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










Publication year range
1.
iScience ; 26(9): 107530, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37664628

ABSTRACT

Ionizing radiation (IR) is a risk factor for acute myeloid leukemia (rAML). Murine rAMLs feature both hemizygous chromosome 2 deletions (Del2) and point mutations (R235) within the hematopoietic regulatory gene Spi1. We generated a heterozygous CBA Spi1 R235 mouse (CBASpm/+) which develops de novo AML with 100% incidence by ∼12 months old and shows a dose-dependent reduction in latency following X-irradiation. These effects are reduced on an AML-resistant C57Bl6 genetic background. CBASpm/Gfp reporter mice show increased Gfp expression, indicating compensation for Spm-induced Spi1 haploinsufficiency. Del2 is always detected in both de novo and rAMLs, indicating that biallelic Spi1 mutation is required for AML. CBASpm/+ mice show that a single Spm modification is sufficient for initiating AML development with complete penetrance, via the "two-hit" mechanism and this is accelerated by IR exposure. Similar SPI1/PU.1 polymorphisms in humans could potentially lead to enhanced susceptibility to IR following medical or environmental exposure.

2.
Antioxidants (Basel) ; 11(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35052515

ABSTRACT

Haematopoietic bone marrow cells are amongst the most sensitive to ionizing radiation (IR), initially resulting in cell death or genotoxicity that may later lead to leukaemia development, most frequently Acute Myeloid Leukaemia (AML). The target cells for radiation-induced Acute Myeloid Leukaemia (rAML) are believed to lie in the haematopoietic stem and progenitor cell (HSPC) compartment. Using the inbred strain CBA/Ca as a murine model of rAML, progress has been made in understanding the underlying mechanisms, characterisation of target cell population and responses to IR. Complex regulatory systems maintain haematopoietic homeostasis which may act to modulate the risk of rAML. However, little is currently known about the role of metabolic factors and diet in these regulatory systems and modification of the risk of AML development. This study characterises cellular proliferative and clonogenic potential as well as metabolic changes within murine HSPCs under oxidative stress and X-ray exposure. Ambient oxygen (normoxia; 20.8% O2) levels were found to increase irradiated HSPC-stress, stimulating proliferative activity compared to low oxygen (3% O2) levels. IR exposure has a negative influence on the proliferative capability of HSPCs in a dose-dependent manner (0-2 Gy) and this is more pronounced under a normoxic state. One Gy x-irradiated HSPCs cultured under normoxic conditions displayed a significant increase in oxygen consumption compared to those cultured under low O2 conditions and to unirradiated HSPCs. Furthermore, mitochondrial analyses revealed a significant increase in mitochondrial DNA (mtDNA) content, mitochondrial mass and membrane potential in a dose-dependent manner under normoxic conditions. Our results demonstrate that both IR and normoxia act as stressors for HSPCs, leading to significant metabolic deregulation and mitochondrial dysfunctionality which may affect long term risks such as leukaemia.

3.
Radiat Environ Biophys ; 60(1): 49-60, 2021 03.
Article in English | MEDLINE | ID: mdl-33221961

ABSTRACT

The effect of low-dose ionizing radiation exposure on leukemia incidence remains poorly understood. Possible dose-response curves for various forms of leukemia are largely based on cohorts of atomic bomb survivors. Animal studies can contribute to an improved understanding of radiation-induced acute myeloid leukemia (rAML) in humans. In male CBA/H mice, incidence of rAML can be described by a two-hit model involving a radiation-induced deletion with Sfpi1 gene copy loss and a point mutation in the remaining Sfpi1 allele. In the present study (historical) mouse data were used and these processes were translated into a mathematical model to study photon-induced low-dose AML incidence in male CBA/H mice following acute exposure. Numerical model solutions for low-dose rAML incidence and diagnosis times could respectively be approximated with a model linear-quadratic in radiation dose and a normal cumulative distribution function. Interestingly, the low-dose incidence was found to be proportional to the modeled number of cells carrying the Sfpi1 deletion present per mouse following exposure. After making only model-derived high-dose rAML estimates available to extrapolate from, the linear-quadratic model could be used to approximate low-dose rAML incidence calculated with our mouse model. The accuracy in estimating low-dose rAML incidence when extrapolating from a linear model using a low-dose effectiveness factor was found to depend on whether a data transformation was used in the curve fitting procedure.


Subject(s)
Disease Models, Animal , Leukemia, Myeloid, Acute , Leukemia, Radiation-Induced , Animals , Dose-Response Relationship, Radiation , Male , Mice, Inbred CBA , Photons , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
4.
Carcinogenesis ; 41(8): 1104-1112, 2020 08 12.
Article in English | MEDLINE | ID: mdl-31646336

ABSTRACT

Therapy-related and more specifically radiotherapy-associated acute myeloid leukaemia (AML) is a well-recognized potential complication of cytotoxic therapy for the treatment of a primary cancer. The CBA mouse model is used to study radiation leukaemogenesis mechanisms with Sfpi1/PU.1 deletion and point mutation already identified as driving events during AML development. To identify new pathways, we analysed 123 mouse radiation-induced AML (rAML) samples for the presence of mutations identified previously in human AML and found three genes to be mutated; Sfpi1 R235 (68%), Flt3-ITD (4%) and Kras G12 (3%), of which G12R was previously unreported. Importantly, a significant decrease in Sfpi1 gene expression is found almost exclusively in rAML samples without an Sfpi1 R235 mutation and is specifically associated with up-regulation of mir-1983 and mir-582-5p. Moreover, this down-regulation of Sfpi1 mRNA is negatively correlated with DNA methylation levels at specific CpG sites upstream of the Sfpi1 transcriptional start site. The down regulation of Sfpi1/PU.1 has also been reported in human AML cases revealing one common pathway of myeloid disruption between mouse and human AML where dysregulation of Sfpi1/PU.1 is a necessary step in AML development.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Experimental/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Carcinogenesis , DNA Methylation/genetics , Down-Regulation , Humans , Mice , Mice, Inbred CBA , MicroRNAs/genetics , Mutation , Promoter Regions, Genetic , fms-Like Tyrosine Kinase 3
5.
Int J Radiat Biol ; 95(4): 452-479, 2019 04.
Article in English | MEDLINE | ID: mdl-29932783

ABSTRACT

PURPOSE: The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS: Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION: The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.


Subject(s)
Diet , Hematopoietic Stem Cells/radiation effects , Leukemia, Radiation-Induced/etiology , Animals , Antioxidants , Autophagy , Cell Differentiation , Epigenesis, Genetic , Gastrointestinal Microbiome , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Mice , Reactive Oxygen Species/metabolism
6.
Leukemia ; 32(6): 1435-1444, 2018 06.
Article in English | MEDLINE | ID: mdl-29556020

ABSTRACT

Epidemiological studies have demonstrated an increased leukemia incidence following ionizing radiation exposure, but to date, the target cells and underlying mechanisms of radiation leukemogenesis remain largely unidentified. We engineered a mouse model carrying a different fluorescent marker on each chromosome 2, located inside the minimum deleted region occurring after radiation exposure and recognized as the first leukemogenic event. Using this tailored model, we report that following radiation exposure, more than half of asymptomatic CBA Sfpi1 GFP/mCh mice presented with expanding clones of preleukemic hematopoietic cells harboring a hemizygous interstitial deletion of chromosome 2. Moreover, following isolation of preleukemic hematopoietic stem and progenitor cells irradiated in their native microenvironment, we identified the presence of Sfpi1 point mutations within a subpopulation of these preleukemic cells expanding rapidly (increasing from 6% to 55% in 21 days in peripheral blood in one case), hence identifying for the first time the presence of such cells within a living animal. Importantly, we also report a previously undescribed gender difference in the phenotype of the preleukemic cells and leukemia, suggesting a gender imbalance in the radiation-induced leukemic target cell. In conclusion, we provide novel insights into the sequence of molecular events occurring during the (radiation-induced) leukemic clonal evolution.


Subject(s)
Leukemia, Radiation-Induced/etiology , Preleukemia/etiology , Animals , Clonal Evolution , Disease Progression , Female , Hematopoietic Stem Cells/radiation effects , Male , Mice , Mice, Inbred CBA , Point Mutation , Proto-Oncogene Proteins/genetics , Sex Characteristics , Trans-Activators/genetics
7.
Cell Mol Life Sci ; 74(23): 4339-4351, 2017 12.
Article in English | MEDLINE | ID: mdl-28667356

ABSTRACT

While the biological effects of high-dose-ionizing radiation on human health are well characterized, the consequences of low-dose radiation exposure remain poorly defined, even though they are of major importance for radiological protection. Lymphocytes are very radiosensitive, and radiation-induced health effects may result from immune cell loss and/or immune system impairment. To decipher the mechanisms of effects of low doses, we analyzed the modulation of the T-cell receptor gene repertoire in mice exposed to a single low (0.1 Gy) or high (1 Gy) dose of radiation. High-throughput T-cell receptor gene profiling was used to visualize T-lymphocyte dynamics over time in control and irradiated mice. Radiation exposure induces "aging-like" effects on the T-cell receptor gene repertoire, detectable as early as 1 month post-exposure and for at least 6 months. Surprisingly, these effects are more pronounced in animals exposed to 0.1 Gy than to 1 Gy, where partial correction occurs over time. Importantly, we found that low-dose radiation effects are partially due to the hematopoietic stem cell impairment. Collectively, our findings show that acute low-dose radiation exposure specifically results in long-term alterations of the T-lymphocyte repertoire.


Subject(s)
Gene Expression/radiation effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/radiation effects , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/radiation effects , Animals , Cellular Senescence/radiation effects , Dose-Response Relationship, Radiation , Gamma Rays , Gene Expression/immunology , Graft Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Male , Mice , Mice, Inbred CBA , Mice, Inbred NOD , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transplantation Chimera , Transplantation, Homologous , V(D)J Recombination/immunology
8.
Int J Mol Sci ; 17(11)2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27827995

ABSTRACT

Scid hematopoietic stem cells (HSCs) have an intrinsic defect in their maintenance within the bone marrow (BM) niche which facilitates HSC transplantation without the absolute requirement of prior conditioning. Nevertheless, NOD scid mice have a significantly altered life span due to early development of thymic lymphomas, which compromises the ability to study the long-term fate of exogenous HSCs and their progeny. Here, we present data on the transplantation of HSCs into NOD scid gamma (NSG) mice to achieve long-term engraftment without prior conditioning. We transplanted allogeneic HSCs constitutively expressing the mCherry fluorescent marker into age-matched NSG mice and assessed donor chimerism 6 months post-transplantation. All transplanted NSG mice showed long-term myeloid and lymphoid cell chimerism. Also, in vivo irradiated HSCs showed long-term engraftment, although overall white blood cell (WBC) donor chimerism was lower compared with non-irradiated HSCs. Using this novel NSG transplantation model, we will be able to study the effects of low dose in vivo X-ray exposure on the long-term fate of HSCs, without the requirement of prior radio-ablation of the recipient, and thus leaving the recipient's BM microenvironment uncompromised. In conclusion, we demonstrated for the first time that allogeneic HSCs from a different inbred strain can compete for niches in the BM compartment of NSG mice.


Subject(s)
Bone Marrow/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Stem Cell Niche/radiation effects , Transplantation Chimera/immunology , Animals , Bone Marrow/pathology , Bone Marrow/radiation effects , Dose-Response Relationship, Radiation , Gene Expression , Genes, Reporter , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/radiation effects , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Stem Cell Niche/immunology , Transplantation, Homologous , X-Rays , Red Fluorescent Protein
9.
Radiat Res ; 186(6): 638-649, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27869555

ABSTRACT

Exposure to ionizing radiation increases the incidence of acute myeloid leukemia (AML), which has been diagnosed in Japanese atomic bombing survivors, as well as patients treated with radiotherapy. The genetic basis for susceptibility to radiation-induced AML is not well characterized. We previously identified a candidate murine gene for susceptibility to radiation-induced AML (rAML): C-terminal binding protein (CTBP)-interacting protein (CTIP)/retinoblastoma binding protein 8 (RBBP8). This gene is essential for embryonic development, double-strand break (DSB) resection in homologous recombination (HR) and tumor suppression. In the 129S2/SvHsd mouse strain, a nonsynonymous single nucleotide polymorphism (nsSNP) in Ctip, Q418P, has been identified. We investigated the role of Q418P in radiation-induced carcinogenesis and its effect on CTIP function in HR. After whole-body exposure to 3 Gy of X rays, 11 out of 113 (9.7%) 129S2/SvHsd mice developed rAML. Furthermore, 129S2/SvHsd mouse embryonic fibroblasts (MEFs) showed lower levels of recruitment of HR factors, Rad51 and replication protein A (RPA) to radiation-induced foci, compared to CBA/H and C57BL/6 MEFs, isolated from rAML-sensitive and resistant strains, respectively. Mitomycin C and alpha particles induced lower levels of sister chromatid exchanges in 129S2/SvHsd cells compared to CBA/H and C57BL/6. Our data demonstrate that Q418P nsSNP influences the efficiency of CTIP function in HR repair of DNA DSBs in vitro and in vivo, and appears to affect susceptibility to rAML.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/radiation effects , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Homologous Recombination/radiation effects , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Polymorphism, Single Nucleotide/radiation effects , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded/radiation effects , Genetic Predisposition to Disease , Mice
10.
Oncotarget ; 7(26): 40461-40480, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27250028

ABSTRACT

A combined transcriptome and proteome analysis of mouse radiation-induced AMLs using two primary AMLs, cell lines from these primaries, another cell line and its in vivo passage is reported. Compared to haematopoietic progenitor and stem cells (HPSC), over 5000 transcriptome alterations were identified, 2600 present in all materials. 55 and 3 alterations were detected in the proteomes of the cell lines and primary/in vivo passage material respectively, with one common to all materials. In cell lines, approximately 50% of the transcriptome changes are related to adaptation to cell culture, and in the proteome this proportion was higher. An AML 'signature' of 17 genes/proteins commonly deregulated in primary AMLs and cell lines compared to HPSCs was identified and validated using human AML transcriptome data. This also distinguishes primary AMLs from cell lines and includes proteins such as Coronin 1, pontin/RUVBL1 and Myeloperoxidase commonly implicated in human AML. C-Myc was identified as having a key role in radiation leukaemogenesis. These data identify novel candidates relevant to mouse radiation AML pathogenesis, and confirm that pathways of leukaemogenesis in the mouse and human share substantial commonality.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasms, Radiation-Induced/metabolism , Proteome , Transcriptome , ATPases Associated with Diverse Cellular Activities/metabolism , Algorithms , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , DNA Helicases/metabolism , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Microfilament Proteins/metabolism , Neoplasms, Radiation-Induced/pathology , Peroxidase/metabolism , Signal Transduction
11.
Article in English | MEDLINE | ID: mdl-26520372

ABSTRACT

Leukaemia is the prevailing neoplastic disorder of the hematopoietic system. Epidemiological analyses of the survivors of the Japanese atomic bombings show that exposure to ionising radiation (IR) can cause leukaemia. Although a clear association between radiation exposure and leukaemia development is acknowledged, the underlying mechanisms remain incompletely understood. A hemizygous deletion on mouse chromosome 2 (del2) is a common feature in several mouse strains susceptible to radiation-induced acute myeloid leukaemia (rAML). The deletion is an early event detectable 24h after exposure in bone marrow cells. Ultimately, 15-25% of exposed animals develop AML with 80-90% of cases carrying del2. Molecular mapping of leukaemic cell genomes identified a minimal deleted region (MDR) on chromosome 2 (chr2) in which a tumour suppressor gene, Sfpi1 is located, encoding the transcription factor PU.1, essential in haematopoiesis. The remaining copy of Sfpi1 has a point mutation in the coding sequence for the DNA-binding domain of the protein in 70% of rAML, which alters a single CpG sequence in the codon for arginine residue R235. In order to identify chr2 deletions and Sfpi.1/PU.1 loss, we performed array comparative genomic hybridization (aCGH) on a unique panel of 79rAMLs. Using a custom made CGH array specifically designed for mouse chr2, we analysed at unprecedentedly high resolution (1.4M array- 148bp resolution) the size of the MDR in low LET and high-LET induced rAMLs (32 X-ray- and 47 neutron-induced). Sequencing of Sfpi1/PU.1DNA binding domain identified the presence of R235 point mutations, showing no influence of radiation quality on R235 type or frequency. We identified for the first time rAML cases with complex del2 in a subset of neutron-induced AMLs. This study allowed us to re-define the MDR to a much smaller 5.5Mb region (still including Sfpi1/PU.1), identical regardless of radiation quality.


Subject(s)
Chromosome Deletion , Chromosomes, Mammalian/radiation effects , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/veterinary , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Chromosomes, Mammalian/genetics , Comparative Genomic Hybridization , Female , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/veterinary , Leukemia, Radiation-Induced/genetics , Male , Mice , Neutrons , Point Mutation , X-Rays
12.
Radiat Res ; 171(4): 474-83, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19397448

ABSTRACT

Since deletion of the PU.1 gene on chromosome 2 is a crucial acute myeloid leukemia (AML) initiating step in the mouse model, we quantified PU.1 deleted cells in the bone marrow of gamma-, X- and 56Fe-ion-irradiated mice at various times postirradiation. Although 56Fe ions were initially some two to three times more effective than X or gamma rays in inducing PU.1 deletions, by 1 month postirradiation, the proportions of cells with PU.1 deletions were similar for the HZE particles and the sparsely ionizing radiations. These results indicate that while 56Fe ions are more effective in inducing PU.1 deletions, they are also more effective in causing collateral damage that removes hit cells from the bone marrow. After X, gamma or 56Fe-ion irradiation, AML-resistant C57BL/6 mice have fewer cells with PU.1 deletions than CBA mice, and those cells do not persist in the bone marrow of the C57B6/6 mice. Our findings suggest that quantification of PU.1 deleted bone marrow cells 1 month postirradiation can be used as surrogate for the incidence of radiation-induced AML measured in large-scale mouse studies. If so, PU.1 loss could be used to systematically assess the potential leukemogenic effects of other ions and energies in the space radiation environment.


Subject(s)
Gene Expression Regulation, Leukemic , Iron , Leukemia/etiology , Leukemia/metabolism , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Chromosomes , Chromosomes, Artificial, Bacterial/metabolism , Dose-Response Relationship, Radiation , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , X-Rays
13.
Mol Carcinog ; 34(2): 64-71, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12112312

ABSTRACT

Cytogenetic and molecular studies have provided evidence of the clustering of chromosome 2 deletion breakpoints in radiation-induced murine acute myeloid leukemia (AML). Moreover, clustering occurs in at least two fragile domains rich in telomere-like arrays. Here we describe a physical map of the distal breakpoint cluster and confirm the presence of inverted head-to-head telomeric sequence arrays. These potentially recombinogenic sequences were not, however, the direct focus for post-irradiation chromosome breakage in AML. Instead, the two arrays bordered a 2.5-kb sequence with properties expected of a nuclear matrix attachment region (MAR). The putative MAR co-localized in the fragile domain with genes important to the hemopoietic system (leukocyte tyrosine kinase, zinc finger protein 106, erythrocyte protein band 4.2, and beta(2)-microglobulin (beta2m)); the beta2m subdomain was a particular focus of breakage. On the basis of these and other data, we suggest that AML-associated chromosome 2 fragility in the mouse is a consequence of domain-specific fragility in genomic domains containing numerous genes critical to the hemopoietic system. Recorded with the permission of the controller of Her Majesty's Stationery Office. Published by Wiley-Liss, Inc.


Subject(s)
Leukemia, Myeloid/genetics , Neoplasms, Radiation-Induced/genetics , Acute Disease , Animals , Base Sequence , Chromosomes, Artificial, Bacterial , Chromosomes, Artificial, Yeast , Cloning, Molecular , DNA, Neoplasm , Leukemia, Myeloid/etiology , Mice , Molecular Sequence Data , Repetitive Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...