Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.837
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 186(9): 1985-2001.e19, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37075754

RESUMO

Aneuploidy, the presence of chromosome gains or losses, is a hallmark of cancer. Here, we describe KaryoCreate (karyotype CRISPR-engineered aneuploidy technology), a system that enables the generation of chromosome-specific aneuploidies by co-expression of an sgRNA targeting chromosome-specific CENPA-binding ɑ-satellite repeats together with dCas9 fused to mutant KNL1. We design unique and highly specific sgRNAs for 19 of the 24 chromosomes. Expression of these constructs leads to missegregation and induction of gains or losses of the targeted chromosome in cellular progeny, with an average efficiency of 8% for gains and 12% for losses (up to 20%) validated across 10 chromosomes. Using KaryoCreate in colon epithelial cells, we show that chromosome 18q loss, frequent in gastrointestinal cancers, promotes resistance to TGF-ß, likely due to synergistic hemizygous deletion of multiple genes. Altogether, we describe an innovative technology to create and study chromosome missegregation and aneuploidy in the context of cancer and beyond.


Assuntos
Centrômero , Técnicas Genéticas , Humanos , Aneuploidia , Centrômero/genética , Deleção Cromossômica , Neoplasias/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas
2.
Cell ; 185(17): 3153-3168.e18, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35926507

RESUMO

The centromere represents a single region in most eukaryotic chromosomes. However, several plant and animal lineages assemble holocentromeres along the entire chromosome length. Here, we compare genome organization and evolution as a function of centromere type by assembling chromosome-scale holocentric genomes with repeat-based holocentromeres from three beak-sedge (Rhynchospora pubera, R. breviuscula, and R. tenuis) and their closest monocentric relative, Juncus effusus. We demonstrate that transition to holocentricity affected 3D genome architecture by redefining genomic compartments, while distributing centromere function to thousands of repeat-based centromere units genome-wide. We uncover a complex genome organization in R. pubera that hides its unexpected octoploidy and describe a marked reduction in chromosome number for R. tenuis, which has only two chromosomes. We show that chromosome fusions, facilitated by repeat-based holocentromeres, promoted karyotype evolution and diploidization. Our study thus sheds light on several important aspects of genome architecture and evolution influenced by centromere organization.


Assuntos
Centrômero , Cyperaceae , Animais , Centrômero/genética , Cyperaceae/genética , Evolução Molecular , Cariótipo , Plantas/genética
3.
Cell ; 185(17): 3083-3085, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35985286

RESUMO

In some plants and animals, microtubules attach across the length of the chromosome in mitosis, forming a holocentromere instead of a single centromeric locus. A new study in Cell shows that in the holocentric beak sedge Rhynchospora, holocentromeres also impact genomic architecture, epigenome organization, and karyotype evolution.


Assuntos
Centrômero , Cyperaceae , Animais , Centrômero/genética , Cyperaceae/genética , Microtúbulos , Mitose , Plantas/genética
4.
Cell ; 178(5): 1132-1144.e10, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31402175

RESUMO

Asymmetric division in female meiosis creates selective pressure favoring selfish centromeres that bias their transmission to the egg. This centromere drive can explain the paradoxical rapid evolution of both centromere DNA and centromere-binding proteins despite conserved centromere function. Here, we define a molecular pathway linking expanded centromeres to histone phosphorylation and recruitment of microtubule destabilizing factors, leading to detachment of selfish centromeres from spindle microtubules that would direct them to the polar body. Exploiting centromere divergence between species, we show that selfish centromeres in two hybrid mouse models use the same molecular pathway but modulate it differently to enrich destabilizing factors. Our results indicate that increasing microtubule destabilizing activity is a general strategy for drive in both models, but centromeres have evolved distinct mechanisms to increase that activity. Furthermore, we show that drive depends on slowing meiotic progression, suggesting that selfish centromeres can be suppressed by regulating meiotic timing.


Assuntos
Centrômero/genética , Meiose , Animais , Segregação de Cromossomos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Oócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
5.
Cell ; 178(3): 624-639.e19, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31348889

RESUMO

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.


Assuntos
Centrômero/metabolismo , Cromossomos Artificiais Humanos/metabolismo , DNA Satélite/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Centrômero/genética , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Proteína B de Centrômero/deficiência , Proteína B de Centrômero/genética , Proteína B de Centrômero/metabolismo , Epigênese Genética , Humanos , Nucleossomos/química , Nucleossomos/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo
6.
Cell ; 175(3): 780-795.e15, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30318142

RESUMO

During mitosis, chromatin condensation shapes chromosomes as separate, rigid, and compact sister chromatids to facilitate their segregation. Here, we show that, unlike wild-type yeast chromosomes, non-chromosomal DNA circles and chromosomes lacking a centromere fail to condense during mitosis. The centromere promotes chromosome condensation strictly in cis through recruiting the kinases Aurora B and Bub1, which trigger the autonomous condensation of the entire chromosome. Shugoshin and the deacetylase Hst2 facilitated spreading the condensation signal to the chromosome arms. Targeting Aurora B to DNA circles or centromere-ablated chromosomes or releasing Shugoshin from PP2A-dependent inhibition bypassed the centromere requirement for condensation and enhanced the mitotic stability of DNA circles. Our data indicate that yeast cells license the chromosome-autonomous condensation of their chromatin in a centromere-dependent manner, excluding from this process non-centromeric DNA and thereby inhibiting their propagation.


Assuntos
Centrômero/genética , Cromossomos Fúngicos/genética , Mitose , Saccharomyces cerevisiae/genética , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/genética , Sirtuína 2/metabolismo
7.
Cell ; 173(4): 839-850.e18, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29628142

RESUMO

Maize abnormal chromosome 10 (Ab10) encodes a classic example of true meiotic drive that converts heterochromatic regions called knobs into motile neocentromeres that are preferentially transmitted to egg cells. Here, we identify a cluster of eight genes on Ab10, called the Kinesin driver (Kindr) complex, that are required for both neocentromere motility and preferential transmission. Two meiotic drive mutants that lack neocentromere activity proved to be kindr epimutants with increased DNA methylation across the entire gene cluster. RNAi of Kindr induced a third epimutant and corresponding loss of meiotic drive. Kinesin gliding assays and immunolocalization revealed that KINDR is a functional minus-end-directed kinesin that localizes specifically to knobs containing 180 bp repeats. Sequence comparisons suggest that Kindr diverged from a Kinesin-14A ancestor ∼12 mya and has driven the accumulation of > 500 Mb of knob repeats and affected the segregation of thousands of genes linked to knobs on all 10 chromosomes.


Assuntos
Centrômero/metabolismo , Cinesinas/metabolismo , Meiose , Proteínas de Plantas/metabolismo , Zea mays/metabolismo , Centrômero/genética , Cromossomos de Plantas , Evolução Molecular , Haplótipos , Hibridização in Situ Fluorescente , Cinesinas/antagonistas & inibidores , Cinesinas/classificação , Cinesinas/genética , Modelos Genéticos , Mutagênese , Filogenia , Proteínas de Plantas/antagonistas & inibidores , Proteínas de Plantas/classificação , Proteínas de Plantas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sequenciamento Completo do Genoma , Zea mays/genética
8.
Annu Rev Cell Dev Biol ; 34: 265-288, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30044650

RESUMO

Constitutive heterochromatin is a major component of the eukaryotic nucleus and is essential for the maintenance of genome stability. Highly concentrated at pericentromeric and telomeric domains, heterochromatin is riddled with repetitive sequences and has evolved specific ways to compartmentalize, silence, and repair repeats. The delicate balance between heterochromatin epigenetic maintenance and cellular processes such as mitosis and DNA repair and replication reveals a highly dynamic and plastic chromatin domain that can be perturbed by multiple mechanisms, with far-reaching consequences for genome integrity. Indeed, heterochromatin dysfunction provokes genetic turmoil by inducing aberrant repeat repair, chromosome segregation errors, transposon activation, and replication stress and is strongly implicated in aging and tumorigenesis. Here, we summarize the general principles of heterochromatin structure and function, discuss the importance of its maintenance for genome integrity, and propose that more comprehensive analyses of heterochromatin roles in tumorigenesis will be integral to future innovations in cancer treatment.


Assuntos
Reparo do DNA/genética , Instabilidade Genômica , Heterocromatina/genética , Mitose/genética , Centrômero/genética , Segregação de Cromossomos/genética , Genoma/genética , Histonas/genética , Humanos , Sequências Repetitivas de Ácido Nucleico/genética , Telômero/genética
9.
Mol Cell ; 84(6): 1003-1020.e10, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38359824

RESUMO

The high incidence of whole-arm chromosome aneuploidy and translocations in tumors suggests instability of centromeres, unique loci built on repetitive sequences and essential for chromosome separation. The causes behind this fragility and the mechanisms preserving centromere integrity remain elusive. We show that replication stress, hallmark of pre-cancerous lesions, promotes centromeric breakage in mitosis, due to spindle forces and endonuclease activities. Mechanistically, we unveil unique dynamics of the centromeric replisome distinct from the rest of the genome. Locus-specific proteomics identifies specialized DNA replication and repair proteins at centromeres, highlighting them as difficult-to-replicate regions. The translesion synthesis pathway, along with other factors, acts to sustain centromere replication and integrity. Prolonged stress causes centromeric alterations like ruptures and translocations, as observed in ovarian cancer models experiencing replication stress. This study provides unprecedented insights into centromere replication and integrity, proposing mechanistic insights into the origins of centromere alterations leading to abnormal cancerous karyotypes.


Assuntos
Centrômero , Sequências Repetitivas de Ácido Nucleico , Humanos , Centrômero/genética , Mitose/genética , Instabilidade Genômica
10.
Mol Cell ; 84(11): 2017-2035.e6, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38795706

RESUMO

Whether and how histone post-translational modifications and the proteins that bind them drive 3D genome organization remains unanswered. Here, we evaluate the contribution of H3K9-methylated constitutive heterochromatin to 3D genome organization in Drosophila tissues. We find that the predominant organizational feature of wild-type tissues is the segregation of euchromatic chromosome arms from heterochromatic pericentromeres. Reciprocal perturbation of HP1a⋅H3K9me binding, using a point mutation in the HP1a chromodomain or replacement of the replication-dependent histone H3 with H3K9R mutant histones, revealed that HP1a binding to methylated H3K9 in constitutive heterochromatin is required to limit contact frequency between pericentromeres and chromosome arms and regulate the distance between arm and pericentromeric regions. Surprisingly, the self-association of pericentromeric regions is largely preserved despite the loss of H3K9 methylation and HP1a occupancy. Thus, the HP1a⋅H3K9 interaction contributes to but does not solely drive the segregation of euchromatin and heterochromatin inside the nucleus.


Assuntos
Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona , Proteínas de Drosophila , Drosophila melanogaster , Heterocromatina , Histonas , Heterocromatina/metabolismo , Heterocromatina/genética , Animais , Histonas/metabolismo , Histonas/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Metilação , Eucromatina/metabolismo , Eucromatina/genética , Centrômero/metabolismo , Centrômero/genética , Ligação Proteica , Genoma de Inseto , Segregação de Cromossomos , Processamento de Proteína Pós-Traducional
11.
Mol Cell ; 83(4): 523-538.e7, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36702125

RESUMO

Centromeres are essential for chromosome segregation in most animals and plants yet are among the most rapidly evolving genome elements. The mechanisms underlying this paradoxical phenomenon remain enigmatic. Here, we report that human centromeres innately harbor a striking enrichment of DNA breaks within functionally active centromere regions. Establishing a single-cell imaging strategy that enables comparative assessment of DNA breaks at repetitive regions, we show that centromeric DNA breaks are induced not only during active cellular proliferation but also de novo during quiescence. Markedly, centromere DNA breaks in quiescent cells are resolved enzymatically by the evolutionarily conserved RAD51 recombinase, which in turn safeguards the specification of functional centromeres. This study highlights the innate fragility of centromeres, which may have been co-opted over time to reinforce centromere specification while driving rapid evolution. The findings also provide insights into how fragile centromeres are likely to contribute to human disease.


Assuntos
Centrômero , DNA , Animais , Humanos , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Recombinação Genética
12.
Mol Cell ; 83(13): 2188-2205.e13, 2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37295434

RESUMO

Kinetochore is an essential protein complex required for accurate chromosome segregation. The constitutive centromere-associated network (CCAN), a subcomplex of the kinetochore, associates with centromeric chromatin and provides a platform for the kinetochore assembly. The CCAN protein CENP-C is thought to be a central hub for the centromere/kinetochore organization. However, the role of CENP-C in CCAN assembly needs to be elucidated. Here, we demonstrate that both the CCAN-binding domain and the C-terminal region that includes the Cupin domain of CENP-C are necessary and sufficient for chicken CENP-C function. Structural and biochemical analyses reveal self-oligomerization of the Cupin domains of chicken and human CENP-C. We find that the CENP-C Cupin domain oligomerization is vital for CENP-C function, centromeric localization of CCAN, and centromeric chromatin organization. These results suggest that CENP-C facilitates the centromere/kinetochore assembly through its oligomerization.


Assuntos
Centrômero , Cinetocoros , Humanos , Cinetocoros/metabolismo , Centrômero/genética , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromatina , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo
13.
Annu Rev Genet ; 56: 279-314, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36055650

RESUMO

Kinetochores are molecular machines that power chromosome segregation during the mitotic and meiotic cell divisions of all eukaryotes. Aristotle explains how we think we have knowledge of a thing only when we have grasped its cause. In our case, to gain understanding of the kinetochore, the four causes correspond to questions that we must ask: (a) What are the constituent parts, (b) how does it assemble, (c) what is the structure and arrangement, and (d) what is the function? Here we outline the current blueprint for the assembly of a kinetochore, how functions are mapped onto this architecture, and how this is shaped by the underlying pericentromeric chromatin. The view of the kinetochore that we present is possible because an almost complete parts list of the kinetochore is now available alongside recent advances using in vitro reconstitution, structural biology, and genomics. In many organisms, each kinetochore binds to multiple microtubules, and we propose a model for how this ensemble-level architecture is organized, drawing on key insights from the simple one microtubule-one kinetochore setup in budding yeast and innovations that enable meiotic chromosome segregation.


Assuntos
Centrômero , Cinetocoros , Centrômero/genética , Segregação de Cromossomos/genética , Microtúbulos/genética , Microtúbulos/metabolismo , Cromatina/genética , Cromatina/metabolismo
14.
Nature ; 629(8010): 136-145, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38570684

RESUMO

Human centromeres have been traditionally very difficult to sequence and assemble owing to their repetitive nature and large size1. As a result, patterns of human centromeric variation and models for their evolution and function remain incomplete, despite centromeres being among the most rapidly mutating regions2,3. Here, using long-read sequencing, we completely sequenced and assembled all centromeres from a second human genome and compared it to the finished reference genome4,5. We find that the two sets of centromeres show at least a 4.1-fold increase in single-nucleotide variation when compared with their unique flanks and vary up to 3-fold in size. Moreover, we find that 45.8% of centromeric sequence cannot be reliably aligned using standard methods owing to the emergence of new α-satellite higher-order repeats (HORs). DNA methylation and CENP-A chromatin immunoprecipitation experiments show that 26% of the centromeres differ in their kinetochore position by >500 kb. To understand evolutionary change, we selected six chromosomes and sequenced and assembled 31 orthologous centromeres from the common chimpanzee, orangutan and macaque genomes. Comparative analyses reveal a nearly complete turnover of α-satellite HORs, with characteristic idiosyncratic changes in α-satellite HORs for each species. Phylogenetic reconstruction of human haplotypes supports limited to no recombination between the short (p) and long (q) arms across centromeres and reveals that novel α-satellite HORs share a monophyletic origin, providing a strategy to estimate the rate of saltatory amplification and mutation of human centromeric DNA.


Assuntos
Centrômero , Evolução Molecular , Variação Genética , Animais , Humanos , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A/metabolismo , Metilação de DNA/genética , DNA Satélite/genética , Cinetocoros/metabolismo , Macaca/genética , Pan troglodytes/genética , Polimorfismo de Nucleotídeo Único/genética , Pongo/genética , Masculino , Feminino , Padrões de Referência , Imunoprecipitação da Cromatina , Haplótipos , Mutação , Amplificação de Genes , Alinhamento de Sequência , Cromatina/genética , Cromatina/metabolismo , Especificidade da Espécie
15.
Nature ; 631(8021): 678-685, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38961301

RESUMO

Pericentric heterochromatin is a critical component of chromosomes marked by histone H3 K9 (H3K9) methylation1-3. However, what recruits H3K9-specific histone methyltransferases to pericentric regions in vertebrates remains unclear4, as does why pericentric regions in different species share the same H3K9 methylation mark despite lacking highly conserved DNA sequences2,5. Here we show that zinc-finger proteins ZNF512 and ZNF512B specifically localize at pericentric regions through direct DNA binding. Notably, both ZNF512 and ZNF512B are sufficient to initiate de novo heterochromatin formation at ectopically targeted repetitive regions and pericentric regions, as they directly recruit SUV39H1 and SUV39H2 (SUV39H) to catalyse H3K9 methylation. SUV39H2 makes a greater contribution to H3K9 trimethylation, whereas SUV39H1 seems to contribute more to silencing, probably owing to its preferential association with HP1 proteins. ZNF512 and ZNF512B from different species can specifically target pericentric regions of other vertebrates, because the atypical long linker residues between the zinc-fingers of ZNF512 and ZNF512B offer flexibility in recognition of non-consecutively organized three-nucleotide triplets targeted by each zinc-finger. This study addresses two long-standing questions: how constitutive heterochromatin is initiated and how seemingly variable pericentric sequences are targeted by the same set of conserved machinery in vertebrates.


Assuntos
Centrômero , Evolução Molecular , Heterocromatina , Histona-Lisina N-Metiltransferase , Histonas , Motivos de Nucleotídeos , Animais , Humanos , Camundongos , Centrômero/genética , Centrômero/metabolismo , Galinhas , Homólogo 5 da Proteína Cromobox , Inativação Gênica , Heterocromatina/metabolismo , Heterocromatina/química , Heterocromatina/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/química , Histonas/metabolismo , Histonas/química , Anfioxos , Metilação , Petromyzon , Proteínas Repressoras/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Serpentes , Xenopus laevis , Peixe-Zebra , Dedos de Zinco
16.
Mol Cell ; 82(11): 1976-1978, 2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35659323

RESUMO

Structures of the reconstituted human inner kinetochore complex by Pesenti et al. (2022) and Yatskevich et al. (2022) raise the question of whether it is the CENP-A nucleosome or the CCAN complex itself that provides the foundation for kinetochore assembly.


Assuntos
Cromatina , Cinetocoros , Centrômero/genética , Proteína Centromérica A/genética , Cromatina/genética , Humanos , Nucleossomos/genética
17.
Mol Cell ; 82(9): 1751-1767.e8, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35320753

RESUMO

Chromosome inheritance depends on centromeres, epigenetically specified regions of chromosomes. While conventional human centromeres are known to be built of long tandem DNA repeats, much of their architecture remains unknown. Using single-molecule techniques such as AFM, nanopores, and optical tweezers, we find that human centromeric DNA exhibits complex DNA folds such as local hairpins. Upon binding to a specific sequence within centromeric regions, the DNA-binding protein CENP-B compacts centromeres by forming pronounced DNA loops between the repeats, which favor inter-chromosomal centromere compaction and clustering. This DNA-loop-mediated organization of centromeric chromatin participates in maintaining centromere position and integrity upon microtubule pulling during mitosis. Our findings emphasize the importance of DNA topology in centromeric regulation and stability.


Assuntos
Centrômero , Proteínas Cromossômicas não Histona , Autoantígenos/genética , Autoantígenos/metabolismo , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Cromatina , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA/genética , Humanos
18.
Annu Rev Genet ; 55: 583-602, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34813350

RESUMO

We are entering a new era in genomics where entire centromeric regions are accurately represented in human reference assemblies. Access to these high-resolution maps will enable new surveys of sequence and epigenetic variation in the population and offer new insight into satellite array genomics and centromere function. Here, we focus on the sequence organization and evolution of alpha satellites, which are credited as the genetic and genomic definition of human centromeres due to their interaction with inner kinetochore proteins and their importance in the development of human artificial chromosome assays. We provide an overview of alpha satellite repeat structure and array organization in the context of these high-quality reference data sets; discuss the emergence of variation-based surveys; and provide perspective on the role of this new source of genetic and epigenetic variation in the context of chromosome biology, genome instability, and human disease.


Assuntos
Centrômero , Genoma , Centrômero/genética , Instabilidade Genômica/genética , Genômica , Humanos
19.
Annu Rev Genet ; 55: 331-348, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34496611

RESUMO

Centromeres are essential to genome inheritance, serving as the site of kinetochore assembly and coordinating chromosome segregation during cell division. Abnormal centromere function is associated with birth defects, infertility, and cancer. Normally, centromeres are assembled and maintained at the same chromosomal location. However, ectopic centromeres form spontaneously at new genomic locations and contribute to genome instability and developmental defects as well as to acquired and congenital human disease. Studies in model organisms have suggested that certain regions of the genome, including pericentromeres, heterochromatin, and regions of open chromatin or active transcription, support neocentromere activation. However, there is no universal mechanism that explains neocentromere formation. This review focuses on recent technological and intellectual advances in neocentromere research and proposes future areas of study. Understanding neocentromere biology will provide a better perspective on chromosome and genome organization and functional context for information generated from the Human Genome Project, ENCODE, and other large genomics consortia.


Assuntos
Centrômero , Cromatina , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Cromatina/genética , Epigênese Genética , Epigenômica , Humanos
20.
Nature ; 617(7960): 335-343, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37165241

RESUMO

The short arms of the human acrocentric chromosomes 13, 14, 15, 21 and 22 (SAACs) share large homologous regions, including ribosomal DNA repeats and extended segmental duplications1,2. Although the resolution of these regions in the first complete assembly of a human genome-the Telomere-to-Telomere Consortium's CHM13 assembly (T2T-CHM13)-provided a model of their homology3, it remained unclear whether these patterns were ancestral or maintained by ongoing recombination exchange. Here we show that acrocentric chromosomes contain pseudo-homologous regions (PHRs) indicative of recombination between non-homologous sequences. Utilizing an all-to-all comparison of the human pangenome from the Human Pangenome Reference Consortium4 (HPRC), we find that contigs from all of the SAACs form a community. A variation graph5 constructed from centromere-spanning acrocentric contigs indicates the presence of regions in which most contigs appear nearly identical between heterologous acrocentric chromosomes in T2T-CHM13. Except on chromosome 15, we observe faster decay of linkage disequilibrium in the pseudo-homologous regions than in the corresponding short and long arms, indicating higher rates of recombination6,7. The pseudo-homologous regions include sequences that have previously been shown to lie at the breakpoint of Robertsonian translocations8, and their arrangement is compatible with crossover in inverted duplications on chromosomes 13, 14 and 21. The ubiquity of signals of recombination between heterologous acrocentric chromosomes seen in the HPRC draft pangenome suggests that these shared sequences form the basis for recurrent Robertsonian translocations, providing sequence and population-based confirmation of hypotheses first developed from cytogenetic studies 50 years ago9.


Assuntos
Centrômero , Cromossomos Humanos , Recombinação Genética , Humanos , Centrômero/genética , Cromossomos Humanos/genética , DNA Ribossômico/genética , Recombinação Genética/genética , Translocação Genética/genética , Citogenética , Telômero/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA