Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
2.
Biol Pharm Bull ; 46(3): 511-516, 2023.
Article in English | MEDLINE | ID: mdl-36858581

ABSTRACT

Pharmacogenetics (PGx) enhances personalized care, often reducing medical costs, and improving patients' QOL. Unlike single variant analysis, multiplex PGx panel tests can result in applying comprehensive PGx-guided medication to maximize drug efficacy and minimize adverse reactions. Among PGx genes, drug-metabolizing enzymes and drug transporters have significant roles in the efficacy and safety of various pharmacotherapies. In this study, a genotyping panel has been developed for the Japanese population called PGx_JPN panel comprising 36 variants in 14 genes for drug-metabolizing enzymes and drug transporters using a mass spectrometry-based genotyping method, in which all the variants could be analyzed in two wells for multiplex analysis. The verification test exhibited good concordance with the results analyzed using the other standard genotyping methods (microarray, TaqMan assay, or another mass spectrometry-based commercial kit). However, copy number variations such as CYP2D6*5 could not apply to this system. In this study, we demonstrated that the mass spectrometry-based multiplex method could be useful for in the simultaneous genotyping of more than 30 variants, which are essential among the Japanese population in two wells, except for copy number variations. Further study is needed to assess our panel to demonstrate the clinical use of pharmacogenomics for precision medicine in the Japanese population.


Subject(s)
DNA Copy Number Variations , Pharmacogenetics , Humans , East Asian People , Quality of Life , Mass Spectrometry , Membrane Transport Proteins
3.
Nature ; 517(7535): 466-71, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25533956

ABSTRACT

The kinetochore is the crucial apparatus regulating chromosome segregation in mitosis and meiosis. Particularly in meiosis I, unlike in mitosis, sister kinetochores are captured by microtubules emanating from the same spindle pole (mono-orientation) and centromeric cohesion mediated by cohesin is protected in the following anaphase. Although meiotic kinetochore factors have been identified only in budding and fission yeasts, these molecules and their functions are thought to have diverged earlier. Therefore, a conserved mechanism for meiotic kinetochore regulation remains elusive. Here we have identified in mouse a meiosis-specific kinetochore factor that we termed MEIKIN, which functions in meiosis I but not in meiosis II or mitosis. MEIKIN plays a crucial role in both mono-orientation and centromeric cohesion protection, partly by stabilizing the localization of the cohesin protector shugoshin. These functions are mediated mainly by the activity of Polo-like kinase PLK1, which is enriched to kinetochores in a MEIKIN-dependent manner. Our integrative analysis indicates that the long-awaited key regulator of meiotic kinetochore function is Meikin, which is conserved from yeasts to humans.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Conserved Sequence , Kinetochores/metabolism , Meiosis , Animals , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Female , Humans , Infertility/genetics , Infertility/metabolism , Male , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Polo-Like Kinase 1
4.
Genes Cells ; 22(6): 552-567, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28497540

ABSTRACT

In meiosis I, sister chromatids are captured by microtubules emanating from the same pole (mono-orientation), and centromeric cohesion is protected throughout anaphase. Shugoshin, which is localized to centromeres depending on the phosphorylation of histone H2A by Bub1 kinase, plays a central role in protecting meiotic cohesin Rec8 from separase cleavage. Another key meiotic kinetochore factor, meikin, may regulate cohesion protection, although the underlying molecular mechanisms remain elusive. Here, we show that fission yeast Moa1 (meikin), which associates stably with CENP-C during meiosis I, recruits Plo1 (polo-like kinase) to the kinetochores and phosphorylates Spc7 (KNL1) to accumulate Bub1. Consequently, in contrast to the transient kinetochore localization of mitotic Bub1, meiotic Bub1 persists at kinetochores until anaphase I. The meiotic Bub1 pool ensures robust Sgo1 (shugoshin) localization and cohesion protection at centromeres by cooperating with heterochromatin protein Swi6, which binds and stabilizes Sgo1. Furthermore, molecular genetic analyses show a hierarchical regulation of centromeric cohesion protection by meikin and shugoshin that is important for establishing meiosis-specific chromosome segregation. We provide evidence that the meiosis-specific Bub1 regulation is conserved in mouse.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Fungal , Meiosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Animals , Cell Adhesion , Cells, Cultured , Centromere/genetics , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/physiology , Kinetochores , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Phosphorylation , Schizosaccharomyces/cytology , Schizosaccharomyces/growth & development , Spermatocytes/cytology , Spermatocytes/metabolism , Polo-Like Kinase 1
5.
Genes Dev ; 24(19): 2169-79, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20889715

ABSTRACT

Shugoshin (Sgo) is a conserved centromeric protein. Mammalian Sgo1 collaborates with protein phosphatase 2A (PP2A) to protect mitotic cohesin from the prophase dissociation pathway. Although another shugoshin-like protein, Sgo2, is required for the centromeric protection of cohesion in germ cells, its precise molecular function remains largely elusive. We demonstrate that hSgo2 plays a dual role in chromosome congression and centromeric protection of cohesion in HeLa cells, while the latter function is exposed only in perturbed mitosis. These functions partly overlap with those of Aurora B, a kinase setting faithful chromosome segregation. Accordingly, we identified the phosphorylation of hSgo2 by Aurora B at the N-terminal coiled-coil region and the middle region, and showed that these phosphorylations separately promote binding of hSgo2 to PP2A and MCAK, factors required for centromeric protection and chromosome congression, respectively. Furthermore, these phosphorylations are essential for localizing PP2A and MCAK to centromeres. This mechanism seems applicable to germ cells as well. Thus, our study identifies Sgo2 as a hitherto unknown crucial cellular substrate of Aurora B in mammalian cells.


Subject(s)
Cell Cycle Proteins/metabolism , Centromere/metabolism , Kinesins/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Aurora Kinase B , Aurora Kinases , Cells, Cultured , HeLa Cells , Humans , Phosphorylation , Protein Transport
6.
Nature ; 467(7316): 719-23, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20739936

ABSTRACT

Successful partition of replicated genomes at cell division requires chromosome attachment to opposite poles of mitotic spindle (bi-orientation). Any defects in this regulation bring about chromosomal instability, which may accelerate tumour progression in humans. To achieve chromosome bi-orientation at prometaphase, the chromosomal passenger complex (CPC), composed of catalytic kinase Aurora B and regulatory components (INCENP, Survivin and Borealin), must be localized to centromeres to phosphorylate kinetochore substrates. Although the CPC dynamically changes the subcellular localization, the regulation of centromere targeting is largely unknown. Here we isolated a fission yeast cyclin B mutant defective specifically in chromosome bi-orientation. Accordingly, we identified Cdk1 (also known as Cdc2)-cyclin-B-dependent phosphorylation of Survivin. Preventing Survivin phosphorylation impairs centromere CPC targeting as well as chromosome bi-orientation, whereas phosphomimetic Survivin suppresses the bi-orientation defect in the cyclin B mutant. Survivin phosphorylation promotes direct binding with shugoshin, which we now define as a conserved centromeric adaptor of the CPC. In human cells, the phosphorylation of Borealin has a comparable role. Thus, our study resolves the conserved mechanisms of CPC targeting to centromeres, highlighting a key role of Cdk1-cyclin B in chromosome bi-orientation.


Subject(s)
CDC2 Protein Kinase/metabolism , Chromosomes, Fungal/metabolism , Chromosomes, Human/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinases , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Substrate Specificity , Survivin
7.
Methods Mol Biol ; 2519: 27-40, 2023.
Article in English | MEDLINE | ID: mdl-36066707

ABSTRACT

Cell cycle-dependent regulation of chromosome is a dynamic event. After replication in S phase, sister chromatids show dynamic behavior including condensation, alignment, and segregation in M phase. These beautiful behaviors of chromosomes observed through the microscope have fascinated people since more than 100 years ago, and now we can sketch the dynamics of regulatory proteins and their posttranscriptional modifications through the fluorescent microscope. The purpose of this chapter is describing the basic methods of immunofluorescence analysis of mitotic cells and chromosomes. Besides, the key ideas for improving the preparation of the specimen are also described. Because the characteristic of the proteins of your interest differs one by one, modifying the method might cause the crucial improvement in the observation.


Subject(s)
Chromatids , Mitosis , Chromosome Segregation , Chromosomes , Humans , Microscopy, Fluorescence
8.
Biochem Biophys Res Commun ; 411(1): 7-13, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21684257

ABSTRACT

The activation of innate immune responses is critical to host defense against microbial infections, wherein nucleic acid-sensing pattern recognition receptors recognize DNA or RNA from viruses or bacteria and activate downstream signaling pathways. In a search for new DNA-sensing molecules that regulate innate immune responses, we identified RNA-binding motif protein 3 (RBM3), whose role has been implicated in the regulation of cell growth. In this study, we generated Rbm3-deficient (Rbm3(-/-)) mice to study the role of RBM3 in immune responses and cell growth. Despite evidence for its interaction with immunogenic DNA in a cell, no overt phenotypic abnormalities were found in cells from Rbm3(-/-) mice for the DNA-mediated induction of cytokine genes. Interestingly, however, Rbm3(-/-) mouse embryonic fibroblasts (MEFs) showed poorer proliferation rates as compared to control MEFs. Further cell cycle analysis revealed that Rbm3(-/-) MEFs have markedly increased number of G2-phase cells, suggesting a hitherto unknown role of RBM3 in the G2-phase control. Thus, these mutant mice and cells may provide new tools with which to study the mechanisms underlying the regulation of cell cycle and oncogenesis.


Subject(s)
Cell Cycle/genetics , Immunity, Innate/genetics , RNA-Binding Proteins/physiology , Animals , Cell Transformation, Neoplastic/genetics , G2 Phase/genetics , Mice , Mice, Mutant Strains , RNA-Binding Proteins/genetics
9.
PLoS One ; 16(11): e0259846, 2021.
Article in English | MEDLINE | ID: mdl-34784358

ABSTRACT

Epigenome research has employed various methods to identify the genomic location of proteins of interest, such as transcription factors and histone modifications. A recently established method called CUT&Tag uses a Protein-A Tn5 transposase fusion protein, which cuts the genome and inserts adapter sequences nearby the target protein. Throughout most of the CUT&Tag procedure, cells are held on concanavalin A (con A)-conjugated magnetic beads. Proper holding of cells would be decisive for the accessibility of Tn5 to the chromatin, and efficacy of the procedure of washing cells. However, BioMag®Plus ConA magnetic beads, used in the original CUT&Tag protocol, often exhibit poor suspendability and severe aggregation. Here, we compared the BioMag beads and Dynabeads® magnetic particles of which conjugation of con A was done by our hands, and examined the performance of these magnetic beads in CUT&Tag. Among tested, one of the Dynabeads, MyOne-T1, kept excessive suspendability in a buffer even after overnight incubation. Furthermore, the MyOne-T1 beads notably improved the sensitivity in CUT&Tag assay for H3K4me3. In conclusion, the arrangement and the selection of MyOne-T1 refine the suspendability of beads, which improves the association of chromatin with Tn5, which enhances the sensitivity in CUT&Tag assay.


Subject(s)
Concanavalin A/administration & dosage , Histones/metabolism , Staphylococcal Protein A/genetics , Transposases/genetics , Animals , Cell Line , Concanavalin A/chemistry , Concanavalin A/pharmacology , Epigenomics , HEK293 Cells , Histone Code , Humans , Immunomagnetic Separation , Magnetic Fields , Methylation , Mice , Particle Size , Recombinant Fusion Proteins/metabolism , Staphylococcal Protein A/metabolism , Transposases/metabolism
10.
J Cell Biol ; 218(10): 3223-3236, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31527146

ABSTRACT

The accurate regulation of phosphorylation at the kinetochore is essential for establishing chromosome bi-orientation. Phosphorylation of kinetochore proteins by the Aurora B kinase destabilizes improper kinetochore-microtubule attachments, whereas the phosphatase PP2A has a counteracting role. Imbalanced phosphoregulation leads to error-prone chromosome segregation and aneuploidy, a hallmark of cancer cells. However, little is known about the molecular events that control the balance of phosphorylation at the kinetochore. Here, we show that localization of SET/TAF1, an oncogene product, to centromeres maintains Aurora B kinase activity by inhibiting PP2A, thereby correcting erroneous kinetochore-microtubule attachment. SET localizes at the inner centromere by interacting directly with shugoshin 2, with SET levels declining at increased distances between kinetochore pairs, leading to establishment of chromosome bi-orientation. Moreover, SET overexpression induces chromosomal instability by disrupting kinetochore-microtubule attachment. Thus, our findings reveal the novel role of SET in fine-tuning the phosphorylation level at the kinetochore by balancing the activities of Aurora B and PP2A.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Histone Chaperones/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans
11.
Curr Biol ; 27(7): 1005-1012, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28343969

ABSTRACT

Sister-chromatid cohesion is established by the cohesin complex in S phase and persists until metaphase, when sister chromatids are captured by microtubules emanating from opposite poles [1]. The Aurora-B-containing chromosome passenger complex (CPC) plays a crucial role in achieving chromosome bi-orientation by correcting erroneous microtubule attachment [2]. The centromeric localization of the CPC relies largely on histone H3-T3 phosphorylation (H3-pT3), which is mediated by the mitotic histone kinase Haspin/Hrk1 [3-5]. Hrk1 localization to centromeres depends largely on the cohesin subunit Pds5 in fission yeast [5]; however, it is unknown how Pds5 regulates Hrk1 localization. Here we identify a conserved Hrk1-interacting motif (HIM) in Pds5 and a Pds5-interacting motif (PIM) in Hrk1 in fission yeast. Mutations in either motif result in the displacement of Hrk1 from centromeres. We also show that the mechanism of Pds5-dependent Hrk1 recruitment is conserved in human cells. Notably, the PIM in Haspin/Hrk1 is reminiscent of the YSR motif found in the mammalian cohesin destabilizer Wapl and stabilizer Sororin, both of which bind PDS5 [6-12]. Similarly, and through the same motifs, fission yeast Pds5 binds to Wpl1/Wapl and acetyltransferase Eso1/Eco1, in addition to Hrk1. Thus, we have identified a protein-protein interaction module in Pds5 that serves as a chromatin platform for regulating sister-chromatid cohesion and chromosome bi-orientation.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Protein Serine-Threonine Kinases/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Transcription Factors/genetics , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromosome Segregation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/metabolism
12.
Science ; 349(6253): 1237-40, 2015 09 11.
Article in English | MEDLINE | ID: mdl-26359403

ABSTRACT

Chromosomal instability (CIN) is a major trait of cancer cells and a potent driver of tumor progression. However, the molecular mechanisms underlying CIN still remain elusive. We found that a number of CIN(+) cell lines have impairments in the integrity of the conserved inner centromere-shugoshin (ICS) network, which coordinates sister chromatid cohesion and kinetochore-microtubule attachment. These defects are caused mostly by the loss of histone H3 lysine 9 trimethylation at centromeres and sometimes by a reduction in chromatin-associated cohesin; both pathways separately sustain centromeric shugoshin stability. Artificial restoration of the ICS network suppresses chromosome segregation errors in a wide range of CIN(+) cells, including RB- and BRCA1-deficient cells. Thus, dysfunction of the ICS network might be a key mechanism underlying CIN in human tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromosomal Instability , Chromosome Segregation , BRCA1 Protein/genetics , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Centromere/genetics , Chromatids/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Histones/metabolism , Humans , Kinetochores/metabolism , Lysine/metabolism , Methylation , Microtubules/metabolism , Retinoblastoma Protein/genetics , Cohesins
14.
J Occup Health ; 55(4): 292-300, 2013.
Article in English | MEDLINE | ID: mdl-23698183

ABSTRACT

OBJECTIVES: The aim of this study was to ascertain the actual state of toner exposure of workers who handle toner. METHODS: Personal exposure measurements were conducted on workers handling toner in which the respirable dust (RD) concentration by work type was determined. Targeted work types consisted of "machine recycling", "toner manufacturing", "toner research/development", "machine design/development" and "servicing." The implementation period lasted from April 2003 to March 2011, and measurements were conducted annually. The measurement method conformed to the Japanese Working Environment Measurement Standards (new standards adopted starting in 2005). RESULTS: Comparing the RD concentrations for fiscal year 2003 by work, significant differences were found between machine recycling and the other four work types, as well as in toner manufacturing and the other four work types. Similarly, based on the new legislative standards applied in Japan from fiscal year 2005, significant differences were found between machine recycling and the other four work types, as well as in toner manufacturing and the other four work types. DISCUSSION: It is clear that workers engaged in machine recycling and toner manufacturing are exposed to toner, and that a certain level of exposure is continuing. Although it cannot be said that workers involved in toner research/development, machine design/development and servicing have no toner exposure, the concentration is of an extremely low level. CONCLUSIONS: At present, toner exposure levels by work type can be divided into two groups-one consisting of machine recycling and toner manufacturing, and the other consisting of toner research/development, machine design/development and servicing.


Subject(s)
Copying Processes , Lung/drug effects , Manufactured Materials/adverse effects , Occupational Exposure/analysis , Occupations , Dust/analysis , Humans , Occupational Exposure/adverse effects
15.
Nat Cell Biol ; 14(7): 746-52, 2012 Jun 03.
Article in English | MEDLINE | ID: mdl-22660415

ABSTRACT

The genomic stability of all organisms depends on the precise partition of chromosomes to daughter cells. The spindle assembly checkpoint (SAC) senses unattached kinetochores and prevents premature entry to anaphase, thus ensuring that all chromosomes attach to opposite spindle poles (bi-orientation) during mitosis. MPS1 is an evolutionarily conserved protein kinase required for the SAC and chromosome bi-orientation. Yet, its primary cellular substrate has remained elusive. We show that fission yeast Mph1 (MPS1 homologue) phosphorylates the kinetochore protein Spc7 (KNL1/Blinkin homologue) at the MELT repeat sequences. This phosphorylation promotes the in vitro binding to the Bub1-Bub3 complex, which is required for kinetochore-based SAC activation (Mad1-Mad2-Mad3 localization) and chromosome alignment. Accordingly, a non-phosphorylatable spc7-12A mutation abolishes kinetochore targeting of Bub1-Bub3, whereas a phospho-mimetic spc7-12E mutation forces them to localize at kinetochores throughout the entire cell cycle, even in the absence of Mph1. Thus, MPS1/Mph1 kinase locating at the unattached kinetochores initially creates a mark, which is crucial for SAC activation and chromosome bi-orientation. This mechanism seems to be conserved in human cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Protein Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Cell Cycle Proteins/metabolism , Mad2 Proteins , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics , Time Factors
16.
Science ; 357(6355): 981, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28874501
17.
Science ; 330(6001): 239-43, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20929775

ABSTRACT

For proper partitioning of chromosomes in mitosis, the chromosomal passenger complex (CPC) including Aurora B and survivin must be localized at the center of paired kinetochores, at the site called the inner centromere. It is largely unknown what defines the inner centromere and how the CPC is targeted to this site. Here, we show that the phosphorylation of histone H3-threonine 3 (H3-pT3) mediated by Haspin cooperates with Bub1-mediated histone 2A-serine 121 (H2A-S121) phosphorylation in targeting the CPC to the inner centromere in fission yeast and human cells. H3-pT3 promotes nucleosome binding of survivin, whereas phosphorylated H2A-S121 facilitates the binding of shugoshin, the centromeric CPC adaptor. Haspin colocalizes with cohesin by associating with Pds5, whereas Bub1 localizes at kinetochores. Thus, the inner centromere is defined by intersection of two histone kinases.


Subject(s)
Centromere/metabolism , Chromosomes, Fungal/physiology , Chromosomes, Human/physiology , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Amino Acid Sequence , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , HeLa Cells , Heterochromatin/metabolism , Humans , Inhibitor of Apoptosis Proteins , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis , Molecular Sequence Data , Mutation , Nucleosomes/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Serine/metabolism , Survivin , Threonine/metabolism , Cohesins
19.
Nat Cell Biol ; 10(1): 42-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18084284

ABSTRACT

Reductional chromosome segregation in germ cells, where sister chromatids are pulled to the same pole, accompanies the protection of cohesin at centromeres from separase cleavage. Here, we show that mammalian shugoshin Sgo2 is expressed in germ cells and is solely responsible for the centromeric localization of PP2A and the protection of cohesin Rec8 in oocytes, proving conservation of the mechanism from yeast to mammals. However, this role of Sgo2 contrasts with its mitotic role in protecting centromeric cohesin only from prophase dissociation, but never from anaphase cleavage. We demonstrate that, in somatic cells, shugoshin colocalizes with cohesin in prophase or prometaphase, but their localizations become separate when centromeres are pulled oppositely at metaphase. Remarkably, if tension is artificially removed from the centromeres at the metaphase-anaphase transition, cohesin at the centromeres can be protected from separase cleavage even in somatic cells, as in germ cells. These results argue for a unified view of centromeric protection by shugoshin in mitosis and meiosis.


Subject(s)
Cell Cycle Proteins/genetics , Centromere/metabolism , Gene Expression Profiling , Oocytes/metabolism , Animals , Blotting, Northern , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Female , HeLa Cells , Humans , Immunohistochemistry , Kinetochores/metabolism , Male , Metaphase/genetics , Mice , Mice, Inbred C57BL , Mitosis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sister Chromatid Exchange , Cohesins
20.
Biochem Biophys Res Commun ; 341(2): 363-8, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16414009

ABSTRACT

Exposure of MDA-MB-468 cells to ionizing radiation (IR) caused biphasic activation of ERK as indicated by its phosphorylation at Thr202/Tyr204. Specific epidermal growth factor receptor (EGFR) inhibitor AG1478 and specific Src inhibitor PP2 inhibited IR-induced ERK1/2 activation but phosphatidylinositol-3 kinase inhibitor wortmannin did not. IR caused EGFR tyrosine phosphorylation, whereas it did not induce EGFR autophosphorylation at Tyr992, Tyr1045, and Tyr1068 or Src-dependent EGFR phosphorylation at Tyr845. SHP-2, which positively regulates EGFR/Ras/ERK signaling cascade, became activated by IR as indicated by its phosphorylation at Tyr542. This activation was inhibited by PP2 not by AG1478, which suggests Src-dependent activation of SHP-2. Src and PTPalpha, which positively regulates Src, became activated as indicated by phosphorylation at Tyr416 and Tyr789, respectively. These data suggest that IR-induced ERK1/2 activation involves EGFR through a Src-dependent pathway that is distinct from EGFR ligand activation.


Subject(s)
Pyrimidines/pharmacology , src-Family Kinases/metabolism , Androstadienes/pharmacology , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Radiation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanosine Triphosphate/metabolism , Humans , Immunoblotting , Immunoprecipitation , Infrared Rays , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Quinazolines , Radiation, Ionizing , Signal Transduction , Temperature , Time Factors , Transcriptional Activation , Tyrosine/chemistry , Tyrphostins/pharmacology , Wortmannin , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL