Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Med Genet ; 40(11): 802-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14627667

ABSTRACT

BACKGROUND: Schwannomas are benign tumours of the nervous system that are usually sporadic but also occur in the inherited disorder neurofibromatosis type 2 (NF2). The NF2 gene is a tumour suppressor on chromosome 22. Loss of expression of the NF2 protein product, merlin, is universal in both sporadic and NF2 related schwannomas. The GTPase signalling molecules RhoA and Rac1 regulate merlin function, but to date only mutation in the NF2 gene has been identified as a causal event in schwannoma formation. METHODS: Comparative genomic hybridisation (CGH) was used to screen 76 vestibular schwannomas from 76 patients (66 sporadic and 10 NF2 related) to identify other chromosome regions that may harbour genes involved in the tumorigenesis. RESULTS: The most common change was loss on chromosome 22, which was more frequent in sporadic than in NF2 related tumours. Importantly, eight tumours (10%) showed gain of copy number on chromosome 9q34. Each of the two NF2 patients who had received stereotactic radiotherapy had non-chromosome 22 changes, whereas only one of eight non-irradiated NF2 patients had any chromosome changes. Three tumours had gain on 17q, which has also been reported in malignant peripheral nerve sheath tumours that are associated with neurofibromatosis type 1. Other sites that were identified in three or fewer tumours were regions on chromosomes 10, 11, 13, 16, 19, 20, X, and Y. CONCLUSIONS: These findings should be verified using techniques that can detect smaller genetic changes, such as microarray-CGH.


Subject(s)
Chromosome Deletion , Gene Amplification/genetics , Neuroma, Acoustic/genetics , Nucleic Acid Hybridization/methods , Adolescent , Adult , Aged , Child , Chromosome Aberrations , Chromosomes, Human, Pair 9/genetics , Female , Genes, Neurofibromatosis 2 , Humans , Loss of Heterozygosity/genetics , Male , Middle Aged , Neurofibromatosis 2/genetics , Recurrence
2.
Hum Mutat ; 21(3): 313-20, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12619118

ABSTRACT

There are now reports of nearly 250 independent germline TP53 (p53) mutations in over 100 publications. Such mutations are typically associated with Li-Fraumeni or Li-Fraumeni-like syndrome, although many have been identified in cohorts of patients with tumors considered to be typical of LFS. In general, the spectrum of mutations that has been detected in the germline reflects that found in tumors, although there are some notable exceptions in certain tumor types. Detailed knowledge of the pedigrees allows a comprehensive analysis of genotype-phenotype correlations and an understanding of the tumors that are associated with germline TP53 mutations. This review will discuss the spectrum of mutations and the methods for mutation detection, the tumors associated with inheritance of a germline mutation, and some of the ethical and clinical problems in patients with a germline TP53 mutation.


Subject(s)
Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Family Health , Genotype , Humans , Phenotype
4.
Br J Cancer ; 86(12): 1920-2, 2002 Jun 17.
Article in English | MEDLINE | ID: mdl-12085187

ABSTRACT

Tumour and normal tissue from 41 male cases of Wilms' tumour were screened to determine the presence of sequence variants in the glypican 3 (GPC3) gene. Two non-conservative single base changes were present in tumour tissue only. These findings imply a possible role for GPC3 in Wilms' tumour development.


Subject(s)
Heparan Sulfate Proteoglycans/genetics , Kidney Neoplasms/genetics , Mutation/genetics , Wilms Tumor/genetics , DNA Primers/chemistry , Glypicans , Humans , Male , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
5.
Radiat Res ; 157(2): 158-65, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11835679

