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










Publication year range
1.
J Biol Chem ; 289(33): 22798-22814, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24990949

ABSTRACT

DHX9 is an ATP-dependent DEXH box helicase with a multitude of cellular functions. Its ability to unwind both DNA and RNA, as well as aberrant, noncanonical polynucleotide structures, has implicated it in transcriptional and translational regulation, DNA replication and repair, and maintenance of genome stability. We report that loss of DHX9 in primary human fibroblasts results in premature senescence, a state of irreversible growth arrest. This is accompanied by morphological defects, elevation of senescence-associated ß-galactosidase levels, and changes in gene expression closely resembling those encountered during replicative (telomere-dependent) senescence. Activation of the p53 signaling pathway was found to be essential to this process. ChIP analysis and investigation of nascent DNA levels revealed that DHX9 is associated with origins of replication and that its suppression leads to a reduction of DNA replication. Our results demonstrate an essential role of DHX9 in DNA replication and normal cell cycle progression.


Subject(s)
Cell Cycle Checkpoints/physiology , Cellular Senescence/physiology , DEAD-box RNA Helicases/metabolism , DNA Replication/physiology , Fibroblasts/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , DEAD-box RNA Helicases/genetics , Diploidy , Fibroblasts/cytology , HEK293 Cells , Humans , Neoplasm Proteins/genetics , Tumor Suppressor Protein p53/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
2.
J Cancer ; 4(5): 358-70, 2013.
Article in English | MEDLINE | ID: mdl-23781282

ABSTRACT

Human origins of DNA replication are specific sequences within the genome whereby DNA replication is initiated. A select group of proteins, known as the pre-replication (pre-RC) complex, in whose formation the Ku protein (Ku70/Ku86) was shown to play a role, bind to replication origins to initiate DNA replication. In this study, we have examined the involvement of Ku in breast tumorigenesis and tumor progression and found that the Ku protein expression levels in human breast metastatic (MCF10AC1a) cells were higher in the chromatin fraction compared to hyperplastic (MCF10AT) and normal (MCF10A) human breast cells, but remained constant in both the nuclear and cytoplasmic fractions. In contrast, in human intestinal cells, the Ku expression level was relatively constant for all cell fractions. Nascent DNA abundance and chromatin association of Ku70/86 revealed that the c-myc origin activity in MCF10AC1a is 2.5 to 5-fold higher than in MCF10AT and MCF10A, respectively, and Ku was bound to the c-myc origin more abundantly in MCF10AC1a, by approximately 1.5 to 4.2-fold higher than in MCF10AT and MCF10A, respectively. In contrast, similar nascent DNA abundance and chromatin association was found for all cell lines for the lamin B2 origin, associated with the constitutively active housekeeping lamin B2 gene. Electrophoretic mobility shift assays (EMSAs) performed on the nuclear extracts (NEs) of the three cell types revealed the presence of protein-DNA replication complexes on both the c-myc and lamin B2 origins, but an increase in binding activity was observed from normal, to transformed, to cancer cells for the c-myc origin, whereas no such difference was seen for the lamin B2 origin. Overall, the results suggest that increased Ku chromatin association, beyond wild type levels, alters cellular processes, which have been implicated in tumorigenesis.

3.
PLoS One ; 8(5): e63190, 2013.
Article in English | MEDLINE | ID: mdl-23696799

ABSTRACT

Epigenetic inactivation of chromatin plays an important role in determining cell phenotype in both normal and cancer cells, but our knowledge is still incomplete with respect to any potential monoallelic nature of the phenomenon. We have genotyped DNA isolated from chromatin of two colorectal cancer-derived lines and a culture of normal human intestinal epithelial cells (HIEC), which was immunoprecipitated with antibodies to acetylated vs. methylated histone H3K9, and presented the data as B allele frequency differences over multiple single-nucleotide polymorphism (SNP) moving window averages. [B allele is an arbitrary term defined as one of the two alleles at any given SNP, named A and B]. Three different validation tests confirmed that peaks exhibiting differences represented monoallelic domains. These complementary tests confirmed the following: 1) genes in the regions of high B allele frequency difference were expressed monoallelically; 2) in normal cells all five imprinting control regions which carried heterozygous SNPs were characterized by B allele difference peaks; and 3) the haplotypes in the B allele difference peaks were faithfully maintained in the chromatin immunoprecipitated with the respective antibodies. In both samples most of the monoallelic domains were found at the boundaries between regions of open and closed chromatin. With respect to the cancer line, this supports the established concept of conformation spreading, but the results from the normal cells were unexpected. Since these cells were polyclonal, the monoallelic structures were probably not determined by random choice as occurs in X-inactivation, so we propose that epigenetic inactivation in some domains may be heritable and polymorphic in normal human cells.


