Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
Add more filters










Publication year range
1.
Chromosome Res ; 21(2): 101-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23580138

ABSTRACT

The first centromeric protein identified in any species was CENP-A, a divergent member of the histone H3 family that was recognised by autoantibodies from patients with scleroderma-spectrum disease. It has recently been suggested to rename this protein CenH3. Here, we argue that the original name should be maintained both because it is the basis of a long established nomenclature for centromere proteins and because it avoids confusion due to the presence of canonical histone H3 at centromeres.


Subject(s)
Autoantigens/genetics , Chromosomal Proteins, Non-Histone/genetics , Histones/genetics , Autoantigens/metabolism , Centromere , Centromere Protein A , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Humans , Kinetochores , Scleroderma, Systemic/genetics , Terminology as Topic
3.
Cell Motil Cytoskeleton ; 63(10): 646-57, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16937396

ABSTRACT

Exposure to the natural mineral fiber asbestos causes severe lung-damaging fibrosis and cancer, yet it continues to be used as an industrial insulating material throughout the world. When cultured human lung cells are exposed to asbestos, individual fibers are engulfed into the cytoplasm where they induce significant mitotic aberrations leading to chromosomal instability and aneuploidy. The mechanisms of how asbestosis ultimately leads to lung cancer remain unclear. However, our experiments indicate that intracellular asbestos fibers induce aneuploidy and chromosome instability by binding to a subset of proteins that include regulators of the cell cycle, cytoskeleton, and mitotic process. Moreover, precoating of fibers with protein complexes efficiently blocked asbestos-induced aneuploidy in human lung cells without affecting their uptake by cells. These results provide new evidence that asbestos fibers can contribute to significant spindle damage and chromosomal instability by binding to proteins needed for the assembly and regulation of the cytoskeleton or the cell cycle.


Subject(s)
Aneuploidy , Asbestos/metabolism , Fibroblasts/metabolism , Lung/metabolism , Cells, Cultured , Chromosome Aberrations , Humans , Protein Binding
5.
Cell Motil Cytoskeleton ; 55(2): 134-46, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12740874

ABSTRACT

Aurora-A kinase, also known as STK15/BTAK kinase, is a member of a serine/threonine kinase superfamily that includes the prototypic yeast Ipl1 and Drosophila aurora kinases as well as other mammalian and non-mammalian aurora kinases involved in the regulation of centrosomes and chromosome segregation. The Aurora-A gene is amplified and overexpressed in a wide variety of human tumors. Aurora-A is centrosome-associated during interphase, and binds the poles and half-spindle during mitosis; its over-expression has been associated with centrosome amplification and multipolar spindles. GFP-Aurora-A was used to mark centrosomes and spindles, and monitor their movements in living cells. Centrosome pairs labeled with GFP-Aurora-A are motile throughout interphase undergoing oscillations and tumbling motions requiring intact microtubules and ATP. Fluorescence recovery after photobleaching (FRAP) was used to examine the relative molecular mobility of GFP-Aurora-A, and GFP-labeled alpha-tubulin, gamma-tubulin, and NuMA. GFP-Aurora-A rapidly exchanges in and out of the centrosome and mitotic spindle (t(1/2) approximately 3 sec); in contrast, both tubulins are relatively immobile indicative of a structural role. GFP-NuMA mobility was intermediate in both interphase nuclei and at the mitotic spindle (t(1/2) approximately 23-30 sec). Deletion mapping identifies a central domain of Aurora-A as essential for its centrosomal localization that is augmented by both the amino and the carboxyl terminal ends of the protein. Interestingly, amino or carboxy terminal deletion mutants that maintained centrosomal targeting exhibited significantly slower molecular exchange. Collectively, these studies contrast the relative cellular dynamics of Aurora-A with other cytoskeletal proteins that share its micro-domains, and identify essential regions required for targeting and dynamics.


