Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer ; 13: 182, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-25092376

ABSTRACT

BACKGROUND: Syndromic forms of osteosarcoma (OS) account for less than 10% of all recorded cases of this malignancy. An individual OS predisposition is also possible by the inheritance of low penetrance alleles of tumor susceptibility genes, usually without evidence of a syndromic condition. Genetic variants involved in such a non-syndromic form of tumor predisposition are difficult to identify, given the low incidence of osteosarcoma cases and the genetic heterogeneity of patients. We recently mapped a major OS susceptibility QTL to mouse chromosome 14 by comparing alpha-radiation induced osteosarcoma in mouse strains which differ in their tumor susceptibility. METHODS: Tumor-specific allelic losses in murine osteosacoma were mapped along chromosome 14 using microsatellite markers and SNP allelotyping. Candidate gene search in the mapped interval was refined using PosMed data mining and mRNA expression analysis in normal osteoblasts. A strain-specific promoter variant in Rb1 was tested for its influence on mRNA expression using reporter assay. RESULTS: A common Rb1 allele derived from the BALB/cHeNhg strain was identified as the major determinant of radiation-induced OS risk at this locus. Increased OS-risk is linked with a hexanucleotide deletion in the promoter region which is predicted to change WT1 and SP1 transcription factor-binding sites. Both in-vitro reporter and in-vivo expression assays confirmed an approx. 1.5 fold reduced gene expression by this promoter variant. Concordantly, the 50% reduction in Rb1 expression in mice bearing a conditional hemizygous Rb1 deletion causes a significant rise of OS incidence following alpha-irradiation. CONCLUSION: This is the first experimental demonstration of a functional and genetic link between reduced Rb1 expression from a common promoter variant and increased tumor risk after radiation exposure. We propose that a reduced Rb1 expression by common variants in regulatory regions can modify the risk for a malignant transformation of bone cells after radiation exposure.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Osteosarcoma/genetics , Osteosarcoma/pathology , Promoter Regions, Genetic , Radiation , Retinoblastoma Protein/genetics , 3' Untranslated Regions/genetics , Allelic Imbalance , Animals , Base Sequence , Binding Sites , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Chromosomes, Mammalian/genetics , Crosses, Genetic , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Hybridization, Genetic , INDEL Mutation/genetics , Male , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors
2.
Radiat Environ Biophys ; 50(1): 135-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21063720

ABSTRACT

Osteosarcoma is the most frequent secondary malignancy following radiotherapy of patients with bilateral retinoblastoma. This suggests that the Rb1 tumour suppressor gene might confer genetic susceptibility towards radiation-induced osteosarcoma. To define the contribution of the Rb1 pathway in the multistep process of radiation carcinogenesis, we evaluated somatic allelic changes affecting the Rb1 gene itself as well as its upstream regulator p16 in murine osteosarcoma induced by (227)Th incorporation. To distinguish between the contribution of germline predisposition and the effect of a 2-hit allelic loss, two mouse models harbouring heterozygote germline Rb1 and p16 defects were tested for the incidence and latency of osteosarcoma following irradiation. We could show that all tumours arising in BALB/c×CBA/CA hybrid mice (wild-type for Rb1 and for p16) carried a somatic allelic loss of either the Rb1 gene (76.5%) or the p16 gene (59%). In none of the tumours, we found concordant retention of heterozygosity at both loci. Heterozygote knock-out mice for Rb1 exhibit a significant increase in the incidence of osteosarcoma following (227)Th incorporation (11/24 [corrected] in Rb1+/- vs. 2/18 in Rb1+/+, p=4×10(-5)), without affecting tumour latency. In contrast, heterozygote knock-out mice for p16 had no significant change in tumour incidence, but a pronounced reduction of latency (LT(50%) =355 days in p16+/- vs. 445 days in p16+/+, p=8×10(-3)). These data suggest that Rb1 germline defects influence early steps of radiation osteosarcomagenesis, whereas alterations in p16 mainly affect later stages of tumour promotion and growth.


Subject(s)
Alpha Particles/adverse effects , Bone Neoplasms/genetics , Neoplasms, Radiation-Induced/genetics , Osteosarcoma/genetics , Retinoblastoma Protein/genetics , Signal Transduction/genetics , Signal Transduction/radiation effects , Allelic Imbalance/radiation effects , Animals , Bone Neoplasms/etiology , Bone Neoplasms/metabolism , Bone and Bones/radiation effects , Female , Genes, p16 , Genetic Predisposition to Disease , Germ-Line Mutation/radiation effects , Mice , Osteosarcoma/etiology , Osteosarcoma/metabolism , Retinoblastoma Protein/metabolism , Thorium/metabolism , Time Factors
3.
Mutat Res ; 597(1-2): 1-4, 2006 May 11.
Article in English | MEDLINE | ID: mdl-16414091

ABSTRACT

The radiation-induced bystander effect challenges the accepted paradigm of direct DNA damage in response to energy deposition driving the biological consequences of radiation exposure. With the bystander response, cells which have not been directly exposed to radiation respond to their neighbours being targeted. In our own studies we have used novel targeted microbeam approaches to specifically irradiate parts of individual cells within a population to quantify the bystander response and obtain mechanistic information. Using this approach it has become clear that energy deposited by radiation in nuclear DNA is not required to trigger the effect, with cytoplasmic irradiation required. Irradiated cells also trigger a bystander response regardless of whether they themselves live or die, suggesting that the phenotype of the targeted cell is not a determining factor. Despite this however, a range of evidence has shown that repair status is important for dealing with the consequences of a bystander signal. Importantly, repair processes involved in the processing of dsb appear to be involved suggesting that the bystander response involves the delayed or indirect production of dsb-type lesions in bystander cells. Whether these are infact true dsb or complexes of oxidised bases in combination with strand breaks and the mechanisms for their formation, remains to be elucidated.


Subject(s)
DNA Damage , DNA Repair , DNA/radiation effects , Animals , Cell Nucleus/radiation effects , Cytoplasm/radiation effects , DNA/genetics , DNA/metabolism , Humans , Mutation , Radiation Tolerance
4.
Int J Cancer ; 118(9): 2132-8, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16331598

ABSTRACT

In a recent study, we presented evidence for genetic predisposition governing radiation osteosarcomagenesis in mice. Following the incorporation of the bone-seeking alpha emitter 227Th, approximately 25% of the variance in osteosarcoma incidence was determined by inherited genetic factors. We have now mapped 5 susceptibility loci in crosses between the more susceptible BALB/c and the more resistant CBA/Ca strains. The major QTL on chromosome 14 overlaps with a locus that was already found in our previous study, using different strains of mice. Here, we investigate the effect by which the major susceptibility locus and 4 minor modifier loci interact to influence osteosarcoma predisposition. Following incorporation of the bone-seeking isotope, 100% of mice that harbour high-risk genotypes at all 5 susceptibility loci develop osteosarcoma with an average of 472 days latency times. In 10 mice inheriting exclusively low-risk genotypes only 1 osteosarcoma was found, arising after 733 days latency time. Inheritance of distinct combinations of BALB/c and CBA/Ca alleles at the susceptibility loci confer more extreme phenotypes in terms of susceptibility or resistance than observed in either of the two parental inbred strains. From the present study, we demonstrate that additive effects of multiple alleles, each making only a minor phenotypic contribution, can combine and significantly alter tumour risk. This mechanism can be of particular importance in genetically heterogeneous populations such as man.


Subject(s)
Alpha Particles/adverse effects , Bone Neoplasms/etiology , Genetic Predisposition to Disease , Osteosarcoma/etiology , Animals , Bone Neoplasms/genetics , Chromosome Mapping , Female , Genotype , Humans , Inheritance Patterns , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Osteosarcoma/genetics , Phenotype , Quantitative Trait Loci , Risk Factors
5.
J Radiol Prot ; 22(3A): A113-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12400958

ABSTRACT

Cancer is a disease of the genome, with the neoplastic phenotype being passed from one cell generation to the other. Radiation-induced cancer has often been considered to represent a unique entity amongst neoplasia, with the energy deposition being held responsible for both direct (gene mutations) and indirect (bystander effects, induced instability etc) alterations to the cellular genome. However, radiogenic tumours in man and experimental animals appear to be physiologically and genetically indistinguishable from their sporadic counterparts, suggesting that the aetiologies of these two tumour types are in fact closely related. We have conducted a general screen of the genetic alterations in radiation-induced mouse osteosarcoma, a tumour that is histopathologically indistinguishable from human sporadic osteosarcoma. Comparison of the two tumour types indicates the existence of a common set of genetic changes, providing additional evidence to support the concept that the molecular pathology of radiation-induced malignancy is no different to that of sporadic cancers.


Subject(s)
Bone Neoplasms/etiology , Bone Neoplasms/genetics , Neoplasms, Radiation-Induced/genetics , Osteosarcoma/etiology , Osteosarcoma/genetics , Allelic Imbalance/radiation effects , Animals , Humans , Mice , Mutation/radiation effects
6.
Oncogene ; 21(38): 5975-80, 2002 Aug 29.
Article in English | MEDLINE | ID: mdl-12185601

ABSTRACT

We have performed a comparative study of allelic imbalances in human and murine osteosarcomas to identify genetic changes critical for osteosarcomagenesis. Two adjacent but discrete loci on mouse chromosome 9 were found to show high levels of allelic imbalance in radiation-induced osteosarcomas arising in (BALB/cxCBA/CA) F1 hybrid mice. The syntenic human chromosomal regions were investigated in 42 sporadic human osteosarcomas. For the distal locus (OSS1) on mouse chromosome 9 the syntenic human locus was identified on chromosome 6q14 and showed allelic imbalance in 77% of the cases. Comparison between the human and mouse syntenic regions narrowed the locus down to a 4 Mbp fragment flanked by the marker genes ME1 and SCL35A1. For the proximal locus (OSS2) on mouse chromosome 9, a candidate human locus was mapped to chromosome 15q21 in a region showing allelic imbalance in 58% of human osteosarcomas. We have used a combination of synteny and microsatellite mapping to identify two potential osteosarcoma suppressor gene loci. This strategy represents a powerful tool for the identification of new genes important for the formation of human tumors.


Subject(s)
Bone Neoplasms/genetics , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 6 , Genes, Tumor Suppressor , Osteosarcoma/genetics , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Microsatellite Repeats , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...