Subject(s)
Chromatin/genetics , Alleles , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation/genetics , Genotype , Histones/metabolism , Humans , Polymorphism, Single Nucleotide/genetics
4.
Blood ; 121(17): 3402-12, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23440244

ABSTRACT

ABT-737 is a promising chemotherapeutic agent that promotes apoptosis by acting as a selective BH3 mimetic to neutralize Bcl-2-like family members. One shortcoming with its use is that Mcl-1, a member of the Bcl-2 family, is poorly inhibited by ABT-737 and thus is a major cause of resistance. We performed a short hairpin RNA (shRNA)-based drop-out screen to identify novel genes and pathways that could reverse resistance to ABT-737 treatment in Eµ-myc/Bcl-2 lymphoma cells engineered to rely on endogenous Mcl-1 for survival. Several drug-sensitive shRNAs were identified that were selectively depleted in the presence of ABT-737. Of these, 2 independent shRNAs targeting the RNA/DNA helicase Dhx9 were found to sensitize lymphomas to ABT-737 to an extent comparable to control Mcl-1 shRNAs. Although Dhx9 suppression sensitized both mouse and human cells to ABT-737 treatment, it did so without altering MCL-1 levels. Rather, loss of Dhx9 appeared to activate a p53-dependent apoptotic program, through aggravation of replicative stress, which was found to be both necessary and sufficient for the ABT-737-shDhx9 synthetic lethal relationship.


Subject(s)
Biphenyl Compounds/pharmacology , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Lymphoma/genetics , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Sulfonamides/pharmacology , Animals , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p16/physiology , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , Genes, Modifier , Humans , Lymphoma/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism
5.
Genes Cancer ; 3(2): 152-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-23050047

ABSTRACT

This study examines the chromatin structure encompassing replication origins in transformed and normal cells. Analysis of the global levels of histone H3 acetylated at K9&14 (open chromatin) and histone H3 trimethylated at K9 (closed chromatin) revealed a higher ratio of open to closed chromatin in the transformed cells. Also, the trithorax and polycomb group proteins, Brg-1 and Bmi-1, respectively, were overexpressed and more abundantly bound to chromatin in the transformed cells. Quantitative comparative analyses of episomal and in situ chromosomal replication origin activity as well as chromatin immunoprecipitation (ChIP) assays, using specific antibodies targeting members of the pre-replication complex (pre-RC) as well as open/closed chromatin markers encompassing both episomal and chromosomal origins, revealed that episomal origins had similar levels of in vivo activity, nascent DNA abundance, pre-RC protein association, and elevated open chromatin structure at the origin in both cell types. In contrast, the chromosomal origins corresponding to 20mer1, 20mer2, and c-myc displayed a 2- to 3-fold higher activity and pre-RC protein abundance as well as higher ratios of open to closed chromatin and of Brg-1 to Bmi-1 in the transformed cells, whereas the origin associated with the housekeeping lamin B2 gene exhibited similar levels of activity, pre-RC protein abundance, and higher ratios of open to closed chromatin and of Brg-1 to Bmi-1 in both cell types. Nucleosomal positioning analysis, using an MNase-Southern blot assay, showed that all the origin regions examined were situated within regions of inconsistently positioned nucleosomes, with the nucleosomes being spaced farther apart from each other prior to the onset of S phase in both cell types. Overall, the results indicate that cellular transformation is associated with differential epigenetic regulation, whereby chromatin structure is more open, rendering replication origins more accessible to initiator proteins, thus allowing increased origin activity.