Subject(s)
Centrosome/enzymology , Neoplasms/enzymology , Protein Kinases/metabolism , Spindle Apparatus/enzymology , Antigens, Nuclear , Aurora Kinase A , Aurora Kinases , Cell Cycle Proteins , Cell Division/genetics , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins , Mutation/genetics , Neoplasms/genetics , Nuclear Matrix-Associated Proteins , Nuclear Proteins/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Protein Transport/physiology , Recombinant Fusion Proteins , Tubulin/metabolism , Xenopus Proteins
6.
Exp Cell Res ; 285(2): 175-88, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12706113

ABSTRACT

A centromere-specific variant of histone H3, centromere protein A (CENP-A), is a critical determinant of centromeric chromatin, and its location on the chromosome may determine centromere identity. To search for factors that direct CENP-A deposition at a specific chromosomal locus, we took advantage of the observation that CENP-A, when expressed at elevated levels, can get incorporated at ectopic sites on the chromosome, in addition to the centromere. As core histone hypoacetylation and DNA replication timing have been implicated as epigenetic factors that may be important for centromere identity, we hypothesized that the sites of preferential CENP-A deposition will be distinguished by these parameters. We found that, on human dicentric chromosomes, ectopically expressed CENP-A preferentially incorporates at the active centromere only, despite the fact that the levels of histone acetylation and replication timing were indistinguishable at the two centromeres. In CHO cells, ectopically expressed CENP-A is preferentially targeted to some, but not all telomeric regions. Again, these regions could not be distinguished from other telomeres by their acetylation levels or replication timing. Thus histone acetylation and replication timing are not sufficient for specifying the sites of CENP-A deposition and likely for centromere identity.


Subject(s)
Autoantigens , Centromere/chemistry , Chromosomal Proteins, Non-Histone/analysis , DNA Replication , Histones/metabolism , 3T3 Cells , Acetylation , Animals , CHO Cells , Cell Line , Centromere Protein A , Chromosomes/chemistry , Cricetinae , HeLa Cells , Humans , Kinetics , Mice , Microscopy, Fluorescence , Telomere/chemistry
7.
Chromosome Res ; 10(4): 267-77, 2002.
Article in English | MEDLINE | ID: mdl-12199140

ABSTRACT

Proper cohesion of sister chromatids is prerequisite for correct segregation of chromosomes during cell division. The cohesin multiprotein complex, conserved in eukaryotes, is required for sister chromatid cohesion. Human cohesion is composed of a stable heterodimer of the structural maintenance of chromosomes (SMC) family proteins, hSMC1 and hSMC3, and non-SMC components, hRAD21 and SA1 (or SA2). In yeast, cohesion associates with chromosomes from late G1 to metaphase and is required for the establishment and maintenance of both chromosome arm and centromeric cohesion. However, in human cells, the majority of cohesion dissociates from chromosomes before mitosis. Although it was recently shown that a small amount of hRAD21 localizes to the centromeres during metaphase, the presence of other cohesion components at the centromere has not been demonstrated in human cells. Here we report the mitosis-specific localization of hSMC1 to the kinetochores. hSMC1 is targeted to the kinetochore region during prophase concomitant with kinetochore assembly and remains through anaphase. Importantly, hSMC1 is targeted only to the active centromere on dicentric chromosomes. These results suggest that hSMC1 is an integral component of the functional kinetochore structure during mitosis.


Subject(s)
Cell Cycle Proteins/analysis , Chromosomal Proteins, Non-Histone/analysis , Kinetochores/chemistry , Antibody Specificity , Burkitt Lymphoma , Cell Cycle Proteins/isolation & purification , Cell Division , Chromosomal Proteins, Non-Histone/isolation & purification , Fluorescent Antibody Technique , HeLa Cells , Humans , Kinetochores/physiology , Mitosis , Tumor Cells, Cultured
8.
J Biol Chem ; 277(8): 6391-8, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11698396

