Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(8): e0182610, 2017.
Article in English | MEDLINE | ID: mdl-28787462

ABSTRACT

A new ovarian near-diploid cell line, OVDM1, was derived from a highly aneuploid serous ovarian metastatic adenocarcinoma. A metastatic tumor was obtained from a 47-year-old Ashkenazi Jewish patient three years after the first surgery removed the primary tumor, both ovaries, and the remaining reproductive organs. OVDM1 was characterized by cell morphology, genotyping, tumorigenic assay, mycoplasma testing, spectral karyotyping (SKY), and molecular profiling of the whole genome by aCGH and gene expression microarray. Targeted sequencing of a panel of cancer-related genes was also performed. Hierarchical clustering of gene expression data clearly confirmed the ovarian origin of the cell line. OVDM1 has a near-diploid karyotype with a low-level aneuploidy, but samples of the original metastatic tumor were grossly aneuploid. A number of single nucleotide variations (SNVs)/mutations were detected in OVDM1 and the metastatic tumor samples. Some of them were cancer-related according to COSMIC and HGMD databases (no founder mutations in BRCA1 and BRCA2 have been found). A large number of focal copy number alterations (FCNAs) were detected, including homozygous deletions (HDs) targeting WWOX and GATA4. Progression of OVDM1 from early to late passages was accompanied by preservation of the near-diploid status, acquisition of only few additional large chromosomal rearrangements and more than 100 new small FCNAs. Most of newly acquired FCNAs seem to be related to localized but massive DNA fragmentation (chromothripsis-like rearrangements). Newly developed near-diploid OVDM1 cell line offers an opportunity to evaluate tumorigenesis pathways/events in a minor clone of metastatic ovarian adenocarcinoma as well as mechanisms of chromothripsis.


Subject(s)
Aneuploidy , Cell Line, Tumor , Diploidy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Adult , Animals , Cell Transformation, Neoplastic , Disease Progression , Female , Gene Expression Profiling , Genomics , Humans , Mice , Neoplasm Metastasis , Neoplasm Staging
2.
Cancer Lett ; 371(2): 214-24, 2016 Feb 28.
Article in English | MEDLINE | ID: mdl-26683771

ABSTRACT

The molecular mechanisms underlying the aggressive behavior of MYCN driven neuroblastoma (NBL) is under intense investigation; however, little is known about the impact of this family of transcription factors on the splicing program. Here we used high-throughput RNA sequencing to systematically study the expression of RNA isoforms in stage 4 MYCN-amplified NBL, an aggressive subtype of metastatic NBL. We show that MYCN-amplified NBL tumors display a distinct gene splicing pattern affecting multiple cancer hallmark functions. Six splicing factors displayed unique differential expression patterns in MYCN-amplified tumors and cell lines, and the binding motifs for some of these splicing factors are significantly enriched in differentially-spliced genes. Direct binding of MYCN to promoter regions of the splicing factors PTBP1 and HNRNPA1 detected by ChIP-seq demonstrates that MYCN controls the splicing pattern by direct regulation of the expression of these key splicing factors. Furthermore, high expression of PTBP1 and HNRNPA1 was significantly associated with poor overall survival of stage4 NBL patients (p ≤ 0.05). Knocking down PTBP1, HNRNPA1 and their downstream target PKM2, an isoform of pro-tumor-growth, result in repressed growth of NBL cells. Therefore, our study reveals a novel role of MYCN in controlling global splicing program through regulation of splicing factors in addition to its well-known role in the transcription program. These findings suggest a therapeutically potential to target the key splicing factors or gene isoforms in high-risk NBL with MYCN-amplification.


Subject(s)
Alternative Splicing , Biomarkers, Tumor/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA, Neoplasm/genetics , Binding Sites , Biomarkers, Tumor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Gene Amplification , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , High-Throughput Nucleotide Sequencing , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , N-Myc Proto-Oncogene Protein , Neoplasm Staging , Neuroblastoma/metabolism , Neuroblastoma/mortality , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Neoplasm/metabolism , Risk Factors , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Transcription, Genetic , Transfection , Thyroid Hormone-Binding Proteins
3.
Oncotarget ; 6(34): 35247-62, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26497213

ABSTRACT

Despite advances in multimodal treatment, neuroblastoma (NB) is often fatal for children with high-risk disease and many survivors need to cope with long-term side effects from high-dose chemotherapy and radiation. To identify new therapeutic targets, we performed an siRNA screen of the druggable genome combined with a small molecule screen of 465 compounds targeting 39 different mechanisms of actions in four NB cell lines. We identified 58 genes as targets, including AURKB, in at least one cell line. In the drug screen, aurora kinase inhibitors (nine molecules) and in particular the AURKB-selective compound, barasertib, were the most discriminatory with regard to sensitivity for MYCN-amplified cell lines. In an expanded panel of ten NB cell lines, those with MYCN-amplification and wild-type TP53 were the most sensitive to low nanomolar concentrations of barasertib. Inhibition of the AURKB kinase activity resulted in decreased phosphorylation of the known target, histone H3, and upregulation of TP53 in MYCN-amplified, TP53 wild-type cells. However, both wild-type and TP53 mutant MYCN-amplified cell lines arrested in G2/M phase upon AURKB inhibition. Additionally, barasertib induced endoreduplication and apoptosis. Treatment of MYCN-amplified/TP53 wild-type neuroblastoma xenografts resulted in profound growth inhibition and tumor regression. Therefore, aurora B kinase inhibition is highly effective in aggressive neuroblastoma and warrants further investigation in clinical trials.


Subject(s)
Aurora Kinase B/antagonists & inhibitors , Neuroblastoma/enzymology , Neuroblastoma/therapy , Animals , Apoptosis/physiology , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Cell Line, Tumor , Female , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
4.
PLoS Genet ; 11(3): e1005075, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25768946

ABSTRACT

To infer the subclonality of rhabdomyosarcoma (RMS) and predict the temporal order of genetic events for the tumorigenic process, and to identify novel drivers, we applied a systematic method that takes into account germline and somatic alterations in 44 tumor-normal RMS pairs using deep whole-genome sequencing. Intriguingly, we find that loss of heterozygosity of 11p15.5 and mutations in RAS pathway genes occur early in the evolutionary history of the PAX-fusion-negative-RMS (PFN-RMS) subtype. We discover several early mutations in non-RAS mutated samples and predict them to be drivers in PFN-RMS including recurrent mutation of PKN1. In contrast, we find that PAX-fusion-positive (PFP) subtype tumors have undergone whole-genome duplication in the late stage of cancer evolutionary history and have acquired fewer mutations and subclones than PFN-RMS. Moreover we predict that the PAX3-FOXO1 fusion event occurs earlier than the whole genome duplication. Our findings provide information critical to the understanding of tumorigenesis of RMS.


Subject(s)
Genome, Human , Rhabdomyosarcoma/genetics , Sequence Analysis, DNA , Adolescent , Child , Child, Preschool , Chromosomes, Human, Pair 11 , Genome-Wide Association Study , Humans , Infant , Loss of Heterozygosity , Oncogene Proteins, Fusion/genetics , Paired Box Transcription Factors/genetics
5.
PLoS Genet ; 10(7): e1004475, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25010205

ABSTRACT

The Ewing sarcoma family of tumors (EFT) is a group of highly malignant small round blue cell tumors occurring in children and young adults. We report here the largest genomic survey to date of 101 EFT (65 tumors and 36 cell lines). Using a combination of whole genome sequencing and targeted sequencing approaches, we discover that EFT has a very low mutational burden (0.15 mutations/Mb) but frequent deleterious mutations in the cohesin complex subunit STAG2 (21.5% tumors, 44.4% cell lines), homozygous deletion of CDKN2A (13.8% and 50%) and mutations of TP53 (6.2% and 71.9%). We additionally note an increased prevalence of the BRCA2 K3326X polymorphism in EFT patient samples (7.3%) compared to population data (OR 7.1, p = 0.006). Using whole transcriptome sequencing, we find that 11% of tumors pathologically diagnosed as EFT lack a typical EWSR1 fusion oncogene and that these tumors do not have a characteristic Ewing sarcoma gene expression signature. We identify samples harboring novel fusion genes including FUS-NCATc2 and CIC-FOXO4 that may represent distinct small round blue cell tumor variants. In an independent EFT tissue microarray cohort, we show that STAG2 loss as detected by immunohistochemistry may be associated with more advanced disease (p = 0.15) and a modest decrease in overall survival (p = 0.10). These results significantly advance our understanding of the genomic and molecular underpinnings of Ewing sarcoma and provide a foundation towards further efforts to improve diagnosis, prognosis, and precision therapeutics testing.


Subject(s)
Antigens, Nuclear/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Sarcoma, Ewing/genetics , Adolescent , Adult , Cell Cycle Proteins , Cell Line, Tumor , Child , Child, Preschool , Disease-Free Survival , Female , Gene Deletion , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Sarcoma, Ewing/etiology , Sarcoma, Ewing/pathology
6.
Nucleic Acids Res ; 42(7): 4546-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493738

ABSTRACT

Between 6 and 30% of human and mouse transcripts are initiated from transposable elements. However, the promoters driving such transcriptional activity are mostly unknown. We experimentally characterized an antisense (AS) promoter in mouse L1 retrotransposons for the first time, oriented antiparallel to the coding strand of L1 open reading frame-1. We found that AS transcription is mediated by RNA polymerase II. Rapid amplification of cDNA ends cloning mapped transcription start sites adjacent to the AS promoter. We identified >100 novel fusion transcripts, of which many were conserved across divergent mouse lineages, suggesting conservation of potential functions. To evaluate whether AS L1 transcription could regulate L1 retrotransposition, we replaced portions of native open reading frame-1 in donor elements by synonymously recoded sequences. The resulting L1 elements lacked AS promoter activity and retrotransposed more frequently than endogenous L1s. Overexpression of AS L1 transcripts also reduced L1 retrotransposition. This suppression of retrotransposition was largely independent of Dicer. Our experiments shed new light on how AS fusion transcripts are initiated from endogenous L1 elements across the mouse genome. Such AS transcription can contribute substantially both to natural transcriptional variation and to endogenous regulation of L1 retrotransposition.


Subject(s)
Long Interspersed Nucleotide Elements , Promoter Regions, Genetic , RNA, Antisense/biosynthesis , RNA-Binding Proteins/genetics , Animals , Base Sequence , Cell Line , Humans , Mice , Molecular Sequence Data , RNA Polymerase II/metabolism , Ribonuclease III/metabolism , Transcription Initiation Site
7.
Cancer Discov ; 4(2): 216-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24436047

ABSTRACT

UNLABELLED: Despite gains in survival, outcomes for patients with metastatic or recurrent rhabdomyosarcoma remain dismal. In a collaboration between the National Cancer Institute, Children's Oncology Group, and Broad Institute, we performed whole-genome, whole-exome, and transcriptome sequencing to characterize the landscape of somatic alterations in 147 tumor/normal pairs. Two genotypes are evident in rhabdomyosarcoma tumors: those characterized by the PAX3 or PAX7 fusion and those that lack these fusions but harbor mutations in key signaling pathways. The overall burden of somatic mutations in rhabdomyosarcoma is relatively low, especially in tumors that harbor a PAX3/7 gene fusion. In addition to previously reported mutations in NRAS, KRAS, HRAS, FGFR4, PIK3CA, and CTNNB1, we found novel recurrent mutations in FBXW7 and BCOR, providing potential new avenues for therapeutic intervention. Furthermore, alteration of the receptor tyrosine kinase/RAS/PIK3CA axis affects 93% of cases, providing a framework for genomics-directed therapies that might improve outcomes for patients with rhabdomyosarcoma. SIGNIFICANCE: This is the most comprehensive genomic analysis of rhabdomyosarcoma to date. Despite a relatively low mutation rate, multiple genes were recurrently altered, including NRAS, KRAS, HRAS, FGFR4, PIK3CA, CTNNB1, FBXW7, and BCOR. In addition, a majority of rhabdomyosarcoma tumors alter the receptor tyrosine kinase/RAS/PIK3CA axis, providing an opportunity for genomics-guided intervention.


Subject(s)
Genomics , Oncogene Proteins, Fusion/genetics , Rhabdomyosarcoma/genetics , Animals , Cell Cycle Proteins/genetics , Chromosome Aberrations , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , DNA Copy Number Variations , Exome , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Gene Regulatory Networks , Genome-Wide Association Study , Genotype , Humans , Mice , Mutation , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Rhabdomyosarcoma/metabolism , Signal Transduction
8.
PLoS One ; 8(10): e76551, 2013.
Article in English | MEDLINE | ID: mdl-24124571

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common childhood soft tissue sarcoma. Despite advances in modern therapy, patients with relapsed or metastatic disease have a very poor clinical prognosis. Fibroblast Growth Factor Receptor 4 (FGFR4) is a cell surface tyrosine kinase receptor that is involved in normal myogenesis and muscle regeneration, but not commonly expressed in differentiated muscle tissues. Amplification and mutational activation of FGFR4 has been reported in RMS and promotes tumor progression. Therefore, FGFR4 is a tractable therapeutic target for patients with RMS. In this study, we used a chimeric Ba/F3 TEL-FGFR4 construct to test five tyrosine kinase inhibitors reported to specifically inhibit FGFRs in the nanomolar range. We found ponatinib (AP24534) to be the most potent FGFR4 inhibitor with an IC50 in the nanomolar range. Ponatinib inhibited the growth of RMS cells expressing wild-type or mutated FGFR4 through increased apoptosis. Phosphorylation of wild-type and mutated FGFR4 as well as its downstream target STAT3 was also suppressed by ponatinib. Finally, ponatinib treatment inhibited tumor growth in a RMS mouse model expressing mutated FGFR4. Therefore, our data suggests that ponatinib is a potentially effective therapeutic agent for RMS tumors that are driven by a dysregulated FGFR4 signaling pathway.


Subject(s)
Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Rhabdomyosarcoma/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Mice , Mutation , Phosphorylation/drug effects , Receptor, Fibroblast Growth Factor, Type 4/genetics , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , STAT3 Transcription Factor/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
9.
Mol Cancer Ther ; 10(7): 1137-48, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21551261

ABSTRACT

Activation of the serine/threonine kinase Akt contributes to the formation, maintenance, and therapeutic resistance of cancer, which is driving development of compounds that inhibit Akt. Phosphatidylinositol ether lipid analogues (PIA) are analogues of the products of phosphoinositide-3-kinase (PI3K) that inhibit Akt activation, translocation, and the proliferation of a broad spectrum of cancer cell types. To gain insight into the mechanism of PIAs, time-dependent transcriptional profiling of five active PIAs and the PI3K inhibitor LY294002 (LY) was conducted in non-small cell lung carcinoma cells using high-density oligonucleotide arrays. Gene ontology analysis revealed that genes involved in apoptosis, wounding response, and angiogenesis were upregulated by PIAs, whereas genes involved in DNA replication, repair, and mitosis were suppressed. Genes that exhibited early differential expression were partitioned into three groups; those induced by PIAs only (DUSP1, KLF6, CENTD2, BHLHB2, and PREX1), those commonly induced by PIAs and LY (TRIB1, KLF2, RHOB, and CDKN1A), and those commonly suppressed by PIAs and LY (IGFBP3, PCNA, PRIM1, MCM3, and HSPA1B). Increased expression of the tumor suppressors RHOB (RhoB), KLF6 (COPEB), and CDKN1A (p21Cip1/Waf1) was validated as an Akt-independent effect that contributed to PIA-induced cytotoxicity. Despite some overlap with LY, active PIAs have a distinct expression signature that contributes to their enhanced cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung , Chromones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lipids/pharmacology , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Chromones/chemistry , Cluster Analysis , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Morpholines/chemistry , Phosphatidylinositols/chemistry , Phosphatidylinositols/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results
10.
Proc Natl Acad Sci U S A ; 107(7): 3001-5, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133699

ABSTRACT

Recent genomewide association studies have found multiple genetic variants on chromosome 8q24 that are significantly associated with an increased susceptibility to prostate, colorectal, and breast cancer. These risk loci are located in a "gene desert," a few hundred kilobases telomeric to the Myc gene. To date, the biological mechanism(s) underlying these associations remain unclear. It has been speculated that these 8q24 genetic variant(s) might affect Myc expression by altering its regulation or amplification status. Here, we show that multiple enhancer elements are present within this region and that they can regulate transcription of Myc. We also demonstrate that one such enhancer element physically interacts with the Myc promoter via transcription factor Tcf-4 binding and acts in an allele specific manner to regulate Myc expression.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Chromatin Immunoprecipitation , Computational Biology , DNA Primers/genetics , Humans , Luciferases , Molecular Sequence Data , Neoplasms/metabolism , Transcription Factor 4 , Transcription Factors/metabolism , beta Catenin/metabolism
11.
Proc Natl Acad Sci U S A ; 107(1): 69-74, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018659

ABSTRACT

Both p53 and the Wnt signaling pathway play important roles in regulating the differentiation of mouse embryonic stem cells (mESCs). However, it is not known whether they directly and/or functionally crosstalk in mESCs. Here we report a surprising antidifferentiation function of p53 in mESCs through directly regulating the Wnt signaling pathway. A chromatin-immunoprecipitation-based microarray (ChIP-chip) and gene expression microarray assays reveal that the Wnt signaling pathway is significantly (P value, 0.000048) overrepresented in p53-regulated genes in mESCs. The expression of five Wnt ligand genes is robustly induced by various genotoxic and nongenotoxic insults in a p53-dependent manner. Moreover, the induction of these Wnt genes is greatly attenuated in mouse embryonic fibroblast (MEF) cells and ESC-derived neural stem/progenitor cells, suggesting that the induction is mESC specific. It is established that the activation of the Wnt signaling pathway inhibits the differentiation of mESCs. Consistent with this notion, we detected an antidifferentiation activity from the conditioned medium (CM) collected from UV (UV)-treated mESCs. This antidifferentiation activity can be lowered by either the addition of Wnt antagonists into the CM or the reduction of p53 levels in UV-treated mESCs. Therefore, reminiscent of its dual functions on death and survival in somatic cells, p53 appears to regulate both prodifferentiation and antidifferentiation programs in mESCs. Our findings uncover a direct and functional connection between p53 and the Wnt signaling pathway, and expand the catalog of p53 regulated genes in mESCs.


Subject(s)
Embryonic Stem Cells/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Cell Differentiation/physiology , Cell Lineage , Cells, Cultured , Culture Media, Conditioned/metabolism , DNA Damage , Doxorubicin/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Nanog Homeobox Protein , Oligonucleotide Array Sequence Analysis , Protein Binding , Tumor Suppressor Protein p53/genetics , Wnt Proteins/genetics
12.
Br J Pharmacol ; 141(5): 795-802, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14769780

ABSTRACT

1. The mechanism of toxicity of sulphur mustard was investigated by examining the biochemical effects of the analog 2-chloroethylethyl sulphide (CEES) in both human Jurkat cells as well as normal human lymphocytes. 2. Exposure of both types of cells to CEES resulted in a marked decrease in the intracellular concentration of the reduced form of glutathione (GSH), and CEES-induced cell death was potentiated by l-buthionine sulphoximine, an inhibitor of GSH synthesis. 3. CEES increased the endogenous production of reactive oxygen species (ROS) in Jurkat cells, and CEES-induced cell death was potentiated by hydrogen peroxide. 4. CEES induced various hallmarks of apoptosis, including collapse of the mitochondrial membrane potential, proteolytic processing and activation of procaspase-3, and cleavage of poly (ADP-ribose) polymerase. 5. The effects of CEES on the accumulation of ROS, the intracellular concentration of GSH, the mitochondrial membrane potential, and caspase-3 activity were all inhibited by pretreatment of cells with the GSH precursor N-acetyl cysteine or with GSH-ethyl ester. Furthermore, CEES-induced cell death was also prevented by these antioxidants. 6. CEES toxicity appears to be mediated, at least in part, by the generation of ROS and consequent depletion of GSH. Given that sulphur mustard is still a potential biohazard, the protective effects of antioxidants against CEES toxicity demonstrated in Jurkat cells and normal human lymphocytes may provide the basis for the development of a therapeutic strategy to counteract exposure to this chemical weapon.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Lymphocytes/drug effects , Mustard Gas/analogs & derivatives , Mustard Gas/toxicity , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Humans , Jurkat Cells , Lymphocytes/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...