Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 291(36): 19079-91, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27422821

ABSTRACT

During the cell cycle, sister-chromatid cohesion tethers sister chromatids together from S phase to the metaphase-anaphase transition and ensures accurate segregation of chromatids into daughter cells. N-terminal acetylation is one of the most prevalent protein covalent modifications in eukaryotes and is mediated by a family of N-terminal acetyltransferases (NAT). Naa50 (also called San) has previously been shown to play a role in sister-chromatid cohesion in metazoans. The mechanism by which Naa50 contributes to cohesion is not understood however. Here, we show that depletion of Naa50 in HeLa cells weakens the interaction between cohesin and its positive regulator sororin and causes cohesion defects in S phase, consistent with a role of Naa50 in cohesion establishment. Strikingly, co-depletion of NatA, a heterodimeric NAT complex that physically interacts with Naa50, rescues the sister-chromatid cohesion defects and the resulting mitotic arrest caused by Naa50 depletion, indicating that NatA and Naa50 play antagonistic roles in cohesion. Purified recombinant NatA and Naa50 do not affect each other's NAT activity in vitro Because NatA and Naa50 exhibit distinct substrate specificity, we propose that they modify different effectors and regulate sister-chromatid cohesion in opposing ways.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatids/enzymology , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/enzymology , Mitosis/physiology , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , S Phase/physiology , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human/genetics , HeLa Cells , Humans , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , Cohesins
2.
J Biol Chem ; 288(23): 16518-16528, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23612983

ABSTRACT

Histone deacetylase 1 (HDAC1) and HDAC2 are components of corepressor complexes that are involved in chromatin remodeling and regulation of gene expression by regulating dynamic protein acetylation. HDAC1 and -2 form homo- and heterodimers, and their activity is dependent upon dimer formation. Phosphorylation of HDAC1 and/or HDAC2 in interphase cells is required for the formation of HDAC corepressor complexes. In this study, we show that during mitosis, HDAC2 and, to a lesser extent, HDAC1 phosphorylation levels dramatically increase. When HDAC1 and -2 are displaced from the chromosome during metaphase, they dissociate from each other, but each enzyme remains in association with components of the HDAC corepressor complexes Sin3, NuRD, and CoREST as homodimers. Enzyme inhibition studies and mutational analyses demonstrated that protein kinase CK2-catalyzed phosphorylation of HDAC1 and -2 is crucial for the dissociation of these two enzymes. These results suggest that corepressor complexes, including HDAC1 or HDAC2 homodimers, might target different cellular proteins during mitosis.


Subject(s)
Casein Kinase I/metabolism , Chromosomes, Human/enzymology , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Mitosis/physiology , Protein Multimerization/physiology , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/genetics , Chromosomes, Human/genetics , Co-Repressor Proteins , HEK293 Cells , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Sin3 Histone Deacetylase and Corepressor Complex/metabolism
4.
Cell Cycle ; 8(9): 1433-43, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19342897

ABSTRACT

Accurate segregation of chromosome, initiated by abrupt and irreversible dissolution of sister-chromatid cohesion at anaphase, is crucial for the faithful inheritance of parental genomes during eukaryotic cell division. The dissolution of sister-chromatid cohesion is catalyzed by separase after the destruction of securin by the anaphase-promoting complex/cyclosome (APC/C). However, separase was localized to the mitotic centrosome, raising the question as how separase hydrolyzes sister-chromatid cohesion of centromere at the anaphase onset. Here we show that separase is associated with mitotic chromosomes and this association is regulated by Aurora B kinase. Using a panel of separase antibodies, we found that separase protein was accumulated in mitosis and degraded at the end of telophase. To study the spatiotemporal distribution of separase in mitosis, we carried out immunofluorescence microscopic analyses. Surprisingly, separase was found to be associated with mitotic chromosomes from prophase to metaphase and dissociated from the chromosomes in anaphase right after sister chromatids separation. Staining of isolated mitotic chromosomes from Nocodazole-arrested cells revealed that separase is concentrated at the centromeric cohesion. To examine if any mitotic kinases are responsible for chromosomal localization of separase in mitosis, we carried out RNAi-mediated knockdown and found that association of separase with mitotic chromosomes was a function of Aurora B. Consistent with the phenotype seen in the Aurora B-repressed cells, inhibition of Aurora B kinase by hersperadin prevents the association of separase with chromosomes. Our results suggest that Aurora B kinase activity helps coordinate the association of separase with chromosome and the initiation of sister-chromatid separation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes, Human/enzymology , Endopeptidases/metabolism , Mitosis , Protein Serine-Threonine Kinases/metabolism , Antibodies , Aurora Kinase B , Aurora Kinases , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , HeLa Cells , Humans , Protein Transport , Reproducibility of Results , Separase , Cohesins
5.
Cell Cycle ; 7(18): 2894-901, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18787396

ABSTRACT

It is well established that B-Raf signaling through the MAP kinase (ERK) pathways plays a prominent role in regulating cell proliferation but how it does this is not completely understood. Here, we show that B-Raf serves a physiological role during mitosis in human somatic cells. Knockdown of B-Raf using short interfering RNA (siRNA) resulted in pleiotropic spindle abnormalities and misaligned chromosomes in over 80% of the mitotic cells analyzed. A second B-Raf siRNA gave similar results suggesting these effects are specific to downregulating B-Raf protein. In agreement with these findings, a portion of B-Raf was detected at the spindle structures including the spindle poles and kinetochores. Knockdown of C-Raf (Raf-1) had no detectable effects on spindle formation or chromosome alignment. Activation of the spindle assembly checkpoint was found to be dependent on B-Raf as evident by the inability of checkpoint proteins Bub1 and Mad2 to localize to unattached kinetochores in HeLa cells treated with B-Raf siRNA. Consistent with this, live-cell imaging microscopy showed that B-Raf-depleted cells exited mitosis earlier than control non-depleted cells. Finally, we provide evidence that B-Raf signaling promotes phosphorylation and kinetochore localization of the mitotic checkpoint kinase Mps1. Blocking B-Raf expression, ERK activity, or phosphorylation at Ser-821 residue perturbed Mps1 localization at unattached kinetochores. Thus, our data implicates a mitotic role for B-Raf in regulating spindle formation and the spindle checkpoint in human somatic cells.


Subject(s)
Fibroblasts/cytology , Fibroblasts/enzymology , Mitosis , Proto-Oncogene Proteins B-raf/deficiency , Spindle Apparatus/enzymology , Cell Cycle Proteins/metabolism , Chromosomes, Human/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Kinetochores/enzymology , Mutation/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Protein-Tyrosine Kinases , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
6.
DNA Repair (Amst) ; 7(10): 1624-35, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18603020

ABSTRACT

Thymidylate deprivation brings about "thymineless death" in prokaryotes and eukaryotes. Although the precise mechanism for thymineless death has remained elusive, inhibition of the enzyme thymidylate synthase (TS), which catalyzes the de novo synthesis of TMP, has served for many years as a basis for chemotherapeutic strategies. Numerous studies have identified a variety of cellular responses to thymidylate deprivation, including disruption of DNA replication and induction of DNA breaks. Since stalled or collapsed replication forks and strand breaks are generally viewed as being recombinogenic, it is not surprising that a link has been demonstrated between recombination induction and thymidylate deprivation in bacteria and lower eukaryotes. A similar connection between recombination and TS inhibition has been suggested by studies done in mammalian cells, but the relationship between recombination and TS inhibition in mammalian cells had not been demonstrated rigorously. To gain insight into the mechanism of thymineless death in mammalian cells, in this work we undertook a direct investigation of recombination in human cells treated with raltitrexed (RTX), a folate analog that is a specific inhibitor of TS. Using a model system to study intrachromosomal homologous recombination in cultured fibroblasts, we provide definitive evidence that treatment with RTX can stimulate accurate recombination events in human cells. Gene conversions not associated with crossovers were specifically enhanced several-fold by RTX. Additional experiments demonstrated that recombination events provoked by a double-strand break (DSB) were not impacted by treatment with RTX, nor was error-prone DSB repair via nonhomologous end-joining. Our work provides evidence that thymineless death in human cells is not mediated by corruption of DSB repair processes and suggests that an increase in chromosomal recombination may be an important element of cellular responses leading to thymineless death.


Subject(s)
Chromosomes, Human/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Folic Acid Antagonists/pharmacology , Quinazolines/pharmacology , Recombination, Genetic/drug effects , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Base Sequence , Blotting, Southern , Cell Line , Chromosomes, Human/enzymology , Crossing Over, Genetic/drug effects , DNA Breaks, Double-Stranded/drug effects , Fibroblasts/enzymology , Gene Conversion/drug effects , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Substrate Specificity/drug effects , Thymidylate Synthase/genetics
7.
Nat Rev Mol Cell Biol ; 8(10): 798-812, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17848966

ABSTRACT

Mitosis and meiosis are remarkable processes during which cells undergo profound changes in their structure and physiology. These events are orchestrated with a precision that is worthy of a classical symphony, with different activities being switched on and off at precise times and locations throughout the cell. One essential 'conductor' of this symphony is the chromosomal passenger complex (CPC), which comprises Aurora-B protein kinase, the inner centromere protein INCENP, survivin and borealin (also known as Dasra-B). Studies of the CPC are providing insights into its functions, which range from chromosome-microtubule interactions to sister chromatid cohesion to cytokinesis, and constitute one of the most dynamic areas of ongoing mitosis and meiosis research.


Subject(s)
Chromosomes, Human/genetics , Meiosis/genetics , Mitosis/genetics , Animals , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/enzymology , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Survivin
8.
Virus Genes ; 35(2): 215-23, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17546494

ABSTRACT

The HIV-1 LTR is regulated by multiple signaling pathways responsive to T cell activation. In this study, we have examined the contribution of the MAPK, calcineurin-NFAT and TNFalpha-NF-kappaB pathways on induction of chromosomally integrated HIV-1 LTR reporter genes. We find that induction by T-cell receptor (CD3) cross-linking and PMA is completely dependent upon a binding site for RBF-2 (USF1/2-TFII-I), known as RBEIII at -120. The MAPK pathway is essential for induction of the wild type LTR by these treatments, as the MEK inhibitors PD98059 and U0126 block induction by both PMA treatment and CD3 cross-linking. Stimulation of cells with ionomycin on its own has no effect on the integrated LTR, indicating that calcineurin-NFAT is incapable of causing induction in the absence of additional signals, but stimulation with both PMA and ionomycin produces a synergistic response. In contrast, stimulation of NF-kappaB by treatment with TNFalpha causes induction of both the wild type and RBEIII mutant LTRs, an effect that is independent of MAPK signaling. USF1, USF2 and TFII-I from unstimulated cells are capable of binding RBEIII in vitro, and furthermore can be observed on the LTR in vivo by chromatin imunoprecipitation from untreated cells. DNA binding activity of USF1/2 is marginally stimulated by PMA/ ionomycin treatment, and all three factors appear to remain associated with the LTR throughout the course of induction. These results implicate major roles for the MAPK pathway and RBF-2 (USF1/2-TFII-I) in coordinating events necessary for transition of latent integrated HIV-1 to active transcription in response to T cell signaling.


Subject(s)
HIV Long Terminal Repeat/genetics , HIV-1/genetics , MAP Kinase Signaling System/physiology , Transcription Factors, TFII/physiology , Upstream Stimulatory Factors/physiology , Virus Integration/genetics , ras Proteins/physiology , Chromosomes, Human/enzymology , Chromosomes, Human/virology , Gene Expression Regulation, Viral/physiology , Humans , Jurkat Cells , Lymphocyte Activation/physiology , Proviruses/enzymology , Proviruses/genetics , Proviruses/metabolism , T-Lymphocytes/enzymology , Transcription, Genetic/physiology
9.
J Cell Sci ; 120(Pt 7): 1245-55, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17356064

ABSTRACT

The assembly of mitotic chromosomes is controlled by condensin complexes. In vertebrates, condensin I binds to chromatin in prometaphase, confers rigidity to chromosomes and enables the release of cohesin complexes from chromosome arms, whereas condensin II associates with chromosomes in prophase and promotes their condensation. Both complexes are essential for chromosome segregation in anaphase. Although the association of condensins with chromatin is important for the assembly and segregation of mitotic chromosomes, it is poorly understood how this process is controlled. Here we show that the mitotic kinase Aurora B regulates the association of condensin I, but not the interaction of condensin II with chromatin. Quantitative time-lapse imaging of cells expressing GFP-tagged condensin subunits revealed that Aurora B is required for efficient loading of condensin I onto chromosomes in prometaphase and for maintenance of the complex on chromosomes in later stages of mitosis. The three non-SMC subunits of condensin I are Aurora B substrates in vitro and their mitosis-specific phosphorylation depends on Aurora B in vivo. Our data indicate that Aurora B contributes to chromosome rigidity and segregation by promoting the binding of condensin I to chromatin. We have also addressed how Aurora B might mediate the dissociation of cohesin from chromosome arms.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes, Human/enzymology , DNA-Binding Proteins/metabolism , Mitosis , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphatases/chemistry , Aurora Kinase B , Aurora Kinases , Chromatin/metabolism , Chromosomes, Human/metabolism , DNA-Binding Proteins/chemistry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Indoles/pharmacology , Kinetics , Microscopy, Video , Multiprotein Complexes/chemistry , Phosphorylation , Protein Subunits/chemistry , Substrate Specificity , Sulfonamides/pharmacology
10.
Oncogene ; 26(7): 945-57, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16909107

ABSTRACT

We previously identified SIRT2, an nicotinamide adenine dinucleotide (NAD)-dependent tubulin deacetylase, as a protein downregulated in gliomas and glioma cell lines, which are characterized by aneuploidy. Other studies reported SIRT2 to be involved in mitotic progression in the normal cell cycle. We herein investigated whether SIRT2 functions in the mitotic checkpoint in response to mitotic stress caused by microtubule poisons. By monitoring chromosome condensation, the exogenously expressed SIRT2 was found to block the entry to chromosome condensation and subsequent hyperploid cell formation in glioma cell lines with a persistence of the cyclin B/cdc2 activity in response to mitotic stress. SIRT2 is thus a novel mitotic checkpoint protein that functions in the early metaphase to prevent chromosomal instability (CIN), characteristics previously reported for the CHFR protein. We further found that histone deacetylation, but not the aberrant DNA methylation of SIRT2 5'untranslated region is involved in the downregulation of SIRT2. Although SIRT2 is normally exclusively located in the cytoplasm, the rapid accumulation of SIRT2 in the nucleus was observed after treatment with a nuclear export inhibitor, leptomycin B and ionizing radiation in normal human fibroblasts, suggesting that nucleo-cytoplasmic shuttling regulates the SIRT2 function. Collectively, our results suggest that the further study of SIRT2 may thus provide new insights into the relationships among CIN, epigenetic regulation and tumorigenesis.


Subject(s)
Chromosomal Instability/physiology , Histone Deacetylases/physiology , Mitosis/physiology , Sirtuins/physiology , Stress, Physiological/enzymology , Cell Line, Tumor , Chromosomal Instability/drug effects , Chromosomal Instability/radiation effects , Chromosomes, Human/drug effects , Chromosomes, Human/enzymology , Chromosomes, Human/radiation effects , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Histone Deacetylase Inhibitors , Humans , Mitosis/drug effects , Mitosis/radiation effects , Nocodazole/pharmacology , Paclitaxel/pharmacology , Polyploidy , Sirtuin 2 , Sirtuins/antagonists & inhibitors , Sirtuins/genetics , Stress, Physiological/chemically induced , Stress, Physiological/pathology , Tubulin/physiology , Ultraviolet Rays , X-Rays
11.
Biochem Biophys Res Commun ; 336(3): 807-12, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16153602

ABSTRACT

A variant of histone H2A, H2AX, is phosphorylated on Ser139 in response to DNA double-strand breaks (DSBs), and clusters of the phosphorylated form of H2AX (gamma-H2AX) in nuclei of DSB-induced cells show foci at breakage sites. Here, we show phosphorylation of H2AX in a cell cycle-dependent manner without any detectable DNA damage response. Western blot and immunocytochemical analyses with the anti-gamma-H2AX antibody revealed that H2AX is phosphorylated at M phase in HeLa cells. In ataxia-telangiectasia cells lacking ATM kinase activity, gamma-H2AX was scarcely detectable in the mitotic chromosomes, suggesting involvement of ATM in M-phase phosphorylation of H2AX. Single-cell gel electrophoresis assay and Western blot analysis with the anti-phospho-p53 (Ser15) antibody indicated that H2AX in human M-phase cells is phosphorylated independently of DSB and DNA damage signaling. Even in the absence of DNA damage, phosphorylation of H2AX in normal cell cycle progression may contribute to maintenance of genomic integrity.


Subject(s)
DNA Damage , Histones/metabolism , Mitosis , Ataxia Telangiectasia Mutated Proteins , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human/enzymology , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
12.
Nucleic Acids Res ; 33(6): 1813-24, 2005.
Article in English | MEDLINE | ID: mdl-15800211

ABSTRACT

Numerous lines of evidence support the role of oxidative stress in different types of cancer. A major DNA lesion, 8-oxo-7,8-dihydroguanine (8-oxoG), is formed by reactive oxygen species in the genome under physiological conditions. 8-OxoG is strongly mutagenic, generating G.C-->T.A transversions, a frequent somatic mutation in cancers. hOGG1 was cloned as a gene encoding a DNA glycosylase that specifically recognizes and removes 8-oxoG from 8-oxoG:C base pairs and suppresses G.C-->T.A transversions. In this study, we investigated the subcellular localization and expression of hOGG1 during the cell cycle. Northern blots showed cell-cycle-dependent mRNA expression of the two major hOGG1 isoforms. By using a cell line constitutively expressing hOGG1 fused to enhanced green fluorescence protein (EGFP), we observed a dynamic relocalization of EGFP-hOGG1 to the nucleoli during the S-phase of the cell cycle, and this localization was shown to be linked to transcription. A C/G change that results in an amino acid substitution from serine to cysteine in codon 326 has been reported as a genetic polymorphism and a risk allele for a variety of cancers. We investigated the cellular localization of the corresponding protein, hOGG1-Cys326, fused to EGFP and observed a dramatic effect on its localization that is explained by a change in the phosphorylation status of hOGG1.


Subject(s)
Cell Nucleolus/enzymology , DNA Glycosylases/genetics , Polymorphism, Single Nucleotide , S Phase , Amino Acid Substitution , Cell Cycle , Chromatin/enzymology , Chromosomes, Human/enzymology , DNA Glycosylases/analysis , DNA Glycosylases/metabolism , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Nuclear Matrix/enzymology , Phosphorylation , Serine/metabolism , Transcription, Genetic
13.
J Cell Sci ; 117(Pt 7): 1047-54, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14996935

ABSTRACT

DNA topoisomerase II (topo II) plays a crucial role in controlling the conformation of both DNA and whole chromosomes. This activity is essential for several cellular events such as DNA replication, transcription, chromosome condensation and segregation. In mammals, two genes code for isoforms of topo II, termed alpha and beta. They are similar in primary structure and have almost identical catalytic properties in vitro. We transfected HeLa cells with small interfering RNAs (siRNAs) targeted against either topo IIalpha or IIbeta, and succeeded in knocking down the expression of the corresponding protein. Chromosomes were condensed and aligned at metaphase in topo IIalpha-knockdown cells. Although some lagging chromosomes were observed, they were still segregated at anaphase despite the absence of topo IIalpha. When both topo IIalpha and topo IIbeta were removed, the segregation of chromosomes was severely arrested, suggesting that topo IIbeta could partially substitute for topo IIalpha. Double-knockdown experiments also revealed that topo II was required for shortening of the chromosome axis.


Subject(s)
DNA Topoisomerases, Type II/metabolism , Antigens, Neoplasm , Chromosome Segregation/physiology , Chromosomes, Human/enzymology , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins , HeLa Cells , Humans , Mitosis , RNA, Small Interfering/genetics , Topoisomerase II Inhibitors , Transfection
14.
Nucleic Acids Res ; 30(23): 5318-27, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12466558

