Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
Cell ; 184(19): 4904-4918.e11, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34433012

ABSTRACT

Selfish centromere DNA sequences bias their transmission to the egg in female meiosis. Evolutionary theory suggests that centromere proteins evolve to suppress costs of this "centromere drive." In hybrid mouse models with genetically different maternal and paternal centromeres, selfish centromere DNA exploits a kinetochore pathway to recruit microtubule-destabilizing proteins that act as drive effectors. We show that such functional differences are suppressed by a parallel pathway for effector recruitment by heterochromatin, which is similar between centromeres in this system. Disrupting the kinetochore pathway with a divergent allele of CENP-C reduces functional differences between centromeres, whereas disrupting heterochromatin by CENP-B deletion amplifies the differences. Molecular evolution analyses using Murinae genomes identify adaptive evolution in proteins in both pathways. We propose that centromere proteins have recurrently evolved to minimize the kinetochore pathway, which is exploited by selfish DNA, relative to the heterochromatin pathway that equalizes centromeres, while maintaining essential functions.


Subject(s)
Centromere Protein B/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Alleles , Amino Acid Sequence , Animals , Biological Evolution , CRISPR-Cas Systems/genetics , Centromere Protein A/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomes, Mammalian/metabolism , Female , Heterochromatin/metabolism , Kinetochores/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Oocytes/metabolism , Protein Domains
2.
Cell ; 178(3): 624-639.e19, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348889

ABSTRACT

Recent breakthroughs with synthetic budding yeast chromosomes expedite the creation of synthetic mammalian chromosomes and genomes. Mammals, unlike budding yeast, depend on the histone H3 variant, CENP-A, to epigenetically specify the location of the centromere-the locus essential for chromosome segregation. Prior human artificial chromosomes (HACs) required large arrays of centromeric α-satellite repeats harboring binding sites for the DNA sequence-specific binding protein, CENP-B. We report the development of a type of HAC that functions independently of these constraints. Formed by an initial CENP-A nucleosome seeding strategy, a construct lacking repetitive centromeric DNA formed several self-sufficient HACs that showed no uptake of genomic DNA. In contrast to traditional α-satellite HAC formation, the non-repetitive construct can form functional HACs without CENP-B or initial CENP-A nucleosome seeding, revealing distinct paths to centromere formation for different DNA sequence types. Our developments streamline the construction and characterization of HACs to facilitate mammalian synthetic genome efforts.


Subject(s)
Centromere/metabolism , Chromosomes, Artificial, Human/metabolism , DNA, Satellite/metabolism , Binding Sites , Cell Line, Tumor , Centromere/genetics , Centromere Protein A/genetics , Centromere Protein A/metabolism , Centromere Protein B/deficiency , Centromere Protein B/genetics , Centromere Protein B/metabolism , Epigenesis, Genetic , Humans , Nucleosomes/chemistry , Nucleosomes/metabolism , Plasmids/genetics , Plasmids/metabolism
3.
Nat Rev Mol Cell Biol ; 16(7): 443-9, 2015 07.
Article in English | MEDLINE | ID: mdl-25991376

ABSTRACT

The kinetochore is a complex molecular machine that directs chromosome segregation during mitosis. It is one of the most elaborate subcellular protein structures in eukaryotes, comprising more than 100 different proteins. Inner kinetochore proteins associate with specialized centromeric chromatin containing the histone H3 variant centromere protein A (CENP-A) in place of H3. Outer kinetochore proteins bind to microtubules and signal to delay anaphase onset when microtubules are absent. Since the first kinetochore proteins were discovered and cloned 30 years ago using autoimmune sera from patients with scleroderma-spectrum disease, much has been learnt about the composition, functions and regulation of this remarkable structure.


Subject(s)
Autoantigens/isolation & purification , Centromere Protein B/isolation & purification , Centromere/metabolism , Chromosomal Proteins, Non-Histone/isolation & purification , Animals , Autoantigens/metabolism , Centromere Protein A , Centromere Protein B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Humans , Kinetochores/metabolism
4.
Genes Dev ; 32(1): 20-25, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29386331

ABSTRACT

We combined classical salt fractionation with chromatin immunoprecipitation to recover human centromeric chromatin under native conditions. We found that >85% of the total centromeric chromatin is insoluble under conditions typically used for native chromatin extraction. To map both soluble and insoluble chromatin in situ, we combined CUT&RUN (cleavage under targets and release using nuclease), a targeted nuclease method, with salt fractionation. Using this approach, we observed unexpected structural and conformational variations of centromere protein A (CENP-A)-containing complexes on different α-satellite dimeric units within highly homogenous arrays. Our results suggest that slight α-satellite sequence differences control the structure and occupancy of the associated centromeric chromatin complex.


Subject(s)
Centromere Protein A/chemistry , Centromere/chemistry , Chromatin/chemistry , Centromere Protein A/isolation & purification , Centromere Protein A/metabolism , Centromere Protein B/chemistry , Centromere Protein B/metabolism , Chemical Fractionation , Chromatin/isolation & purification , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA, Satellite/chemistry , Humans , K562 Cells , Solubility
5.
Chromosoma ; 133(3): 169-181, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38856923

ABSTRACT

Centromeres are chromatin structures specialized in sister chromatid cohesion, kinetochore assembly, and microtubule attachment during chromosome segregation. The regional centromere of vertebrates consists of long regions of highly repetitive sequences occupied by the Histone H3 variant CENP-A, and which are flanked by pericentromeres. The three-dimensional organization of centromeric chromatin is paramount for its functionality and its ability to withstand spindle forces. Alongside CENP-A, key contributors to the folding of this structure include components of the Constitutive Centromere-Associated Network (CCAN), the protein CENP-B, and condensin and cohesin complexes. Despite its importance, the intricate architecture of the regional centromere of vertebrates remains largely unknown. Recent advancements in long-read sequencing, super-resolution and cryo-electron microscopy, and chromosome conformation capture techniques have significantly improved our understanding of this structure at various levels, from the linear arrangement of centromeric sequences and their epigenetic landscape to their higher-order compaction. In this review, we discuss the latest insights on centromere organization and place them in the context of recent findings describing a bipartite higher-order organization of the centromere.


Subject(s)
Centromere , Chromatin , Chromosomal Proteins, Non-Histone , Vertebrates , Centromere/metabolism , Centromere/ultrastructure , Animals , Chromatin/metabolism , Chromatin/genetics , Chromatin/ultrastructure , Chromatin/chemistry , Humans , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Vertebrates/genetics , Centromere Protein A/metabolism , Centromere Protein A/genetics , Cohesins , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Centromere Protein B/metabolism , Centromere Protein B/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/ultrastructure , Adenosine Triphosphatases
6.
EMBO J ; 39(20): e105505, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32945564

ABSTRACT

Centromeres are built on repetitive DNA sequences (CenDNA) and a specific chromatin enriched with the histone H3 variant CENP-A, the epigenetic mark that identifies centromere position. Here, we interrogate the importance of CenDNA in centromere specification by developing a system to rapidly remove and reactivate CENP-A (CENP-AOFF/ON ). Using this system, we define the temporal cascade of events necessary to maintain centromere position. We unveil that CENP-B bound to CenDNA provides memory for maintenance on human centromeres by promoting de novo CENP-A deposition. Indeed, lack of CENP-B favors neocentromere formation under selective pressure. Occasionally, CENP-B triggers centromere re-activation initiated by CENP-C, but not CENP-A, recruitment at both ectopic and native centromeres. This is then sufficient to initiate the CENP-A-based epigenetic loop. Finally, we identify a population of CENP-A-negative, CENP-B/C-positive resting CD4+ T cells capable to re-express and reassembles CENP-A upon cell cycle entry, demonstrating the physiological importance of the genetic memory.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Centromere Protein A/metabolism , Centromere Protein B/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Nucleosomes/genetics , CD4-Positive T-Lymphocytes/cytology , CRISPR-Cas Systems , Cell Cycle , Cell Line, Tumor , Centromere/genetics , Chromosome Segregation/genetics , Computational Biology , Epigenesis, Genetic , Gene Targeting , Humans , In Situ Hybridization, Fluorescence , Nucleosomes/metabolism , RNA, Small Interfering
7.
Cell Biol Int ; 48(8): 1212-1222, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38946594

ABSTRACT

JRK is a DNA-binding protein of the pogo superfamily of transposons, which includes the well-known centromere binding protein B (CENP-B). Jrk null mice exhibit epilepsy, and growth and reproductive disorders, consistent with its relatively high expression in the brain and reproductive tissues. Human JRK DNA variants and gene expression levels are implicated in cancers and neuropsychiatric disorders. JRK protein modulates ß-catenin-TCF activity but little is known of its cellular functions. Based on its homology to CENP-B, we determined whether JRK binds centromeric or other satellite DNAs. We show that human JRK binds satellite III DNA, which is abundant at the chromosome 9q12 juxtacentromeric region and on Yq12, both sites of nuclear stress body assembly. Human JRK-GFP overexpressed in HeLa cells strongly localises to 9q12. Using an anti-JRK antiserum we show that endogenous JRK co-localises with a subset of centromeres in non-stressed cells, and with heat shock factor 1 following heat shock. Knockdown of JRK in HeLa cells proportionately reduces heat shock protein gene expression in heat-shocked cells. A role for JRK in regulating the heat shock response is consistent with the mouse Jrk null phenotype and suggests that human JRK may act as a modifier of diseases with a cellular stress component.


Subject(s)
DNA, Satellite , DNA-Binding Proteins , Heat-Shock Response , Animals , Humans , Mice , Centromere/metabolism , Centromere Protein B/metabolism , Centromere Protein B/genetics , DNA, Satellite/genetics , DNA, Satellite/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , HeLa Cells , Protein Binding
8.
J Biol Chem ; 297(4): 101213, 2021 10.
Article in English | MEDLINE | ID: mdl-34547289

ABSTRACT

The centromere is a chromosomal locus that is essential for the accurate segregation of chromosomes during cell division. Transcription of noncoding RNA (ncRNA) at the centromere plays a crucial role in centromere function. The zinc-finger transcriptional regulator ZFAT binds to a specific 8-bp DNA sequence at the centromere, named the ZFAT box, to control ncRNA transcription. However, the precise molecular mechanisms by which ZFAT localizes to the centromere remain elusive. Here we show that the centromeric protein CENP-B is required for the centromeric localization of ZFAT to regulate ncRNA transcription. The ectopic expression of CENP-B induces the accumulation of both endogenous and ectopically expressed ZFAT protein at the centromere in human cells, suggesting that the centromeric localization of ZFAT requires the presence of CENP-B. Coimmunoprecipitation analysis reveals that ZFAT interacts with the acidic domain of CENP-B, and depletion of endogenous CENP-B reduces the centromeric levels of ZFAT protein, further supporting that CENP-B is required for the centromeric localization of ZFAT. In addition, knockdown of CENP-B significantly decreased the expression levels of ncRNA at the centromere where ZFAT regulates the transcription, suggesting that CENP-B is involved in the ZFAT-regulated centromeric ncRNA transcription. Thus, we concluded that CENP-B contributes to the establishment of the centromeric localization of ZFAT to regulate ncRNA transcription.


Subject(s)
Centromere Protein B/metabolism , Centromere/metabolism , RNA, Untranslated/biosynthesis , Transcription Factors/metabolism , Transcription, Genetic , Animals , Centromere/genetics , Centromere Protein B/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , NIH 3T3 Cells , RNA, Untranslated/genetics , Transcription Factors/genetics
9.
Mol Biol Evol ; 38(12): 5576-5587, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34464971

ABSTRACT

Human centromeres are mainly composed of alpha satellite DNA hierarchically organized as higher-order repeats (HORs). Alpha satellite dynamics is shown by sequence homogenization in centromeric arrays and by its transfer to other centromeric locations, for example, during the maturation of new centromeres. We identified during prenatal aneuploidy diagnosis by fluorescent in situ hybridization a de novo insertion of alpha satellite DNA from the centromere of chromosome 18 (D18Z1) into cytoband 15q26. Although bound by CENP-B, this locus did not acquire centromeric functionality as demonstrated by the lack of constriction and the absence of CENP-A binding. The insertion was associated with a 2.8-kbp deletion and likely occurred in the paternal germline. The site was enriched in long terminal repeats and located ∼10 Mbp from the location where a centromere was ancestrally seeded and became inactive in the common ancestor of humans and apes 20-25 million years ago. Long-read mapping to the T2T-CHM13 human genome assembly revealed that the insertion derives from a specific region of chromosome 18 centromeric 12-mer HOR array in which the monomer size follows a regular pattern. The rearrangement did not directly disrupt any gene or predicted regulatory element and did not alter the methylation status of the surrounding region, consistent with the absence of phenotypic consequences in the carrier. This case demonstrates a likely rare but new class of structural variation that we name "alpha satellite insertion." It also expands our knowledge on alphoid DNA dynamics and conveys the possibility that alphoid arrays can relocate near vestigial centromeric sites.