6.
J Cell Biochem ; 113(4): 1333-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22134836

ABSTRACT

This study examines the abundance of the major protein constituents of the pre-replication complex (pre-RC), both genome-wide and in association with specific replication origins, namely the lamin B2, c-myc, 20mer1, and 20mer2 origins. Several pre-RC protein components, namely ORC1-6, Cdc6, Cdt1, MCM4, MCM7, as well as additional replication proteins, such as Ku70/86, 14-3-3, Cdc45, and PCNA, were comparatively and quantitatively analyzed in both transformed and normal cells. The results show that these proteins are overexpressed and more abundantly bound to chromatin in the transformed compared to normal cells. Interestingly, the 20mer1, 20mer2, and c-myc origins exhibited a two- to threefold greater origin activity and a two- to threefold greater in vivo association of the pre-RC proteins with these origins in the transformed cells, whereas the origin associated with the housekeeping lamin B2 gene exhibited both similar levels of activity and in vivo association of these pre-RC proteins in both cell types. Overall, the results indicate that cellular transformation is associated with an overexpression and increased chromatin association of the pre-RC proteins. This study is significant, because it represents the most systematic comprehensive analysis done to date, using multiple replication proteins and different replication origins in both normal and transformed cell lines.


Subject(s)
Proteins/metabolism , Replication Origin , Blotting, Western , Cell Line , Cell Line, Transformed , Chromatin/metabolism , DNA/isolation & purification , Gene Dosage , Humans , Immunoprecipitation , Real-Time Polymerase Chain Reaction
7.
J Cell Biochem ; 110(2): 281-7, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20336691

ABSTRACT

Tumor suppressor genes are frequently inactivated in cancer by large-scale deletion events or epigenetic silencing, and experimental demonstration of such inactivation has historically been considered as support for assigning tumor suppressive function to a given gene. However, the discovery of a number of chromosomal domains wherein large deletions naturally occur at frequencies up to 100 times the average for the genome as a whole leads us to reevaluate the significance of sporadic deletions found within genes associated with these hotspots. Similarly, our recent demonstration that epigenetic chromatin silencing frequently spreads in cancer cells from gene-poor into gene-rich regions with apparent indifference to the gene content of the affected domain raises questions about the pertinence of inactivation as a criterion for ascribing tumor suppressor function to a given gene. We suggest that a number of putative suppressor genes for which inactivation and/or deletion events have been documented may simply be victims of collateral damage when these events occur, and the implication that these genes are being selected against during cancer progression should in some cases be reassessed.


Subject(s)
Gene Deletion , Genes, Tumor Suppressor , Chromatin/metabolism , Chromosomes, Human, Pair 3 , Epigenesis, Genetic , Gene Silencing , Humans , Neoplasms/genetics
8.
Nucleic Acids Res ; 38(7): 2314-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20064876

ABSTRACT

Using libraries of replication origins generated previously, we identified three clones that supported the autonomous replication of their respective plasmids in transformed, but not in normal cells. Assessment of their in vivo replication activity by in situ chromosomal DNA replication assays revealed that the chromosomal loci corresponding to these clones coincided with chromosomal replication origins in all cell lines, which were more active by 2-3-fold in the transformed by comparison to the normal cells. Evaluation of pre-replication complex (pre-RC) protein abundance at these origins in transformed and normal cells by chromatin immunoprecipitation assays, using anti-ORC2, -cdc6 and -cdt1 antibodies, showed that they were bound by these pre-RC proteins in all cell lines, but a 2-3-fold higher abundance was observed in the transformed by comparison to the normal cells. Electrophoretic mobility shift assays (EMSAs) performed on the most efficiently replicating clone, using nuclear extracts from the transformed and normal cells, revealed the presence of a DNA replication complex in transformed cells, which was barely detectable in normal cells. Subsequent supershift EMSAs suggested the presence of transformation-specific complexes. Mass spectrometric analysis of these complexes revealed potential new protein players involved in DNA replication that appear to correlate with cellular transformation.