ABSTRACT

The essential Aurora B kinase is a chromosomal passenger protein that is required for mitotic chromosome alignment and segregation. Aurora B function is dependent on the chromosome passenger, INCENP. INCENP, in turn, requires sister chromatid cohesion for its appropriate behaviour. Relatively few substrates have been identified for Aurora B, so that the precise role it plays in controlling mitosis remains to be elucidated. To identify potential novel mitotic substrates of Aurora B, extracted chromosomes were prepared from mitotically-arrested HeLa S3 cells and incubated with recombinant human Aurora B in the presence of radioactive ATP. Immunoblot analysis confirmed the HeLa scaffold fraction to be enriched for known chromosomal proteins including CENP-A, CENP-B, CENP-C, ScII and INCENP. Mass spectrometry of bands excised from one-dimensional polyacrylamide gels further defined the protein composition of the extracted chromosome fraction. Cloning, fluorescent tagging and expression in HeLa cells of the putative GTP-binding protein NGB/CRFG demonstrated it to be a novel mitotic chromosome protein, with a perichromosomal localisation. Identi fication of the protein bands corresponding to those phosphorylated by Aurora B revealed topoisomerase II alpha (topo IIalpha) as a potential Aurora B substrate. Purified recombinant human topo IIalpha was phosphorylated by Aurora B in vitro, confirming this proteomic approach as a valid method for the initial definition of candidate substrates of key mitotic kinases.


Subject(s)
Chromosomes, Human/enzymology , DNA Topoisomerases, Type II/metabolism , Protein Serine-Threonine Kinases/metabolism , Antigens, Neoplasm , Aurora Kinase B , Aurora Kinases , Chromosomes, Human/chemistry , DNA Topoisomerases, Type II/analysis , DNA Topoisomerases, Type II/immunology , DNA-Binding Proteins , Fluorescent Antibody Technique , GTP-Binding Proteins/analysis , HeLa Cells , Humans , Metaphase , Nuclear Proteins/analysis , Nuclear Proteins/classification , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/immunology , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Biochem Biophys Res Commun ; 278(2): 385-9, 2000 Nov 19.
Article in English | MEDLINE | ID: mdl-11097846

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) takes part mainly in regulation of DNA repair, thereby maintaining genomic stability in the nucleus. However, what role PARP plays in mitotic cells is not known. Centrosomes play an important role in maintaining the fidelity of chromosome distribution during cell division. Loss of these functions might cause chromosomal instability and aneuploidy. p53 and BRCA1 were recently found to localize to the centrosome at mitosis. We found that PARP is localized to the centrosomes and the chromosomes at cell-division phase and interphase by indirect immunofluorescence. Furthermore, by analysis of isolated centrosomes PARP protein was found to associate with the centrosomes during mitosis. These data suggest that PARP may be involved in maintenance of chromosomal stability.


Subject(s)
Centrosome/enzymology , Chromosomes, Human/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Cell Line , Fluorescent Antibody Technique, Indirect , Humans , Mitosis , Tumor Cells, Cultured
18.
Exp Cell Res ; 247(1): 29-37, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10047445

ABSTRACT

Telomere maintenance is thought to be essential for immortalization of human cancer cells to compensate for the loss of DNA from the ends of chromosomes and to prevent chromosome fusion. We have investigated telomere dynamics in the telomerase-positive squamous cell carcinoma cell line SCC-61 by marking the ends of chromosomes with integrated plasmid sequences so that changes in the length of individual telomeres could be monitored. Despite having very short telomeres, SCC-61 has a relatively stable genome and few telomere associations. The marked telomeres in different SCC-61 clones have similar mean lengths which show little change with increasing time in culture. Thus, each marked telomere is maintained at a specific length, which we term the equilibrium mean length (EML). The Gaussian distribution in the length of the marked telomeres demonstrates that telomeres continuously fluctuate in length. Consistent with this observation, the mean lengths of the marked telomere in subclones of these cell lines initially differ, but then gradually return to the EML of the original clone with increasing time in culture. The analysis of a clone with two marked telomeres demonstrated that changes in telomere length can occur on each marked telomere independently or coordinately on both telomeres. These results suggest that the short telomeres in many tumor cell lines do not result from an inability to properly maintain telomeres at a specific length.


Subject(s)
Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Telomere/chemistry , Carcinoma, Squamous Cell/enzymology , Chromosomes, Human/enzymology , Chromosomes, Human/genetics , Clone Cells/chemistry , Clone Cells/enzymology , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Telomerase/chemistry , Telomere/enzymology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...