Subject(s)
Centromere , Chromosomal Proteins, Non-Histone , Centromere/genetics , Centromere/metabolism , Centromere Protein B/genetics , Centromere Protein B/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA, Satellite/genetics , Humans , In Situ Hybridization, Fluorescence
10.
Exp Cell Res ; 389(2): 111900, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32044309

ABSTRACT

The centromere is a specialized chromosomal locus required for accurate chromosome segregation. Heterochromatin also assembles around centromere chromatin and forms a base that supports sister chromatid cohesion until anaphase begins. Both centromere chromatin and heterochromatin assemble on a centromeric DNA sequence, a highly repetitive sequence called alphoid DNA (α-satellite DNA) in humans. Alphoid DNA can form a de novo centromere and subsequent human artificial chromosome (HAC) when introduced into the human culture cells HT1080. HAC is maintained stably as a single chromosome independent of other human chromosomes. For de novo centromere assembly and HAC formation, the centromere protein CENP-B and its binding sites, CENP-B boxes, are required in the repeating units of alphoid DNA. CENP-B has multiple roles in de novo centromere chromatin assembly and stabilization and in heterochromatin formation upon alphoid DNA introduction into the cells. Here we review recent progress in human artificial chromosome construction and centromere/heterochromatin assembly and maintenance, focusing on the involvement of human centromere DNA and CENP-B protein.


Subject(s)
Centromere Protein B/metabolism , Centromere/genetics , Chromatin Assembly and Disassembly , Chromosome Segregation , Chromosomes, Artificial, Human , DNA, Satellite/genetics , Centromere Protein B/genetics , Epigenesis, Genetic , Humans
11.
Exp Cell Res ; 390(2): 111959, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32173469

ABSTRACT

The centromere is the nucleoproteic chromosomal structure necessary for accurate chromosome segregation during cell division. One of the earliest centromeric proteins to be discovered was CENP-B, the only one capable of recognizing a specific centromeric DNA binding motif. The phylogenetic history of this protein and of its DNA binding site shows independent events of function acquisition across different species and raises questions on the evolutionary dynamics of CENP-B, including what may be the selective advantage provided by its role at the centromere. Recent results have provided insight into potential functions of CENP-B in chromosome dynamics, however, its function is still object of debate. The recurrent appearance of CENP-B centromeric activity along phylogenesis, together with its dispensability, represent strictly intertwined facets of this controversy. This chapter focuses on the evolution, function and homeostasis of CENP-B and its importance in centromere biology.


Subject(s)
Centromere Protein B/genetics , Centromere/metabolism , DNA/genetics , Eukaryota/genetics , Evolution, Molecular , Animals , Binding Sites , Cell Division , Centromere/ultrastructure , Centromere Protein B/metabolism , Chromosome Segregation , DNA/metabolism , Eukaryota/classification , Eukaryota/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Gene Expression , Humans , Nucleotide Motifs , Phylogeny , Protein Binding
12.
Arch Toxicol ; 95(2): 703-713, 2021 02.
Article in English | MEDLINE | ID: mdl-33057863

ABSTRACT

1-Methylpyrene (1-MP) is a common environmental pollutant and animal carcinogen. After sequential activation by cytochromes P450 and sulfotransferases, it induced gene mutations and micronuclei in mammalian cells. The type of micronuclei formed, entire chromosomes or fragments, was not analysed. In this study, 1-MP and its primary metabolite, 1-hydroxymethylpyrene (1-HMP), were investigated for the induction of centromere-positive and -negative micronuclei in the human hepatoma cell line HepG2 and its derivative C3A, expressing relevant enzymes at higher levels. Under a short-exposure (9 h)/long-recovery regime (2 cell cycles in total), 1-MP and 1-HMP provided negative test results in HepG2 cells. However, they induced micronuclei in C3A cells, the effect being blocked by 1-aminobenzotriazole (inhibitor of cytochromes P450s) and reduced by pentachlorophenol (inhibitor of sulfotransferases). Immunofluorescence staining of centromere protein B in the micronuclei revealed purely clastogenic effects under this regime. Unexpectedly, 1-MP and 1-HMP at concentrations 1/5-1/4 of that required for micronuclei formation led to mitotic arrest and spindle aberrations, as detected by immunofluorescence staining of ß- and γ-tubulin. Following extended exposure (72 h, 2 cell cycles, no recovery), damage to the spindle apparatus and centrosomes was detected at even lower concentrations, with concurrent formation of micronuclei. At low concentrations (1-8 µM 1-MP, 0.25-0.5 µM 1-HMP), the micronuclei induced were unexceptionally centromere-positive. Thus, the chromosome-damaging mechanism of 1-MP was regime and concentration dependent: potently aneugenic under persistent exposure, while clastogenic at higher concentrations following a short-exposure/long-recovery regime. This is a convincing evidence for the existence of metabolic activation-dependent aneugens.