ABSTRACT

Knockout mice with only one Trp53 allele (+/- genotype) are highly susceptible to radiation-induced cancers, possibly through numerical chromosome changes. Patients with the Li-Fraumeni syndrome, having heterozygous TP53 germline mutations (+/mut genotype), are also susceptible to spontaneous and radiogenic cancers. We have investigated the susceptibility of six Li-Fraumeni syndrome +/mut and six normal fibroblast strains to induced numerical and unstable structural aberrations at six population doublings after exposure to 3 or 6 Gy gamma rays. Four of the irradiated Li-Fraumeni syndrome strains showed small increases in both aberration types, similar to those seen in the normal strains. In two irradiated Li-Fraumeni syndrome strains, there were high levels of induced structural changes, and one of these showed a modest increase in hyperploidy. We suggest that enhanced sensitivity to delayed radiation-induced chromosome changes in Li-Fraumeni syndrome cells requires other genetic alterations in addition to TP53 heterozygosity, apparently in contrast to the situation in Trp53 heterozygous null mice. If such additional alterations occur in vivo in Li-Fraumeni syndrome patients, they may predispose them to radiogenic cancers, mainly through enhanced structural rather than numerical chromosome changes. Our findings raise questions about the validity of quantitative extrapolation of cytogenetic data from Trp53-defective mice to radiogenic cancer risk in humans.


Subject(s)
Chromosome Breakage , Chromosomes, Human/radiation effects , Fibroblasts/radiation effects , Gamma Rays/adverse effects , Li-Fraumeni Syndrome/genetics , Aneuploidy , Animals , Cells, Cultured/radiation effects , Chromosome Aberrations , Chromosomes, Human/ultrastructure , Dose-Response Relationship, Radiation , Fibroblasts/ultrastructure , Genes, p53 , Genetic Predisposition to Disease , Genotype , Humans , Li-Fraumeni Syndrome/pathology , Loss of Heterozygosity , Mice , Mice, Knockout , Radiation Tolerance/genetics , Species Specificity , Time Factors
6.
Cancer Res ; 61(22): 8062-7, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719428

ABSTRACT

Li Fraumeni Syndrome (LFS) is a multicancer phenotype, most commonly associated with germ-line mutations in TP53. In a kindred with LFS without an inherited TP53 mutation, we have previously reported a truncating mutation (1100delC) in CHK2, encoding a kinase that phosphorylates p53 on Ser(20). Here, we describe a CHK2 missense mutation (R145W) in another LFS family. This mutation destabilizes the encoded protein, reducing its half-life from >120 min to 30 min. This effect is abrogated by treatment of cells with a proteosome inhibitor, suggesting that CHK2(R145W) is targeted through this degradation pathway. Both 1100delC and R145W germ-line mutations in CHK2 are associated with loss of the wild-type allele in the corresponding tumor specimens, and neither tumor harbors a somatic TP53 mutation. Our observations support the functional significance of CHK2 mutations in rare cases of LFS and suggest that such mutations may substitute for inactivation of TP53.


Subject(s)
Li-Fraumeni Syndrome/genetics , Mutation, Missense , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Adult , Base Sequence , Checkpoint Kinase 2 , Colonic Neoplasms/genetics , DNA, Complementary/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, p53/genetics , Humans , Li-Fraumeni Syndrome/enzymology , Loss of Heterozygosity , Male , Molecular Sequence Data , Pedigree , Protein Kinases/metabolism , Tumor Cells, Cultured
7.
Cancer Genet Cytogenet ; 129(1): 85-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520573

ABSTRACT

Many polymorphisms have been reported in the TP53 gene. Some of these are within the coding region, and may affect the function of the p53 protein, others are within introns or non-coding regions, and their significance is unclear. Recently, a number of publications have claimed that polymorphisms within intron 6 are responsible for inherited predisposition to childhood malignancies, familial breast cancer, and Li-Fraumeni syndrome (LFS). We find no evidence for intron 6 sequence variants predisposing to LFS in our cohort of families and, furthermore, we show that some of the conclusions of other groups cannot be supported by data from our analysis.


Subject(s)
Genes, p53 , Introns , Li-Fraumeni Syndrome/genetics , Genetic Predisposition to Disease , Humans , Polymorphism, Genetic
8.
Oncogene ; 20(34): 4621-8, 2001 Aug 02.
Article in English | MEDLINE | ID: mdl-11498785

ABSTRACT

The spectrum and frequency of cancers associated with germline TP53 mutations are uncertain. To address this issue a cohort of individuals from 28 families with Li-Fraumeni syndrome, segregating germline TP53 mutations was established. Predicted cancers were estimated by applying age, morphology, site and sex-specific UK cancer statistics to person-years at risk. Observed and predicted cancers were compared and two-sided P-values calculated. Cancer types occurring to excess and showing P-values <0.02, were designated strongly associated with germline TP53 mutations. These were removed from the data and a second round of analyses performed. Cancer types with P-values <0.02 and 0.02-0.05 in the second round analyses were considered moderately and weakly associated respectively. Strongly associated cancers were: breast carcinoma, soft tissue sarcomas, osteosarcoma, brain tumours, adrenocortical carcinoma, Wilms' tumour and phyllodes tumour. Carcinoma of pancreas was moderately associated. Leukaemia and neuroblastoma were weakly associated. Other common carcinomas including lung, colon, bladder, prostate, cervix and ovary did not occur to excess. Although breast carcinoma and sarcomas were numerically most frequent, the greatest increases relative to general population rates were in adrenocortical carcinoma and phyllodes tumour. We conclude that germline TP53 mutations do not simply increase general cancer risk. There are tissue-specific effects.


Subject(s)
Germ-Line Mutation , Li-Fraumeni Syndrome/epidemiology , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Family Health , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Li-Fraumeni Syndrome/pathology , Male , Middle Aged
10.
Br J Cancer ; 85(2): 293-6, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11461092

ABSTRACT

We previously showed that cultured fibroblasts from patients with the cancer-prone Li-Fraumeni (LF) syndrome, having heterozygous germline TP53 mutations, sustain less ionizing radiation-induced permanent G(1)arrest than normal fibroblasts. In contrast, fibroblast strains from LF patients without TP53 mutations showed normal G(1)arrest. We have now investigated the relationship between the extent of G(1)arrest and the level of structural chromosome damage (mainly dicentrics, rings and acentric fragments) in cells at their first mitosis after G(1)irradiation, in 9 LF strains with TP53 mutations, 6 without TP53 mutations and 7 normal strains. Average levels of damage in the mutant strains were 50% higher than in normals, whereas in non-mutant LF strains they were 100% higher. DNA double strand breaks (dsb) are known to act as a signal for p53-dependent G(1)arrest and to be the lesions from which chromosome aberrations arise. These results suggest that a minimal level of dsb is required before the signal for arrest is activated and that p53-defective cells have a higher signal threshold than p53-proficient cells. Dsb that do not cause G(1)blockage can progress to mitosis and appear as simple deletions or interact to form exchange aberrations. The elevated levels in the non-mutant strains may arise from defects in the extent or accuracy of dsb repair. In LF cells with or without TP53 mutations, the reduced capacity to eliminate or repair chromosomal damage of the type induced by ionising radiation, may contribute to cancer predisposition in this syndrome.


Subject(s)
Chromosome Aberrations , G1 Phase/radiation effects , Genes, p53 , Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Humans , Li-Fraumeni Syndrome/pathology
11.
Oncogene ; 20(21): 2647-54, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11420676

ABSTRACT

Germline TP53 splicing mutations have been described infrequently (>2%) in the literature, however in a series of 40 patients and families identified by our group in which there are germline TP53 mutations, seven affect splicing (18%). The low figure reported in the literature might reflect the method of mutation detection, which in many studies does not include all splice junctions. These data indicate that a significant proportion of TP53 germline mutations are currently unrecognized. We have carried out detailed studies of the effects of the different mutations on splicing, and see distinct variations in the effects of the same mutation in different patients. Furthermore we have identified the usage of a non-consensus splice donor site in four families with an intron 4 splice donor mutation.


Subject(s)
Alternative Splicing/physiology , Genes, p53/genetics , Germ-Line Mutation/physiology , Alternative Splicing/genetics , Cell Line , Fibroblasts/physiology , Germ-Line Mutation/genetics , Humans , Immunohistochemistry , Introns , Li-Fraumeni Syndrome/genetics , Loss of Heterozygosity , Lymphocytes/physiology , Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
12.
Br J Cancer ; 84(3): 321-8, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11161395

ABSTRACT

Mutation of human homologues of DNA mismatch repair (MMR) genes in tumours has been shown to be associated with the phenomenon of microsatellite instability (MSI). Several studies have reported the occurrence of MSI in bladder cancer, but evidence of involvement of MMR genes in the pathogenesis of this cancer is still unclear. We therefore utilized quantitative immunohistochemical (IHC) image analysis and PCR-based allelotype analysis to determine hMLH1 and hMSH2 genes alteration in a cohort of Egyptian bladder cancer samples. IHC analysis of 24 TCC and 12 SCC revealed marked- intra and intertumour heterogeneity in the levels of expression of the two MMR proteins. One TCC lost MLH1 expression and one lost MSH2, (1/24, 4%), and one SCC lost MSH2 (1/12, 8%). A large proportion of analysed tumours revealed a percentage positivity of less than 50% for MLH1 and MSH2 expression (44% and 69%, respectively). Complete loss of heterozygosity in three dinucleotide repeats lying within, or in close proximity to, hMLH1 and hMSH2 was rare (2/57, (4%) for MLH1; and 1/55, (2%) for MSH2), however allelic imbalance was detected in 11/57 (hMLH1) and 10/55 (hMSH2) at any of the informative microsatellite loci. These alterations in structure and expression of DNA MMR genes suggest their possible involvement in the tumorigenesis and/or progression of bladder cancer.


Subject(s)
DNA-Binding Proteins , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Urinary Bladder Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Alleles , Allelic Imbalance , Carrier Proteins , DNA, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Microsatellite Repeats/genetics , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/analysis , Nuclear Proteins , Polymerase Chain Reaction , Proto-Oncogene Proteins/analysis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
13.
Br J Cancer ; 83(4): 467-72, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10945493

ABSTRACT

Studies of Li-Fraumeni syndrome fibroblasts heterozygous for germline TP53 mutations have shown that loss of heterozygosity (LOH) occurs during passaging and is associated with genomic instability, such as chromosomal aberrations and aneuploidy to investigate the genomic changes associated with LOH in Li-Fraumeni (LF) fibroblasts, we have analysed cell strains at increasing population doublings (PD) using Comparative Genomic Hybridization (CGH). We have looked at three groups of cell strains: LF mutation-carrying strains which showed LOH for TP53, LF mutation-carrying strains which did not show LOH, and strains from normal individuals. Using CGH, we have detected loss of distinct chromosomal regions associated with LOH in 4 out of 5 mutation-carrying strains. In particular we have found loss of chromosomal regions containing genes involved in cell cycle control or senescence, including loss of 9p and 17p in these strains. Other recurrent changes included loss of chromosomes 4q and 6q, regions shown to contain one or more tumour suppressor genes. No genomic alterations were detected at cumulative PD in the normal strains or in the LF mutation-carrying strains which did not show LOH for TP53. We have also analysed the three groups of strains for microsatellite instability and somatic TP53 mutations, and have found genetic alterations in only one strain.


Subject(s)
Genes, p53/genetics , Li-Fraumeni Syndrome/genetics , Loss of Heterozygosity/genetics , Cells, Cultured , Chromosome Deletion , Fibroblasts/diagnostic imaging , Fibroblasts/physiology , Germ-Line Mutation/genetics , Humans , Li-Fraumeni Syndrome/pathology , Microsatellite Repeats/genetics , Nucleic Acid Hybridization , Ultrasonography
15.
Biochim Biophys Acta ; 1491(1-3): 75-92, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10760572