ABSTRACT

Protein phosphatase 4 (PP4, previously named protein phosphatase X (PPX)), a PP2A-related serine/threonine phosphatase, has been shown to be involved in essential cellular processes, such as microtubule growth and nuclear factor kappa B activation. We provide evidence that PP4 is involved in tumor necrosis factor (TNF)-alpha signaling in human embryonic kidney 293T (HEK293T) cells. Treatment of HEK293T cells with TNF-alpha resulted in time-dependent activation of endogenous PP4, peaking at 10 min, as well as increased serine and threonine phosphorylation of PP4. We also found that PP4 is involved in relaying the TNF-alpha signal to c-Jun N-terminal kinase (JNK) as indicated by the ability of PP4-RL, a dominant-negative PP4 mutant, to block TNF-alpha-induced JNK activation. Moreover, the response of JNK to TNF-alpha was inhibited in HEK293 cells stably expressing PP4-RL in comparison to parental HEK293 cells. The involvement of PP4 in JNK signaling was further demonstrated by the specific activation of JNK, but not p38 and ERK2, by PP4 in transient transfection assays. However, no direct PP4-JNK interaction was detected, suggesting that PP4 exerts its positive regulatory effect on JNK in an indirect manner. Taken together, these data indicate that PP4 is a signaling component of the JNK cascade and involved in relaying the TNF-alpha signal to the JNK pathway.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Phosphoprotein Phosphatases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Kidney , Kinetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mutagenesis , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , p38 Mitogen-Activated Protein Kinases
9.
Biochem Biophys Res Commun ; 289(1): 173-80, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11708796

ABSTRACT

The c-Jun N-terminal kinase (JNK), a subgroup of the mitogen-activated protein kinase (MAPK) family of serine/threonine kinases, has established functions in cell growth and apoptosis. While the mechanisms are unclear, JNK has also been also implicated in signaling pathways that initiate cell cycle checkpoints and cell cycle progression. By following the localization of active and inactive JNK during the cell cycle, we have found that the majority of cellular JNK is soluble and present in the cytoplasm and the nucleus. Interestingly, insoluble fractions of JNK are also localized in nuclear and cytoplasmic speckles, and to the centrosomes. While JNK is associated with the centrosome throughout the cell cycle, it is only active at the centrosome from S phase through anaphase. This novel localization of centrosomal JNK is a possible link between JNK-activating stimuli and centrosome or cell cycle events.


Subject(s)
Cell Cycle/physiology , Centrosome/enzymology , Mitogen-Activated Protein Kinases/metabolism , Animals , Antibody Specificity , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/immunology , Rabbits , Signal Transduction , Solubility
10.
Mol Biol Cell ; 12(11): 3527-37, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11694586

ABSTRACT

Condensin is a conserved 13S heteropentamer composed of two nonidentical structural maintenance of chromosome (SMC) family proteins, in Xenopus XCAP-C and XCAP-E, and three regulatory subunits, XCAP-D2, XCAP-G, and XCAP-H. Both biochemical and genetic analyses have demonstrated an essential role for the 13S condensin complex in mitotic chromosome condensation. Further, a potential requirement for condensin in completion of chromatid arm separation in early anaphase is demonstrated by the mutational phenotypes of the Drosophila homologues of XCAP-H, barren and XCAP-C, DmSMC4. In this study we have investigated the expression and subcellular distribution of hCAP-H, the human homolog of XCAP-H, in order to better understand its cellular functions. Transcription of hCAP-H was restricted to proliferating cells with highest expression during the G(2) phase of the cell cycle. In contrast, cellular hCAP-H protein levels were constant throughout the cell cycle. hCAP-H was found to be associated with mitotic chromosomes exhibiting a nonuniform but symmetric distribution along sister chromatids. The symmetry of hCAP-H association with sister chromatids suggests that there are sequence-dependent domains of condensin aggregation. During interphase hCAP-H, -C, and -E, have distinct punctate nucleolar localization, suggesting that condensin may associate with and modulate the conformation and function of rDNA. hCAP-H association with condensed chromatin was not observed in the early phase of chromosome condensation when histone H3 phosphorylation has already taken place. This finding is consistent with the hypothesis that histone H3 phosphorylation precedes condensin-mediated condensation.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Nucleolus/metabolism , Gene Expression , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Cycle , Cell Line, Transformed , Cells, Cultured , Chromatin/metabolism , Conserved Sequence , Evolution, Molecular , HL-60 Cells , HeLa Cells , Histones/metabolism , Humans , Interphase , Jurkat Cells , K562 Cells , Mitosis , Molecular Sequence Data , Nuclear Proteins/genetics , Phosphorylation , Rabbits , Sequence Homology, Amino Acid
11.
J Cell Sci ; 114(Pt 19): 3529-42, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11682612

