Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cancer Res ; 77(7): 1697-1708, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28108507

ABSTRACT

Acquired copy neutral LOH (CN-LOH) is a frequent occurrence in myeloid malignancies and is often associated with resistance to standard therapeutic modalities and poor survival. Here, we show that constitutive signaling driven by mutated FLT3 and JAK2 confers interchromosomal homologous recombination (iHR), a precedent for CN-LOH. Using a targeted recombination assay, we determined significant iHR activity in internal tandem duplication FLT3 (FLT3-ITD) and JAK2V617F-mutated cells. Sister chromatid exchanges, a surrogate measure of iHR, was significantly elevated in primary FLT3-ITD normal karyotype acute myeloid leukemia (NK-AML) compared with wild-type FLT3 NK-AML. HR was harmonized to S phase of the cell cycle to repair broken chromatids and prevent iHR. Increased HR activity in G0 arrested primary FLT3-ITD NK-AML in contrast to wild-type FLT3 NK-AML. Cells expressing mutated FLT3-ITD demonstrated a relative increase in mutation frequency as detected by thymidine kinase (TK) gene mutation assay. Moreover, resistance was associated with CN-LOH at the TK locus. Treatment of FLT3-ITD- and JAK2V617F-mutant cells with the antioxidant N-acetylcysteine diminished reactive oxygen species (ROS), restoring iHR and HR levels. Our findings show that mutated FLT3-ITD and JAK2 augment ROS production and HR, shifting the cellular milieu toward illegitimate recombination events such as iHR and CN-LOH. Therapeutic reduction of ROS may thus prevent leukemic progression and relapse in myeloid malignancies. Cancer Res; 77(7); 1697-708. ©2017 AACR.


Subject(s)
Homologous Recombination , Janus Kinase 2/genetics , Leukemia, Myeloid, Acute/genetics , Loss of Heterozygosity , Mutation , fms-Like Tyrosine Kinase 3/genetics , Acetylcysteine/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Child , Humans , Leukemia, Myeloid, Acute/metabolism , Middle Aged , Rad51 Recombinase/physiology , Reactive Oxygen Species/metabolism , Sister Chromatid Exchange
3.
Haematologica ; 98(9): 1397-406, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23349304

ABSTRACT

Inactivation of the DNA mismatch repair pathway manifests as microsatellite instability, an accumulation of mutations that drives carcinogenesis. Here, we determined whether microsatellite instability in acute myeloid leukemia and myelodysplastic syndrome correlated with chromosomal instability and poly (ADP-ribose) polymerase (PARP) inhibitor sensitivity through disruption of DNA repair function. Acute myeloid leukemia cell lines (n=12) and primary cell samples (n=18), and bone marrow mononuclear cells from high-risk myelodysplastic syndrome patients (n=63) were profiled for microsatellite instability using fluorescent fragment polymerase chain reaction. PARP inhibitor sensitivity was performed using cell survival, annexin V staining and cell cycle analysis. Homologous recombination was studied using immunocytochemical analysis. SNP karyotyping was used to study chromosomal instability. RNA silencing, Western blotting and gene expression analysis was used to study the functional consequences of mutations. Acute myeloid leukemia cell lines (4 of 12, 33%) and primary samples (2 of 18, 11%) exhibited microsatellite instability with mono-allelic mutations in CtIP and MRE11. These changes were associated with reduced expression of mismatch repair pathway components, MSH2, MSH6 and MLH1. Both microsatellite instability positive primary acute myeloid leukemia samples and cell lines demonstrated a downregulation of homologous recombination DNA repair conferring marked sensitivity to PARP inhibitors. Similarly, bone marrow mononuclear cells from 11 of 56 (20%) patients with de novo high-risk myelodysplastic syndrome exhibited microsatellite instability. Significantly, all 11 patients with microsatellite instability had cytogenetic abnormalities with 4 of them (36%) possessing a mono-allelic microsatellite mutation in CtIP. Furthermore, 50% reduction in CtIP expression by RNA silencing also down-regulated homologous recombination DNA repair responses conferring PARP inhibitor sensitivity, whilst CtIP differentially regulated the expression of homologous recombination modulating RecQ helicases, WRN and BLM. In conclusion, microsatellite instability dependent mutations in DNA repair genes, CtIP and MRE11 are detected in myeloid malignancies conferring hypersensitivity to PARP inhibitors. Microsatellite instability is significantly correlated with chromosomal instability in myeloid malignancies.