Subject(s)
Cell Transformation, Neoplastic , DNA Replication , Replication Origin , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Transformed , Cloning, Molecular , DNA/analysis , DNA Copy Number Variations , Genetic Loci , Genome, Human , Humans , Mass Spectrometry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Plasmids/genetics
9.
J Cell Sci ; 121(Pt 5): 590-600, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18252799

ABSTRACT

The Ku protein (Ku70-Ku80) is involved in various genome-maintenance processes such as DNA replication and repair, telomere maintenance, and chromosomal stability. We previously found that Ku80 is implicated in the loading of members of the pre-replicative complex (pre-RC) onto replication origins. Here, we report that acute reduction of Ku80 to 10% of its normal levels leads to impaired DNA replication and activation of a replication stress checkpoint. In the absence of Ku80, decreased levels of the initiator proteins Orc1 and Orc6 as well as reduced chromatin binding of Orc1, Orc4 and Cdc45 were observed, leading to decreased origin firing, whereas Orc2 and Orc3 were unaffected. Prolonged perturbation of DNA replication caused the block of cell-cycle progression in late G1 phase with low Cdk2 activity due to increased p21 expression and decreased Cdc25A and Cdk2 levels. The data suggest the interplay between the DNA-replication and cell-cycle machineries and shed light on a new role of Ku in G1-S transition.


Subject(s)
Antigens, Nuclear/metabolism , Cell Cycle Proteins/genetics , Cell Proliferation , DNA Replication/genetics , DNA-Binding Proteins/metabolism , G1 Phase/genetics , S Phase/genetics , Antigens, Nuclear/genetics , Cell Cycle/genetics , Cell Division/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Gene Expression Regulation/genetics , Genes, cdc/physiology , HeLa Cells , Humans , Ku Autoantigen , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , RNA Interference/physiology
10.
J Cell Biochem ; 104(3): 773-84, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18181156

ABSTRACT

DNA replication initiates at origins within the genome. The late-firing murine adenosine deaminase (mAdA) origin is located within a 2 kb fragment of DNA, making it difficult to examine by realtime technology. In this study, fine mapping of the mAdA region by measuring the abundance of nascent strand DNA identified two origins, mAdA-1 and mAdA-C, located 397 bp apart from each other. Both origins conferred autonomous replication to plasmids transfected in murine embryonic fibroblasts (MEFs), and exhibited similar activities in vivo and in vitro. Furthermore, both were able to recruit the DNA replication initiator proteins Cdc6 and Ku in vitro, similar to other bona fide replication origins. When tested in a murine Ku80(-/-) cell line, both origins exhibited replication activities comparable to those observed in wildtype cells, as did the hypoxanthine-guanine phosphoribosyltransferase (HPRT) and c-myc origins. This contrasts with previously published studies using Ku80-deficient human cells lines and suggests differences in the mechanism of initiation of DNA replication between the murine and human systems.


Subject(s)
Adenosine Deaminase/metabolism , Fibroblasts/cytology , Adenosine Deaminase/genetics , Animals , Antigens, Nuclear/biosynthesis , Base Sequence , Cell Line , Chromosome Mapping/methods , DNA Replication , DNA-Binding Proteins/biosynthesis , Fibroblasts/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Ku Autoantigen , Mice , Molecular Sequence Data , Proto-Oncogene Proteins c-myc/metabolism , Replication Origin , Reverse Transcriptase Polymerase Chain Reaction
11.
J Cell Biochem ; 99(6): 1606-15, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16823771

ABSTRACT

We previously isolated from African green monkey (CV-1) cells a replication origin, ors8, that is active at the onset of S-phase. Here, its homologous sequence (hors8, accession number: DQ230978) was amplified from human cells, using the monkey-ors8-specific primers. Sequence alignment between the monkey and the human fragment revealed a 92% identity. Nascent DNA abundance analysis, involving quantification by real-time PCR, indicated that hors8 is an active replication origin, as the abundance of nascent DNA from a genomic region containing it was 97-fold higher relative to a non-origin region in the same locus. Furthermore, the data showed that the hors8 fragment is capable of supporting the episomal replication of its plasmid, when cloned into pBlueScript (pBS), as assayed by the DpnI resistance assay after transfection of HeLa cells. A quantitative chromatin immunoprecipitation (ChIP) assay, using antibodies against Ku, Orc2, and Cdc6, showed that these DNA replication initiator proteins were associated in vivo with the human ors8 (hors8). Finally, nascent DNA abundance experiments from human cells synchronized at different phases of the cell cycle revealed that hors8 is a late-firing origin of DNA replication, having the highest activity 8 h after release from late G(1).