ABSTRACT

The mechanisms that specify precisely where mammalian kinetochores form within arrays of centromeric heterochromatin remain largely unknown. Localization of CENP-A exclusively beneath kinetochore plates suggests that this distinctive histone might direct kinetochore formation by altering the structure of heterochromatin within a sub-region of the centromere. To test this hypothesis, we experimentally mistargeted CENP-A to non-centromeric regions of chromatin and determined whether other centromere-kinetochore components were recruited. CENP-A-containing non-centromeric chromatin assembles a subset of centromere-kinetochore components, including CENP-C, hSMC1, and HZwint-1 by a mechanism that requires the unique CENP-A N-terminal tail. The sequence-specific DNA-binding protein CENP-B and the microtubule-associated proteins CENP-E and HZW10 were not recruited, and neocentromeric activity was not detected. Experimental mistargeting of CENP-A to inactive centromeres or to acentric double-minute chromosomes was also not sufficient to assemble complete kinetochore activity. The recruitment of centromere-kinetochore proteins to chromatin appears to be a unique function of CENP-A, as the mistargeting of other components was not sufficient for assembly of the same complex. Our results indicate at least two distinct steps in kinetochore assembly: (1) precise targeting of CENP-A, which is sufficient to assemble components of a centromere-prekinetochore scaffold; and (2) targeting of kinetochore microtubule-associated proteins by an additional mechanism present only at active centromeres.


Subject(s)
Autoantigens , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Kinetochores/metabolism , Amino Acid Sequence , Animals , CHO Cells , Centromere Protein A , Centromere Protein B , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cricetinae , Gene Expression , HeLa Cells , Histones , Humans , Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Molecular Sequence Data , Protein Structure, Tertiary , Transfection
12.
Trends Cell Biol ; 11(1): 18-21, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11146294

ABSTRACT

Aneuploid tumor cells can arise through multipolar mitosis caused by supernumerary centrosomes. Multipolar spindles, however, are antagonistic to cell viability. Thus, most cells derived from such an aberrant mitosis would be eliminated by apoptosis. A rare daughter cell, through chance acquisition of an appropriate chromosome complement and/or gene dosage, could survive and contribute to a clone of aneuploid tumor cells. Survival and perpetuation of the clone, however, requires an additional step - the resumption of mitotic stability through the assembly of a bipolar, not multipolar, spindle. Either selective inactivation of the extra centrosomes or their coalescence into two functional spindle poles corrects the problem of centrosome excess. Current data support coalescence as a mechanism for regulating the number of functional centrosomes in tumor cells.


Subject(s)
Aneuploidy , Centrosome , Neoplasms/genetics , Animals , Cell Division , Humans , Neoplasms/pathology , Tumor Cells, Cultured
13.
FASEB J ; 14(14): 2221-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053243

ABSTRACT

Mammary epithelial cells from p53 null mice have been shown recently to exhibit an increased risk for tumor development. Hormonal stimulation markedly increased tumor development in p53 null mammary cells. Here we demonstrate that mammary tumors arising in p53 null mammary cells are highly aneuploid, with greater than 70% of the tumor cells containing altered chromosome number and a mean chromosome number of 56. Normal mammary cells of p53 null genotype and aged less than 14 wk do not exhibit aneuploidy in primary cell culture. Significantly, the hormone progesterone, but not estrogen, increases the incidence of aneuploidy in morphologically normal p53 null mammary epithelial cells. Such cells exhibited 40% aneuploidy and a mean chromosome number of 54. The increase in aneuploidy measured in p53 null tumor cells or hormonally stimulated normal p53 null cells was not accompanied by centrosome amplification. These results suggest that normal levels of progesterone can facilitate chromosomal instability in the absence of the tumor suppressor gene, p53. The results support the emerging hypothesis based both on human epidemiological and animal model studies that progesterone markedly enhances mammary tumorigenesis.


Subject(s)
Aneuploidy , Epithelial Cells/drug effects , Mammary Glands, Animal/drug effects , Progesterone/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Cells, Cultured , Centrosome/drug effects , Centrosome/metabolism , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Mutation , Tumor Cells, Cultured
14.
Curr Top Dev Biol ; 49: 331-42, 2000.
Article in English | MEDLINE | ID: mdl-11005026

ABSTRACT

Because of the well-known role of the centrosome and mitotic apparatus in genome partitioning in normal cells, defects in pathways essential for mitotic regulation are likely implicated in the cascade of events leading to aneuploidy and neoplasia. Exogenous overexpression of AIM-1, for example, produces multinuclearity in human cells and increased ploidy as well as aneuploidy (Tatsuka et al., 1998). Overexpression in colorectal tumor cell lines is thought to have a causal relationship with multinuclearity and increased ploidy. Cytokinesis error caused by AIM-1 overexpression is a major factor in the predisposition to cancer. As previously mentioned, the involvement of BTAK/aur2/AIK in centrosome amplification and its oncogenic activity are compelling. Aur2 has also been implicated in oncogenesis, and defects in kinetochore function leading to chromosome instability in human tumors should not be minimized (Farruggio et al., 1999). Further studies are needed to provide a clearer definition of how these kinetic proteins are linked and regulated in normal mitosis and cancer. Thus, Boveri appears to have been correct in formulating his early hypothesis that a defective mitotic apparatus and centrosome number were central and causative in chromosome missegregation and cancer. One hundred years later, at the onset of a new millennium and with light-years of advanced technology in our favor, we are just now beginning to piece together the enzymes, substrates, and signaling pathways that support and explain his long-ignored but prophetic claim.


Subject(s)
Cell Cycle Proteins/physiology , Centrosome/physiology , Protein Serine-Threonine Kinases/physiology , Humans
15.
Oncogene ; 19(32): 3623-31, 2000 Jul 27.
Article in English | MEDLINE | ID: mdl-10951568

ABSTRACT

Mutations in the transforming growth factor beta type II receptor (TGFbetaRII) have been found in various malignant tumors, suggesting that loss of TGFbeta signaling plays a causal role in late-stage cancer development. To test whether loss of TGFbetaRII is involved in early-stage carcinogenesis, we have generated transgenic mice expressing a dominant negative TGFbetaRII (deltabetaRII) in the epidermis. These mice exhibited an increased susceptibility to chemical carcinogenesis protocols at both early and late stages. In the current study, parameters for cell cycle progression and chromosome instability were analysed in deltabetaRII tumors. DeltabetaRII papillomas showed an increased S phase in flow cytometry. Bromodeoxyuridine (BrdU) labeling and mitotic indices in deltabetaRII papillomas also showed a threefold increase compared to papillomas developing in non-transgenic mice. When papillomas further progressed to squamous cell carcinomas (SCC), both control and deltabetaRII SCC showed similar BrdU labeling indices and percentages of S phase cells. However, deltabetaRII SCC cells showed a sixfold increase in the G2/M population. Mitotic indices in deltabetaRII SCC also showed a threefold increase compared to non-transgenic SCC. Consistent with a perturbed cell cycle, deltabetaRII papillomas and SCC showed reduced expression of the TGFbeta target genes p15 (INK4b), p21 (WAF-1) and p27 (Kip1), inhibitors of cyclin-dependent kinases (cdks). However, most deltabetaRII papilloma cells exhibited normal centrosome numbers, and deltabetaRII SCC exhibited a similar extent of centrosome abnormalities compared to control SCC (35-40% cells). Most of deltabetaRII SCC exhibited diploid chromosome profiles. These data indicate that inactivation of TGFbetaRII accelerates skin tumorigenesis at early stages by the acceleration of loss of cell cycle control, but not by increased chromosome instability.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16 , Epidermal Cells , Papilloma/pathology , Receptors, Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins , 9,10-Dimethyl-1,2-benzanthracene/adverse effects , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Bromodeoxyuridine/pharmacokinetics , Carcinogens/adverse effects , Carcinogens/pharmacology , Carcinoma, Squamous Cell/chemically induced , Carrier Proteins/genetics , Cell Cycle , Centrosome , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p27 , Gene Expression , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Mitosis , Papilloma/chemically induced , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Tetradecanoylphorbol Acetate/adverse effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
16.
Mol Biol Cell ; 11(4): 1305-13, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10749931

ABSTRACT

This work describes BRN1, the budding yeast homologue of Drosophila Barren and Xenopus condensin subunit XCAP-H. The Drosophila protein is required for proper chromosome segregation in mitosis, and Xenopus protein functions in mitotic chromosome condensation. Mutant brn1 cells show a defect in mitotic chromosome condensation and sister chromatid separation and segregation in anaphase. Chromatid cohesion before anaphase is properly maintained in the mutants. Some brn1 mutant cells apparently arrest in S-phase, pointing to a possible function for Brn1p at this stage of the cell cycle. Brn1p is a nuclear protein with a nonuniform distribution pattern, and its level is up-regulated at mitosis. Temperature-sensitive mutations of BRN1 can be suppressed by overexpression of a novel gene YCG1, which is homologous to another Xenopus condensin subunit, XCAP-G. Overexpression of SMC2, a gene necessary for chromosome condensation, and a homologue of the XCAP-E condensin, does not suppress brn1, pointing to functional specialization of components of the condensin complex.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes/physiology , Drosophila Proteins , Mitosis/physiology , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Chromatids/physiology , DNA, Fungal/analysis , In Situ Hybridization, Fluorescence , Mitosis/genetics , Mutation , Nuclear Proteins/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics
17.
Nucleic Acids Res ; 27(15): 3001-8, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10454593

ABSTRACT

Phenotypes produced by gene overexpression may provide important clues to gene function. Here, we have performed a search for genes that affect chromo-some stability when overexpressed in the budding yeast Saccharomyces cerevisiae. We have obtained clones encompassing 30 different genes. Twenty-four of these genes have been previously characterized. Most of them are involved in chromatin dynamics, cell cycle control, DNA replication or mitotic chromosome segregation. Six novel genes obtained in this screen were named CST (chromosome stability). Based on the pattern of genomic instability, inter-action with checkpoint mutations and sensitivity to chromosome replication or segregation inhibitors, we conclude that overexpression of CST4 specifically interferes with mitotic chromosome segregation, and CST6 affects some aspect of DNA metabolism. The other CST genes had complex pleiotropic phenotypes. We have created deletions of five genes obtained in this screen, CST9, CST13, NAT1, SBA1 and FUN30. None of these genes is essential for viability, and deletions of NAT1 and SBA1 cause chromosome instability, a phenotype not previously associated with these genes. This work shows that analysis of dosage effects is complementary to mutational analysis of chromosome transmission fidelity, as it allows the identification of chromosome stability genes that have not been detected in mutational screens.


Subject(s)
Aneuploidy , Chromosome Segregation/genetics , Gene Dosage , Genes, Fungal/physiology , Genome, Fungal , Saccharomyces cerevisiae/genetics , Cell Cycle , Cloning, Molecular , Conserved Sequence/genetics , DNA Damage/genetics , DNA Replication/genetics , DNA, Fungal/biosynthesis , DNA, Fungal/metabolism , Gene Deletion , Gene Expression , Genes, Essential , Genes, Fungal/genetics , Humans , Nondisjunction, Genetic , Phenotype , Recombination, Genetic/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Temperature
18.
Cell Motil Cytoskeleton ; 43(2): 99-113, 1999.
Article in English | MEDLINE | ID: mdl-10379835

ABSTRACT

Platelet-activating factor (PAF), a phospholipid signaling molecule found in brain, modulates several neural functions and is implicated in the human developmental brain disorder Miller-Dieker Lissencephaly (MDL). Exposure to PAF, and a non-hydrolyzable analogue, methyl carbamyl PAF (mc-PAF), produces the following rapid, reversible effects upon cultured hippocampal neurites: growth cone collapse, neurite retraction, and neurite varicosity formation. In this study, the cytoskeletal alterations that mediate these shape changes were investigated by comparing the effects of mc-PAF with other cytoskeletal-altering drugs, through the fluorescent labeling of cytoskeletal proteins and mitochondria, and by electron microscopy. Results indicate that rearrangements of microtubules (MTs), F-actin, and mitochondria underlie the neurite shape changes produced by mc-PAF. Evidence for MT alteration was obtained by comparing the effects of mc-PAF with nocodozole and taxol. Exposure to nocodazole, a MT-depolymerizing agent, produced growth cone collapse and neurite varicosity formation similar to mc-PAF, whereas pre-incubation of neurites in taxol, a MT-stabilizing drug, was effective in blocking mc-PAF-induced neurite effects. Immunofluorescent labeling and EM revealed MT splaying and unbundling within neurite varicosities following mc-PAF treatment. Immunofluorescent labeling also revealed that F-actin shifted from concentration in the growth cone to a diffuse distribution along the neurite shaft following mc-PAF exposure. Fluorescent labeling and EM also revealed retrograde movement and morphological alterations of mitochondria following mc-PAF exposure, resulting in mitochondrial aggregates within neurite varicosities. These cytoskeletal rearrangements may provide insights into the mechanisms by which PAF influences neuronal activity, and could have important implications for the impairment of neuronal motility observed in MDL.


Subject(s)
Cytoskeleton/drug effects , Neurons/drug effects , Phospholipid Ethers/pharmacology , Platelet Activating Factor/analogs & derivatives , Actins/metabolism , Animals , Cells, Cultured , Cytochalasin D/pharmacology , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Fluorescent Antibody Technique, Indirect , Microtubules/drug effects , Microtubules/physiology , Mitochondria/drug effects , Mitochondria/physiology , Neurites/drug effects , Neurites/ultrastructure , Neurons/physiology , Neurons/ultrastructure , Nocodazole/pharmacology , Paclitaxel/pharmacology , Phospholipid Ethers/chemistry , Polymers , Rats , Rats, Sprague-Dawley
19.
FASEB J ; 13 Suppl 2: S216-20, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10619130

ABSTRACT

The centromere-kinetochore complex can be divided into distinct domains based on structure and function. Previous work has used CREST auto-antibodies with various microscopic techniques to map the locations of proteins within the centromere-kinetochore complex and to analyze the maturation of prekinetochores before mitosis. Here we have focused on the centromere-specific histone Centromere Protein (CENP)-A and its spatial relationship to other histones and histone modifications found in condensed chromatin. We demonstrate that the phosphorylation of histone H3 is essentially excluded from a specific region of centromeric chromatin, defined by the presence of CENP-A. Interspersion of CENP-B with phosphorylated H3 in the inner centromere indicates that the exclusion of H3 modification is not a general property of alpha-satellite DNA. We also demonstrate that these regions are functionally distinct by fragmenting mitotic chromatin into motile centromere-kinetochore fragments that contain CENP-A with little or no phosphorylated H3 and nonmotile fragments that contain exclusively phosphorylated H3. The sequence of CENP-A diverges from H3 in a number of key residues involved in chromosome condensation and in transcription, potentially allowing a more specialized chromatin structure within centromeric heterochromatin, on which kinetochore plates may nucleate and mature. This specialized centromere subdomain would be predicted to have a very tight and static nucleosome structure as a result of the absence of H3 phosphorylation and acetylation.


Subject(s)
Autoantigens , Centromere , Chromosomal Proteins, Non-Histone , Amino Acid Sequence , Animals , CREST Syndrome/immunology , Centromere/chemistry , Centromere/physiology , Centromere Protein A , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/physiology , Histones/chemistry , Histones/physiology , Mammals/genetics , Molecular Sequence Data , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...