Subject(s)
Carrier Proteins/genetics , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Microsatellite Instability , Mutation/genetics , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Adult , Aged , Aged, 80 and over , Cell Survival/genetics , Endodeoxyribonucleases , Female , Gene Knockdown Techniques/methods , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , MRE11 Homologue Protein , Male , Middle Aged , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Poly(ADP-ribose) Polymerase Inhibitors
4.
Haematologica ; 94(5): 638-46, 2009 May.
Article in English | MEDLINE | ID: mdl-19407318

ABSTRACT

UNLABELLED: Background Aberrant or impaired repair of double-strand DNA breaks is a common feature of de novo acute myeloid leukemia and myelodysplastic syndromes. Since poly (ADP-ribose) polymerase (PARP) inhibitors have been recently shown to selectively target cells with defects in double-strand DNA repair, the aim of this study was to explore the possibility of exploiting defects in DNA repair in leukemic cells using PARP inhibitors. DESIGN AND METHODS: Leukemic cell lines were exposed to various PARP inhibitors alone and in combination with non-cytotoxic concentrations of DNA methyltransferase inhibitor, 5' aza-2'-deoxycytidine and/or the histone deacetylase inhibitor, MS275, to test for potentiation of apoptosis with these agents. RESULTS: PARP inhibitors, KU-0058948 and PJ34, induced cell cycle arrest and apoptosis of primary myeloid leukemic cells and myeloid leukemic cell lines in vitro. Immunofluorescence analysis also revealed that PARP inhibitor sensitivity in these leukemic cells was due to a defect in homologous recombination DNA repair. Addition of 5' aza-2'-deoxycytidine failed to increase the cytotoxicity of PARP inhibitors. In contrast, MS275 potentiated the cytotoxic effect of KU-0058948 and PJ34 in all PARP inhibitor-sensitive leukemic cells. Immunofluorescence analysis supported the idea that histone deacetylase inhibitors potentiate cytotoxicity by inhibiting DNA repair processes. Conclusions On the basis of the data presented here, we suggest that PARP inhibitors can potentially exploit defects in double-strand DNA break repair in leukemic cells, paving the way for testing the therapeutic potential of these agents in myelodysplastic syndromes and acute myeloid leukemia.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , DNA Repair/drug effects , Poly(ADP-ribose) Polymerase Inhibitors , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Benzamides/pharmacology , Butyrates/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Drug Synergism , Flow Cytometry , Fluorescent Antibody Technique , Fluorobenzenes/pharmacology , HL-60 Cells , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , K562 Cells , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Phenanthrenes/pharmacology , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Pyridines/pharmacology , U937 Cells
5.
Haematologica ; 93(12): 1886-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18838476

ABSTRACT

Poly ADP-ribose polymerase inhibitors have been shown to target cells with homologous recombination DNA repair defects. We report that poly ADP-ribose polymerase inhibitors induces apoptosis in cells deficient in other key DNA repair components. Chromosomal instability disorders, Fanconi Anemia and Bloom's syndrome have dysfunctional DNA repair and an increased likelihood of leukemic transformation. PI addition to Fanconi Anemia and Bloom's syndrome cells resulted in significant apoptosis. Furthermore, poly ADP-ribose polymerase inhibitors induced apoptosis in DNA repair signaling defective ATM(-/-) and NBS(-/-) fibroblasts. Immunocytochemistry showed homologous recombination was abrogated in NBS(-/-) and ATM(-/-) fibroblasts, compromised in Fanconi anemia and normal in Bloom's syndrome cells in response to poly ADP-ribose polymerase inhibitors. Strikingly, poly ADP-ribose polymerase inhibitors increases non-homologous end joining repair activity, whilst non-homologous end joining deficient cells are extremely sensitive to poly ADP-ribose polymerase inhibitors. These data suggest poly ADP-ribose polymerase inhibitors target cells with DNA repair and signaling defects rather than solely defects in homologous recombination improving the potential of poly ADP-ribose polymerase inhibitors therapy in a wider range of cancers.


Subject(s)
DNA Repair-Deficiency Disorders/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Apoptosis/drug effects , Bloom Syndrome/drug therapy , Bloom Syndrome/genetics , Bloom Syndrome/pathology , Cells, Cultured , Chromosomal Instability/drug effects , DNA Repair , Enzyme Inhibitors/therapeutic use , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Humans , Syndrome
6.
J Biol Chem ; 283(33): 22565-72, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18524763

ABSTRACT

The BCL6 oncogenic transcriptional repressor is required for development of germinal center centroblasts, which undergo simultaneous genetic recombination and massive clonal expansion. Although BCL6 is required for survival of centroblasts, its expression in earlier B-cells is toxic. Understanding these opposing effects could provide critical insight into normal B-cell biology and lymphomagenesis. We examined the transcriptional and biological effects of BCL6 in various primary cells. BCL6 repression of ATR was previously shown to play a critical role in the centroblast phenotype. Likewise, we found that BCL6 could impose an ATR-dependent phenotype of attenuated DNA damage sensing and repair in primary fibroblasts and B-cells. BCL6 induced true genomic instability because DNA repair was delayed and was qualitatively impaired, which could be critical for BCL6-induced lymphomagenesis. Although BCL6 can directly repress TP53 in centroblasts, BCL6 induced TP53 expression in primary fibroblasts and B-cells, and these cells underwent p53-dependent growth arrest and senescence in the presence of physiological levels of BCL6. This differential ability to trigger a functional p53 response explains at least in part the different biological response to BCL6 expression in centroblasts versus other cells. The data suggest that targeted re-activation of TP53 could be of therapeutic value in centroblast-derived lymphomas.


Subject(s)
DNA Damage , Proto-Oncogene Proteins c-bcl-6/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Chromatin/physiology , DNA/radiation effects , Gamma Rays , Humans , Palatine Tonsil/physiology , Phosphorylation , Tonsillectomy
7.
Cancer Res ; 67(18): 8762-71, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875717

ABSTRACT

Myelodysplastic syndromes (MDS) comprise a heterogeneous group of disorders characterized by ineffective hematopoiesis, with an increased propensity to develop acute myelogenous leukemia (AML). The molecular basis for MDS progression is unknown, but a key element in MDS disease progression is loss of chromosomal material (genomic instability). Using our two-step mouse model for myeloid leukemic disease progression involving overexpression of human mutant NRAS and BCL2 genes, we show that there is a stepwise increase in the frequency of DNA damage leading to an increased frequency of error-prone repair of double-strand breaks (DSB) by nonhomologous end-joining. There is a concomitant increase in reactive oxygen species (ROS) in these transgenic mice with disease progression. Importantly, RAC1, an essential component of the ROS-producing NADPH oxidase, is downstream of RAS, and we show that ROS production in NRAS/BCL2 mice is in part dependent on RAC1 activity. DNA damage and error-prone repair can be decreased or reversed in vivo by N-acetyl cysteine antioxidant treatment. Our data link gene abnormalities to constitutive DNA damage and increased DSB repair errors in vivo and provide a mechanism for an increase in the error rate of DNA repair with MDS disease progression. These data suggest treatment strategies that target RAS/RAC pathways and ROS production in human MDS/AML.


Subject(s)
DNA Damage , DNA Repair , Genomic Instability , Leukemia, Myeloid/genetics , Reactive Oxygen Species/metabolism , Animals , Disease Models, Animal , Disease Progression , Genes, bcl-2 , Genes, ras , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Transgenic
8.
Mol Cancer Res ; 4(8): 563-73, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877702

ABSTRACT

Histone deacetylase inhibitors (HDI) increase gene expression through induction of histone acetylation. However, it remains unclear whether increases in specific gene expression events determine the apoptotic response following HDI administration. Herein, we show that a variety of HDI trigger in hematopoietic cells not only widespread histone acetylation and DNA damage responses but also actual DNA damage, which is significantly increased in leukemic cells compared with normal cells. Thus, increase in H2AX and ataxia telangiectasia mutated (ATM) phosphorylation, early markers of DNA damage, occurs rapidly following HDI administration. Activation of the DNA damage and repair response following HDI treatment is further emphasized by localizing DNA repair proteins to regions of DNA damage. These events are followed by subsequent apoptosis of neoplastic cells but not normal cells. Our data indicate that induction of apoptosis by HDI may result predominantly through accumulation of excessive DNA damage in leukemia cells, leading to activation of apoptosis.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , Histone Deacetylase Inhibitors , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins , Butyrates/pharmacology , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/drug effects , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , Gamma Rays , HL-60 Cells , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , K562 Cells , Mice , Mice, Transgenic , Organ Specificity/drug effects , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Staurosporine/pharmacology , Transfection , Tumor Suppressor Proteins/metabolism
9.
Cancer Res ; 63(8): 1798-805, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12702565

ABSTRACT

Double strand breaks (DSBs) are considered the most lethal form of DNA damage for eukaryotic cells, and misrepair of DSB can cause cell death, chromosome instability, and cancer. Nonhomologous end-joining (NHEJ) is a major mechanism for the repair of DSBs. We previously reported that the cancer predisposition Bloom's syndrome and myeloid leukemias demonstrate increased NHEJ activity and consequent misrepair. In this study, we link this increased NHEJ activity and infidelity to ongoing or induced DNA damage at sites that recruit key NHEJ proteins. We show here that in myeloid leukemia cells and normal hemopoietic cells, agents that induce DSBs produce an up to 2-fold increase in this DSB misrepair activity, whereas an alkylating agent produces little or no increases. Furthermore, NHEJ overactivity after induction of DSBs is dependent on the presence of Ku70/Ku86. We also present data to explain the constitutively activated NHEJ in myeloid leukemias. Using an immunofluorescence-based assay for DNA damage, myeloid leukemias demonstrate constitutive DNA damage in the absence of treatment with DSB-inducing agents compared with normal hemopoietic cells. Importantly, damaged foci from myeloid leukemia and normal cells colocalize with NHEJ proteins Ku70 and Ku86. These data suggest that the generation of increased constitutive DNA damage may be a common pathway for the creation of NHEJ-dependent genomic instability.