Subject(s)
Micronuclei, Chromosome-Defective/drug effects , Mitosis/drug effects , Pyrenes/toxicity , Activation, Metabolic/drug effects , Aneugens/metabolism , Aneugens/toxicity , Cell Line, Tumor , Centromere Protein B/metabolism , Centrosome/drug effects , Hep G2 Cells , Humans , Micronucleus Tests , Microscopy, Fluorescence , Mutagens , Pyrenes/metabolism , Spindle Apparatus/drug effects
13.
Genome Res ; 27(12): 2040-2049, 2017 12.
Article in English | MEDLINE | ID: mdl-29141960

ABSTRACT

The centromere is the structural unit responsible for the faithful segregation of chromosomes. Although regulation of centromeric function by epigenetic factors has been well-studied, the contributions of the underlying DNA sequences have been much less well defined, and existing methodologies for studying centromere genomics in biology are laborious. We have identified specific markers in the centromere of 23 of the 24 human chromosomes that allow for rapid PCR assays capable of capturing the genomic landscape of human centromeres at a given time. Use of this genetic strategy can also delineate which specific centromere arrays in each chromosome drive the recruitment of epigenetic modulators. We further show that, surprisingly, loss and rearrangement of DNA in centromere 21 is associated with trisomy 21. This new approach can thus be used to rapidly take a snapshot of the genetics and epigenetics of each specific human centromere in nondisjunction disorders and other biological settings.


Subject(s)
Centromere , Genomics/methods , Real-Time Polymerase Chain Reaction/methods , Base Sequence , Centromere Protein B/metabolism , Chromosomal Instability , Chromosomes, Human, Pair 21 , DNA , DNA, Satellite , Down Syndrome/genetics , Epigenesis, Genetic , Female , Gene Rearrangement , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Karyotype , Male , Sequence Deletion
14.
J Proteome Res ; 16(9): 3433-3442, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28704058

ABSTRACT

The centromere is the chromosomal locus where the kinetochore forms and is critical for ensuring proper segregation of sister chromatids during cell division. A substantial amount of effort has been devoted to understanding the characteristic features and roles of the centromere, yet some fundamental aspects of the centromere, such as the complete list of elements that define it, remain obscure. It is well-known that human centromeres include a highly repetitive class of DNA known as alpha satellite, or alphoid, DNA. We present here the first DNA-centric examination of human protein-alpha satellite interactions, employing an approach known as HyCCAPP (hybridization capture of chromatin-associated proteins for proteomics) to identify the protein components of alphoid chromatin in a human cell line. Using HyCCAPP, cross-linked alpha satellite chromatin was isolated from cell lysate, and captured proteins were analyzed via mass spectrometry. After being compared to proteins identified in control pulldown experiments, 90 proteins were identified as enriched at alphoid DNA. This list included many known centromere-binding proteins in addition to multiple novel alpha satellite-binding proteins, such as LRIF1, a heterochromatin-associated protein. The ability of HyCCAPP to reveal both known as well as novel alphoid DNA-interacting proteins highlights the validity and utility of this approach.


Subject(s)
Centromere/metabolism , Chromatin/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , In Situ Hybridization, Fluorescence/methods , Antibodies, Monoclonal/chemistry , Centromere/ultrastructure , Centromere Protein B/genetics , Centromere Protein B/metabolism , Chromatin/ultrastructure , Chromatin Immunoprecipitation , DNA/genetics , DNA-Binding Proteins/genetics , Gene Expression , Humans , K562 Cells , Mass Spectrometry/methods
15.
Biochim Biophys Acta ; 1859(10): 1314-21, 2016 10.
Article in English | MEDLINE | ID: mdl-27345571