ABSTRACT

Gene identification studies, centred on a region of overlapping loss of heterozygosity in breast tumours within band 1p31.1, lead to the characterisation of LPHH1, a novel human 7TM gene. The coding sequence of LPHH1 extends over a 60 kb region and comprises in excess of 28 exons. Alternative splicing occurs minimally at five positions, four of which are within the coding sequence. The fifth region of alternative splicing occurs at the extreme 5' end of the transcript. A clear tissue specific bias in alternative exon selection is observed to some degree at all five positions, including the extreme 5' region, which raises the possibility of multiple and perhaps tissue specific promoters. One such putative promoter region, which appears to be utilised predominantly in breast cancer cells, has been identified. LPHH1 is highly evolutionarily conserved, with the simplest (19 exon) gene product being 95% identical between human and rat. Comparison of the alternatively spliced exons between three species, where data are available, has so far revealed 100% identity in the encoded peptide sequences, suggesting conservation of a functional aspect of this splicing. Gene expression has been observed in all tissues and cell lines tested, with the exception of lymphoblastoid and multiple myeloma lines, where there appears to be only a very low level of transcription. LPHH1 also appears to be downregulated in human bone marrow. These data are consistent with a role for the gene products in adhesion-mediated signalling. Analysis of a panel of breast tumour cell lines revealed that a number apparently overexpressed the gene whilst others showed very low levels of transcription. In one case, the overexpression correlated with a low level increase in gene copy number in the tumour line. In addition to differences in the overall levels of expression, LPHH1 mRNAs were alternatively spliced to varying degrees with shifts in the major gene product to truncated or altered forms in some lines. No somatic LPHH1 mutations were detected through sequence analysis of four primary breast tumours that showed loss of the adjacent 1p31.1 marker D1S207.


Subject(s)
Membrane Proteins/genetics , Alternative Splicing , Animals , Base Sequence , Brain/metabolism , Breast Neoplasms , Cattle , DNA Primers , Exons , Gene Expression , Gene Library , Humans , Introns , Molecular Sequence Data , Mutation , Open Reading Frames , Rats , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured
16.
Oncogene ; 18(52): 7599-601, 1999 Dec 09.
Article in English | MEDLINE | ID: mdl-10602520

ABSTRACT

A proportion of extraskeletal myxoid chondrosarcomas (EMC) have been shown to have a characteristic translocation t(9;22)(q22;q12) involving the EWS gene at 22q12 and the CHN orphan nuclear receptor gene at 9q22. This translocation appears to be largely specific for EMC, but has not been detected in all such tumours. We report here a case of EMC with a t(9;17)(q22;q11.2) as the sole chromosome abnormality. We have determined that the translocation results in the fusion of the entire coding region of CHN to the N-terminal transactivation domain of RBP56/hTAFII68. This is the first report of a translocation involving RBP56/hTAFII 68, a protein with sequence homology to both EWS and TLS/FUS. The involvement of RBP56/hTAFII68 may explain some unusual features of the tumour.


Subject(s)
Artificial Gene Fusion , Chondrosarcoma/genetics , DNA-Binding Proteins/genetics , Nerve Tissue Proteins , Nuclear Proteins/genetics , TATA-Binding Protein Associated Factors , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 9 , Humans , Molecular Sequence Data , Receptors, Steroid , Receptors, Thyroid Hormone , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic
17.
Br J Cancer ; 81(2): 300-4, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10496356

ABSTRACT

We have examined 11 cases of childhood adrenocortical tumours for copy number changes using comparative genomic hybridization (CGH). The changes seen are highly consistent between cases, and are independent of tumour type (carcinoma versus adenoma) or the presence of a germline TP53 mutation. The regions of chromosomal gain and loss identified in this study indicate the location of genes that are potentially important in the development and progression of childhood adrenocortical tumours. Finally, the copy number changes identified in childhood tumours are distinctly different to those seen in adult cases (Kjellman et al (1996) Cancer Res 56: 4219-4223), and we propose that this indicates that childhood tumours are of embryonal origin.


Subject(s)
Adenoma/genetics , Adrenal Cortex Neoplasms/genetics , Carcinoma/genetics , Chromosome Aberrations , Adolescent , Child , Child, Preschool , DNA, Neoplasm/analysis , Female , Genes, p53 , Humans , Infant , Karyotyping , Male , Mutation , Nucleic Acid Hybridization
18.
Am J Hum Genet ; 65(4): 995-1006, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10486318

ABSTRACT

We have analyzed a panel of 14 cases of childhood adrenocortical tumors unselected for family history and have identified germline TP53 mutations in >80%, making this the highest known incidence of a germline mutation in a tumor-suppressor gene in any cancer. The spectrum of germline TP53 mutations detected is remarkably limited. Analysis of tumor tissue for loss of constitutional heterozygosity, with respect to the germline mutant allele and the occurrence of other somatic TP53 mutations, indicates complex sequences of genetic events in a number of tumors. None of the families had cancer histories that conformed to the criteria for Li-Fraumeni syndrome, but, in some families, we were able to demonstrate that the mutation had been inherited. In these families there were gene carriers unaffected in their 40s and 50s, and there were others with relatively late-onset cancers. These data provide evidence that certain TP53 alleles confer relatively low penetrance for predisposition to the development of cancer, and they imply that deleterious TP53 mutations may be more frequent in the population than has been estimated previously. Our findings have considerable implications for the clinical management of children with andrenocortical tumors and their parents, in terms of both genetic testing and the early detection and treatment of tumors.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Alleles , DNA-Binding Proteins , Genes, p53/genetics , Genetic Predisposition to Disease/genetics , Penetrance , Adaptor Proteins, Signal Transducing , Adolescent , Adrenal Cortex Neoplasms/diagnosis , Adrenal Cortex Neoplasms/epidemiology , Adrenal Cortex Neoplasms/metabolism , Adult , Age of Onset , Aged , Carrier Proteins , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Testing , Germ-Line Mutation/genetics , Humans , Immunohistochemistry , Li-Fraumeni Syndrome/genetics , Loss of Heterozygosity/genetics , Male , Microsatellite Repeats/genetics , Middle Aged , Molecular Sequence Data , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/analysis , Nuclear Proteins , Polymorphism, Single-Stranded Conformational , Proto-Oncogene Proteins/analysis
19.
Br J Cancer ; 80(1-2): 9-10, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10389970

ABSTRACT

We have analysed Li-Fraumeni syndrome families, previously shown to be negative for mutations in TP53, for mutations to the tumour suppressor genes PTEN and CDKN2. These genes function in cell cycle progression or are mutated in a variety of tumours. We have detected no mutations in the family members tested.


Subject(s)
Genes, Tumor Suppressor/genetics , Genes, p16 , Li-Fraumeni Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Proteins , DNA Mutational Analysis , Genes, p53 , Humans , Mutation , PTEN Phosphohydrolase , Pedigree
20.
Br J Cancer ; 79(11-12): 1657-64, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10206274

ABSTRACT

Radiation-induced G1 arrest was studied in four classes of early passage skin fibroblasts comprising 12 normals, 12 heterozygous (mut/wt) TP53 mutation-carriers, two homozygous (mut/-) TP53 mutation-carriers and 16 strains from nine Li-Fraumeni syndrome or Li-Fraumeni-like families in which no TP53 mutation has been found, despite sequencing of all exons, exon-intron boundaries, 3' and 5' untranslated regions and promoter regions. In an assay of p53 allelic expression in yeast, cDNAs from these non-mutation strains behaved as wild-type p53. Using two different assays, we found G1 arrest was reduced in heterozygous strains with mis-sense mutations and one truncation mutation, when compared to the range established for the normal cells. Heterozygous strains with mutations at splice sites behaved like normal cells, whilst homozygous (mut/-) strains showed either extremely reduced, or no, arrest. Strains from all nine non-mutation families gave responses within the normal range. Exceptions to the previously reported inverse correlation between G1 arrest and clonogenic radiation resistance were observed, indicating that these phenotypes are not strictly interdependent.


Subject(s)
Fibroblasts/radiation effects , G1 Phase/radiation effects , Li-Fraumeni Syndrome/genetics , Alleles , Colony-Forming Units Assay , Fibroblasts/cytology , Genetic Carrier Screening , Humans , Li-Fraumeni Syndrome/pathology , Mutation , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL