukemia include a sentence in Introduction, that the molecular pathology of the disease "is still largely unknown", the amount of accumulated information is impressive and enables to create the first models of the overall genesis of this "most frequent leukemia in the Western world". Since many studies have confirmed that B-CLL lymphocytes in peripheral blood are anchored in G0/G1-phase of the cell cycle, the recent general opinion is, that CLL is primarily caused by defects in apoptosis--lymphocytes are slowly accumulating, being not able to "die properly". However, it becomes evident, that in the microenvironment appropriate for the cell growth, i.e. in the bone marrow and lymph nodes, B-CLL lymphocytes proliferate and they are subsequently accumulated in peripheral blood. This review summarizes namely the knowledge about status and expression of key genes regulating apoptosis and cell cycle in B-CLL lymphocytes, including p53, ATM, MDM2, Bcl-2/Bax, caspase-3, CDK-inhibitor p27, cyclins D2 and D3. Relationship between some of these genes and the standard therapy is discussed and prospective therapeutic alternatives resulting from the new molecular-genetic findings are presented.


Subject(s)
Apoptosis , Cell Cycle , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Genes, Tumor Suppressor , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy
13.
Cell Mol Life Sci ; 58(9): 1333-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11577989

ABSTRACT

Activation of the p53 tumour suppressor protein by distinct forms of stress leads to inhibition of cellular proliferation by inducing cell cycle arrest or apoptosis. The cyclin-dependent kinase inhibitor roscovitine has been shown to induce nuclear accumulation of wild-type p53 in human untransformed and tumour-derived cells. We analyzed the response of different human tumour cell lines to roscovitine treatment with respect to their p53 status. Striking induction of wild-type p53 protein and dramatic enhancement of p53-dependent transcription, coinciding with p21WAF1 induction, was observed in wildtype, but not mutant, p53-bearing tumour cells after treatment with roscovitine. The transcriptional activity of p53 was substantially higher in roscovitine-treated cells than in cells irradiated with ultraviolet C or ionizing radiation, even though all these agents induced a similar amount of p53 accumulation. These results highlight the therapeutic potential of roscovitine as an anticancer drug, especially in tumours retaining a functional wild-type p53 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Purines/pharmacology , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Female , Fibroblasts , Humans , Kinetics , Melanoma , Mice , Osteosarcoma , Recombinant Proteins/metabolism , Roscovitine , Transfection , Tumor Cells, Cultured
14.
Hum Genet ; 109(1): 117-20, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11479743

ABSTRACT

Galactosemia is a metabolic disorder caused by a defect in the galactose-1-phosphate uridyltransferase (GALT) enzyme. In previous studies, we have shown that the presence of a deletion in the 5' upstream (promoter) region of the GALT gene is associated with the Duarte (D2) allele. In the present study, by using a promoter fusion assay we provide direct evidence that a GTCA deletion located in position -119/-116 of the GALT gene (considered in relation to the translational start site) decreases transcription of a reporter gene to about 55% compared with a normal "healthy" promoter transfected into human hepatocyte HepG2 cells. This result coincides well with previously published biochemical data showing 50% GALT-gene activity in Duarte (D2) galactosemia patients. By transfecting the same promoters (normal and deleted) into mouse NIH/3T3 cells, we show that the GTCA motif in the promoter region of the GALT gene was conserved throughout evolution. We conclude that the -119/-116delGTCA promoter mutation is a crucial factor in reduction of Duarte allele enzyme activity.


Subject(s)
Galactosemias/enzymology , Galactosemias/genetics , Promoter Regions, Genetic , Sequence Deletion , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics , 3T3 Cells , Alleles , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Genes, Reporter , Humans , Luciferases/genetics , Mice , Transfection
16.
Tumour Biol ; 22(2): 59-66, 2001.
Article in English | MEDLINE | ID: mdl-11125276

ABSTRACT

Most p53 mutations occur in the central part of the p53 gene that codes for the DNA-binding domain. Missense mutations are prevalent. However, 10-25% of all mutations occur outside exons 5-8 and include a prevalence of frameshift, nonsense and splice site mutations. Functional analysis of p53 transactivation ability in yeast (FASAY) was used to screen for p53 mutations in tumors and a mutant p53 protein retaining partial activity was identified. We characterized this somatic p53 mutation in codon 337: transition C-->T, changing codon CGC to TGC and causing substitution of arginine for cysteine in exon 10, which codes for the tetramerization domain of p53. We detected high accumulation of this mutant p53 protein within the tumor tissue and found that it cannot be immunoprecipitated by either a wild-type p53-specific antibody (PAb1620) or by a mutant p53-specific antibody (PAb240). We confirmed the somatic origin of the mutation by analysis of p53 status in peripheral leukocytes.


Subject(s)
Breast Neoplasms/genetics , Genes, p53 , Mutation , Alleles , Codon , Female , Humans , Immunoblotting , Immunohistochemistry , Leukocytes/metabolism , Middle Aged , Plasmids/metabolism , Polymerase Chain Reaction , Precipitin Tests , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Yeasts/genetics
17.
Folia Biol (Praha) ; 43(1): 19-24, 1997.
Article in English | MEDLINE | ID: mdl-9158945

ABSTRACT

The genotoxic effects of N-nitroso-N-methylurea (MNU) and acetone oxime (ACOX) were tested in the Somatic Mutation and Recombination Test (SMART) in Drosophila melanogaster. We have performed the same assay on transgenic flies expressing the human gene encoding a glutathione S-transferase alpha subunit (HGST). The SMART assay is used here to demonstrate genotoxicity and to determine the effect of human glutathione S-transferase on the genotoxic response. Three types of Drosophila strains were used: non-transgenic strains first described by Szabad (1986), transgenic strains derived from the Szabad strains but expressing the bacterial lacZ gene, and similarly derived transgenic strains expressing the HGST gene. MNU was highly genotoxic in both transgenic and non-transgenic flies. The non-transgenic lies were significantly more sensitive to the genotoxic effects of MNU compared to both types of transgenic flies. There were statistically significant differences between the transgenic HGST crosses and transgenic lacZ and non-transgenic control crosses but there was no significant difference between the genotoxic response to MNU in flies from the transgenic cross with lacZ and from the cross carrying three copies of HGST. ACOX also proved to be genotoxic to both non-transgenic and transgenic flies. However, flies carrying three copies of the gene were significantly more resistant to the genotoxic effect of ACOX than those transgenic flies with two or no copies of the human gene.


Subject(s)
Drosophila melanogaster/genetics , Glutathione Transferase/biosynthesis , Methylnitrosourea/toxicity , Mutagens/toxicity , Oximes/toxicity , Animals , Animals, Genetically Modified , Crosses, Genetic , Female , Glutathione Transferase/genetics , Humans , Larva , Macromolecular Substances , Male , Mutagenesis , Recombinant Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...