ABSTRACT

It is well established that eukaryotic genomes are pervasively transcribed producing cryptic unstable transcripts (CUTs). However, the mechanisms regulating pervasive transcription are not well understood. Here, we report that the fission yeast CENP-B homolog Abp1 plays an important role in preventing pervasive transcription. We show that loss of abp1 results in the accumulation of CUTs, which are targeted for degradation by the exosome pathway. These CUTs originate from different types of genomic features, but the highest increase corresponds to Tf2 retrotransposons and rDNA repeats, where they map along the entire elements. In the absence of abp1, increased RNAPII-Ser5P occupancy is observed throughout the Tf2 coding region and, unexpectedly, RNAPII-Ser5P is enriched at rDNA repeats. Loss of abp1 also results in Tf2 derepression and increased nucleolus size. Altogether these results suggest that Abp1 prevents pervasive RNAPII transcription of repetitive DNA elements (i.e., Tf2 and rDNA repeats) from internal cryptic sites.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , RNA Polymerase II/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Transcription, Genetic , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Centromere/metabolism , Centromere/ultrastructure , Centromere Protein B/genetics , Centromere Protein B/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , DNA-Binding Proteins/deficiency , Heterochromatin/metabolism , Heterochromatin/ultrastructure , RNA Polymerase II/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroelements , Schizosaccharomyces/metabolism , Schizosaccharomyces/ultrastructure , Schizosaccharomyces pombe Proteins/metabolism
16.
Chromosoma ; 125(4): 645-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27376724

ABSTRACT

The kinetochore is an essential structure for the chromosome segregation machinery in eukaryotes; it serves as a bridge between the spindle microtubules and chromosomes. The kinetochore consists of multiple interconnecting components on the centromere; therefore, understanding its formation, molecular function, and regulation has remained an ongoing challenge. Recent studies have provided new insights into centromere identity, kinetochore assembly, and function. In this review, we discuss recent advances in our understanding of the function and regulation of key kinetochore components. We highlight the reciprocal localization dependencies of the different sub-complexes of the kinetochore and describe their regulation during the cell cycle.


Subject(s)
Cell Cycle/genetics , Cell Division/physiology , Centromere/metabolism , Chromosome Segregation/physiology , Chromosomes, Human/metabolism , Kinetochores/metabolism , Saccharomyces cerevisiae/genetics , Spindle Apparatus/metabolism , Autoantigens/metabolism , Centromere Protein A , Centromere Protein B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Humans , Microtubules/metabolism
17.
Nature ; 469(7328): 112-5, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21151105

ABSTRACT

Centromere-binding protein B (CENP-B) is a widely conserved DNA binding factor associated with heterochromatin and centromeric satellite repeats. In fission yeast, CENP-B homologues have been shown to silence long terminal repeat (LTR) retrotransposons by recruiting histone deacetylases. However, CENP-B factors also have unexplained roles in DNA replication. Here we show that a molecular function of CENP-B is to promote replication-fork progression through the LTR. Mutants have increased genomic instability caused by replication-fork blockage that depends on the DNA binding factor switch-activating protein 1 (Sap1), which is directly recruited by the LTR. The loss of Sap1-dependent barrier activity allows the unhindered progression of the replication fork, but results in rearrangements deleterious to the retrotransposon. We conclude that retrotransposons influence replication polarity through recruitment of Sap1 and transposition near replication-fork blocks, whereas CENP-B counteracts this activity and promotes fork stability. Our results may account for the role of LTR in fragile sites, and for the association of CENP-B with pericentromeric heterochromatin and tandem satellite repeats.


Subject(s)
Centromere Protein B/metabolism , DNA Replication/genetics , Genome, Fungal/genetics , Genomic Instability/genetics , Retroelements/genetics , Schizosaccharomyces/genetics , Terminal Repeat Sequences/genetics , Centromere Protein B/deficiency , Centromere Protein B/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Conserved Sequence/genetics , DNA Damage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Recombination, Genetic , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
18.
Nucleic Acids Res ; 43(10): 4909-22, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25916850

