123 results on '"Obuse C"'
Search Results
2. Inhibition of RIF1 by SCAI Allows BRCA1-Mediated Repair
- Author
-
Isobe, S. Y., Nagao, K., Nozaki, N., Kimura, Hiroshi, and Obuse, C.
- Subjects
0301 basic medicine ,Genome instability ,DNA Repair ,DNA damage ,Telomere-Binding Proteins ,HDR ,Biology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,RIF1 ,law ,Humans ,DNA double-strand breaks ,DNA Breaks, Double-Stranded ,Phosphorylation ,lcsh:QH301-705.5 ,NHEJ ,Genetics ,Reporter gene ,BRCA1 Protein ,BRCA1 ,resection-dependent NHEJ ,genomic instability ,53BP1 ,Cell biology ,Kinetics ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,alternative NHEJ ,Cancer cell ,Suppressor ,SCAI ,Tumor Suppressor p53-Binding Protein 1 ,Function (biology) ,DNA ,DNA Damage ,Protein Binding ,Transcription Factors - Abstract
DNA double-strand breaks (DSBs) are repaired by either the homology-directed repair (HDR) or the non-homologous end-joining (NHEJ) pathway. RIF1 (RAP1-interacting factor homolog) was recently shown to stimulate NHEJ through an interaction with 53BP1 (p53-binding protein 1) phosphorylated at S/TQ sites, but the molecular mechanism underlying pathway choice remains unclear. Here, we show that SCAI (suppressor of cancer cell invasion) binds to 53BP1 phosphorylated at S/TP sites and facilitates HDR. Upon DNA damage, RIF1 immediately accumulates at damage sites and then gradually dissociates from 53BP1 and is subsequently replaced with SCAI. Depletion of SCAI reduces both the accumulation of HDR factors, including BRCA1 (breast cancer susceptibility gene 1), at damage sites and the efficiency of HDR, as detected by a reporter assay system. These data suggest that SCAI inhibits RIF1 function to allow BRCA1-mediated repair, which possibly includes alt-NHEJ and resection-dependent NHEJ in G1, as well as HDR in S/G2.
- Published
- 2017
3. Histone H3K36 trimethylation is essential for multiple silencing mechanisms in fission yeast
- Author
-
Suzuki, S., Kato, H., Suzuki, Y., Chikashige, Y., Hiraoka, Y., Kimura, Hiroshi, Nagao, K., Obuse, C., Takahata, S., and Murakami, Y.
- Subjects
0301 basic medicine ,Transcription, Genetic ,Heterochromatin ,RNA Stability ,Genes, Fungal ,RNA polymerase II ,Methylation ,Histones ,03 medical and health sciences ,Transcription (biology) ,Gene Expression Regulation, Fungal ,Schizosaccharomyces ,Genetics ,Gene silencing ,Protein Interaction Domains and Motifs ,Gene Silencing ,biology ,Gene regulation, Chromatin and Epigenetics ,Histone-Lysine N-Methyltransferase ,Telomere ,Yeast ,030104 developmental biology ,Histone ,biology.protein ,Histone deacetylase complex ,RNA Polymerase II ,Schizosaccharomyces pombe Proteins ,Chromosomes, Fungal ,Protein Processing, Post-Translational - Abstract
In budding yeast, Set2 catalyzes di- and trimethylation of H3K36 (H3K36me2 and H3K36me3) via an interaction between its Set2-Rpb1 interaction (SRI) domain and C-terminal repeats of RNA polymerase II (Pol2) phosphorylated at Ser(2) and Ser(5) (CTD-S2,5-P). H3K36me2 is sufficient for recruitment of the Rpd3S histone deacetylase complex to repress cryptic transcription from transcribed regions. In fission yeast, Set2 is also responsible for H3K36 methylation, which represses a subset of RNAs including heterochromatic and subtelomeric RNAs, at least in part via recruitment of Clr6 complex II, a homolog of Rpd3S. Here, we show that CTD-S2P-dependent interaction of fission yeast Set2 with Pol2 via the SRI domain is required for formation of H3K36me3, but not H3K36me2. H3K36me3 silenced heterochromatic and subtelomeric transcripts mainly through post-transcriptional and transcriptional mechanisms, respectively, whereas H3K36me2 was not enough for silencing. Clr6 complex II appeared not to be responsible for heterochromatic silencing by H3K36me3. Our results demonstrate that H3K36 methylation has multiple outputs in fission yeast; these findings provide insights into the distinct roles of H3K36 methylation in metazoans, which have different enzymes for synthesis of H3K36me1/2 and H3K36me3.
- Published
- 2016
4. Ser7 of RNAPII-CTD facilitates heterochromatin formation by linking ncRNA to RNAi
- Author
-
Kajitani, T., Kato, H., Chikashige, Y., Tsutsumi, C., Hiraoka, Y., Kimura, Hiroshi, Ohkawa, Y., Obuse, C., Hermand, D., and Murakami, Y.
- Subjects
0301 basic medicine ,Heterochromatin ,NcRNA transcription ,Cell Cycle Proteins ,RNA polymerase II ,RNA Polymerase II/genetics ,Noncoding RNA ,noncoding RNA ,Chromodomain ,03 medical and health sciences ,Long Noncoding/genetics ,Protein Domains ,RNA interference ,Schizosaccharomyces/genetics ,Schizosaccharomyces ,Serine ,Gene silencing ,RNA, Long Noncoding/genetics ,RNA, Small Interfering ,RNA, Small Interfering/genetics ,Cell Cycle Proteins/genetics ,Serine/genetics ,Fungal/genetics ,Multidisciplinary ,biology ,Chemistry ,fungi ,heterochromatin ,RNA, Fungal ,Heterochromatin/genetics ,Non-coding RNA ,Chromatin ,Cell biology ,RNA, Fungal/genetics ,030104 developmental biology ,PNAS Plus ,Small Interfering/genetics ,RNAi ,biology.protein ,RNA ,Schizosaccharomyces pombe Proteins/genetics ,RNA, Long Noncoding ,Schizosaccharomyces pombe Proteins - Abstract
Some long noncoding RNAs (ncRNAs) transcribed by RNA polymerase II (RNAPII) are retained on chromatin, where they regulate RNAi and chromatin structure. The molecular basis of this retention remains unknown. We show that in fission yeast serine 7 (Ser7) of the C-terminal domain (CTD) of RNAPII is required for efficient siRNA generation for RNAi-dependent heterochromatin formation. Surprisingly, Ser7 facilitates chromatin retention of nascent heterochromatic RNAs (hRNAs). Chromatin retention of hRNAs and siRNA generation requires both Ser7 and an RNA-binding activity of the chromodomain of Chp1, a subunit of the RNA-induced transcriptional silencing (RITS) complex. Furthermore, RITS associates with RNAPII in a Ser7-dependent manner. We propose that Ser7 promotes cotranscriptional chromatin retention of hRNA by recruiting the RNA-chromatin connector protein Chp1, which facilitates RNAi-dependent heterochromatin formation. Our findings reveal a function of the CTD code: linking ncRNA transcription to RNAi for heterochromatin formation.
- Published
- 2017
5. Human inactive X chromosome is compacted through a PRC2-independent SMCHD1-HBiX1 pathway
- Author
-
Nozawa, R. S., Nagao, K., Igami, K. T., Shibata, S., Shirai, N., Nozaki, N., Sado, T., Kimura, Hiroshi, and Obuse, C.
- Subjects
Chromosomes, Human, X ,biology ,Chromosomal Proteins, Non-Histone ,Barr body ,Histones/metabolism ,macromolecular substances ,Plasma protein binding ,Chromosomal Proteins, Non-Histone/*metabolism ,Molecular biology ,X-inactivation ,Histones ,Histone ,Chromobox Protein Homolog 5 ,Structural Biology ,biology.protein ,Humans ,RNA, Long Noncoding ,Heterochromatin protein 1 ,XIST ,Chromosomes, Human, X/*metabolism ,PRC2 ,RNA, Long Noncoding/metabolism ,Molecular Biology ,X chromosome ,Protein Binding - Abstract
Human inactive X chromosome (Xi) forms a compact structure called the Barr body, which is enriched in repressive histone modifications such as trimethylation of histone H3 Lys9 (H3K9me3) and Lys27 (H3K27me3). These two histone marks are distributed in distinct domains, and X-inactive specific transcript (XIST) preferentially colocalizes with H3K27me3 domains. Here we show that Xi compaction requires HBiX1, a heterochromatin protein 1 (HP1)-binding protein, and structural maintenance of chromosomes hinge domain-containing protein 1 (SMCHD1), both of which are enriched throughout the Xi chromosome. HBiX1 localization to H3K9me3 and XIST-associated H3K27me3 (XIST-H3K27me3) domains was mediated through interactions with HP1 and SMCHD1, respectively. Furthermore, HBiX1 was required for SMCHD1 localization to H3K9me3 domains. Depletion of HBiX1 or SMCHD1, but not Polycomb repressive complex 2 (PRC2), resulted in Xi decompaction, similarly to XIST depletion. Thus, the molecular network involving HBiX1 and SMCHD1 links the H3K9me3 and XIST-H3K27me3 domains to organize the compact Xi structure.
- Published
- 2013
6. Histone H4 lysine 20 acetylation is associated with gene repression in human cells
- Author
-
Kaimori, J. Y., Maehara, K., Hayashi-Takanaka, Y., Harada, A., Fukuda, M., Yamamoto, S., Ichimaru, N., Umehara, T., Yokoyama, S., Matsuda, R., Ikura, T., Nagao, K., Obuse, C., Nozaki, N., Takahara, S., Takao, T., Ohkawa, Y., Kimura, Hiroshi, and Isaka, Y.
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Biology ,SAP30 ,Methylation ,Article ,Histone H4 ,Histones ,03 medical and health sciences ,Histone H3 ,Mice ,0302 clinical medicine ,Histone H2A ,Nucleosome ,Histone code ,Animals ,Humans ,Transcription Factors/metabolism ,Histone octamer ,Antibodies, Monoclonal/immunology ,Chromatography, High Pressure Liquid ,Multidisciplinary ,Binding Sites ,Lysine ,Antibodies, Monoclonal ,Acetylation ,Peptides/analysis ,Molecular biology ,030104 developmental biology ,Microscopy, Fluorescence ,Histone methyltransferase ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Transcription Initiation Site ,Peptides ,Histones/immunology/*metabolism ,030217 neurology & neurosurgery ,Lysine/metabolism ,HeLa Cells ,Transcription Factors - Abstract
Histone acetylation is generally associated with gene activation and chromatin decondensation. Recent mass spectrometry analysis has revealed that histone H4 lysine 20, a major methylation site, can also be acetylated. To understand the function of H4 lysine 20 acetylation (H4K20ac), we have developed a specific monoclonal antibody and performed ChIP-seq analysis using HeLa-S3 cells. H4K20ac was enriched around the transcription start sites (TSSs) of minimally expressed genes and in the gene body of expressed genes, in contrast to most histone acetylation being enriched around the TSSs of expressed genes. The distribution of H4K20ac showed little correlation with known histone modifications, including histone H3 methylations. A motif search in H4K20ac-enriched sequences, together with transcription factor binding profiles based on ENCODE ChIP-seq data, revealed that most transcription activators are excluded from H4K20ac-enriched genes and a transcription repressor NRSF/REST co-localized with H4K20ac. These results suggest that H4K20ac is a unique acetylation mark associated with gene repression.
- Published
- 2016
- Full Text
- View/download PDF
7. Human POGZ modulates dissociation of HP1alpha from mitotic chromosome arms through Aurora B activation
- Author
-
Nozawa, R. S., Nagao, K., Masuda, H. T., Iwasaki, O., Hirota, T., Nozaki, N., Kimura, Hiroshi, and Obuse, C.
- Subjects
Chromosomal Proteins, Non-Histone ,Immunoprecipitation ,Blotting, Western ,Aurora B kinase ,Mitosis ,Transposases ,Plasma protein binding ,Chromatids ,Protein Serine-Threonine Kinases ,Biology ,Mass Spectrometry ,Cell Line ,Chromatids/genetics/metabolism ,Chromatin/metabolism ,Aurora Kinases ,Two-Hybrid System Techniques ,Aurora Kinase B ,Humans ,Kinetochores ,Zinc finger ,Mitosis/genetics/*physiology ,Fluorescence recovery after photobleaching ,Cell Biology ,Molecular biology ,Chromatin ,Cell biology ,Kinetochores/metabolism ,Microscopy, Fluorescence ,Chromobox Protein Homolog 5 ,Chromosomal Proteins, Non-Histone/genetics/*metabolism ,Protein-Serine-Threonine Kinases/*metabolism ,Heterochromatin protein 1 ,RNA Interference ,Transposases/genetics/*metabolism ,Fluorescence Recovery After Photobleaching ,HeLa Cells ,Protein Binding - Abstract
Heterochromatin protein 1 (HP1) has an essential role in heterochromatin formation and mitotic progression through its interaction with various proteins. We have identified a unique HP1alpha-binding protein, POGZ (pogo transposable element-derived protein with zinc finger domain), using an advanced proteomics approach. Proteins generally interact with HP1 through a PxVxL (where x is any amino-acid residue) motif; however, POGZ was found to bind to HP1alpha through a zinc-finger-like motif. Binding by POGZ, mediated through its zinc-finger-like motif, competed with PxVxL proteins and destabilized the HP1alpha-chromatin interaction. Depletion experiments confirmed that the POGZ HP1-binding domain is essential for normal mitotic progression and dissociation of HP1alpha from mitotic chromosome arms. Furthermore, POGZ is required for the correct activation and dissociation of Aurora B kinase from chromosome arms during M phase. These results reveal POGZ as an essential protein that links HP1alpha dissociation with Aurora B kinase activation during mitosis.
- Published
- 2010
8. Vertebrate Spt2 is a novel nucleolar histone chaperone that assists in ribosomal DNA transcription
- Author
-
Osakabe, A., Tachiwana, H., Takaku, M., Hori, T., Obuse, C., Kimura, Hiroshi, Fukagawa, T., and Kurumizaka, H.
- Subjects
Yeasts/genetics ,Transcription, Genetic ,Histones/metabolism ,RNA polymerase I ,RNA polymerase II ,Chromatin remodeling ,Cell Line ,Protein Binding/genetics ,Histones ,Protein Structure, Tertiary/genetics ,RNA Polymerase I/metabolism ,Yeasts ,Sequence Deletion/genetics ,Histone H2A ,Histone methylation ,Nucleoli ,Animals ,Humans ,Histone chaperone ,Nucleosome ,Histone code ,Histone Chaperones ,Cell Nucleus/*metabolism ,RNA polymerase II holoenzyme ,Sequence Deletion ,Cell Nucleus ,biology ,General transcription factor ,Chromatin Assembly and Disassembly/genetics ,DNA-Binding Proteins/genetics/isolation & purification/*metabolism ,Protein Transport/genetics ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,Protein Structure, Tertiary ,Cell biology ,DNA-Binding Proteins ,Protein Transport ,Histone Chaperones/genetics/isolation & purification/*metabolism ,biology.protein ,Ribosomes/genetics/metabolism ,Ribosomes ,Chickens ,Transcription ,Protein Binding - Abstract
In eukaryotes, transcription occurs in the chromatin context with the assistance of histone binding proteins, such as chromatin/nucleosome remodeling factors and histone chaperones. However, it is unclear how each remodeling factor or histone chaperone functions in transcription. Here, we identified a novel histone-binding protein, Spt2, in higher eukaryotes. Recombinant human Spt2 binds to histones and DNA, and promotes nucleosome assembly in vitro. Spt2 accumulates in nucleoli and interacts with RNA polymerase I in chicken DT40 cells, suggesting its involvement in ribosomal RNA transcription. Consistently, Spt2-deficient chicken DT40 cells are sensitive to RNA polymerase I inhibitors and exhibit decreased transcription activity, based on a transcription run-on assay. Domain analyses of Spt2 revealed that the C-terminal region, containing the region homologous to yeast Spt2, is responsible for histone binding, while the central region is essential for nucleolar localization and DNA binding. Based on these results, we conclude that vertebrate Spt2 is a novel histone chaperone with a separate DNA binding domain, facilitating ribosomal DNA transcription through chromatin remodeling during transcription.
- Published
- 2013
9. Genetically encoded system to track histone modification in vivo
- Author
-
Sato, Y., Mukai, M., Ueda, J., Muraki, M., Stasevich, T. J., Horikoshi, N., Kujirai, T., Kita, H., Kimura, T., Hira, S., Okada, Y., Hayashi-Takanaka, Y., Obuse, C., Kurumizaka, H., Kawahara, A., Yamagata, K., Nozaki, N., and Kimura, Hiroshi
- Subjects
Histones/*metabolism ,Drosophila Proteins/metabolism ,Green Fluorescent Proteins ,Molecular Sequence Data ,Intracellular Space ,Embryonic Development ,SAP30 ,Biology ,Article ,Cell Line ,Histones ,Mice ,Histone H2A ,Drosophila Proteins ,Histone code ,Animals ,Humans ,Amino Acid Sequence ,Zebrafish ,Green Fluorescent Proteins/metabolism ,Genetics ,Histone deacetylase 5 ,Multidisciplinary ,HDAC11 ,Histone deacetylase 2 ,Lysine ,Intracellular Space/metabolism ,Acetylation ,HDAC4 ,Cell biology ,Drosophila melanogaster ,Genetic Techniques ,Protein Processing, Post-Translational ,Histone methyltransferase ,Single-Chain Antibodies/metabolism ,Single-Chain Antibodies ,Drosophila melanogaster/embryology/metabolism ,Lysine/metabolism - Abstract
Post-translational histone modifications play key roles in gene regulation, development, and differentiation, but their dynamics in living organisms remain almost completely unknown. To address this problem, we developed a genetically encoded system for tracking histone modifications by generating fluorescent modification-specific intracellular antibodies (mintbodies) that can be expressed in vivo. To demonstrate, an H3 lysine 9 acetylation specific mintbody (H3K9ac-mintbody) was engineered and stably expressed in human cells. In good agreement with the localization of its target acetylation, H3K9ac-mintbody was enriched in euchromatin, and its kinetics measurably changed upon treatment with a histone deacetylase inhibitor. We also generated transgenic fruit fly and zebrafish stably expressing H3K9ac-mintbody for in vivo tracking. Dramatic changes in H3K9ac-mintbody localization during Drosophila embryogenesis could highlight enhanced acetylation at the start of zygotic transcription around mitotic cycle 7. Together, this work demonstrates the broad potential of mintbody and lays the foundation for epigenetic analysis in vivo.
- Published
- 2013
10. The Human Homologue of Fission Yeast cdc27, p66, Is a Component of Active Human DNA Polymerase ,
- Author
-
Shikata, K., primary, Ohta, S., additional, Yamada, K., additional, Obuse, C., additional, Yoshikawa, H., additional, and Tsurimoto, T., additional
- Published
- 2001
- Full Text
- View/download PDF
11. Interaction of transcription factor YY1 with a replication-enhancing element, REE1, in an autonomously replicating human chromosome fragment
- Author
-
Obuse, C., primary, Okazaki, T., additional, and Masukata, H., additional
- Published
- 1998
- Full Text
- View/download PDF
12. Characterization of a novel CDC gene (ORC1) partly homologous to CDC6 of Saccharomyces cerevisiae.
- Author
-
Hori, Y, primary, Shirahige, K, additional, Obuse, C, additional, Tsurimoto, T, additional, and Yoshikawa, H, additional
- Published
- 1996
- Full Text
- View/download PDF
13. A replication-enhancing element with transcriptional silencer activity in autonomously replicating human chromosomal fragments.
