Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 999
Filter
Add more filters

Publication year range
1.
Cell ; 187(12): 3006-3023.e26, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38744280

ABSTRACT

Centromeres are scaffolds for the assembly of kinetochores that ensure chromosome segregation during cell division. How vertebrate centromeres obtain a three-dimensional structure to accomplish their primary function is unclear. Using super-resolution imaging, capture-C, and polymer modeling, we show that vertebrate centromeres are partitioned by condensins into two subdomains during mitosis. The bipartite structure is found in human, mouse, and chicken cells and is therefore a fundamental feature of vertebrate centromeres. Super-resolution imaging and electron tomography reveal that bipartite centromeres assemble bipartite kinetochores, with each subdomain binding a distinct microtubule bundle. Cohesin links the centromere subdomains, limiting their separation in response to spindle forces and avoiding merotelic kinetochore-spindle attachments. Lagging chromosomes during cancer cell divisions frequently have merotelic attachments in which the centromere subdomains are separated and bioriented. Our work reveals a fundamental aspect of vertebrate centromere biology with implications for understanding the mechanisms that guarantee faithful chromosome segregation.


Subject(s)
Centromere , Cohesins , Kinetochores , Mitosis , Animals , Humans , Mice , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chickens , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosome Segregation , Kinetochores/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism
2.
Cell ; 186(9): 1985-2001.e19, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37075754

ABSTRACT

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.


Subject(s)
Centromere , Genetic Techniques , Humans , Aneuploidy , Centromere/genetics , Chromosome Deletion , Neoplasms/genetics , Clustered Regularly Interspaced Short Palindromic Repeats
3.
Cell ; 185(17): 3153-3168.e18, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35926507

ABSTRACT

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.


Subject(s)
Centromere , Cyperaceae , Animals , Centromere/genetics , Cyperaceae/genetics , Evolution, Molecular , Karyotype , Plants/genetics
4.
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
5.
Cell ; 178(5): 1132-1144.e10, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31402175

ABSTRACT

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.


Subject(s)
Centromere/genetics , Meiosis , Animals , Chromosome Segregation , Female , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism
6.
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
7.
Cell ; 175(3): 780-795.e15, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30318142

ABSTRACT

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.


Subject(s)
Centromere/genetics , Chromosomes, Fungal/genetics , Mitosis , Saccharomyces cerevisiae/genetics , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism
8.
Cell ; 171(1): 72-84.e13, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938124

ABSTRACT

The ring-shaped cohesin complex brings together distant DNA domains to maintain, express, and segregate the genome. Establishing specific chromosomal linkages depends on cohesin recruitment to defined loci. One such locus is the budding yeast centromere, which is a paradigm for targeted cohesin loading. The kinetochore, a multiprotein complex that connects centromeres to microtubules, drives the recruitment of high levels of cohesin to link sister chromatids together. We have exploited this system to determine the mechanism of specific cohesin recruitment. We show that phosphorylation of the Ctf19 kinetochore protein by a conserved kinase, DDK, provides a binding site for the Scc2/4 cohesin loading complex, thereby directing cohesin loading to centromeres. A similar mechanism targets cohesin to chromosomes in vertebrates. These findings represent a complete molecular description of targeted cohesin loading, a phenomenon with wide-ranging importance in chromosome segregation and, in multicellular organisms, transcription regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Saccharomyces cerevisiae/metabolism , Centromere/metabolism , Cytoskeletal Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphorylation , Phylogeny , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/metabolism , X-Ray Diffraction , Cohesins
9.
Mol Cell ; 84(6): 1003-1020.e10, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38359824

ABSTRACT

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.


Subject(s)
Centromere , Repetitive Sequences, Nucleic Acid , Humans , Centromere/genetics , Mitosis/genetics , Genomic Instability
10.
Mol Cell ; 84(9): 1783-1801.e7, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38614097

ABSTRACT

Liquid-liquid phase separation (LLPS) of putative assembly scaffolds has been proposed to drive the biogenesis of membraneless compartments. LLPS scaffolds are usually identified through in vitro LLPS assays with single macromolecules (homotypic), but the predictive value of these assays remains poorly characterized. Here, we apply a strategy to evaluate the robustness of homotypic LLPS assays. When applied to the chromosomal passenger complex (CPC), which undergoes LLPS in vitro and localizes to centromeres to promote chromosome biorientation, LLPS propensity in vitro emerged as an unreliable predictor of subcellular localization. In vitro CPC LLPS in aqueous buffers was enhanced by commonly used crowding agents. Conversely, diluted cytomimetic media dissolved condensates of the CPC and of several other proteins. We also show that centromeres do not seem to nucleate LLPS, nor do they promote local, spatially restrained LLPS of the CPC. Our strategy can be adapted to purported LLPS scaffolds of other membraneless compartments.


Subject(s)
Centromere , Centromere/metabolism , Macromolecular Substances/metabolism , Macromolecular Substances/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Humans , Phase Separation
11.
Cell ; 167(4): 1028-1040.e15, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27881301

ABSTRACT

Kinetochores, multisubunit protein assemblies, connect chromosomes to spindle microtubules to promote chromosome segregation. The 10-subunit KMN assembly (comprising KNL1, MIS12, and NDC80 complexes, designated KNL1C, MIS12C, and NDC80C) binds microtubules and regulates mitotic checkpoint function through NDC80C and KNL1C, respectively. MIS12C, on the other hand, connects the KMN to the chromosome-proximal domain of the kinetochore through a direct interaction with CENP-C. The structural basis for this crucial bridging function of MIS12C is unknown. Here, we report crystal structures of human MIS12C associated with a fragment of CENP-C and unveil the role of Aurora B kinase in the regulation of this interaction. The structure of MIS12:CENP-C complements previously determined high-resolution structures of functional regions of NDC80C and KNL1C and allows us to build a near-complete structural model of the KMN assembly. Our work illuminates the structural organization of essential chromosome segregation machinery that is conserved in most eukaryotes.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Crystallography, X-Ray , Kinetochores/chemistry , Multiprotein Complexes/chemistry , Animals , Aurora Kinase B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytoskeletal Proteins , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Models, Chemical , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism
12.
Cell ; 165(2): 396-409, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27020753

ABSTRACT

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian/metabolism , Metabolic Networks and Pathways , Oocytes/metabolism , Animals , Argonaute Proteins/metabolism , Base Sequence , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/metabolism , Cell Division , Embryo, Nonmammalian/cytology , Embryonic Development , Female , Kinesins/metabolism , Microtubules/metabolism , Molecular Sequence Data , RNA Processing, Post-Transcriptional
13.
Cell ; 167(4): 1014-1027.e12, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27881300

ABSTRACT

Kinetochores connect centromeric nucleosomes with mitotic-spindle microtubules through conserved, cross-interacting protein subassemblies. In budding yeast, the heterotetrameric MIND complex (Mtw1, Nnf1, Nsl1, Dsn1), ortholog of the metazoan Mis12 complex, joins the centromere-proximal components, Mif2 and COMA, with the principal microtubule-binding component, the Ndc80 complex (Ndc80C). We report the crystal structure of Kluyveromyces lactis MIND and examine its partner interactions, to understand the connection from a centromeric nucleosome to a much larger microtubule. MIND resembles an elongated, asymmetric Y; two globular heads project from a coiled-coil shaft. An N-terminal extension of Dsn1 from one head regulates interactions of the other head, blocking binding of Mif2 and COMA. Dsn1 phosphorylation by Ipl1/Aurora B relieves this autoinhibition, enabling MIND to join an assembling kinetochore. A C-terminal extension of Dsn1 recruits Ndc80C to the opposite end of the shaft. The structure and properties of MIND show how it integrates phospho-regulatory inputs for kinetochore assembly and disassembly.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Fungal Proteins/chemistry , Kinetochores/chemistry , Kluyveromyces/chemistry , Multiprotein Complexes/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , Fungal Proteins/metabolism , Kinetochores/metabolism , Kluyveromyces/cytology , Kluyveromyces/metabolism , Multiprotein Complexes/metabolism
14.
Mol Cell ; 83(13): 2188-2205.e13, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37295434

ABSTRACT

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.


Subject(s)
Centromere , Kinetochores , Humans , Kinetochores/metabolism , Centromere/genetics , Centromere/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromatin , Centromere Protein A/genetics , Centromere Protein A/metabolism
15.
Annu Rev Genet ; 56: 279-314, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36055650

ABSTRACT

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.


Subject(s)
Centromere , Kinetochores , Centromere/genetics , Chromosome Segregation/genetics , Microtubules/genetics , Microtubules/metabolism , Chromatin/genetics , Chromatin/metabolism
16.
Mol Cell ; 82(21): 4018-4032.e9, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332605

ABSTRACT

Kinetochore assembly on centromeres is central for chromosome segregation, and defects in this process cause mitotic errors and aneuploidy. Besides the well-established protein network, emerging evidence suggests the involvement of regulatory RNA in kinetochore assembly; however, it has remained elusive about the identity of such RNA, let alone its mechanism of action in this critical process. Here, we report CCTT, a previously uncharacterized long non-coding RNA (lncRNA) transcribed from the arm of human chromosome 17, which plays a vital role in kinetochore assembly. We show that CCTT highly localizes to all centromeres via the formation of RNA-DNA triplex and specifically interacts with CENP-C to help engage this blueprint protein in centromeres, and consequently, CCTT loss triggers extensive mitotic errors and aneuploidy. These findings uncover a non-centromere-derived lncRNA that recruits CENP-C to centromeres and shed critical lights on the function of centromeric DNA sequences as anchor points for kinetochore assembly.


Subject(s)
RNA, Long Noncoding , Humans , Aneuploidy , Centromere Protein A/metabolism , DNA , Kinetochores/metabolism , RNA, Long Noncoding/genetics , Centromere
17.
Mol Cell ; 82(11): 2113-2131.e8, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35525244

ABSTRACT

Centromeres are specialized chromosome loci that seed the kinetochore, a large protein complex that effects chromosome segregation. A 16-subunit complex, the constitutive centromere associated network (CCAN), connects between the specialized centromeric chromatin, marked by the histone H3 variant CENP-A, and the spindle-binding moiety of the kinetochore. Here, we report a cryo-electron microscopy structure of human CCAN. We highlight unique features such as the pseudo GTPase CENP-M and report how a crucial CENP-C motif binds the CENP-LN complex. The CCAN structure has implications for the mechanism of specific recognition of the CENP-A nucleosome. A model consistent with our structure depicts the CENP-C-bound nucleosome as connected to the CCAN through extended, flexible regions of CENP-C. An alternative model identifies both CENP-C and CENP-N as specificity determinants but requires CENP-N to bind CENP-A in a mode distinct from the classical nucleosome octamer.


Subject(s)
Kinetochores , Nucleosomes , Centromere/metabolism , Centromere Protein A/metabolism , Cryoelectron Microscopy , Humans , Kinetochores/metabolism , Nucleosomes/genetics
18.
Mol Cell ; 82(9): 1751-1767.e8, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35320753

ABSTRACT

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.


Subject(s)
Centromere , Chromosomal Proteins, Non-Histone , Autoantigens/genetics , Autoantigens/metabolism , Centromere/genetics , Centromere/metabolism , Centromere Protein A/genetics , Centromere Protein A/metabolism , Chromatin , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/genetics , Humans
19.
Annu Rev Genet ; 55: 583-602, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34813350

ABSTRACT

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.


Subject(s)
Centromere , Genome , Centromere/genetics , Genomic Instability/genetics , Genomics , Humans
20.
Annu Rev Genet ; 55: 401-425, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34813351

ABSTRACT

Repeat-enriched genomic regions evolve rapidly and yet support strictly conserved functions like faithful chromosome transmission and the preservation of genome integrity. The leading resolution to this paradox is that DNA repeat-packaging proteins evolve adaptively to mitigate deleterious changes in DNA repeat copy number, sequence, and organization. Exciting new research has tested this model of coevolution by engineering evolutionary mismatches between adaptively evolving chromatin proteins of one species and the DNA repeats of a close relative. Here, we review these innovative evolution-guided functional analyses. The studies demonstrate that vital, chromatin-mediated cellular processes, including transposon suppression, faithful chromosome transmission, and chromosome retention depend on species-specific versions of chromatin proteins that package species-specific DNA repeats. In many cases, the ever-evolving repeats are selfish genetic elements, raising the possibility that chromatin is a battleground of intragenomic conflict.


Subject(s)
Centromere , Chromatin , Chromatin/genetics , Evolution, Molecular , Genome , Genomics
SELECTION OF CITATIONS
SEARCH DETAIL