Subject(s)
Replication Origin , Animals , Base Sequence , Chlorocebus aethiops , Chromatin Immunoprecipitation , DNA Primers , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
12.
Cancer Res ; 66(10): 5094-103, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707432

ABSTRACT

Previously, a degenerate 36 bp human consensus sequence was identified as a determinant of autonomous replication in eukaryotic cells. Random mutagenesis analyses further identified an internal 20 bp of the 36 bp consensus sequence as sufficient for acting as a core origin element. Here, we have located six versions of the 20 bp consensus sequence (20mer) on human chromosome 19q13 over a region spanning approximately 211 kb and tested them for ectopic and in situ replication activity by transient episomal replication assays and nascent DNA strand abundance analyses, respectively. The six versions of the 20mer alone were capable of supporting autonomous replication of their respective plasmids, unlike random genomic sequence of the same length. Furthermore, comparative analyses of the endogenous replication activity of these 20mers at their respective chromosomal sites, in five tumor/transformed and two normal cell lines, done by in situ chromosomal DNA replication assays, involving preparation of nascent DNA by the lambda exonuclease method and quantification by real-time PCR, showed that these sites coincided with chromosomal origins of DNA replication in all cell lines. Moreover, a 2- to 3-fold higher origin activity in the tumor/transformed cells by comparison to the normal cells was observed, suggesting a higher activation of these origins in tumor/transformed cell lines.


Subject(s)
DNA Replication/physiology , DNA, Neoplasm/biosynthesis , Neoplasms/genetics , Base Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomes, Human, Pair 19/genetics , Consensus Sequence , DNA, Neoplasm/genetics , Gene Dosage , HCT116 Cells , HeLa Cells , Humans , Neoplasms/metabolism , Plasmids/genetics
13.
Mol Cell Biol ; 25(9): 3814-30, 2005 May.
Article in English | MEDLINE | ID: mdl-15831485

ABSTRACT

The human stress-activated protein kin17 accumulates in the nuclei of proliferating cells with predominant colocalization with sites of active DNA replication. The distribution of kin17 protein is in equilibrium between chromatin-DNA and the nuclear matrix. An increased association with nonchromatin nuclear structure is observed in S-phase cells. We demonstrated here that kin17 protein strongly associates in vivo with DNA fragments containing replication origins in both human HeLa and monkey CV-1 cells. This association was 10-fold higher than that observed with nonorigin control DNA fragments in exponentially growing cells. In addition, the association of kin17 protein to DNA fragments containing replication origins was also analyzed as a function of the cell cycle. High binding of kin17 protein was found at the G(1)/S border and throughout the S phase and was negligible in both G(0) and M phases. Specific monoclonal antibodies against kin17 protein induced a threefold inhibition of in vitro DNA replication of a plasmid containing a minimal replication origin that could be partially restored by the addition of recombinant kin17 protein. Immunoelectron microscopy confirmed the colocalization of kin17 protein with replication proteins like RPA, PCNA, and DNA polymerase alpha. A two-step chromatographic fractionation of nuclear extracts from HeLa cells revealed that kin17 protein localized in vivo in distinct protein complexes of high molecular weight. We found that kin17 protein purified within an approximately 600-kDa protein complex able to support in vitro DNA replication by means of two different biochemical methods designed to isolate replication complexes. In addition, the reduced in vitro DNA replication activity of the multiprotein replication complex after immunodepletion for kin17 protein highlighted for a direct role in DNA replication at the origins.


Subject(s)
DNA Replication/physiology , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Replication Origin/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Cycle/physiology , Cell Nucleus/chemistry , Chromatin/metabolism , DNA Polymerase I/analysis , DNA Polymerase I/metabolism , DNA Replication/drug effects , DNA Replication/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Haplorhini , Humans , Microscopy, Immunoelectron , Multiprotein Complexes/physiology , Multiprotein Complexes/ultrastructure , Nuclear Proteins/analysis , Nuclear Proteins/antagonists & inhibitors , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/metabolism , RNA-Binding Proteins , Replication Protein A
SELECTION OF CITATIONS
SEARCH DETAIL
...