- Author
-
Obuse, C, primary, Okuno, Y, additional, Okazaki, T, additional, and Masukata, H, additional
- Published
- 1996
- Full Text
- View/download PDF
14. Autonomous replication of human chromosomal DNA fragments in human cells.
- Author
-
Masukata, H, primary, Satoh, H, additional, Obuse, C, additional, and Okazaki, T, additional
- Published
- 1993
- Full Text
- View/download PDF
15. Association of human origin recognition complex 1 with chromatin DNA and nuclease-resistant nuclear structures.
- Author
-
Tatsumi, Y, Tsurimoto, T, Shirahige, K, Yoshikawa, H, and Obuse, C
- Abstract
An origin recognition complex (ORC) consisting of six polypeptides has been identified as a DNA replication origin-binding factor in Saccharomyces cerevisiae. Homologues of ORC subunits have been discovered among eukaryotes, and we have prepared monoclonal antibodies against a human homologue of ORC1 (hORC1) to study its localization in human cells. It was thus found to associate with nuclei throughout the cell cycle and to be resistant to nonionic detergent treatment, in contrast to MCM proteins, which are other replication factors, the association of which with nuclei is clearly dependent on the phase of the cell cycle. A characteristic feature of hORC1 is dissociation by NaCl in a narrow concentration range around 0.25 M, suggesting interaction with some specific partner(s) in nuclei. Nuclease treatment experiments and UV cross-linking experiments further indicated interaction with both nuclease-resistant nuclear structures and chromatin DNA. Although its DNA binding was unaffected, some variation in the cell cycle was apparent, the association with nuclear structures being less stable in the M phase. Interestingly, the less stable association occurred concomitantly with hyperphosphorylation of hORC1, suggesting that this hyperphosphorylation may be involved in M phase changes.
- Published
- 2000
16. Distribution of histone H4 modifications as revealed by a panel of specific monoclonal antibodies
- Author
-
Hayashi-Takanaka, Y., Maehara, K., Harada, A., Umehara, T., Yokoyama, S., Obuse, C., Ohkawa, Y., Nozaki, N., and Kimura, Hiroshi
- Subjects
Monoclonal antibody ,Chromatin Immunoprecipitation ,Blotting, Western ,Methylation ,Article ,Epigenesis, Genetic ,Histone H4 ,Histones ,Mice ,Histone H2A ,Histone methylation ,Genetics ,Histone code ,Animals ,Humans ,Epigenomics ,Cell Line, Transformed ,biology ,Antibodies, Monoclonal ,High-Throughput Nucleotide Sequencing ,Acetylation ,Immunohistochemistry ,Chromatin ,Histone ,Biochemistry ,Histone methyltransferase ,Immunoglobulin G ,biology.protein ,Epigenetics ,Histone modification ,Chromatin immunoprecipitation ,Protein Processing, Post-Translational ,HeLa Cells - Abstract
Post-translational histone modifications play a critical role in genome functions such as epigenetic gene regulation and genome maintenance. The tail of the histone H4 N-terminus contains several amino acids that can be acetylated and methylated. Some of these modifications are known to undergo drastic changes during the cell cycle. In this study, we generated a panel of mouse monoclonal antibodies against histone H4 modifications, including acetylation at K5, K8, K12, and K16, and different levels of methylation at K20. Their specificity was evaluated by ELISA and immunoblotting using synthetic peptide and recombinant proteins that harbor specific modifications or amino acid substitutions. Immunofluorescence confirmed the characteristic distributions of target modifications. An H4K5 acetylation (H4K5ac)-specific antibody CMA405 reacted with K5ac only when the neighboring K8 was unacetylated. This unique feature allowed us to detect newly assembled H4, which is diacetylated at K5 and K12, and distinguish it from hyperacetylated H4, where K5 and K8 are both acetylated. Chromatin immunoprecipiation combined with deep sequencing (ChIP-seq) revealed that acetylation of both H4K8 and H4K16 were enriched around transcription start sites. These extensively characterized and highly specific antibodies will be useful for future epigenetics and epigenome studies.
- Full Text
- View/download PDF
17. Structural evidence for protein-protein interaction between the non-canonical methyl-CpG-binding domain of SETDB proteins and C11orf46.
- Author
-
Mahana Y, Ariyoshi M, Nozawa RS, Shibata S, Nagao K, Obuse C, and Shirakawa M
- Subjects
- Humans, Cysteine metabolism, DNA metabolism, DNA Methylation, DNA-Binding Proteins chemistry, Transcription Factors metabolism
- Abstract
SETDB1 and SETDB2 mediate trimethylation of histone H3 lysine 9 (H3K9), an epigenetic hallmark of repressive chromatin. They contain a non-canonical methyl-CpG-binding domain (MBD) and bifurcated SET domain, implying interplay between H3K9 trimethylation and DNA methylation in SETDB functions. Here, we report the crystal structure of human SETDB2 MBD bound to the cysteine-rich domain of a zinc-binding protein, C11orf46. SETDB2 MBD comprises the conserved MBD core and a unique N-terminal extension. Although the MBD core has the conserved basic concave surface for DNA binding, it utilizes it for recognition of the cysteine-rich domain of C11orf46. This interaction involves the conserved arginine finger motif and the unique N-terminal extension of SETDB2 MBD, with a contribution from intermolecular β-sheet formation. Thus, the non-canonical MBD of SETDB1/2 seems to have lost methylated DNA-binding ability but gained a protein-protein interaction surface. Our findings provide insight into the molecular assembly of SETDB-associated repression complexes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Functional domains of nuclear long noncoding RNAs: Insights into gene regulation and intracellular architecture.
- Author
-
Obuse C and Hirose T
- Subjects
- Animals, Gene Expression Regulation, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Mammals metabolism, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism
- Abstract
Recent functional research on long noncoding RNAs (lncRNAs) has revealed their significant regulatory roles in gene expression and intracellular architecture. Well-characterized examples of such lncRNAs include Xist and NEAT1_2, which play critical roles in heterochromatin formation of inactive X-chromosomes and paraspeckle assembly, in mammalian cells. Both lncRNAs possess modular domain structures with multiple functionally distinct domains that serve as platforms for specific RNA-binding proteins (RBPs), which dictate the function of each lncRNA. Some of these RBPs bind characteristic RNA structures, which can be targeted by small chemical compounds that modulate lncRNA function by perturbing the interaction of RBPs with the RNA structures. Therefore, RNA structures hidden in lncRNAs represent a novel and potent type of therapeutic target., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this article., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. Replication dynamics identifies the folding principles of the inactive X chromosome.
- Author
-
Poonperm R, Ichihara S, Miura H, Tanigawa A, Nagao K, Obuse C, Sado T, and Hiratani I
- Subjects
- X Chromosome Inactivation, DNA Replication, Heterochromatin genetics, X Chromosome genetics
- Abstract
Chromosome-wide late replication is an enigmatic hallmark of the inactive X chromosome (Xi). How it is established and what it represents remains obscure. By single-cell DNA replication sequencing, here we show that the entire Xi is reorganized to replicate rapidly and uniformly in late S-phase during X-chromosome inactivation (XCI), reflecting its relatively uniform structure revealed by 4C-seq. Despite this uniformity, only a subset of the Xi became earlier replicating in SmcHD1-mutant cells. In the mutant, these domains protruded out of the Xi core, contacted each other and became transcriptionally reactivated. 4C-seq suggested that they constituted the outermost layer of the Xi even before XCI and were rich in escape genes. We propose that this default positioning forms the basis for their inherent heterochromatin instability in cells lacking the Xi-binding protein SmcHD1 or exhibiting XCI escape. These observations underscore the importance of 3D genome organization for heterochromatin stability and gene regulation., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. Dominant-negative variants in CBX1 cause a neurodevelopmental disorder.
- Author
-
Kuroda Y, Iwata-Otsubo A, Dias KR, Temple SEL, Nagao K, De Hayr L, Zhu Y, Isobe SY, Nishibuchi G, Fiordaliso SK, Fujita Y, Rippert AL, Baker SW, Leung ML, Koboldt DC, Harman A, Keena BA, Kazama I, Subramanian GM, Manickam K, Schmalz B, Latsko M, Zackai EH, Edwards M, Evans CA, Dulik MC, Buckley MF, Yamashita T, O'Brien WT, Harvey RJ, Obuse C, Roscioli T, and Izumi K
- Subjects
- Animals, Mice, Chromatin genetics, Chromosomal Proteins, Non-Histone genetics, Histones genetics, Histones metabolism, Chromobox Protein Homolog 5, Heterochromatin
- Abstract
Purpose: This study aimed to establish variants in CBX1, encoding heterochromatin protein 1β (HP1β), as a cause of a novel syndromic neurodevelopmental disorder., Methods: Patients with CBX1 variants were identified, and clinician researchers were connected using GeneMatcher and physician referrals. Clinical histories were collected from each patient. To investigate the pathogenicity of identified variants, we performed in vitro cellular assays and neurobehavioral and cytological analyses of neuronal cells obtained from newly generated Cbx1 mutant mouse lines., Results: In 3 unrelated individuals with developmental delay, hypotonia, and autistic features, we identified heterozygous de novo variants in CBX1. The identified variants were in the chromodomain, the functional domain of HP1β, which mediates interactions with chromatin. Cbx1 chromodomain mutant mice displayed increased latency-to-peak response, suggesting the possibility of synaptic delay or myelination deficits. Cytological and chromatin immunoprecipitation experiments confirmed the reduction of mutant HP1β binding to heterochromatin, whereas HP1β interactome analysis demonstrated that the majority of HP1β-interacting proteins remained unchanged between the wild-type and mutant HP1β., Conclusion: These collective findings confirm the role of CBX1 in developmental disabilities through the disruption of HP1β chromatin binding during neurocognitive development. Because HP1β forms homodimers and heterodimers, mutant HP1β likely sequesters wild-type HP1β and other HP1 proteins, exerting dominant-negative effects., Competing Interests: Conflict of Interest The authors declare no conflicts of interest., (Copyright © 2023 American College of Medical Genetics and Genomics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Inner nuclear membrane proteins Lem2 and Bqt4 interact with different lipid synthesis enzymes in fission yeast.
- Author
-
Hirano Y, Kinugasa Y, Kubota Y, Obuse C, Haraguchi T, and Hiraoka Y
- Subjects
- Nuclear Envelope metabolism, Nuclear Proteins metabolism, Membrane Proteins metabolism, Lipids, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins metabolism
- Abstract
The nuclear envelope (NE) is a double-membrane structure consisting of inner and outer membranes that spatially separate the nucleus from the cytoplasm, and its function is critical for cellular functions such as genome maintenance. In the fission yeast, Schizosaccharomyces pombe, the inner nuclear membrane proteins, Lem2 and Bqt4, play pivotal roles in maintaining the NE structure. We previously found that the double deletion of lem2+ and bqt4+ causes a synthetic lethal defect associated with severe NE rupture, and overexpression of Elo2, a solo very-long-chain fatty acid elongase, suppresses this defect by restoring the NE. However, the molecular basis of this restoration remains elusive. To address this, we identified Lem2- and Bqt4-binding proteins via immunoprecipitation and mass spectrometry in this study. Forty-five and 23 proteins were identified as Lem2- and Bqt4-binding proteins, respectively. Although these binding proteins partially overlapped, Lem2 and Bqt4 interacted with different types of lipid metabolic enzymes: Cho2, Ole1 and Erg11 for Lem2 and Cwh43 for Bqt4. These enzymes are known to be involved in various lipid synthesis processes, suggesting that Lem2 and Bqt4 may contribute to the regulation of lipid synthesis by binding to these enzymes., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Japanese Biochemical Society. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Polycomb repressive complexes 1 and 2 are each essential for maintenance of X inactivation in extra-embryonic lineages.
- Author
-
Masui O, Corbel C, Nagao K, Endo TA, Kezuka F, Diabangouaya P, Nakayama M, Kumon M, Koseki Y, Obuse C, Koseki H, and Heard E
- Subjects
- Female, Mice, Animals, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Polycomb-Group Proteins genetics, X Chromosome genetics, X Chromosome metabolism, Mammals metabolism, Polycomb Repressive Complex 1 genetics, Polycomb Repressive Complex 1 metabolism, X Chromosome Inactivation genetics
- Abstract
In female mammals, one of the two X chromosomes becomes inactivated during development by X-chromosome inactivation (XCI). Although Polycomb repressive complex (PRC) 1 and PRC2 have both been implicated in gene silencing, their exact roles in XCI during in vivo development have remained elusive. To this end, we have studied mouse embryos lacking either PRC1 or PRC2. Here we demonstrate that the loss of either PRC has a substantial impact on maintenance of gene silencing on the inactive X chromosome (Xi) in extra-embryonic tissues, with overlapping yet different genes affected, indicating potentially independent roles of the two complexes. Importantly, a lack of PRC1 does not affect PRC2/H3K27me3 accumulation and a lack of PRC2 does not impact PRC1/H2AK119ub1 accumulation on the Xi. Thus PRC1 and PRC2 contribute independently to the maintenance of XCI in early post-implantation extra-embryonic lineages, revealing that both Polycomb complexes can be directly involved and differently deployed in XCI., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
23. RNaseH2A downregulation drives inflammatory gene expression via genomic DNA fragmentation in senescent and cancer cells.
- Author
-
Sugawara S, Okada R, Loo TM, Tanaka H, Miyata K, Chiba M, Kawasaki H, Katoh K, Kaji S, Maezawa Y, Yokote K, Nakayama M, Oshima M, Nagao K, Obuse C, Nagayama S, Takubo K, Nakanishi A, Kanemaki MT, Hara E, and Takahashi A
- Subjects
- Animals, Mice, Cellular Senescence genetics, DNA Fragmentation, Down-Regulation, Gene Expression, Genomics, Ligands, Nucleotides, Phenotype, Humans, Cell Line, DNA, Neoplasms genetics, Neoplasms metabolism
- Abstract
Cellular senescence caused by oncogenic stimuli is associated with the development of various age-related pathologies through the senescence-associated secretory phenotype (SASP). SASP is mediated by the activation of cytoplasmic nucleic acid sensors. However, the molecular mechanism underlying the accumulation of nucleotide ligands in senescent cells is unclear. In this study, we revealed that the expression of RNaseH2A, which removes ribonucleoside monophosphates (rNMPs) from the genome, is regulated by E2F transcription factors, and it decreases during cellular senescence. Residual rNMPs cause genomic DNA fragmentation and aberrant activation of cytoplasmic nucleic acid sensors, thereby provoking subsequent SASP factor gene expression in senescent cells. In addition, RNaseH2A expression was significantly decreased in aged mouse tissues and cells from individuals with Werner syndrome. Furthermore, RNaseH2A degradation using the auxin-inducible degron system induced the accumulation of nucleotide ligands and induction of certain tumourigenic SASP-like factors, promoting the metastatic properties of colorectal cancer cells. Our results indicate that RNaseH2A downregulation provokes SASP through nucleotide ligand accumulation, which likely contributes to the pathological features of senescent, progeroid, and cancer cells., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. SmcHD1 underlies the formation of H3K9me3 blocks on the inactive X chromosome in mice.
- Author
-
Ichihara S, Nagao K, Sakaguchi T, Obuse C, and Sado T
- Subjects
- Animals, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Female, Fibroblasts metabolism, Germ Layers metabolism, Mammals genetics, Mice, X Chromosome genetics, X Chromosome metabolism, Histones metabolism, X Chromosome Inactivation genetics
- Abstract
Stable silencing of the inactive X chromosome (Xi) in female mammals is crucial for the development of embryos and their postnatal health. SmcHD1 is essential for stable silencing of the Xi, and its functional deficiency results in derepression of many X-inactivated genes. Although SmcHD1 has been suggested to play an important role in the formation of higher-order chromatin structure of the Xi, the underlying mechanism is largely unknown. Here, we explore the epigenetic state of the Xi in SmcHD1-deficient epiblast stem cells and mouse embryonic fibroblasts in comparison with their wild-type counterparts. The results suggest that SmcHD1 underlies the formation of H3K9me3-enriched blocks on the Xi, which, although the importance of H3K9me3 has been largely overlooked in mice, play a crucial role in the establishment of the stably silenced state. We propose that the H3K9me3 blocks formed on the Xi facilitate robust heterochromatin formation in combination with H3K27me3, and that the substantial loss of H3K9me3 caused by SmcHD1 deficiency leads to aberrant distribution of H3K27me3 on the Xi and derepression of X-inactivated genes., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
25. Chromatin loading of MCM hexamers is associated with di-/tri-methylation of histone H4K20 toward S phase entry.
- Author
-
Hayashi-Takanaka Y, Hayashi Y, Hirano Y, Miyawaki-Kuwakado A, Ohkawa Y, Obuse C, Kimura H, Haraguchi T, and Hiraoka Y
- Subjects
- DNA Replication, HeLa Cells, Humans, Methylation, Cell Cycle, DNA metabolism, Histones metabolism
- Abstract
DNA replication is a key step in initiating cell proliferation. Loading hexameric complexes of minichromosome maintenance (MCM) helicase onto DNA replication origins during the G1 phase is essential for initiating DNA replication. Here, we examined MCM hexamer states during the cell cycle in human hTERT-RPE1 cells using multicolor immunofluorescence-based, single-cell plot analysis, and biochemical size fractionation. Experiments involving cell-cycle arrest at the G1 phase and release from the arrest revealed that a double MCM hexamer was formed via a single hexamer during G1 progression. A single MCM hexamer was recruited to chromatin in the early G1 phase. Another single hexamer was recruited to form a double hexamer in the late G1 phase. We further examined relationship between the MCM hexamer states and the methylation levels at lysine 20 of histone H4 (H4K20) and found that the double MCM hexamer state was correlated with di/trimethyl-H4K20 (H4K20me2/3). Inhibiting the conversion from monomethyl-H4K20 (H4K20me1) to H4K20me2/3 retained the cells in the single MCM hexamer state. Non-proliferative cells, including confluent cells or Cdk4/6 inhibitor-treated cells, also remained halted in the single MCM hexamer state. We propose that the single MCM hexamer state is a halting step in the determination of cell cycle progression., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
26. Protein phosphatase 1 acts as a RIF1 effector to suppress DSB resection prior to Shieldin action.
- Author
-
Isobe SY, Hiraga SI, Nagao K, Sasanuma H, Donaldson AD, and Obuse C
- Subjects
- BRCA1 Protein metabolism, Base Sequence, Endodeoxyribonucleases metabolism, HeLa Cells, Homologous Recombination drug effects, Humans, Multiprotein Complexes metabolism, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Protein Binding drug effects, Cell Cycle Proteins metabolism, DNA Breaks, Double-Stranded drug effects, DNA-Binding Proteins metabolism, Protein Phosphatase 1 metabolism, Telomere-Binding Proteins metabolism
- Abstract
DNA double-strand breaks (DSBs) are repaired mainly by non-homologous end joining (NHEJ) or homologous recombination (HR). RIF1 negatively regulates resection through the effector Shieldin, which associates with a short 3' single-stranded DNA (ssDNA) overhang by the MRN (MRE11-RAD50-NBS1) complex, to prevent further resection and HR repair. In this study, we show that RIF1, but not Shieldin, inhibits the accumulation of CtIP at DSB sites immediately after damage, suggesting that RIF1 has another effector besides Shieldin. We find that protein phosphatase 1 (PP1), a known RIF1 effector in replication, localizes at damage sites dependent on RIF1, where it suppresses downstream CtIP accumulation and limits the resection by the MRN complex. PP1 therefore acts as a RIF1 effector distinct from Shieldin. Furthermore, PP1 deficiency in the context of Shieldin depletion elevates HR immediately after irradiation. We conclude that PP1 inhibits resection before the action of Shieldin to prevent precocious HR in the early phase of the damage response., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Mapping replication timing domains genome wide in single mammalian cells with single-cell DNA replication sequencing.
- Author
-
Miura H, Takahashi S, Shibata T, Nagao K, Obuse C, Okumura K, Ogata M, Hiratani I, and Takebayashi SI
- Subjects
- Animals, Cell Line, Humans, S Phase genetics, DNA Replication, Genomics methods, Sequence Analysis, DNA methods, Single-Cell Analysis methods
- Abstract
Replication timing (RT) domains are stable units of chromosome structure that are regulated in the context of development and disease. Conventional genome-wide RT mapping methods require many S-phase cells for either the effective enrichment of replicating DNA through bromodeoxyuridine (BrdU) immunoprecipitation or the determination of copy-number differences during S-phase, which precludes their application to non-abundant cell types and single cells. Here, we provide a simple, cost-effective, and robust protocol for single-cell DNA replication sequencing (scRepli-seq). The scRepli-seq methodology relies on whole-genome amplification (WGA) of genomic DNA (gDNA) from single S-phase cells and next-generation sequencing (NGS)-based determination of copy-number differences that arise between replicated and unreplicated DNA. Haplotype-resolved scRepli-seq, which distinguishes pairs of homologous chromosomes within a single cell, is feasible by using single-nucleotide polymorphism (SNP)/indel information. We also provide computational pipelines for quality control, normalization, and binarization of the scRepli-seq data. The experimental portion of this protocol (before sequencing) takes 3 d.
- Published
- 2020
- Full Text
- View/download PDF
28. Regulation of an adaptor protein STING by Hsp90β to enhance innate immune responses against microbial infections.
- Author
-
Sato S, Li K, Sakurai N, Hashizume M, Baidya S, Nonaka H, Noguchi K, Ishikawa K, Obuse C, and Takaoka A
- Subjects
- Animals, DNA, Viral immunology, HEK293 Cells, HSP90 Heat-Shock Proteins metabolism, Herpesvirus 1, Human genetics, Herpesvirus 1, Human immunology, Humans, Immune Evasion immunology, Immunity, Innate, Listeria monocytogenes genetics, Listeria monocytogenes immunology, Membrane Proteins metabolism, Mice, RAW 264.7 Cells, Signal Transduction, Viral Proteins metabolism, HSP90 Heat-Shock Proteins immunology, Membrane Proteins immunology
- Abstract
Stimulator of interferon genes (STING) plays important roles in the DNA-mediated innate immune responses. However, the regulatory mechanism of STING in terms of stabilization is not fully understood. Here, we identified the chaperone protein Hsp90s as novel STING interacting proteins. Treatment with an Hsp90 inhibitor 17-AAG and knockdown of Hsp90β but not Hsp90α reduced STING at protein level, resulted in the suppression of IFN induction in response to stimulation with cGAMP, and infections with HSV-1 and Listeria monocytogenes. Collectively, our results suggest that the control of STING protein by Hsp90β is a critical biological process in the DNA sensing pathways., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
29. Rare variant of the epigenetic regulator SMCHD1 in a patient with pituitary hormone deficiency.
- Author
-
Kinjo K, Nagasaki K, Muroya K, Suzuki E, Ishiwata K, Nakabayashi K, Hattori A, Nagao K, Nozawa RS, Obuse C, Miyado K, Ogata T, Fukami M, and Miyado M
- Subjects
- Adolescent, Computer Simulation, Epigenesis, Genetic, Female, Genetic Association Studies, Genetic Testing, Humans, Male, Middle Aged, Pedigree, Exome Sequencing, Young Adult, Amino Acid Substitution, Chromosomal Proteins, Non-Histone genetics, DNA Methylation, Hypogonadism genetics, Hypopituitarism genetics
- Abstract
Isolated hypogonadotropic hypogonadism (IHH), combined pituitary hormone deficiency (CPHD), and septo-optic dysplasia (SOD) constitute a disease spectrum whose etiology remains largely unknown. This study aimed to clarify whether mutations in SMCHD1, an epigenetic regulator gene, might underlie this disease spectrum. SMCHD1 is a causative gene for Bosma arhinia microphthalmia syndrome characterized by arhinia, microphthalmia and IHH. We performed mutation screening of SMCHD1 in patients with etiology-unknown IHH (n = 31) or CPHD (n = 43, 19 of whom also satisfied the SOD diagnostic criteria). Rare variants were subjected to in silico analyses and classified according to the American College of Medical Genetics and Genomics guidelines. Consequently, a rare likely pathogenic variant, p.Asp398Asn, was identified in one patient. The patient with p.Asp398Asn exhibited CPHD, optic nerve hypoplasia, and a thin retinal nerve fiber layer, and therefore satisfied the criteria of SOD. This patient showed a relatively low DNA methylation level of the 52 SMCHD1-target CpG sites at the D4Z4 locus. Exome sequencing for the patient excluded additional variants in other IHH/CPHD-causative genes. In vitro assays suggested functional impairment of the p.Asp398Asn variant. These results provide the first indication that SMCHD1 mutations represent a rare genetic cause of the HH-related disease spectrum.
- Published
- 2020
- Full Text
- View/download PDF
30. Homozygous nonsense variant in LRIF1 associated with facioscapulohumeral muscular dystrophy.
- Author
-
Hamanaka K, Šikrová D, Mitsuhashi S, Masuda H, Sekiguchi Y, Sugiyama A, Shibuya K, Lemmers RJLF, Goossens R, Ogawa M, Nagao K, Obuse C, Noguchi S, Hayashi YK, Kuwabara S, Balog J, Nishino I, and van der Maarel SM
- Subjects
- Biopsy, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cells, Cultured, Chromatin ultrastructure, Chromosomal Proteins, Non-Histone metabolism, Chromosomes, Human, Pair 4 genetics, Codon, Nonsense, Consanguinity, Fibroblasts, Frameshift Mutation, Gene Duplication, Gene Expression Regulation, Homeodomain Proteins biosynthesis, Homeodomain Proteins genetics, Homozygote, Humans, Male, Middle Aged, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal pathology, Pedigree, Protein Isoforms genetics, Repetitive Sequences, Nucleic Acid, Cell Cycle Proteins genetics, Muscular Dystrophy, Facioscapulohumeral genetics
- Abstract
Objective: Facioscapulohumeral muscular dystrophy (FSHD) is a heterogenetic disorder predominantly characterized by progressive facial and scapular muscle weakness. Patients with FSHD either have a contraction of the D4Z4 repeat on chromosome 4q35 or mutations in D4Z4 chromatin modifiers SMCHD1 and DNMT3B, both causing D4Z4 chromatin relaxation and inappropriate expression of the D4Z4-encoded DUX4 gene in skeletal muscle. In this study, we tested the hypothesis whether LRIF1 , a known SMCHD1 protein interactor, is a disease gene for idiopathic FSHD2., Methods: Clinical examination of a patient with idiopathic FSHD2 was combined with pathologic muscle biopsy examination and with genetic, epigenetic, and molecular studies., Results: A homozygous LRIF1 mutation was identified in a patient with a clinical phenotype consistent with FSHD. This mutation resulted in the absence of the long isoform of LRIF1 protein, D4Z4 chromatin relaxation, and DUX4 and DUX4 target gene expression in myonuclei, all molecular and epigenetic hallmarks of FSHD. In concordance, LRIF1 was shown to bind to the D4Z4 repeat, and knockdown of the LRIF1 long isoform in muscle cells results in DUX4 and DUX4 target gene expression., Conclusion: LRIF1 is a bona fide disease gene for FSHD2. This study further reinforces the unifying genetic mechanism, which postulates that FSHD is caused by D4Z4 chromatin relaxation, resulting in inappropriate DUX4 expression in skeletal muscle., (© 2020 American Academy of Neurology.)
- Published
- 2020
- Full Text
- View/download PDF
31. Subacute cutaneous lupus erythematosus with melanocyte elimination induced by pembrolizumab.
- Author
-
Ogawa-Momohara M, Muro Y, Goto K, Obuse C, Satoh M, Kono M, and Akiyama M
- Subjects
- Aged, 80 and over, Autoantibodies blood, Autoantibodies immunology, Autoantigens immunology, Facial Neoplasms drug therapy, Facial Neoplasms immunology, Facial Neoplasms pathology, Humans, Lupus Erythematosus, Cutaneous chemically induced, Lupus Erythematosus, Cutaneous drug therapy, Lupus Erythematosus, Cutaneous immunology, Male, Melanocytes immunology, Melanocytes pathology, Melanoma drug therapy, Melanoma immunology, Melanoma secondary, Prednisolone therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, RNA, Small Cytoplasmic immunology, Ribonucleoproteins immunology, Skin drug effects, Skin immunology, Skin pathology, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Skin Neoplasms pathology, Antibodies, Monoclonal, Humanized adverse effects, Immune Checkpoint Inhibitors adverse effects, Lupus Erythematosus, Cutaneous diagnosis, Melanocytes drug effects
- Published
- 2020
- Full Text
- View/download PDF
32. KDM2A-dependent reduction of rRNA transcription on glucose starvation requires HP1 in cells, including triple-negative breast cancer cells.
- Author
-
Okamoto K, Tanaka Y, Ogasawara S, Obuse C, Nakayama JI, Yano H, and Tsuneoka M
- Abstract
Triple-negative breast cancer (TNBC) is very aggressive and lacks specific therapeutic targets. Ribosome RNAs (rRNAs) are central components of ribosomes and transcribed in nucleoli, and the level of rRNA transcription greatly affects ribosome production and cell proliferation. We have reported that an epigenetic protein, KDM2A, exists in nucleoli and reduces rRNA transcription on glucose starvation. However, the molecular mechanism is still unclear. The purpose of this study is to examine the KDM2A-dependent regulation mechanism of rRNA transcription. In this study, we turned our attention to the nucleolar accumulation of KDM2A. We found that KDM2A had multiple regions for its nucleolar localization, and one of the regions was directly bound by heterochromatin protein 1γ (HP1γ) using valine 801 in the LxVxL motif of KDM2A. A knockdown of HP1γ or a point mutation of valine 801 in KDM2A decreased the nucleolar accumulation of KDM2A, and suppressed the reduction of rRNA transcription on glucose starvation. These results uncovered a novel function of HP1γ: the regulation of rRNA transcription, and suggested that HP1γ stimulates the nucleolar accumulation of KDM2A to support the KDM2A-dependent regulation of rRNA transcription. HP1γ was expressed in cancer cells in all breast carcinoma tissues examined, including TNBC tissues. A knockdown of HP1γ in a TNBC cell line, MDA-MB-231 cells, reduced the nucleolar accumulation of KDM2A, and suppressed the reductions of rRNA transcription and cell proliferation on glucose starvation. These results suggest that the KDM2A-dependent regulation of rRNA transcription requires HP1γ, and thus may be applicable to the treatment of TNBC., Competing Interests: CONFLICTS OF INTEREST None
- Published
- 2019
- Full Text
- View/download PDF
33. Asymmetrical localization of Nup107-160 subcomplex components within the nuclear pore complex in fission yeast.
- Author
-
Asakawa H, Kojidani T, Yang HJ, Ohtsuki C, Osakada H, Matsuda A, Iwamoto M, Chikashige Y, Nagao K, Obuse C, Hiraoka Y, and Haraguchi T
- Subjects
- Active Transport, Cell Nucleus genetics, Cell Cycle genetics, Cell Division genetics, Cell Nucleus genetics, Cell Nucleus ultrastructure, Cytoplasm genetics, Cytoplasm ultrastructure, Humans, Meiosis genetics, Microscopy, Fluorescence, Nuclear Envelope genetics, Nuclear Pore ultrastructure, Protein Binding genetics, Saccharomyces cerevisiae genetics, Schizosaccharomyces genetics, Nuclear Pore genetics, Nuclear Pore Complex Proteins genetics, Schizosaccharomyces pombe Proteins genetics
- Abstract
The nuclear pore complex (NPC) forms a gateway for nucleocytoplasmic transport. The outer ring protein complex of the NPC (the Nup107-160 subcomplex in humans) is a key component for building the NPC. Nup107-160 subcomplexes are believed to be symmetrically localized on the nuclear and cytoplasmic sides of the NPC. However, in S. pombe immunoelectron and fluorescence microscopic analyses revealed that the homologous components of the human Nup107-160 subcomplex had an asymmetrical localization: constituent proteins spNup132 and spNup107 were present only on the nuclear side (designated the spNup132 subcomplex), while spNup131, spNup120, spNup85, spNup96, spNup37, spEly5 and spSeh1 were localized only on the cytoplasmic side (designated the spNup120 subcomplex), suggesting the complex was split into two pieces at the interface between spNup96 and spNup107. This contrasts with the symmetrical localization reported in other organisms. Fusion of spNup96 (cytoplasmic localization) with spNup107 (nuclear localization) caused cytoplasmic relocalization of spNup107. In this strain, half of the spNup132 proteins, which interact with spNup107, changed their localization to the cytoplasmic side of the NPC, leading to defects in mitotic and meiotic progression similar to an spNup132 deletion strain. These observations suggest the asymmetrical localization of the outer ring spNup132 and spNup120 subcomplexes of the NPC is necessary for normal cell cycle progression in fission yeast., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
34. Genome-wide stability of the DNA replication program in single mammalian cells.
- Author
-
Takahashi S, Miura H, Shibata T, Nagao K, Okumura K, Ogata M, Obuse C, Takebayashi SI, and Hiratani I
- Subjects
- Animals, Cell Differentiation genetics, Cell Line, DNA Copy Number Variations genetics, DNA Replication Timing genetics, Embryonic Stem Cells physiology, Genome genetics, Genome-Wide Association Study methods, Genomic Instability genetics, Humans, Mice, Mouse Embryonic Stem Cells physiology, S Phase genetics, X Chromosome genetics, DNA genetics, DNA Replication genetics, Mammals genetics
- Abstract
Here, we report a single-cell DNA replication sequencing method, scRepli-seq, a genome-wide methodology that measures copy number differences between replicated and unreplicated DNA. Using scRepli-seq, we demonstrate that replication-domain organization is conserved among individual mouse embryonic stem cells (mESCs). Differentiated mESCs exhibited distinct profiles, which were also conserved among cells. Haplotype-resolved scRepli-seq revealed similar replication profiles of homologous autosomes, while the inactive X chromosome was clearly replicated later than its active counterpart. However, a small degree of cell-to-cell replication-timing heterogeneity was present, which was smallest at the beginning and the end of S phase. In addition, developmentally regulated domains were found to deviate from others and showed a higher degree of heterogeneity, thus suggesting a link to developmental plasticity. Moreover, allelic expression imbalance was found to strongly associate with replication-timing asynchrony. Our results form a foundation for single-cell-level understanding of DNA replication regulation and provide insights into three-dimensional genome organization.
- Published
- 2019
- Full Text
- View/download PDF
35. Publisher Correction: Exosomes maintain cellular homeostasis by excreting harmful DNA from cells.
- Author
-
Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S, Takasugi M, Watanabe S, Kanemaki MT, Obuse C, and Hara E
- Abstract
This Article contains errors in Fig. 4. In panel d, the lanes of the western blot should have been labeled '1.05','1.06, '1.09', '1.11' '1.13', '1.16', '1.19', '1.22', '1.24', '1.25'. The correct version of Figure 4 appears in the associated Publisher Correction.
- Published
- 2018
- Full Text
- View/download PDF
36. Functional characterization of lysosomal interaction of Akt with VRK2.
- Author
-
Hirata N, Suizu F, Matsuda-Lennikov M, Tanaka T, Edamura T, Ishigaki S, Donia T, Lithanatudom P, Obuse C, Iwanaga T, and Noguchi M
- Subjects
- Autophagy, Cell Line, Cell Proliferation, Humans, Protein Binding, Lysosomes metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Serine-threonine kinase Akt (also known as PKB, protein kinase B), a core intracellular mediator of cell survival, is involved in various human cancers and has been suggested to play an important role in the regulation of autophagy in mammalian cells. Nonetheless, the physiological function of Akt in the lysosomes is currently unknown. We have reported previously that PtdIns(3)P-dependent lysosomal accumulation of the Akt-Phafin2 complex is a critical step for autophagy induction. Here, to characterize the molecular function of activated Akt in the lysosomes in the process of autophagy, we searched for the molecules that interact with the Akt complex at the lysosomes after induction of autophagy. By time-of-flight-mass spectrometry (TOF/MS) analysis, kinases of the VRK family, a unique serine-threonine family of kinases in the human kinome, were identified. VRK2 interacts with Akt1 and Akt2, but not with Akt3; the C terminus of Akt and the N terminus of VRK2 facilitate the interaction of Akt and VRK2 in mammalian cells. The kinase-dead form of VRK2A (KD VRK2A) failed to interact with Akt in coimmunoprecipitation assays. Bimolecular fluorescence complementation (BiFC) experiments showed that, in the lysosomes, Akt interacted with VRK2A but not with VRK2B or KD VRK2A. Immunofluorescent assays revealed that VRK2 and phosphorylated Akt accumulated in the lysosomes after autophagy induction. WT VRK2A, but not KD VRK2A or VRK2B, facilitated accumulation of phosphorylated Akt in the lysosomes. Downregulation of VRK2 abrogated the lysosomal accumulation of phosphorylated Akt and impaired nuclear localization of TFEB; these events coincided to inhibition of autophagy induction. The VRK2-Akt complex is required for control of lysosomal size, acidification, bacterial degradation, and for viral replication. Moreover, lysosomal VRK2-Akt controls cellular proliferation and mitochondrial outer-membrane stabilization. Given the roles of autophagy in the pathogenesis of human cancer, the current study provides a novel insight into the oncogenic activity of VRK2-Akt complexes in the lysosomes via modulation of autophagy.
- Published
- 2018
- Full Text
- View/download PDF
37. Role of SmcHD1 in establishment of epigenetic states required for the maintenance of the X-inactivated state in mice.
- Author
-
Sakakibara Y, Nagao K, Blewitt M, Sasaki H, Obuse C, and Sado T
- Subjects
- Animals, Cells, Cultured, Chromosomal Proteins, Non-Histone genetics, Embryo, Mammalian embryology, Female, Fibroblasts cytology, Histones genetics, Histones metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Chromosomal Proteins, Non-Histone metabolism, Epigenesis, Genetic genetics, Genes, X-Linked genetics, X Chromosome Inactivation genetics
- Abstract
X inactivation in mammals is regulated by epigenetic modifications. Functional deficiency of SmcHD1 has been shown to cause de-repression of X-inactivated genes in post-implantation female mouse embryos, suggesting a role of SmcHD1 in the maintenance of X inactivation. Here, we show that de-repression of X-inactivated genes accompanied a local reduction in the enrichment of H3K27me3 in mouse embryonic fibroblasts deficient for SmcHD1. Furthermore, many of these genes overlapped with those having a significantly lower enrichment of H3K27me3 at the blastocyst stage in wild type. Intriguingly, however, depletion of SmcHD1 did not compromise the X-inactivated state in immortalized female mouse embryonic fibroblasts, in which X inactivation had been established and maintained. Taking all these findings together, we suggest that SmcHD1 facilitates the incorporation of H3K27me3 and perhaps other epigenetic modifications at gene loci that are silenced even with the lower enrichment of H3K27me3 at the early stage of X inactivation. The epigenetic state at these loci would, however, remain as it is at the blastocyst stage in the absence of SmcHD1 after implantation, which would eventually compromise the maintenance of the X-inactivated state at later stages., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
38. Nucleosomes around a mismatched base pair are excluded via an Msh2-dependent reaction with the aid of SNF2 family ATPase Smarcad1.
- Author
-
Terui R, Nagao K, Kawasoe Y, Taki K, Higashi TL, Tanaka S, Nakagawa T, Obuse C, Masukata H, and Takahashi TS
- Subjects
- Animals, Base Pair Mismatch genetics, Chromatin Assembly and Disassembly genetics, DNA genetics, DNA metabolism, DNA Helicases genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Transcription Factors genetics, Transcription Factors metabolism, Xenopus laevis, Base Pair Mismatch physiology, DNA Helicases metabolism, DNA Mismatch Repair genetics, MutS Homolog 2 Protein metabolism, Nucleosomes metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Post-replicative correction of replication errors by the mismatch repair (MMR) system is critical for suppression of mutations. Although the MMR system may need to handle nucleosomes at the site of chromatin replication, how MMR occurs in the chromatin environment remains unclear. Here, we show that nucleosomes are excluded from a >1-kb region surrounding a mismatched base pair in Xenopus egg extracts. The exclusion was dependent on the Msh2-Msh6 mismatch recognition complex but not the Mlh1-containing MutL homologs and counteracts both the HIRA- and CAF-1 (chromatin assembly factor 1)-mediated chromatin assembly pathways. We further found that the Smarcad1 chromatin remodeling ATPase is recruited to mismatch-carrying DNA in an Msh2-dependent but Mlh1-independent manner to assist nucleosome exclusion and that Smarcad1 facilitates the repair of mismatches when nucleosomes are preassembled on DNA. In budding yeast, deletion of FUN30 , the homolog of Smarcad1, showed a synergistic increase of spontaneous mutations in combination with MSH6 or MSH3 deletion but no significant increase with MSH2 deletion. Genetic analyses also suggested that the function of Fun30 in MMR is to counteract CAF-1. Our study uncovers that the eukaryotic MMR system has an ability to exclude local nucleosomes and identifies Smarcad1/Fun30 as an accessory factor for the MMR reaction., (© 2018 Terui et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
39. Lem2 is retained at the nuclear envelope through its interaction with Bqt4 in fission yeast.
- Author
-
Hirano Y, Kinugasa Y, Asakawa H, Chikashige Y, Obuse C, Haraguchi T, and Hiraoka Y
- Subjects
- DNA, Fungal genetics, DNA, Fungal metabolism, Nuclear Envelope genetics, Protein Interaction Domains and Motifs, Schizosaccharomyces growth & development, Telomere genetics, DNA-Binding Proteins metabolism, Membrane Proteins metabolism, Nuclear Envelope metabolism, Nuclear Proteins metabolism, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins metabolism, Telomere metabolism
- Abstract
Inner nuclear membrane (INM) proteins are thought to play important roles in modulating nuclear organization and function through their interactions with chromatin. However, these INM proteins share redundant functions in metazoans that pose difficulties for functional studies. The fission yeast Schizosaccharomyces pombe exhibits a relatively small number of INM proteins, and molecular genetic tools are available to separate their redundant functions. In S. pombe, it has been reported that among potentially redundant INM proteins, Lem2 displays a unique genetic interaction with another INM protein, Bqt4, which is involved in anchoring telomeres to the nuclear envelope. Double mutations in the lem2 and bqt4 genes confer synthetic lethality during vegetative growth. Here, we show that Lem2 is retained at the nuclear envelope through its interaction with Bqt4, as the loss of Bqt4 results in the exclusive accumulation of Lem2 to the spindle pole body (SPB). An N-terminal nucleoplasmic region of Lem2 bears affinity to both Bqt4 and the SPB in a competitive manner. In contrast, the synthetic lethality of the lem2 bqt4 double mutant is suppressed by the C-terminal region of Lem2. These results indicate that the N-terminal and C-terminal domains of Lem2 show independent functions with respect to Bqt4., (© 2018 The Authors. Genes to Cells published by Molecular Biology Society of Japan and John Wiley & Sons Australia, Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
40. Defects in dosage compensation impact global gene regulation in the mouse trophoblast.
- Author
-
Sakata Y, Nagao K, Hoki Y, Sasaki H, Obuse C, and Sado T
- Subjects
- Alleles, Animals, Cells, Cultured, Dosage Compensation, Genetic genetics, Embryonic Stem Cells metabolism, Female, Fluorescent Antibody Technique, Mice, X Chromosome genetics, X Chromosome Inactivation genetics, Dosage Compensation, Genetic physiology, Gene Expression Regulation, Developmental genetics, Trophoblasts metabolism
- Abstract
Xist RNA, which is responsible for X inactivation, is a key epigenetic player in the embryogenesis of female mammals. Of the several repeats conserved in Xist RNA, the A-repeat has been shown to be essential for its silencing function in differentiating embryonic stem cells. Here, we introduced a new Xist allele into mouse that produces mutated Xist RNA lacking the A-repeat ( Xist
CAGΔ5' ). XistCAGΔ5' RNA expressed in the embryo coated the X chromosome but failed to silence it. Although imprinted X inactivation was substantially compromised upon paternal transmission, allele-specific RNA-seq in the trophoblast revealed that XistCAGΔ5' RNA still retained some silencing ability. Furthermore, the failure of imprinted X inactivation had more significant impacts than expected on genome-wide gene expression. It is likely that dosage compensation is required not only for equalizing X-linked gene expression between the sexes but also for proper global gene regulation in differentiated female somatic cells., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)- Published
- 2017
- Full Text
- View/download PDF
41. Reconstitution of the oocyte nucleolus in mice through a single nucleolar protein, NPM2.
- Author
-
Ogushi S, Yamagata K, Obuse C, Furuta K, Wakayama T, Matzuk MM, and Saitou M
- Subjects
- Amino Acid Sequence, Animals, Cell Nucleolus metabolism, Chromatin metabolism, Female, Mice, Oocytes cytology, Nucleoplasmins metabolism, Oocytes metabolism
- Abstract
The mammalian oocyte nucleolus, the most prominent subcellular organelle in the oocyte, is vital in early development, yet its key functions and constituents remain unclear. We show here that the parthenotes/zygotes derived from enucleolated oocytes exhibited abnormal heterochromatin formation around parental pericentromeric DNAs, which led to a significant mitotic delay and frequent chromosome mis-segregation upon the first mitotic division. A proteomic analysis identified nucleoplasmin 2 (NPM2) as a dominant component of the oocyte nucleolus. Consistently, Npm2 -deficient oocytes, which lack a normal nucleolar structure, showed chromosome segregation defects similar to those in enucleolated oocytes, suggesting that nucleolar loss, rather than micromanipulation-related damage to the genome, leads to a disorganization of higher-order chromatin structure in pronuclei and frequent chromosome mis-segregation during the first mitosis. Strikingly, expression of NPM2 alone sufficed to reconstitute the nucleolar structure in enucleolated embryos, and rescued their first mitotic division and full-term development. The nucleolus rescue through NPM2 required the pentamer formation and both the N- and C-terminal domains. Our findings demonstrate that the NPM2-based oocyte nucleolus is an essential platform for parental chromatin organization in early embryonic development., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
42. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells.
- Author
-
Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S, Takasugi M, Watanabe S, Kanemaki MT, Obuse C, and Hara E
- Subjects
- Animals, Apoptosis, Cell Cycle Checkpoints, Cell Line, Cell Line, Tumor, Cells, Cultured, Cytoplasm genetics, HEK293 Cells, HeLa Cells, Humans, Mice, Models, Biological, Reactive Oxygen Species metabolism, Cytoplasm metabolism, DNA metabolism, Exosomes metabolism, Homeostasis
- Abstract
Emerging evidence is revealing that exosomes contribute to many aspects of physiology and disease through intercellular communication. However, the biological roles of exosome secretion in exosome-secreting cells have remained largely unexplored. Here we show that exosome secretion plays a crucial role in maintaining cellular homeostasis in exosome-secreting cells. The inhibition of exosome secretion results in the accumulation of nuclear DNA in the cytoplasm, thereby causing the activation of cytoplasmic DNA sensing machinery. This event provokes the innate immune response, leading to reactive oxygen species (ROS)-dependent DNA damage response and thus induce senescence-like cell-cycle arrest or apoptosis in normal human cells. These results, in conjunction with observations that exosomes contain various lengths of chromosomal DNA fragments, indicate that exosome secretion maintains cellular homeostasis by removing harmful cytoplasmic DNA from cells. Together, these findings enhance our understanding of exosome biology, and provide valuable new insights into the control of cellular homeostasis.
- Published
- 2017
- Full Text
- View/download PDF
43. Compositionally distinct nuclear pore complexes of functionally distinct dimorphic nuclei in the ciliate Tetrahymena .
- Author
-
Iwamoto M, Osakada H, Mori C, Fukuda Y, Nagao K, Obuse C, Hiraoka Y, and Haraguchi T
- Subjects
- Conserved Sequence, Macronucleus metabolism, Micronucleus, Germline metabolism, Models, Biological, Nuclear Pore Complex Proteins chemistry, Permeability, Protein Domains, Protein Structure, Secondary, Protozoan Proteins chemistry, Protozoan Proteins metabolism, Sequence Homology, Amino Acid, Nuclear Pore metabolism, Nuclear Pore Complex Proteins metabolism, Tetrahymena thermophila metabolism
- Abstract
The nuclear pore complex (NPC), a gateway for nucleocytoplasmic trafficking, is composed of ∼30 different proteins called nucleoporins. It remains unknown whether the NPCs within a species are homogeneous or vary depending on the cell type or physiological condition. Here, we present evidence for compositionally distinct NPCs that form within a single cell in a binucleated ciliate. In Tetrahymena thermophila , each cell contains both a transcriptionally active macronucleus (MAC) and a germline micronucleus (MIC). By combining in silico analysis, mass spectrometry analysis for immuno-isolated proteins and subcellular localization analysis of GFP-fused proteins, we identified numerous novel components of MAC and MIC NPCs. Core members of the Nup107-Nup160 scaffold complex were enriched in MIC NPCs. Strikingly, two paralogs of Nup214 and of Nup153 localized exclusively to either the MAC or MIC NPCs. Furthermore, the transmembrane components Pom121 and Pom82 localize exclusively to MAC and MIC NPCs, respectively. Our results argue that functional nuclear dimorphism in ciliates is likely to depend on the compositional and structural specificity of NPCs., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
44. Usp7-dependent histone H3 deubiquitylation regulates maintenance of DNA methylation.
- Author
-
Yamaguchi L, Nishiyama A, Misaki T, Johmura Y, Ueda J, Arita K, Nagao K, Obuse C, and Nakanishi M
- Subjects
- Animals, Chromatin metabolism, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA Replication physiology, HeLa Cells, Humans, Ovum, Xenopus, DNA Methylation, Histones metabolism, Ubiquitin-Specific Peptidase 7 metabolism, Ubiquitination
- Abstract
Uhrf1-dependent histone H3 ubiquitylation plays a crucial role in the maintenance of DNA methylation via the recruitment of the DNA methyltransferase Dnmt1 to DNA methylation sites. However, the involvement of deubiquitylating enzymes (DUBs) targeting ubiquitylated histone H3 in the maintenance of DNA methylation is largely unknown. With the use of Xenopus egg extracts, we demonstrate here that Usp7, a ubiquitin carboxyl-terminal hydrolase, forms a stable complex with Dnmt1 and is recruited to DNA methylation sites during DNA replication. Usp7 deubiquitylates ubiquitylated histone H3 in vitro. Inhibition of Usp7 activity or its depletion in egg extracts results in enhanced and extended binding of Dnmt1 to chromatin, suppressing DNA methylation. Depletion of Usp7 in HeLa cells causes enhanced histone H3 ubiquitylation and enlargement of Dnmt1 nuclear foci during DNA replication. Our results thus suggest that Usp7 is a key factor that regulates maintenance of DNA methylation.
- Published
- 2017
- Full Text
- View/download PDF
45. Human RIF1 and protein phosphatase 1 stimulate DNA replication origin licensing but suppress origin activation.
- Author
-
Hiraga SI, Ly T, Garzón J, Hořejší Z, Ohkubo YN, Endo A, Obuse C, Boulton SJ, Lamond AI, and Donaldson AD
- Subjects
- Cell Cycle, Cell Cycle Proteins metabolism, Chromatin genetics, Chromatin metabolism, Humans, Minichromosome Maintenance Proteins metabolism, Phosphorylation, Proteasome Endopeptidase Complex metabolism, Protein Binding, Protein Interaction Domains and Motifs, Protein Phosphatase 1 chemistry, Protein Serine-Threonine Kinases metabolism, Proteolysis, Telomere-Binding Proteins chemistry, DNA Replication, Protein Phosphatase 1 metabolism, Replication Origin, Telomere-Binding Proteins metabolism
- Abstract
The human RIF1 protein controls DNA replication, but the molecular mechanism is largely unknown. Here, we demonstrate that human RIF1 negatively regulates DNA replication by forming a complex with protein phosphatase 1 (PP1) that limits phosphorylation-mediated activation of the MCM replicative helicase. We identify specific residues on four MCM helicase subunits that show hyperphosphorylation upon RIF1 depletion, with the regulatory N-terminal domain of MCM4 being particularly strongly affected. In addition to this role in limiting origin activation, we discover an unexpected new role for human RIF1-PP1 in mediating efficient origin licensing. Specifically, during the G1 phase of the cell cycle, RIF1-PP1 protects the origin-binding ORC1 protein from untimely phosphorylation and consequent degradation by the proteasome. Depletion of RIF1 or inhibition of PP1 destabilizes ORC1, thereby reducing origin licensing. Consistent with reduced origin licensing, RIF1-depleted cells exhibit increased spacing between active origins. Human RIF1 therefore acts as a PP1-targeting subunit that regulates DNA replication positively by stimulating the origin licensing step, and then negatively by counteracting replication origin activation., (© 2017 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2017
- Full Text
- View/download PDF
46. BRCA1 Directs the Repair Pathway to Homologous Recombination by Promoting 53BP1 Dephosphorylation.
- Author
-
Isono M, Niimi A, Oike T, Hagiwara Y, Sato H, Sekine R, Yoshida Y, Isobe SY, Obuse C, Nishi R, Petricci E, Nakada S, Nakano T, and Shibata A
- Subjects
- Ataxia Telangiectasia Mutated Proteins metabolism, Carrier Proteins metabolism, DNA Breaks, Double-Stranded, DNA Repair Enzymes metabolism, Endodeoxyribonucleases, Exodeoxyribonucleases metabolism, G2 Phase, Humans, MRE11 Homologue Protein metabolism, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism, Phosphorylation, S Phase, Telomere-Binding Proteins metabolism, BRCA1 Protein metabolism, DNA Repair, Homologous Recombination, Signal Transduction, Tumor Suppressor p53-Binding Protein 1 metabolism
- Abstract
BRCA1 promotes homologous recombination (HR) by activating DNA-end resection. By contrast, 53BP1 forms a barrier that inhibits DNA-end resection. Here, we show that BRCA1 promotes DNA-end resection by relieving the 53BP1-dependent barrier. We show that 53BP1 is phosphorylated by ATM in S/G
2 phase, promoting RIF1 recruitment, which inhibits resection. 53BP1 is promptly dephosphorylated and RIF1 released, despite remaining unrepaired DNA double-strand breaks (DSBs). When resection is impaired by CtIP/MRE11 endonuclease inhibition, 53BP1 phosphorylation and RIF1 are sustained due to ongoing ATM signaling. BRCA1 depletion also sustains 53BP1 phosphorylation and RIF1 recruitment. We identify the phosphatase PP4C as having a major role in 53BP1 dephosphorylation and RIF1 release. BRCA1 or PP4C depletion impairs 53BP1 repositioning, EXO1 recruitment, and HR progression. 53BP1 or RIF1 depletion restores resection, RAD51 loading, and HR in PP4C-depleted cells. Our findings suggest that BRCA1 promotes PP4C-dependent 53BP1 dephosphorylation and RIF1 release, directing repair toward HR., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
47. Corrigendum to "Clinical, muscle pathological, and genetic features of Japanese facioscapulohumeral muscular dystrophy 2 (FSHD2) patients with SMCHD1 mutations": [Neuromuscular Disorders 26/4-5 (2016) 300-308].
- Author
-
Hamanaka K, Goto K, Arai M, Nagao K, Obuse C, Noguchi S, Hayashi YK, Mitsuhashi S, and Nishino I
- Published
- 2016
- Full Text
- View/download PDF
48. The dataset of proteins specifically interacted with activated TICAM-1.
- Author
-
Funami K, Matsumoto M, Oshiumi H, Obuse C, and Seya T
- Abstract
The presented data are related with our paper entitled "14-3-3-zeta participates in TLR3-mediated TICAM-1 signal-platform formation" (Funami et al., 2016) [1]. These data show the proteins which specifically bind to the activated (oligomerized) TICAM-1. Fifty-three proteins were identified as specifically interacted with oligomerized TICAM-1. Mutant TICAM-1 cannot form the active oligomer, so the proteins interacted with mutant TICAM-1 are dispensable for TICAM-1-signaling. Among 53 proteins, 14-3-3-zeta specifically interacts with oligomerized TICAM-1 to corroborate TICAM-1 signalosome.
- Published
- 2016
- Full Text
- View/download PDF
49. 14-3-3-zeta participates in TLR3-mediated TICAM-1 signal-platform formation.
- Author
-
Funami K, Matsumoto M, Obuse C, and Seya T
- Subjects
- Gene Knockdown Techniques, HEK293 Cells, Humans, Immunoprecipitation, Mass Spectrometry, Microscopy, Confocal, Real-Time Polymerase Chain Reaction, 14-3-3 Proteins immunology, Adaptor Proteins, Vesicular Transport immunology, Immunity, Innate immunology, Signal Transduction immunology, Toll-Like Receptor 3 immunology
- Abstract
Recognition of pathogen-associated molecular patterns (PAMPs) by pattern-recognition receptors (PRRs) is important in innate immune signaling. Toll-like receptors (TLRs) are well-characterized PRRs and are pivotal in antiviral and antitumor host defense. TIR domain-containing adaptor molecule 1 (TICAM-1, also called TRIF) is an adapter molecule in TLR3- and TLR4-mediated IRF3 activation, late-phase NF-κB activation and MAPK-mediated AP-1 activation. When a TLR3 ligand is added to TLR3-positive cells, TICAM-1 transiently interacts with TLR3 and forms multimers in the cytosol. However, the precise mechanism of TICAM-1 multimer formation remains unknown. In this study, we identified 14-3-3-zeta as a molecule that functions in TLR3-mediated signaling. Knockdown of 14-3-3-zeta reduced production of type I interferon and inflammatory cytokines, nuclear translocation of IRF3 and phosphorylation of IκB via the TLR3-TICAM-1 pathway. Furthermore, TICAM-1 multimerization by ligand stimulation was prohibited by 14-3-3-zeta knockdown. These results suggest that 14-3-3-zeta is involved in the TLR3-TICAM-1 pathway in promoting multimerization of TICAM-1 for the formation of a TICAM-1 signalosome., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Clinical, muscle pathological, and genetic features of Japanese facioscapulohumeral muscular dystrophy 2 (FSHD2) patients with SMCHD1 mutations.
- Author
-
Hamanaka K, Goto K, Arai M, Nagao K, Obuse C, Noguchi S, Hayashi YK, Mitsuhashi S, and Nishino I
- Subjects
- Adolescent, Adult, Biopsy, DNA Methylation, DNA Mutational Analysis, Family, Female, Humans, Japan, Male, Middle Aged, Muscular Dystrophy, Facioscapulohumeral pathology, Mutation, Sequence Homology, Amino Acid, Chromosomal Proteins, Non-Histone genetics, Muscle, Skeletal pathology, Muscular Dystrophy, Facioscapulohumeral genetics, Muscular Dystrophy, Facioscapulohumeral physiopathology
- Abstract
Facioscapulohumeral muscular dystrophy 2 (FSHD2) is a genetic muscular disorder characterized by DNA hypomethylation on the 4q-subtelomeric macrosatellite repeat array, D4Z4. FSHD2 is caused by heterozygous mutations in the gene encoding structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1). Because there has been no study on FSHD2 in Asian populations, it is not known whether this disease mechanism is widely seen. To identify FSHD2 patients with SMCHD1 mutations in the Japanese population, bisulfite pyrosequencing was used to measure DNA methylation on the D4Z4 repeat array, and in patients with DNA hypomethylation, the SMCHD1 gene was sequenced by the Sanger method. Twenty patients with D4Z4 hypomethylation were identified. Of these, 13 patients from 11 unrelated families had ten novel and one reported SMCHD1 mutations: four splice-site, two nonsense, two in-frame deletion, two out-of-frame deletion, and one missense mutations. One of the splice-site mutations was homozygous in the single patient identified with this. In summary, we identified novel SMCHD1 mutations in a Japanese cohort of FSHD2 patients, confirming the presence of this disease in a wider population than previously known., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.