Subject(s)
Antigens, Nuclear/metabolism , DNA Damage , DNA Helicases , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Leukemia, Myeloid/genetics , Antibodies/pharmacology , Antigens, Nuclear/immunology , Aphidicolin/pharmacology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA, Neoplasm/radiation effects , DNA-Binding Proteins/immunology , HL-60 Cells , Humans , K562 Cells , Ku Autoantigen , Leukemia, Myeloid/metabolism
10.
Cancer Res ; 62(10): 2791-7, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019155

ABSTRACT

Human myeloid leukemias are characterized by chromosomal abnormalities, including translocations, deletions, and allelic loss. These alterations are known to disrupt the function of genes that contribute to tumor initiation and progression. The mechanism underlying the appearance of these chromosomal alterations is poorly understood. Recent evidence suggests that altered nonhomologous end joining (NHEJ) is associated with the incidence of chromosome abnormalities in mutant rodent cells. This pathway is thought to provide a major mechanism for the repair of double-strand breaks (DSB) in higher eukaryotes. Here, we show that in an in vitro assay for DSB end ligation, nuclear extracts prepared from cultured and primary myeloid leukemia cells show a 2-7-fold increase in end-ligation efficiency as compared with mobilized peripheral CD34+ blood progenitor cells (CD34+) and interleukin-2-stimulated peripheral blood lymphocytes from normal healthy donors (P < 0.001). Furthermore, using an in vitro plasmid LacZ gene reactivation assay to determine DSB repair fidelity, nuclear extracts prepared from myeloid leukemia cells showed an increased frequency of misrepair compared with normal control cells (P < 0.001). Most importantly, this misrepair in myeloid leukemia cells is associated with large deletions (30-400 bp) within the test plasmids used in our assay. These deletions were not observed using normal hematopoietic cells (<28 bp). Strikingly, we show that the NHEJ proteins, Ku70 and 86, are required for the deletions in myeloid leukemias because preincubating nuclear extracts from leukemic cells with antisera against Ku86 and Ku70 inhibits plasmid reactivation and restores the frequency and size of deletions to control levels. Our findings suggest that an overactive NHEJ system and, specifically, aberrant Ku70/86 activity is a candidate mechanism for chromosomal instability in myeloid leukemias.


Subject(s)
Antigens, Nuclear , DNA Damage , DNA Helicases , DNA Repair , DNA, Neoplasm/metabolism , DNA-Binding Proteins/physiology , Leukemia, Myeloid/genetics , Nuclear Proteins/physiology , Chromosome Aberrations , DNA, Neoplasm/genetics , Dimerization , HL-60 Cells , Humans , K562 Cells , Ku Autoantigen , Leukemia, Myeloid/metabolism
11.
Oncogene ; 21(16): 2525-33, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11971187

ABSTRACT

BS is an inherited cancer predisposition disorder caused by inactivation of the RecQ family helicase, BLM. One of the defining features of cells from BS individuals is chromosomal instability, characterized by elevated sister chromatid exchanges (SCEs), as well as chromosomal breaks, deletions, and rearrangements. Although the basis for chromosomal instability is poorly understood, there is evidence that chromosomal abnormalities can arise through an alteration in the efficiency or fidelity of DNA double strand break (DSB) repair. Here, we show that BS cells demonstrate aberrant DSB repair mediated by the non-homologous end-joining (NHEJ) pathway for DNA repair, one of the two main pathways for the repair of DSBs in mammalian cells. Through a comparison of BS cell lines, and a derivative in which the BS phenotype has been reverted by expression of the BLM cDNA, we show that BS cells display aberrant end-joining of DSBs. Importantly, DNA end-joining in BS cells is highly error-prone and frequently results in DNA ligation at distant sites of microhomology, creating large DNA deletions. This aberrant repair is dependent upon the presence of the Ku70/86 heterodimer, a key component in the NHEJ pathway. We propose that aberrant NHEJ is a candidate mechanism for the generation of chromosomal instability in BS.


Subject(s)
Antigens, Nuclear , Bloom Syndrome/genetics , Chromosome Aberrations , DNA Helicases , DNA Repair , Antibodies/immunology , Base Sequence , Bloom Syndrome/metabolism , Cell Line , Cells, Cultured , DNA-Binding Proteins/analysis , DNA-Binding Proteins/immunology , DNA-Binding Proteins/physiology , Humans , Ku Autoantigen , Nuclear Proteins/analysis , Nuclear Proteins/immunology , Nuclear Proteins/physiology , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...