Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
PLoS Genet ; 10(6): e1004299, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24901438

ABSTRACT

Homologous recombination (HR) is critical for the repair of double strand breaks and broken replication forks. Although HR is mostly error free, inherent or environmental conditions that either suppress or induce HR cause genomic instability. Despite its importance in carcinogenesis, due to limitations in our ability to detect HR in vivo, little is known about HR in mammalian tissues. Here, we describe a mouse model in which a direct repeat HR substrate is targeted to the ubiquitously expressed Rosa26 locus. In the Rosa26 Direct Repeat-GFP (RaDR-GFP) mice, HR between two truncated EGFP expression cassettes can yield a fluorescent signal. In-house image analysis software provides a rapid method for quantifying recombination events within intact tissues, and the frequency of recombinant cells can be evaluated by flow cytometry. A comparison among 11 tissues shows that the frequency of recombinant cells varies by more than two orders of magnitude among tissues, wherein HR in the brain is the lowest. Additionally, de novo recombination events accumulate with age in the colon, showing that this mouse model can be used to study the impact of chronic exposures on genomic stability. Exposure to N-methyl-N-nitrosourea, an alkylating agent similar to the cancer chemotherapeutic temozolomide, shows that the colon, liver and pancreas are susceptible to DNA damage-induced HR. Finally, histological analysis of the underlying cell types reveals that pancreatic acinar cells and liver hepatocytes undergo HR and also that HR can be specifically detected in colonic somatic stem cells. Taken together, the RaDR-GFP mouse model provides new understanding of how tissue and age impact susceptibility to HR, and enables future studies of genetic, environmental and physiological factors that modulate HR in mammals.


Subject(s)
Aging , DNA Repair/genetics , Green Fluorescent Proteins/genetics , Homologous Recombination/genetics , RNA, Untranslated/genetics , Age Factors , Animals , Bacterial Proteins/genetics , Brain/cytology , Colon/cytology , DNA Breaks, Double-Stranded , Genomic Instability/genetics , Liver/cytology , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreas/cytology
2.
Sci Rep ; 8(1): 12108, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108260

ABSTRACT

Homologous recombination (HR) events are key drivers of cancer-promoting mutations, and the ability to visualize these events in situ provides important information regarding mutant cell type, location, and clonal expansion. We have previously created the Rosa26 Direct Repeat (RaDR) mouse model wherein HR at an integrated substrate gives rise to a fluorescent cell. To fully leverage this in situ approach, we need better ways to quantify rare fluorescent cells deep within tissues. Here, we present a robust, automated event quantification algorithm that uses image intensity and gradient features to detect fluorescent cells in deep tissue specimens. To analyze the performance of our algorithm, we simulate fluorescence behavior in tissue using Monte Carlo methods. Importantly, this approach reduces the potential for bias in manual counting and enables quantification of samples with highly dense HR events. Using this approach, we measured the relative frequency of HR within a chromosome and between chromosomes and found that HR within a chromosome is more frequent, which is consistent with the close proximity of sister chromatids. Our approach is both objective and highly rapid, providing a powerful tool, not only to researchers interested in HR, but also to many other researchers who are similarly using fluorescence as a marker for understanding mammalian biology in tissues.


Subject(s)
Chromosomes, Mammalian/genetics , Image Processing, Computer-Assisted/methods , Models, Genetic , Molecular Imaging/methods , Optical Imaging/methods , Animals , Carcinogenesis/genetics , Chromatids/genetics , Chromatids/metabolism , Chromosomes, Mammalian/metabolism , Computer Simulation , Fluorescence , Genes, Reporter/genetics , Homologous Recombination , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Monte Carlo Method , Mutation , Neoplasms/diagnostic imaging , Neoplasms/genetics , Neoplasms/pathology , Pancreas/diagnostic imaging , Pancreas/pathology , Repetitive Sequences, Nucleic Acid/genetics , Support Vector Machine
3.
DNA Repair (Amst) ; 10(12): 1294-9, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-21993421

ABSTRACT

The tumor suppressor p53 is a transcription factor whose function is critical for maintaining genomic stability in mammalian cells. In response to DNA damage, p53 initiates a signaling cascade that results in cell cycle arrest, DNA repair or, if the damage is severe, programmed cell death. In addition, p53 interacts with repair proteins involved in homologous recombination. Mitotic homologous recombination (HR) plays an essential role in the repair of double-strand breaks (DSBs) and broken replication forks. Loss of function of either p53 or HR leads to an increased risk of cancer. Given the importance of both p53 and HR in maintaining genomic integrity, we analyzed the effect of p53 on HR in vivo using Fluorescent Yellow Direct Repeat (FYDR) mice as well as with the sister chromatid exchange (SCE) assay. FYDR mice carry a direct repeat substrate in which an HR event can yield a fluorescent phenotype. Here, we show that p53 status does not significantly affect spontaneous HR in adult pancreatic cells in vivo or in primary fibroblasts in vitro when assessed using the FYDR substrate and SCEs. In addition, primary fibroblasts from p53 null mice do not show increased susceptibility to DNA damage-induced HR when challenged with mitomycin C. Taken together, the FYDR assay and SCE analysis indicate that, for some tissues and cell types, p53 status does not greatly impact HR.


Subject(s)
Homologous Recombination/genetics , Sequence Deletion , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Animals , Fibroblasts/metabolism , Mice , Pancreas/cytology , Pancreas/metabolism , Sister Chromatid Exchange/genetics
SELECTION OF CITATIONS
SEARCH DETAIL