ABSTRACT

CENP-A and CENP-B are major components of centromeric chromatin. CENP-A is the histone H3 variant, which forms the centromere-specific nucleosome. CENP-B specifically binds to the CENP-B box DNA sequence on the centromere-specific repetitive DNA. In the present study, we found that the CENP-A nucleosome more stably retains human CENP-B than the H3.1 nucleosome in vitro. Specifically, CENP-B forms a stable complex with the CENP-A nucleosome, when the CENP-B box sequence is located at the proximal edge of the nucleosome. Surprisingly, the CENP-B binding was weaker when the CENP-B box sequence was located in the distal linker region of the nucleosome. This difference in CENP-B binding, depending on the CENP-B box location, was not observed with the H3.1 nucleosome. Consistently, we found that the DNA-binding domain of CENP-B specifically interacted with the CENP-A-H4 complex, but not with the H3.1-H4 complex, in vitro. These results suggested that CENP-B forms a more stable complex with the CENP-A nucleosome through specific interactions with CENP-A, if the CENP-B box is located proximal to the CENP-A nucleosome. Our in vivo assay also revealed that CENP-B binding in the vicinity of the CENP-A nucleosome substantially stabilizes the CENP-A nucleosome on alphoid DNA in human cells.


Subject(s)
Autoantigens/metabolism , Centromere Protein B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nucleosomes/metabolism , Autoantigens/chemistry , Cell Line, Tumor , Centromere/chemistry , Centromere Protein A , Chromosomal Proteins, Non-Histone/chemistry , DNA/chemistry , DNA/metabolism , Histones/metabolism , Humans , Protein Interaction Domains and Motifs
19.
Biol Lett ; 12(3): 20150817, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27029836

ABSTRACT

Centromere protein B (CENP-B) is one of the major proteins involved in centromere formation, binding to centromeric repetitive DNA by recognizing a 17 bp motif called the CENP-B box. Hominids (humans and great apes) carry large numbers of CENP-B boxes in alpha satellite DNA (AS, the major centromeric repetitive DNA of simian primates). Only negative results have been reported regarding the presence of the CENP-B box in other primate taxa. Consequently, it is widely believed that the CENP-B box is confined, within primates, to the hominids. We report here that the common marmoset, a New World monkey, contains an abundance of CENP-B boxes in its AS. First, in a long contig sequence we constructed and analysed, we identified the motif in 17 of the 38 alpha satellite repeat units. We then sequenced terminal regions of additional clones and found the motif in many of them. Immunostaining of marmoset cells demonstrated that CENP-B binds to DNA in the centromeric regions of chromosomes. Therefore, functional CENP-B boxes are not confined to hominids. Our results indicate that the efficiency of identification of the CENP-B box may depend largely on the sequencing methods used, and that the CENP-B box in centromeric repetitive DNA may be more common than researchers previously thought.


Subject(s)
Callithrix/genetics , Centromere Protein B/genetics , Centromere/metabolism , Nucleotide Motifs , Animals , Base Sequence , Callithrix/metabolism , Centromere Protein B/metabolism
20.
EMBO J ; 30(8): 1444-58, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21399610

ABSTRACT

Wnt/Wingless signal transduction directs fundamental developmental processes, and upon hyperactivation triggers colorectal adenoma/carcinoma formation. Responses to Wnt stimulation are cell specific and diverse; yet, how cell context modulates Wnt signalling outcome remains obscure. In a Drosophila genetic screen for components that promote Wingless signalling, we identified Earthbound 1 (Ebd1), a novel member in a protein family containing Centromere Binding Protein B (CENPB)-type DNA binding domains. Ebd1 is expressed in only a subset of Wingless responsive cell types, and is required for only a limited number of Wingless-dependent processes. In addition, Ebd1 shares sequence similarity and can be functionally replaced with the human CENPB domain protein Jerky, previously implicated in juvenile myoclonic epilepsy development. Both Jerky and Ebd1 interact directly with the Wnt/Wingless pathway transcriptional co-activators ß-catenin/Armadillo and T-cell factor (TCF). In colon carcinoma cells, Jerky facilitates Wnt signalling by promoting association of ß-catenin with TCF and recruitment of ß-catenin to chromatin. These findings indicate that tissue-restricted transcriptional co-activators facilitate cell-specific Wnt/Wingless signalling responses by modulating ß-catenin-TCF activity.


Subject(s)
Centromere Protein B/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Nuclear Proteins/metabolism , TCF Transcription Factors/metabolism , Trans-Activators/metabolism , Wnt Proteins/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome , Blotting, Western , Cells, Cultured , Centromere Protein B/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA-Binding Proteins , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Luciferases/metabolism , Nuclear Proteins/genetics , Protein Binding , RNA, Messenger/genetics , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TCF Transcription Factors/genetics , Trans-Activators/genetics , Wnt Proteins/genetics , Wnt1 Protein/genetics , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL