Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Cell ; 62(4): 558-71, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27132940

ABSTRACT

Histone H2B monoubiquitination (H2Bub1) is centrally involved in gene regulation. The deubiquitination module (DUBm) of the SAGA complex is a major regulator of global H2Bub1 levels, and components of this DUBm are linked to both neurodegenerative diseases and cancer. Unexpectedly, we find that ablation of USP22, the enzymatic center of the DUBm, leads to a reduction, rather than an increase, in global H2bub1 levels. In contrast, depletion of non-enzymatic components, ATXN7L3 or ENY2, results in increased H2Bub1. These observations led us to discover two H2Bub1 DUBs, USP27X and USP51, which function independently of SAGA and compete with USP22 for ATXN7L3 and ENY2 for activity. Like USP22, USP51 and USP27X are required for normal cell proliferation, and their depletion suppresses tumor growth. Our results reveal that ATXN7L3 and ENY2 orchestrate activities of multiple deubiquitinating enzymes and that imbalances in these activities likely potentiate human diseases including cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Deubiquitinating Enzymes/metabolism , Histones/metabolism , Transcription Factors/metabolism , Tumor Burden , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genotype , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phenotype , RNA Interference , Signal Transduction , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Time Factors , Transcription Factors/genetics , Transfection , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
2.
Development ; 146(4)2019 02 22.
Article in English | MEDLINE | ID: mdl-30718289

ABSTRACT

USP22, a component of the SAGA complex, is overexpressed in highly aggressive cancers, but the normal functions of this deubiquitinase are not well defined. We determined that loss of USP22 in mice results in embryonic lethality due to defects in extra-embryonic placental tissues and failure to establish proper vascular interactions with the maternal circulatory system. These phenotypes arise from abnormal gene expression patterns that reflect defective kinase signaling, including TGFß and several receptor tyrosine kinase pathways. USP22 deletion in endothelial cells and pericytes that are induced from embryonic stem cells also hinders these signaling cascades, with detrimental effects on cell survival and differentiation as well as on the ability to form vessels. Our findings provide new insights into the functions of USP22 during development that may offer clues to its role in disease states.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation, Developmental , Placenta/metabolism , Signal Transduction , Animals , Cardiovascular System/metabolism , Cell Differentiation , Cell Survival , Chorioallantoic Membrane/metabolism , Ear, Inner/embryology , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Female , Gene Expression , Gene Expression Profiling , Mice , Phenotype , Pregnancy , Protein Processing, Post-Translational , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquitin Thiolesterase
3.
Mol Cell ; 35(3): 352-64, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683498

ABSTRACT

Histone acetyltransferases (HATs) play important roles in gene regulation and DNA repair by influencing the accessibility of chromatin to transcription factors and repair proteins. Here, we show that deletion of Gcn5 leads to telomere dysfunction in mouse and human cells. Biochemical studies reveal that depletion of Gcn5 or ubiquitin-specific protease 22 (Usp22), which is another bona fide component of the Gcn5-containing SAGA complex, increases ubiquitination and turnover of TRF1, a primary component of the telomeric shelterin complex. Inhibition of the proteasome or overexpression of USP22 opposes this effect. The USP22 deubiquitinating module requires association with SAGA complexes for activity, and we find that depletion of Gcn5 compromises this association in mammalian cells. Thus, our results indicate that Gcn5 regulates TRF1 levels through effects on Usp22 activity and SAGA integrity.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Thiolester Hydrolases/metabolism , p300-CBP Transcription Factors/physiology , Animals , Cells, Cultured , Chromosome Aberrations , DNA Breaks, Double-Stranded , DNA Repair/genetics , Gene Deletion , Humans , Mice , Models, Biological , Proteasome Inhibitors , Protein Stability , Shelterin Complex , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 1/genetics , Thiolester Hydrolases/genetics , Ubiquitin Thiolesterase , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
4.
Life Sci Alliance ; 7(3)2024 03.
Article in English | MEDLINE | ID: mdl-38182161

ABSTRACT

Neurodevelopmental disorders with intellectual disability (ND/ID) are a heterogeneous group of diseases driving lifelong deficits in cognition and behavior with no definitive cure. X-linked intellectual disability disorder 105 (XLID105, #300984; OMIM) is a ND/ID driven by hemizygous variants in the USP27X gene encoding a protein deubiquitylase with a role in cell proliferation and neural development. Currently, only four genetically diagnosed individuals from two unrelated families have been described with limited clinical data. Furthermore, the mechanisms underlying the disorder are unknown. Here, we report 10 new XLID105 individuals from nine families and determine the impact of gene variants on USP27X protein function. Using a combination of clinical genetics, bioinformatics, biochemical, and cell biology approaches, we determined that XLID105 variants alter USP27X protein biology via distinct mechanisms including changes in developmentally relevant protein-protein interactions and deubiquitylating activity. Our data better define the phenotypic spectrum of XLID105 and suggest that XLID105 is driven by USP27X functional disruption. Understanding the pathogenic mechanisms of XLID105 variants will provide molecular insight into USP27X biology and may create the potential for therapy development.


Subject(s)
Intellectual Disability , Mental Retardation, X-Linked , Humans , Cell Proliferation , Computational Biology , Intellectual Disability/genetics , Neurogenesis , Mental Retardation, X-Linked/genetics
5.
EMBO Rep ; 12(9): 924-30, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21779003

ABSTRACT

Ubiquitin-specific protease 22 (USP22) edits the histone code by deubiquitinating H2A and H2B as part of the mammalian SAGA (Spt-Ada-Gcn5) complex, and is required for transcriptional regulation and normal cell-cycle progression. Here, we show that USP22 affects the expression of p21 by altering far upstream element (FUSE)-binding protein 1 (FBP1) ubiquitination, as ablation of USP22 leads to increased FBP1 ubiquitination and decreased FBP1 protein occupancy at the p21 gene. Surprisingly, increased polyubiquitination of FBP1 does not alter its protein stability, but instead modulates the stable recruitment of FBP1 to target loci. Our results indicate a mechanism by which USP22 regulates cell proliferation and tumorigenesis.


Subject(s)
Cell Proliferation , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Thiolester Hydrolases/metabolism , Ubiquitination , Cell Cycle , Cell Line , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Protein Processing, Post-Translational , RNA-Binding Proteins/metabolism , Thiolester Hydrolases/genetics , Transcription, Genetic , Ubiquitin Thiolesterase
6.
Cell Chem Biol ; 29(4): 541-543, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35452621

ABSTRACT

In this issue of Cell Chemical Biology, Morgan et al. (2021) show that cyclic peptides can be potent and highly specific inhibitors for deubiquitinating enzymes. This study identifies the first selective inhibitors of the cancer-associated ubiquitin-specific protease 22 (USP22).


Subject(s)
Peptides, Cyclic , Peptides, Cyclic/pharmacology
7.
Mol Cancer Res ; 20(12): 1751-1762, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36001804

ABSTRACT

Cyclin D1 (CCND1) is a critical regulator of cell proliferation and its overexpression has been linked to the development and progression of several malignancies. CCND1 overexpression is recognized as a major mechanism of therapy resistance in several cancers; tumors that rely on CCND1 overexpression to evade cancer therapy are extremely sensitive to its ablation. Therefore, targeting CCND1 is a promising strategy for preventing tumor progression and combating therapy resistance in cancer patients. Although CCND1 itself is not a druggable target, it can be targeted indirectly by inhibiting its regulators. CCND1 steady-state levels are tightly regulated by ubiquitin-mediated degradation, and defects in CCND1 ubiquitination are associated with increased CCND1 protein levels in cancer. Here, we uncover a novel function of ubiquitin-specific protease 27X (USP27X), a deubiquitinating enzyme (DUB), in regulating CCND1 degradation in cancer. USP27X binds to and stabilizes CCND1 in a catalytically dependent manner by negatively regulating its ubiquitination. USP27X expression levels correlate with the levels of CCND1 in several HER2 therapy-resistant breast cancer cell lines, and its ablation leads to a severe reduction of CCND1 protein levels, inhibition of tumor growth, and resensitization to targeted therapy. Together, the results presented in our study are the first to expose USP27X as a major CCND1 deubiquitinase and provide a mechanistic explanation for how this DUB fosters tumor growth. IMPLICATIONS: As a deubiquitinating enzyme, USP27X is a druggable target. Our study illuminates new avenues for therapeutic intervention in CCND1-driven cancers.


Subject(s)
Breast Neoplasms , Cyclin D1 , Humans , Female , Cyclin D1/genetics , Cyclin D1/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Proteolysis , Cell Line, Tumor
8.
Cancers (Basel) ; 13(17)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34503086

ABSTRACT

Usp22 overexpression is observed in several human cancers and is correlated with poor patient outcomes. The molecular basis underlying this correlation is not clear. Usp22 is the catalytic subunit of the deubiquitylation module in the SAGA histone-modifying complex, which regulates gene transcription. Our previous work demonstrated that the loss of Usp22 in mice leads to decreased expression of several components of receptor tyrosine kinase and TGFß signaling pathways. To determine whether these pathways are upregulated when Usp22 is overexpressed, we created a mouse model that expresses high levels of Usp22 in all tissues. Phenotypic characterization of these mice revealed over-branching of the mammary glands in females. Transcriptomic analyses indicate the upregulation of key pathways involved in mammary gland branching in mammary epithelial cells derived from the Usp22-overexpressing mice, including estrogen receptor, ERK/MAPK, and TGFß signaling. However, Usp22 overexpression did not lead to increased tumorigenesis in any tissue. Our findings indicate that elevated levels of Usp22 are not sufficient to induce tumors, but it may enhance signaling abnormalities associated with oncogenesis.

9.
Cells ; 9(7)2020 07 16.
Article in English | MEDLINE | ID: mdl-32708614

ABSTRACT

Eukaryotic cells are constantly exposed to both endogenous and exogenous stressors that promote the induction of DNA damage. Of this damage, double strand breaks (DSBs) are the most lethal and must be efficiently repaired in order to maintain genomic integrity. Repair of DSBs occurs primarily through one of two major pathways: non-homologous end joining (NHEJ) or homologous recombination (HR). The choice between these pathways is in part regulated by histone post-translational modifications (PTMs) including ubiquitination. Ubiquitinated histones not only influence transcription and chromatin architecture at sites neighboring DSBs but serve as critical recruitment platforms for repair machinery as well. The reversal of these modifications by deubiquitinating enzymes (DUBs) is increasingly being recognized in a number of cellular processes including DSB repair. In this context, DUBs ensure proper levels of ubiquitin, regulate recruitment of downstream effectors, dictate repair pathway choice, and facilitate appropriate termination of the repair response. This review outlines the current understanding of histone ubiquitination in response to DSBs, followed by a comprehensive overview of the DUBs that catalyze the removal of these marks.


Subject(s)
DNA Breaks, Double-Stranded , Histones/metabolism , Ubiquitination , Animals , DNA Repair/genetics , Deubiquitinating Enzymes/metabolism , Gene Expression Regulation , Humans
10.
Cancer Res ; 79(1): 33-46, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30341066

ABSTRACT

In cancer cells, epithelial-to-mesenchymal transition (EMT) is controlled by Snail1, a transcriptional factor also required for the activation of cancer-associated fibroblasts (CAF). Snail1 is short-lived in normal epithelial cells as a consequence of its coordinated and continuous ubiquitination by several F-box-specific E3 ligases, but its degradation is prevented in cancer cells and in activated fibroblasts. Here, we performed an siRNA screen and identified USP27X as a deubiquitinase that increases Snail1 stability. Expression of USP27X in breast and pancreatic cancer cell lines and tumors positively correlated with Snail1 expression levels. Accordingly, downregulation of USP27X decreased Snail1 protein in several tumor cell lines. USP27X depletion impaired Snail1-dependent cell migration and invasion and metastasis formation and increased cellular sensitivity to cisplatin. USP27X was upregulated by TGFß during EMT and was required for TGFß-induced expression of Snail1 and other mesenchymal markers in epithelial cells and CAF. In agreement with this, depletion of USP27X prevented TGFß-induced EMT and fibroblast activation. Collectively, these results indicate that USP27X is an essential protein controlling Snail1 expression and function and may serve as a target for inhibition of Snail1-dependent tumoral invasion and chemoresistance. SIGNIFICANCE: These findings show that inhibition of USP27X destabilizes Snail1 to impair EMT and renders tumor cells sensitive to chemotherapy, thus opening new strategies for the inhibition of Snail1 expression and its protumoral actions.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/1/33/F1.large.jpg.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Drug Resistance, Neoplasm , Snail Family Transcription Factors/chemistry , Transforming Growth Factor beta/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Protein Stability , RNA, Small Interfering/genetics , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
11.
Free Radic Biol Med ; 42(4): 530-40, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17275685

ABSTRACT

The establishment of oxidants as mediators of signal transduction has renewed the interest of investigators in oxidant production and metabolism. In particular, H(2)O(2) has been demonstrated to play pivotal roles in mediating cell differentiation, proliferation, and death. Intracellular concentrations of H(2)O(2) are modulated by its rate of production and its rate of decomposition by catalase and peroxidases. In inflammation and infection, some of the H(2)O(2) is converted to hypochlorous acid, a key mediator of the host immune response against pathogens. In vivo HOCl production is mediated by myeloperoxidase, which uses excess H(2)O(2) to oxidize Cl(-). Mashino and Fridovich (Biochim. Biophys. Acta 956:63-69; 1988) observed that a high excess of HOCl over catalase inactivated the enzyme by mechanisms that remain unclear. The potential relevance of this as an alternative mechanism for catalase activity control and its potential impact on H(2)O(2)-mediated signaling and HOCl production compelled us to explore in depth the HOCl-mediated catalase inactivation pathways. Here, we demonstrate that HOCl induces formation of catalase protein radicals and carbonyls, which are temporally correlated with catalase aggregation. Hypochlorite-induced catalase aggregation and free radical formation that paralleled the enzyme loss of function in vitro were also detected in mouse hepatocytes treated with the oxidant. Interestingly, the novel immuno-spin-trapping technique was applied to image radical production in the cells. Indeed, in HOCl-treated hepatocytes, catalase and protein-DMPO nitrone adducts were colocalized in the cells' peroxisomes. In contrast, when hepatocytes from catalase-knockout mice were treated with hypochlorous acid, there was extensive production of free radicals in the plasma membrane. Because free radicals are short-lived species with fundamental roles in biology, the possibility of their detection and localization to cell compartments is expected to open new and stimulating research venues in the interface of chemistry, biology, and medicine.


Subject(s)
Catalase/metabolism , Free Radicals/metabolism , Hepatocytes/drug effects , Hypochlorous Acid/pharmacology , Animals , Catalase/antagonists & inhibitors , Chromatography, High Pressure Liquid , Electron Spin Resonance Spectroscopy , Hepatocytes/enzymology , Immunoprecipitation , Mice , Spectrometry, Mass, Electrospray Ionization
12.
Cell Rep ; 17(9): 2382-2393, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880911

ABSTRACT

Decreased expression of the USP44 deubiquitinase has been associated with global increases in H2Bub1 levels during mouse embryonic stem cell (mESC) differentiation. However, whether USP44 directly deubiquitinates histone H2B or how its activity is targeted to chromatin is not known. We identified USP44 as an integral subunit of the nuclear receptor co-repressor (N-CoR) complex. USP44 within N-CoR deubiquitinates H2B in vitro and in vivo, and ablation of USP44 impairs the repressive activity of the N-CoR complex. Chromatin immunoprecipitation (ChIP) experiments confirmed that USP44 recruitment reduces H2Bub1 levels at N-CoR target loci. Furthermore, high expression of USP44 correlates with reduced levels of H2Bub1 in the breast cancer cell line MDA-MB-231. Depletion of either USP44 or TBL1XR1 impairs the invasiveness of MDA-MB-231 cells in vitro and causes an increase of global H2Bub1 levels. Our findings indicate that USP44 contributes to N-CoR functions in regulating gene expression and is required for efficient invasiveness of triple-negative breast cancer cells.


Subject(s)
Histones/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Cell Line, Tumor , HEK293 Cells , Humans , Neoplasm Invasiveness , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Subunits/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transducin/chemistry , Transducin/metabolism , Ubiquitin Thiolesterase , WD40 Repeats
13.
Mol Cell Biol ; 36(22): 2855-2866, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27601583

ABSTRACT

The SAGA complex contains two enzymatic modules, which house histone acetyltransferase (HAT) and deubiquitinase (DUB) activities. USP22 is the catalytic subunit of the DUB module, but two adaptor proteins, ATXN7L3 and ENY2, are necessary for DUB activity toward histone H2Bub1 and other substrates. ATXN7L3B shares 74% identity with the N-terminal region of ATXN7L3, but the functions of ATXN7L3B are not known. Here we report that ATXN7L3B interacts with ENY2 but not other SAGA components. Even though ATXN7L3B localizes in the cytoplasm, ATXN7L3B overexpression increases H2Bub1 levels, while overexpression of ATXN7L3 decreases H2Bub1 levels. In vitro, ATXN7L3B competes with ATXN7L3 to bind ENY2, and in vivo, knockdown of ATXN7L3B leads to concomitant loss of ENY2. Unlike the ATXN7L3 DUB complex, a USP22-ATXN7L3B-ENY2 complex cannot deubiquitinate H2Bub1 efficiently in vitro Moreover, ATXN7L3B knockdown inhibits migration of breast cancer cells in vitro and limits expression of ER target genes. Collectively, our studies suggest that ATXN7L3B regulates H2Bub1 levels and SAGA DUB activity through competition for ENY2 binding.


Subject(s)
Breast Neoplasms/metabolism , Cytoplasm/metabolism , Histones/metabolism , Thiolester Hydrolases/metabolism , Transcription Factors/metabolism , Cell Movement , Cell Nucleus/enzymology , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , MCF-7 Cells , Transcription Factors/genetics , Ubiquitin Thiolesterase , Up-Regulation
14.
FEBS Lett ; 585(13): 2016-23, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-20974139

ABSTRACT

Post-translational modifications of the histones are centrally involved in the regulation of all DNA-templated processes, including gene transcription, DNA replication, recombination, and repair. These modifications are often dynamic, and their removal is just as important as their addition in proper regulation of cellular functions. Although histone acetylation/deacetylation and histone methylation/demethylation are highly studied, the functions and regulation of histone ubiquitination and deubiquitination are less well understood. This review highlights our current understanding of how histone ubiquitination impacts gene transcription, DNA repair, and cell cycle progression, and stresses the importance of deubiquitinases to normal cellular functions as well as to disease states such as cancer.


Subject(s)
Chromatin/metabolism , Endopeptidases/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Histones/metabolism , Humans , Ubiquitination/genetics , Ubiquitination/physiology
15.
Genes Chromosomes Cancer ; 44(4): 429-37, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16127665

ABSTRACT

The role of the region encoded by exon 27 of the Brca2 gene in DNA repair was studied in cells and tissues from Brca2Delta27/Delta27 mice. The COOH-terminal truncated Brca2 localized to the nucleus in primary mouse embryo fibroblasts from Brca2Delta27/Delta27 mice. Fluorescence-activated cell sorting (FACS) analysis demonstrated that these fibroblasts were hypersensitive to mitomycin C-induced cross-links, but not to double-strand breaks (DSBs) induced by irradiation. The gammaH2AX appearance kinetics and comet assay showed that DSBs were repaired through non-homologous end joining pathways, while interstrand cross-links were not repaired due to deficient homologous recombination pathways. Immunoprecipitation experiments showed that Fancd2 did not coprecipitate with the mutated Brca2. There were also no detectable Rad51-positive foci formed in these cells after damage. On the other hand, we did not find any difference during gametogenesis in mice harboring exon 27 truncating mutation of the Brca2 gene and control mice, and in both cases, Rad51 localized to the recombination foci. Our results suggest that exon 27 of murine Brca2 is crucial for the interaction of Brca2 and Fancd2 in Rad51-mediated recombination in response to DNA damage, but that this interaction is not taking place in the homologous recombination during meiosis.


Subject(s)
BRCA2 Protein/metabolism , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/metabolism , Germ-Line Mutation , Meiosis , Recombination, Genetic , Animals , BRCA2 Protein/chemistry , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Comet Assay , Embryo, Mammalian , Exons , Female , Fibroblasts/drug effects , Fibroblasts/radiation effects , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Histones/metabolism , Kinetics , Mice , Mitomycin/pharmacology , Rad51 Recombinase/metabolism , Radiation, Ionizing
16.
Cancer Detect Prev ; 27(1): 24-9, 2003.
Article in English | MEDLINE | ID: mdl-12600414

ABSTRACT

We have developed fast, reliable and simple fluorescent method to assess and compare repair capacity of cells. To this end plasmid pEGFP containing the gene for the enhanced green fluorescent protein was damaged in vitro by genotoxic agents and introduced into cells by transfection. The repair capacity of the cells was determined from the number of fluorescent cells counted with a fluorescent microscope 24 h after transfection. The ability of four human tumor cell lines--HEK293, HeLa, Namalwa and K562 to repair DNA lesions inflicted by cis-diamminedichloroplatinum(II), UV light, 8-methoxypsoralen and 4',5'-8-trimethylpsoralen were determined and compared to the survival rates of the cells after treatment with the same genotoxic agents. In most but not all cases, there was a good correlation between repair capacity and cell survival. This finding indicates that the DNA repair capacity could be used as a biomarker in risk assessment and/or drug resistance assays.


Subject(s)
DNA Repair/physiology , Spectrometry, Fluorescence/methods , Animals , Cell Death/drug effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , Dose-Response Relationship, Drug , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mutagens/pharmacology , Plasmids/drug effects , Plasmids/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL