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.
Leuk Res ; 37(10): 1374-82, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23806234

ABSTRACT

The CBA/H mouse model of radiation-induced acute myeloid leukaemia (rAML) has been studied for decades to bring to light the molecular mechanisms associated with multistage carcinogenesis. A specific interstitial deletion of chromosome 2 found in a high proportion of rAML is recognised as the initiating event. The deletion leads to the loss of Sfpi, a gene essential for haematopoietic development. Its product, the transcription factor PU.1 acts as a tumour suppressor in this model. Although the deletion can be detected early following ionising radiation exposure by cytogenetic techniques, precise characterisation of the haematopoietic cells carrying the deletion and the study of their fate in vivo cannot be achieved. Here, using a genetically engineered C57BL/6 mouse model expressing the GFP fluorescent molecule under the control of the Sfpi1 promoter, which we have bred onto the rAML-susceptible CBA/H strain, we demonstrate that GFP expression did not interfere with X-ray induced leukaemia incidence and that GFP fluorescence in live leukaemic cells is a surrogate marker of radiation-induced chromosome 2 deletions with or without point mutations on the remaining allele of the Sfpi1 gene. This study presents the first experimental evidence for the detection of this leukaemia initiating event in live leukemic cells.


Subject(s)
Chromosome Deletion , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Disease Models, Animal , Exons , Female , Flow Cytometry , Gene Deletion , Gene Expression , Genes, Reporter , Genetic Predisposition to Disease , Immunophenotyping , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Radiation-Induced/metabolism , Mice , Mutation , Transcription, Genetic
2.
Mutat Res ; 756(1-2): 119-26, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23665297

ABSTRACT

Exposure to ionising radiation can lead to an increased risk of cancer, particularly leukaemia. In radiation-induced acute myeloid leukaemia (rAML), a partial hemizygous deletion of mouse chromosome 2 is a common feature in several susceptible strains. The deletion is an early event detectable 24h after exposure in bone marrow cells using cytogenetic techniques. Expanding clones of bone marrow cells with chromosome 2 deletions can be detected less than a year after exposure to ionising radiation in around half of the irradiated mice. Ultimately, 15-25% of exposed animals develop AML. It is generally assumed that leukaemia originates in an early progenitor cell or haematopoietic stem cell, but it is unknown whether the original chromosome damage occurs at a similar frequency in committed progenitors and stem cells. In this study, we monitored the frequency of chromosome 2 deletions in immature bone marrow cells (Lin(-)) and haematopoietic stem cells/multipotent progenitor cells (LSK) by several techniques, fluorescent in situ hybridisation (FISH) and through use of a reporter gene model, flow cytometry and colony forming units in spleen (CFU-S) following ex vivo or in vivo exposure. We showed that partial chromosome 2 deletions are present in the LSK subpopulation, but cannot be detected in Lin(-) cells and CFU-S12 cells. Furthermore, we transplanted irradiated Lin(-) or LSK cells into host animals to determine whether specific irradiated cell populations acquire an increased proliferative advantage compared to unirradiated cells. Interestingly, the irradiated LSK subpopulation containing cells carrying chromosome 2 deletions does not appear to repopulate as well as the unirradiated population, suggesting that the chromosomal deletion does not provide an advantage for growth and in vivo repopulation, at least at early stages following occurrence.


Subject(s)
Bone Marrow/pathology , Chromosome Deletion , Chromosomes/genetics , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/genetics , Animals , Antigens, Ly/metabolism , Bone Marrow/metabolism , Cell Lineage , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Immunomagnetic Separation , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Proto-Oncogene Proteins c-kit/metabolism , X-Rays
4.
Leuk Res ; 35(1): 126-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20638124

ABSTRACT

Radiation-induced acute myeloid leukaemias (AMLs) in mice are characterised by deletions and point mutations in the Sfpi1/PU.1 transcription factor. Six AML cell lines were used to examine the impact of three previously described R235 point mutations. AML cells carry myeloid and stem cell markers and the R235 mutations differentially affect mRNA and protein abundance. Expression of Sfpi1/PU.1 target genes was deregulated in a broadly similar fashion irrespective of R235 mutation including Flt3, which is frequently subject to activating mutations in human myeloid leukaemias. While R235 mutations differentially affect protein abundance they resulted in similar disruption of Sfpi1/PU.1 functions.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/genetics , Mutation , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Trans-Activators/genetics , Transcription, Genetic/genetics , Animals , Cell Line, Tumor , Cell Lineage , Chromosomes, Artificial, Bacterial , Gene Expression Regulation, Neoplastic , Immunohistochemistry , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/pathology , Mice , Reverse Transcriptase Polymerase Chain Reaction
5.
Carcinogenesis ; 27(2): 311-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16093251

ABSTRACT

The mapping of genes which affect individual cancer risk is an important but complex challenge. A surrogate assay of susceptibility to radiation-induced acute myeloid leukaemia (AML) in the mouse based on chromosomal radiosensitivity has been developed and validated. This assay was applied to the mapping of radiation-induced AML risk modifier loci by association with microsatellite markers. A region on chromosome (chr) 18 with strong association is identified and confirmed by backcross analysis. Additional loci on chrs 8 and 13 show significant association. A key candidate gene Rbbp8 on chr18 is identified. Rbbp8 is shown to be upregulated in response to X-irradiation in the AML sensitive CBA strain but not AML resistant C57BL/6 strain. This study demonstrates the strength of utilizing surrogate endpoints of cancer susceptibility in the mapping of mouse loci and identifies additional loci that may affect radiation cancer risk.


Subject(s)
Genetic Predisposition to Disease , Leukemia, Myeloid/genetics , Leukemia, Radiation-Induced/genetics , Multigene Family , Acute Disease , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Chromosome Mapping , Endodeoxyribonucleases , Endonucleases , Endpoint Determination , Genetic Markers , Inheritance Patterns , Leukemia, Myeloid/veterinary , Leukemia, Radiation-Induced/veterinary , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Microsatellite Repeats , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Phenotype , Up-Regulation
6.
Genes Chromosomes Cancer ; 31(4): 373-81, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11433528

ABSTRACT

Previous studies have identified five lymphoma-related tumour suppressor gene regions on murine chromosome 4. Using detailed allelotype analysis on a range of lympho-haematopoietic tumour types arising in F1 hybrid mice, we now show a consistent pattern of loss of heterozygosity (LOH) which identifies a common region of loss delineated by microsatellites D4Mit21 and D4Mit53 on proximal chromosome 4. This critical segment corresponds to the thymic lymphoma tumour suppressor region 5 (TLSR5) identified in an earlier study. Tumours of this type have also been reported as showing allelic loss from the Trp53 and Ikaros regions on chromosome 11. In the present study, only a small fraction of tumours showed LOH in the Ikaros region, while a minority of lymphomas, but not acute myeloid leukaemias, showed allelic loss of the chromosome 11 segment encoding Trp53. These and other data indicate strongly that the genomic regions identified as showing recurrent LOH depend on the genetic background of the mice. Overall, the results indicate a key role for a tumour suppressor gene(s) encoded in an approximately 3 cM segment on proximal chromosome 4 and provide an experimental basis for the further investigation of the functional role of candidate genes which include Pax5 and Tgfbr1.


Subject(s)
Leukemia, Experimental/genetics , Leukemia, Radiation-Induced/genetics , Loss of Heterozygosity/genetics , Lymphoma/genetics , Animals , Chromosomes/radiation effects , Crosses, Genetic , Female , Genetic Markers/radiation effects , Loss of Heterozygosity/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
SELECTION OF CITATIONS
SEARCH DETAIL
...