Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 137(3): 409-12, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19410539

RESUMO

CENP-A is a histone H3-like protein specific to centromeres that is essential for kinetochore formation and accurate chromosome segregation in eukaryotes. Recent studies (Dunleavy et al., 2009; Foltz et al., 2009; Perpelescu et al., 2009; Pidoux et al., 2009; Williams et al., 2009) analyze CENP-A binding proteins required for the recruitment of CENP-A to centromeres in humans and in fission yeast, bringing us closer to understanding how centromere identity is faithfully propagated.


Assuntos
Autoantígenos/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , Schizosaccharomyces/metabolismo , Autoantígenos/genética , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Transativadores/genética , Transativadores/metabolismo
2.
PLoS Biol ; 17(5): e3000241, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31086362

RESUMO

Centromeres are essential chromosomal regions that mediate kinetochore assembly and spindle attachments during cell division. Despite their functional conservation, centromeres are among the most rapidly evolving genomic regions and can shape karyotype evolution and speciation across taxa. Although significant progress has been made in identifying centromere-associated proteins, the highly repetitive centromeres of metazoans have been refractory to DNA sequencing and assembly, leaving large gaps in our understanding of their functional organization and evolution. Here, we identify the sequence composition and organization of the centromeres of Drosophila melanogaster by combining long-read sequencing, chromatin immunoprecipitation for the centromeric histone CENP-A, and high-resolution chromatin fiber imaging. Contrary to previous models that heralded satellite repeats as the major functional components, we demonstrate that functional centromeres form on islands of complex DNA sequences enriched in retroelements that are flanked by large arrays of satellite repeats. Each centromere displays distinct size and arrangement of its DNA elements but is similar in composition overall. We discover that a specific retroelement, G2/Jockey-3, is the most highly enriched sequence in CENP-A chromatin and is the only element shared among all centromeres. G2/Jockey-3 is also associated with CENP-A in the sister species D. simulans, revealing an unexpected conservation despite the reported turnover of centromeric satellite DNA. Our work reveals the DNA sequence identity of the active centromeres of a premier model organism and implicates retroelements as conserved features of centromeric DNA.


Assuntos
Centrômero/genética , Drosophila/genética , Retroelementos/genética , Animais , Proteína Centromérica A/genética , Cromatina/metabolismo , Elementos de DNA Transponíveis/genética , DNA Satélite/genética , Drosophila/embriologia , Proteínas de Drosophila/genética , Embrião não Mamífero/metabolismo , Genoma de Inseto , Sequências Repetidas Terminais/genética
3.
J Biol Chem ; 294(38): 14119-14134, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31366733

RESUMO

The successful assembly and regulation of the kinetochore are critical for the equal and accurate segregation of genetic material during the cell cycle. CENP-C (centromere protein C), a conserved inner kinetochore component, has been broadly characterized as a scaffolding protein and is required for the recruitment of multiple kinetochore proteins to the centromere. At its C terminus, CENP-C harbors a conserved cupin domain that has an established role in protein dimerization. Although the crystal structure of the Saccharomyces cerevisiae Mif2CENP-C cupin domain has been determined, centromeric organization and kinetochore composition vary greatly between S. cerevisiae (point centromere) and other eukaryotes (regional centromere). Therefore, whether the structural and functional role of the cupin domain is conserved throughout evolution requires investigation. Here, we report the crystal structures of the Schizosaccharomyces pombe and Drosophila melanogaster CENP-C cupin domains at 2.52 and 1.81 Å resolutions, respectively. Although the central jelly roll architecture is conserved among the three determined CENP-C cupin domain structures, the cupin domains from organisms with regional centromeres contain additional structural features that aid in dimerization. Moreover, we found that the S. pombe Cnp3CENP-C jelly roll fold harbors an inner binding pocket that is used to recruit the meiosis-specific protein Moa1. In summary, our results unveil the evolutionarily conserved and unique features of the CENP-C cupin domain and uncover the mechanism by which it functions as a recruitment factor.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/ultraestrutura , Animais , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Proteína Centromérica A/metabolismo , Cristalografia por Raios X/métodos , Proteínas de Ligação a DNA/metabolismo , Dimerização , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/ultraestrutura , Drosophila melanogaster/metabolismo , Histonas/metabolismo , Cinetocoros/metabolismo , Cinetocoros/ultraestrutura , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
4.
Trends Genet ; 33(2): 101-117, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28069312

RESUMO

Centromeres are essential chromosomal structures that mediate the accurate distribution of genetic material during meiotic and mitotic cell divisions. In most organisms, centromeres are epigenetically specified and propagated by nucleosomes containing the centromere-specific H3 variant, centromere protein A (CENP-A). Although centromeres perform a critical and conserved function, CENP-A and the underlying centromeric DNA are rapidly evolving. This paradox has been explained by the centromere drive hypothesis, which proposes that CENP-A is undergoing an evolutionary tug-of-war with selfish centromeric DNA. Here, we review our current understanding of CENP-A evolution in relation to centromere drive and discuss classical and recent advances, including new evidence implicating CENP-A chaperones in this conflict.


Assuntos
Proteína Centromérica A/genética , Centrômero/genética , DNA/genética , Nucleossomos/genética , Cromatina , Proteínas de Ligação a DNA/genética , Histonas/genética , Humanos , Chaperonas Moleculares/genética
5.
PLoS Genet ; 7(5): e1002068, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21589899

RESUMO

Semi-conservative segregation of nucleosomes to sister chromatids during DNA replication creates gaps that must be filled by new nucleosome assembly. We analyzed the cell-cycle timing of centromeric chromatin assembly in Drosophila, which contains the H3 variant CID (CENP-A in humans), as well as CENP-C and CAL1, which are required for CID localization. Pulse-chase experiments show that CID and CENP-C levels decrease by 50% at each cell division, as predicted for semi-conservative segregation and inheritance, whereas CAL1 displays higher turnover. Quench-chase-pulse experiments demonstrate that there is a significant lag between replication and replenishment of centromeric chromatin. Surprisingly, new CID is recruited to centromeres in metaphase, by a mechanism that does not require an intact mitotic spindle, but does require proteasome activity. Interestingly, new CAL1 is recruited to centromeres before CID in prophase. Furthermore, CAL1, but not CENP-C, is found in complex with pre-nucleosomal CID. Finally, CENP-C displays yet a different pattern of incorporation, during both interphase and mitosis. The unusual timing of CID recruitment and unique dynamics of CAL1 identify a distinct centromere assembly pathway in Drosophila and suggest that CAL1 is a key regulator of centromere propagation.


Assuntos
Centrômero , Cromatina/metabolismo , Mitose , Animais , Ciclina A/metabolismo , Drosophila , Humanos , Metáfase , Microtúbulos/metabolismo , Prófase , Complexo de Endopeptidases do Proteassoma/metabolismo
6.
Chromosome Res ; 20(5): 493-504, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22820845

RESUMO

Centromeres are essential cis-elements on chromosomes that are crucial for the stable transmission of genetic information during mitotic and meiotic cell divisions. Different species employ a variety of centromere configurations, from small genetically defined centromeres in budding yeast to holocentric centromeres that occupy entire chromosomes in Caenorhabditis, yet the incorporation of nucleosomes containing the essential centromere-specific histone H3 variant CENP-A is a common feature of centromeres in all eukaryotes. In vertebrates and fungi, CENP-A is specifically deposited at centromeres by a conserved chaperone, called HJURP or Scm3, respectively. Surprisingly, homologs of these proteins have not been identified in Drosophila, Caenorhabditis, or plants. How CENP-A is targeted to centromeres in these organisms is not known. The Drosophila centromeric protein CAL1, found only in the Diptera genus, is essential for CENP-A localization, is recruited to centromeres at a similar time as CENP-A, and interacts with CENP-A in both chromatin and pre-nucleosomal complexes, making it a strong candidate for a CENP-A chaperone in this lineage. Here, we discuss the conservation and evolution of this essential centromere factor and report the identification of a "Scm3-domain"-like region with similarity to the corresponding region of fungal Scm3 as well as a shared predicted alpha-helical structure. Given the lack of common ancestry between Scm3 and CAL1, we propose that an optimal CENP-A binding region was independently acquired by CAL1, which caused the loss of an ancestral Scm3 protein from the Diptera lineage.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Evolução Molecular , Histonas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteína Centromérica A , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Códon , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Estrutura Secundária de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Seleção Genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
7.
Proc Natl Acad Sci U S A ; 106(43): 18213-8, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19822757

RESUMO

Ubiquitination by the anaphase-promoting complex (APC/C) is essential for proliferation in all eukaryotes. The human APC/C promotes the degradation of mitotic regulators by assembling K11-linked ubiquitin chains, the formation of which is initiated by its E2 UbcH10. Here, we identify the conserved Ube2S as a K11-specific chain elongating E2 for human and Drosophila APC/C. Ube2S depends on the cell cycle-dependent association with the APC/C activators Cdc20 and Cdh1 for its activity. While depletion of Ube2S already inhibits APC/C in cells, the loss of the complete UbcH10/Ube2S-module leads to dramatic stabilization of APC/C substrates, severe spindle defects, and a strong mitotic delay. Ube2S and UbcH10 are tightly co-regulated in the cell cycle by APC/C-dependent degradation. We conclude that UbcH10 and Ube2S constitute a physiological E2-module for APC/C, the activity of which is required for spindle assembly and cell division.


Assuntos
Drosophila melanogaster/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Biocatálise , Células HeLa , Humanos , Mitose , Ligação Proteica , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética
8.
Curr Biol ; 32(18): R947-R949, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-36167041

RESUMO

Crosses between closely related species of frogs result in inviable embryos that die following catastrophic cell divisions. A new study identifies defects in centromere maintenance and DNA replication stress as key forces driving this incompatibility.


Assuntos
Centrômero , Replicação do DNA , Animais , Centrômero/genética , Xenopus laevis/genética
9.
Genome Biol Evol ; 14(5)2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35441684

RESUMO

Centromeres are essential chromosomal regions that mediate the accurate inheritance of genetic information during eukaryotic cell division. Despite their conserved function, centromeres do not contain conserved DNA sequences and are instead epigenetically marked by the presence of the centromere-specific histone H3 variant centromeric protein A. The functional contribution of centromeric DNA sequences to centromere identity remains elusive. Previous work found that dyad symmetries with a propensity to adopt noncanonical secondary DNA structures are enriched at the centromeres of several species. These findings lead to the proposal that noncanonical DNA structures may contribute to centromere specification. Here, we analyze the predicted secondary structures of the recently identified centromere DNA sequences of Drosophila melanogaster. Although dyad symmetries are only enriched on the Y centromere, we find that other types of noncanonical DNA structures, including melted DNA and G-quadruplexes, are common features of all D. melanogaster centromeres. Our work is consistent with previous models suggesting that noncanonical DNA secondary structures may be conserved features of centromeres with possible implications for centromere specification.


Assuntos
Centrômero , Drosophila melanogaster , Animais , Centrômero/genética , Proteína Centromérica A/genética , Cromatina , DNA , Drosophila melanogaster/genética , Histonas/genética
10.
Nat Commun ; 13(1): 3981, 2022 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-35810185

RESUMO

Pairing of homologous chromosomes in somatic cells provides the opportunity of interchromosomal interaction between homologous gene regions. In the Drosophila male germline, the Stat92E gene is highly expressed in a germline stem cell (GSC) and gradually downregulated during the differentiation. Here we show that the pairing of Stat92E is always tight in GSCs and immediately loosened in differentiating daughter cells, gonialblasts (GBs). Disturbance of Stat92E pairing by relocation of one locus to another chromosome or by knockdown of global pairing/anti-pairing factors both result in a failure of Stat92E downregulation, suggesting that the pairing is required for the decline in transcription. Furthermore, the Stat92E enhancer, but not its transcription, is required for the change in pairing state, indicating that pairing is not a consequence of transcriptional changes. Finally, we show that the change in Stat92E pairing is dependent on asymmetric histone inheritance during the asymmetric division of GSCs. Taken together, we propose that the changes in Stat92E pairing status is an intrinsically programmed mechanism for enabling prompt cell fate switch during the differentiation of stem cells.


Assuntos
Proteínas de Drosophila , Alelos , Animais , Diferenciação Celular/genética , Drosophila/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Células Germinativas
11.
Curr Biol ; 31(22): R1491-R1504, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34813757

RESUMO

The centromere performs a universally conserved function, to accurately partition genetic information upon cell division. Yet, centromeres are among the most rapidly evolving regions of the genome and are bound by a varying assortment of centromere-binding factors that are themselves highly divergent at the protein-sequence level. A common thread in most species is the dependence on the centromere-specific histone variant CENP-A for the specification of the centromere site. However, CENP-A is not universally required in all species or cell types, making the identification of a general mechanism for centromere specification challenging. In this review, we examine our current understanding of the mechanisms of centromere specification in CENP-A-dependent and independent systems, focusing primarily on recent work.


Assuntos
Centrômero , Proteínas Cromossômicas não Histona , Autoantígenos/metabolismo , Centrômero/metabolismo , Proteína Centromérica A/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo
13.
PLoS Genet ; 3(7): e121, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17677001

RESUMO

The histone H3 variant CENP-A assembles into chromatin exclusively at centromeres. The process of CENP-A chromatin assembly is epigenetically regulated. Fission yeast centromeres are composed of a central kinetochore domain on which CENP-A chromatin is assembled, and this is flanked by heterochromatin. Marker genes are silenced when placed within kinetochore or heterochromatin domains. It is not known if fission yeast CENP-A(Cnp1) chromatin is confined to specific sequences or whether histone H3 is actively excluded. Here, we show that fission yeast CENP-A(Cnp1) can assemble on noncentromeric DNA when it is inserted within the central kinetochore domain, suggesting that in fission yeast CENP-A(Cnp1) chromatin assembly is driven by the context of a sequence rather than the underlying DNA sequence itself. Silencing in the central domain is correlated with the amount of CENP-A(Cnp1) associated with the marker gene and is also affected by the relative level of histone H3. Our analyses indicate that kinetochore integrity is dependent on maintaining the normal ratio of H3 and H4. Excess H3 competes with CENP-A(Cnp1) for assembly into central domain chromatin, resulting in less CENP-A(Cnp1) and other kinetochore proteins at centromeres causing defective kinetochore function, which is manifest as aberrant mitotic chromosome segregation. Alterations in the levels of H3 relative to H4 and CENP-A(Cnp1) influence the extent of DNA at centromeres that is packaged in CENP-A(Cnp1) chromatin and the composition of this chromatin. Thus, CENP-A(Cnp1) chromatin assembly in fission yeast exhibits plasticity with respect to the underlying sequences and is sensitive to the levels of CENP-A(Cnp1) and other core histones.


Assuntos
Autoantígenos/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Schizosaccharomyces/metabolismo , Centrômero/metabolismo , Proteína Centromérica A , Cromatina/metabolismo , DNA/química , Primers do DNA/química , Proteínas Fúngicas/química , Genoma Fúngico , Cinetocoros/metabolismo , Modelos Biológicos , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Fuso Acromático/metabolismo
14.
Dev Cell ; 52(3): 379-394.e7, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32049040

RESUMO

Centromeres are essential for accurate chromosome segregation and are marked by centromere protein A (CENP-A) nucleosomes. Mis-targeted CENP-A chromatin has been shown to seed centromeres at non-centromeric DNA. However, the requirements for such de novo centromere formation and transmission in vivo remain unknown. Here, we employ Drosophila melanogaster and the LacI/lacO system to investigate the ability of targeted de novo centromeres to assemble and be inherited through development. De novo centromeres form efficiently at six distinct genomic locations, which include actively transcribed chromatin and heterochromatin, and cause widespread chromosomal instability. During tethering, de novo centromeres sometimes prevail, causing the loss of the endogenous centromere via DNA breaks and HP1-dependent epigenetic inactivation. Transient induction of de novo centromeres and chromosome healing in early embryogenesis show that, once established, these centromeres can be maintained through development. Our results underpin the ability of CENP-A chromatin to establish and sustain mitotic centromere function in Drosophila.


Assuntos
Plasticidade Celular , Proteína Centromérica A/metabolismo , Centrômero , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Epigênese Genética , Histonas/metabolismo , Animais , Proteína Centromérica A/genética , Cromatina/genética , Segregação de Cromossomos , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Feminino , Histonas/genética , Masculino
15.
Curr Opin Genet Dev ; 13(2): 191-8, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12672497

RESUMO

The centromere is the locus responsible for the segregation of chromosomes during mitosis and meiosis. The number of newly characterised centromere-associated proteins continues to increase. The kinetochore complex assembles at this site and in many organisms is visible as the primary constriction. In several systems the location of the site of kinetochore assembly is known to vary and the site is not specified by a strict cis-acting primary sequence. It is proposed that tension between bioriented sister centromeres may act to imprint the site.


Assuntos
Autoantígenos , Centrômero/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Proteína Centromérica A , Cinetocoros/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiologia
16.
Curr Biol ; 13(20): 1748-57, 2003 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-14561399

RESUMO

BACKGROUND: Centromeric domains often consist of repetitive elements that are assembled in specialized chromatin, characterized by hypoacetylation of histones H3 and H4 and methylation of lysine 9 of histone H3 (K9-MeH3). Perturbation of this underacetylated state by transient treatment with histone deacetylase inhibitors leads to defective centromere function, correlating with delocalization of the heterochromatin protein Swi6/HP1. Likewise, deletion of the K9-MeH3 methyltransferase Clr4/Suvar39 causes defective chromosome segregation. Here, we create fission yeast strains retaining one histone H3 and H4 gene; the creation of these strains allows mutation of specific N-terminal tail residues and their role in centromeric silencing and chromosome stability to be investigated. RESULTS: Reduction of H3/H4 gene dosage to one-third does not affect cell viability or heterochromatin formation. Mutation of lysines 9 or 14 or serine 10 within the amino terminus of histone H3 impairs centromere function, leading to defective chromosome segregation and Swi6 delocalization. Surprisingly, silent centromeric chromatin does not require the conserved lysine 8 and 16 residues of histone H4. CONCLUSIONS: To date, mutation of conserved N-terminal residues in endogenous histone genes has only been performed in budding yeast, which lacks the Clr4/Suvar39 histone methyltransferase and Swi6/HP1. We demonstrate the importance of conserved residues within the histone H3 N terminus for the maintenance of centromeric heterochromatin in fission yeast. In sharp contrast, mutation of two conserved lysines within the histone H4 tail has no impact on the integrity of centromeric heterochromatin. Our data highlight the striking divergence between the histone tail requirements for the fission yeast and budding yeast silencing pathways.


Assuntos
Acetiltransferases/metabolismo , Centrômero , Segregação de Cromossomos , Inativação Gênica , Histonas/metabolismo , Schizosaccharomyces , Acetilação , Anticorpos Monoclonais , Western Blotting , Divisão Celular/genética , Centrômero/genética , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cruzamentos Genéticos , Heterocromatina/metabolismo , Histonas/genética , Lisina/metabolismo , Mutação , Testes de Precipitina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
17.
Mol Biol Cell ; 27(21): 3189-3191, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27799491

RESUMO

It is an honor to become a part of the talented group of cell biologists who have received this award before me. While running a research group certainly has its ups and downs, I love being a faculty member and am continuously excited by the prospect of scientific discoveries yet to be made. I have benefited from the support of many people over the years and hope to be able to do the same for others through my mentoring and teaching.


Assuntos
Mentores , Distinções e Prêmios , Centrômero , Humanos , Pesquisa
18.
Dev Cell ; 37(5): 389-90, 2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27270035

RESUMO

Heterochromatin is incompatible with centromeric chromatin assembly and propagation. In this issue of Developmental Cell, Ohzeki et al. (2016) reveal that a critical role of the Mis18 complex is to transiently recruit the lysine acetyltransferase KAT7 to centromeres to facilitate the removal of H3K9me3 and the deposition of CENP-A.


Assuntos
Centrômero/metabolismo , Histonas/metabolismo , Lisina Acetiltransferases/metabolismo , Acetilação , Cromatina/metabolismo , Células HeLa , Humanos , Modelos Biológicos
19.
J Cell Biol ; 214(1): 13-24, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27377247

RESUMO

All eukaryotic genomes are packaged into basic units of DNA wrapped around histone proteins called nucleosomes. The ability of histones to specify a variety of epigenetic states at defined chromatin domains is essential for cell survival. The most distinctive type of chromatin is found at centromeres, which are marked by the centromere-specific histone H3 variant CENP-A. Many of the factors that regulate CENP-A chromatin have been identified; however, our understanding of the mechanisms of centromeric nucleosome assembly, maintenance, and reorganization remains limited. This review discusses recent insights into these processes and draws parallels between centromeric and noncentromeric chromatin assembly mechanisms.


Assuntos
Centrômero/metabolismo , Montagem e Desmontagem da Cromatina , Cromossomos/metabolismo , Animais , Autoantígenos , Proteína Centromérica A , Proteínas Cromossômicas não Histona , Replicação do DNA , Humanos , Transcrição Gênica
20.
Dev Cell ; 37(2): 136-47, 2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-27093083

RESUMO

Centromeres mediate the conserved process of chromosome segregation, yet centromeric DNA and the centromeric histone, CENP-A, are rapidly evolving. The rapid evolution of Drosophila CENP-A loop 1 (L1) is thought to modulate the DNA-binding preferences of CENP-A to counteract centromere drive, the preferential transmission of chromosomes with expanded centromeric satellites. Consistent with this model, CENP-A from Drosophila bipectinata (bip) cannot localize to Drosophila melanogaster (mel) centromeres. We show that this result is due to the inability of the mel CENP-A chaperone, CAL1, to deposit bip CENP-A into chromatin. Co-expression of bip CENP-A and bip CAL1 in mel cells restores centromeric localization, and similar findings apply to other Drosophila species. We identify two co-evolving regions, CENP-A L1 and the CAL1 N terminus, as critical for lineage-specific CENP-A incorporation. Collectively, our data show that the rapid evolution of L1 modulates CAL1-mediated CENP-A assembly, suggesting an alternative mechanism for the suppression of centromere drive.


Assuntos
Centrômero/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histonas/metabolismo , Animais , Proteína Centromérica A , Cromatina/metabolismo , Segregação de Cromossomos/fisiologia , Mitose/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA