Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Genet ; 56(5): 900-912, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38388848

RESUMO

Whole chromosome and arm-level copy number alterations occur at high frequencies in tumors, but their selective advantages, if any, are poorly understood. Here, utilizing unbiased whole chromosome genetic screens combined with in vitro evolution to generate arm- and subarm-level events, we iteratively selected the fittest karyotypes from aneuploidized human renal and mammary epithelial cells. Proliferation-based karyotype selection in these epithelial lines modeled tissue-specific tumor aneuploidy patterns in patient cohorts in the absence of driver mutations. Hi-C-based translocation mapping revealed that arm-level events usually emerged in multiples of two via centromeric translocations and occurred more frequently in tetraploids than diploids, contributing to the increased diversity in evolving tetraploid populations. Isogenic clonal lineages enabled elucidation of pro-tumorigenic mechanisms associated with common copy number alterations, revealing Notch signaling potentiation as a driver of 1q gain in breast cancer. We propose that intrinsic, tissue-specific proliferative effects underlie tumor copy number patterns in cancer.


Assuntos
Aneuploidia , Humanos , Feminino , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Variações do Número de Cópias de DNA , Neoplasias/genética , Neoplasias/patologia , Translocação Genética , Evolução Molecular , Proliferação de Células/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Especificidade de Órgãos/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia
2.
Life Sci Alliance ; 7(2)2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37989525

RESUMO

The genome is organized in functional compartments and structural domains at the sub-megabase scale. How within these domains interactions between numerous cis-acting enhancers and promoters regulate transcription remains an open question. Here, we determined chromatin folding and composition over several hundred kb around estrogen-responsive genes in human breast cancer cell lines after hormone stimulation. Modeling of 5C data at 1.8 kb resolution was combined with quantitative 3D analysis of multicolor FISH measurements at 100 nm resolution and integrated with ChIP-seq data on transcription factor binding and histone modifications. We found that rapid estradiol induction of the progesterone gene expression occurs in the context of preexisting, cell type-specific chromosomal architectures encompassing the 90 kb progesterone gene coding region and an enhancer-spiked 5' 300 kb upstream genomic region. In response to estradiol, interactions between estrogen receptor α (ERα) bound regulatory elements are reinforced. Whereas initial enhancer-gene contacts coincide with RNA Pol 2 binding and transcription initiation, sustained hormone stimulation promotes ERα accumulation creating a regulatory hub stimulating transcript synthesis. In addition to implications for estrogen receptor signaling, we uncover that preestablished chromatin architectures efficiently regulate gene expression upon stimulation without the need for de novo extensive rewiring of long-range chromatin interactions.


Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Humanos , Feminino , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Progesterona , Elementos Facilitadores Genéticos/genética , Cromatina/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Estradiol/farmacologia
3.
Cell Rep ; 42(9): 113132, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37708024

RESUMO

Uveal melanoma (UM) is a rare cancer resulting from the transformation of melanocytes in the uveal tract. Integrative analysis has identified four molecular and clinical subsets of UM. To improve our molecular understanding of UM, we performed extensive multi-omics characterization comparing two aggressive UM patient-derived xenograft models with normal choroidal melanocytes, including DNA optical mapping, specific histone modifications, and DNA topology analysis using Hi-C. Our gene expression and cytogenetic analyses suggest that genomic instability is a hallmark of UM. We also identified a recurrent deletion in the BAP1 promoter resulting in loss of expression and associated with high risk of metastases in UM patients. Hi-C revealed chromatin topology changes associated with the upregulation of PRAME, an independent prognostic biomarker in UM, and a potential therapeutic target. Our findings illustrate how multi-omics approaches can improve our understanding of tumorigenesis and reveal two distinct mechanisms of gene expression dysregulation in UM.


Assuntos
Melanoma , Multiômica , Humanos , Melanoma/patologia , Melanócitos/metabolismo , DNA , Antígenos de Neoplasias/genética
4.
bioRxiv ; 2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37398486

RESUMO

Protein arginine methyltransferase 5 (Prmt5) is an essential regulator of embryonic development and adult progenitor cell functions. Prmt5 expression is mis-regulated in many cancers, and the development of Prmt5 inhibitors as cancer therapeutics is an active area of research. Prmt5 functions via effects on gene expression, splicing, DNA repair, and other critical cellular processes. We examined whether Prmt5 functions broadly as a genome-wide regulator of gene transcription and higher-order chromatin interactions during the initial stages of adipogenesis using ChIP-Seq, RNA-seq, and Hi-C using 3T3-L1 cells, a frequently utilized model for adipogenesis. We observed robust genome-wide Prmt5 chromatin-binding at the onset of differentiation. Prmt5 localized to transcriptionally active genomic regions, acting as both a positive and a negative regulator. A subset of Prmt5 binding sites co-localized with mediators of chromatin organization at chromatin loop anchors. Prmt5 knockdown decreased insulation strength at the boundaries of topologically associating domains (TADs) adjacent to sites with Prmt5 and CTCF co-localization. Genes overlapping such weakened TAD boundaries showed transcriptional dysregulation. This study identifies Prmt5 as a broad regulator of gene expression, including regulation of early adipogenic factors, and reveals an unappreciated requirement for Prmt5 in maintaining strong insulation at TAD boundaries and overall chromatin organization.

5.
Mol Cell ; 83(14): 2434-2448.e7, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37402370

RESUMO

Insertions and deletions (indels) are common sources of structural variation, and insertions originating from spontaneous DNA lesions are frequent in cancer. We developed a highly sensitive assay called insertion and deletion sequencing (Indel-seq) to monitor rearrangements in human cells at the TRIM37 acceptor locus that reports indels stemming from experimentally induced and spontaneous genome instability. Templated insertions, which derive from sequences genome wide, require contact between donor and acceptor loci, require homologous recombination, and are stimulated by DNA end-processing. Insertions are facilitated by transcription and involve a DNA/RNA hybrid intermediate. Indel-seq reveals that insertions are generated via multiple pathways. The broken acceptor site anneals with a resected DNA break or invades the displaced strand of a transcription bubble or R-loop, followed by DNA synthesis, displacement, and then ligation by non-homologous end joining. Our studies identify transcription-coupled insertions as a critical source of spontaneous genome instability that is distinct from cut-and-paste events.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Humanos , Reparo do DNA por Junção de Extremidades , DNA/genética , Instabilidade Genômica , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/metabolismo
6.
Nat Struct Mol Biol ; 30(1): 38-51, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36550219

RESUMO

The relationships between chromosomal compartmentalization, chromatin state and function are poorly understood. Here by profiling long-range contact frequencies in HCT116 colon cancer cells, we distinguish three silent chromatin states, comprising two types of heterochromatin and a state enriched for H3K9me2 and H2A.Z that exhibits neutral three-dimensional interaction preferences and which, to our knowledge, has not previously been characterized. We find that heterochromatin marked by H3K9me3, HP1α and HP1ß correlates with strong compartmentalization. We demonstrate that disruption of DNA methyltransferase activity greatly remodels genome compartmentalization whereby domains lose H3K9me3-HP1α/ß binding and acquire the neutrally interacting state while retaining late replication timing. Furthermore, we show that H3K9me3-HP1α/ß heterochromatin is permissive to loop extrusion by cohesin but refractory to CTCF binding. Together, our work reveals a dynamic structural and organizational diversity of the silent portion of the genome and establishes connections between the regulation of chromatin state and chromosome organization, including an interplay between DNA methylation, compartmentalization and loop extrusion.


Assuntos
Cromatina , Heterocromatina , Metilação , Histonas/metabolismo , Homólogo 5 da Proteína Cromobox , Fatores de Transcrição/metabolismo
7.
Nat Commun ; 12(1): 6241, 2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34716321

RESUMO

Precise control of gene expression during differentiation relies on the interplay of chromatin and nuclear structure. Despite an established contribution of nuclear membrane proteins to developmental gene regulation, little is known regarding the role of inner nuclear proteins. Here we demonstrate that loss of the nuclear scaffolding protein Matrin-3 (Matr3) in erythroid cells leads to morphological and gene expression changes characteristic of accelerated maturation, as well as broad alterations in chromatin organization similar to those accompanying differentiation. Matr3 protein interacts with CTCF and the cohesin complex, and its loss perturbs their occupancy at a subset of sites. Destabilization of CTCF and cohesin binding correlates with altered transcription and accelerated differentiation. This association is conserved in embryonic stem cells. Our findings indicate Matr3 negatively affects cell fate transitions and demonstrate that a critical inner nuclear protein impacts occupancy of architectural factors, culminating in broad effects on chromatin organization and cell differentiation.


Assuntos
Cromatina/química , Leucemia Eritroblástica Aguda/patologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/fisiologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Embrionárias/fisiologia , Células Eritroides/patologia , Leucemia Eritroblástica Aguda/metabolismo , Camundongos Knockout , Proteínas Associadas à Matriz Nuclear/genética , Proteínas de Ligação a RNA/genética , Coesinas
8.
Elife ; 102021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34406118

RESUMO

DNA loop extrusion by condensins and decatenation by DNA topoisomerase II (topo II) are thought to drive mitotic chromosome compaction and individualization. Here, we reveal that the linker histone H1.8 antagonizes condensins and topo II to shape mitotic chromosome organization. In vitro chromatin reconstitution experiments demonstrate that H1.8 inhibits binding of condensins and topo II to nucleosome arrays. Accordingly, H1.8 depletion in Xenopus egg extracts increased condensins and topo II levels on mitotic chromatin. Chromosome morphology and Hi-C analyses suggest that H1.8 depletion makes chromosomes thinner and longer through shortening the average loop size and reducing the DNA amount in each layer of mitotic loops. Furthermore, excess loading of condensins and topo II to chromosomes by H1.8 depletion causes hyper-chromosome individualization and dispersion. We propose that condensins and topo II are essential for chromosome individualization, but their functions are tuned by the linker histone to keep chromosomes together until anaphase.


Assuntos
Cromatina/metabolismo , Cromossomos/genética , DNA Topoisomerases Tipo II/genética , Histonas/genética , Adenosina Trifosfatases/metabolismo , Animais , Extratos Celulares/química , Cromossomos/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Feminino , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Oócitos/química , Oócitos/metabolismo , Fuso Acromático/genética , Fuso Acromático/patologia , Fuso Acromático/ultraestrutura , Xenopus laevis
9.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33351783

RESUMO

The cohesin complex plays an essential role in chromosome maintenance and transcriptional regulation. Recurrent somatic mutations in the cohesin complex are frequent genetic drivers in cancer, including myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Here, using genetic dependency screens of stromal antigen 2-mutant (STAG2-mutant) AML, we identified DNA damage repair and replication as genetic dependencies in cohesin-mutant cells. We demonstrated increased levels of DNA damage and sensitivity of cohesin-mutant cells to poly(ADP-ribose) polymerase (PARP) inhibition. We developed a mouse model of MDS in which Stag2 mutations arose as clonal secondary lesions in the background of clonal hematopoiesis driven by tet methylcytosine dioxygenase 2 (Tet2) mutations and demonstrated selective depletion of cohesin-mutant cells with PARP inhibition in vivo. Finally, we demonstrated a shift from STAG2- to STAG1-containing cohesin complexes in cohesin-mutant cells, which was associated with longer DNA loop extrusion, more intermixing of chromatin compartments, and increased interaction with PARP and replication protein A complex. Our findings inform the biology and therapeutic opportunities for cohesin-mutant malignancies.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Reparo do DNA/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Animais , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Feminino , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Camundongos Transgênicos , Síndromes Mielodisplásicas/tratamento farmacológico , Proteínas Nucleares/genética , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto , Coesinas
10.
Nat Cell Biol ; 21(11): 1393-1402, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31685986

RESUMO

Chromosome folding is modulated as cells progress through the cell cycle. During mitosis, condensins fold chromosomes into helical loop arrays. In interphase, the cohesin complex generates loops and topologically associating domains (TADs), while a separate process of compartmentalization drives segregation of active and inactive chromatin. We used synchronized cell cultures to determine how the mitotic chromosome conformation transforms into the interphase state. Using high-throughput chromosome conformation capture (Hi-C) analysis, chromatin binding assays and immunofluorescence, we show that, by telophase, condensin-mediated loops are lost and a transient folding intermediate is formed that is devoid of most loops. By cytokinesis, cohesin-mediated CTCF-CTCF loops and the positions of TADs emerge. Compartment boundaries are also established early, but long-range compartmentalization is a slow process and proceeds for hours after cells enter G1. Our results reveal the kinetics and order of events by which the interphase chromosome state is formed and identify telophase as a critical transition between condensin- and cohesin-driven chromosome folding.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Complexos Multiproteicos/genética , Telófase , Adenosina Trifosfatases/metabolismo , Compartimento Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Cromatina/ultraestrutura , Proteínas Cromossômicas não Histona/metabolismo , Mapeamento Cromossômico , Citocinese/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Células HeLa , Humanos , Interfase , Complexos Multiproteicos/metabolismo , Fase S , Coesinas
11.
Cell Stem Cell ; 25(5): 682-696.e8, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31495782

RESUMO

Transcriptional regulators, including the cohesin complex member STAG2, are recurrently mutated in cancer. The role of STAG2 in gene regulation, hematopoiesis, and tumor suppression remains unresolved. We show that Stag2 deletion in hematopoietic stem and progenitor cells (HSPCs) results in altered hematopoietic function, increased self-renewal, and impaired differentiation. Chromatin immunoprecipitation (ChIP) sequencing revealed that, although Stag2 and Stag1 bind a shared set of genomic loci, a component of Stag2 binding sites is unoccupied by Stag1, even in Stag2-deficient HSPCs. Although concurrent loss of Stag2 and Stag1 abrogated hematopoiesis, Stag2 loss alone decreased chromatin accessibility and transcription of lineage-specification genes, including Ebf1 and Pax5, leading to increased self-renewal and reduced HSPC commitment to the B cell lineage. Our data illustrate a role for Stag2 in transformation and transcriptional dysregulation distinct from its shared role with Stag1 in chromosomal segregation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Autorrenovação Celular/genética , Cromatina/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Linfócitos B/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem da Célula/genética , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica/genética , Técnicas de Inativação de Genes , Células-Tronco Hematopoéticas/citologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Proteínas Nucleares/genética , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX5/metabolismo , RNA-Seq , Mutações Sintéticas Letais/genética , Transativadores/genética , Transativadores/metabolismo
12.
Genome Biol ; 20(1): 57, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30890172

RESUMO

BACKGROUND: Hi-C is currently the most widely used assay to investigate the 3D organization of the genome and to study its role in gene regulation, DNA replication, and disease. However, Hi-C experiments are costly to perform and involve multiple complex experimental steps; thus, accurate methods for measuring the quality and reproducibility of Hi-C data are essential to determine whether the output should be used further in a study. RESULTS: Using real and simulated data, we profile the performance of several recently proposed methods for assessing reproducibility of population Hi-C data, including HiCRep, GenomeDISCO, HiC-Spector, and QuASAR-Rep. By explicitly controlling noise and sparsity through simulations, we demonstrate the deficiencies of performing simple correlation analysis on pairs of matrices, and we show that methods developed specifically for Hi-C data produce better measures of reproducibility. We also show how to use established measures, such as the ratio of intra- to interchromosomal interactions, and novel ones, such as QuASAR-QC, to identify low-quality experiments. CONCLUSIONS: In this work, we assess reproducibility and quality measures by varying sequencing depth, resolution and noise levels in Hi-C data from 13 cell lines, with two biological replicates each, as well as 176 simulated matrices. Through this extensive validation and benchmarking of Hi-C data, we describe best practices for reproducibility and quality assessment of Hi-C experiments. We make all software publicly available at http://github.com/kundajelab/3DChromatin_ReplicateQC to facilitate adoption in the community.


Assuntos
Genômica/normas , Sequenciamento de Nucleotídeos em Larga Escala/normas , Neoplasias/genética , Controle de Qualidade , Software , Humanos , Reprodutibilidade dos Testes , Células Tumorais Cultivadas
13.
Nat Genet ; 50(10): 1388-1398, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30202056

RESUMO

Structural variants (SVs) can contribute to oncogenesis through a variety of mechanisms. Despite their importance, the identification of SVs in cancer genomes remains challenging. Here, we present a framework that integrates optical mapping, high-throughput chromosome conformation capture (Hi-C), and whole-genome sequencing to systematically detect SVs in a variety of normal or cancer samples and cell lines. We identify the unique strengths of each method and demonstrate that only integrative approaches can comprehensively identify SVs in the genome. By combining Hi-C and optical mapping, we resolve complex SVs and phase multiple SV events to a single haplotype. Furthermore, we observe widespread structural variation events affecting the functions of noncoding sequences, including the deletion of distal regulatory sequences, alteration of DNA replication timing, and the creation of novel three-dimensional chromatin structural domains. Our results indicate that noncoding SVs may be underappreciated mutational drivers in cancer genomes.


Assuntos
Genoma Humano , Variação Estrutural do Genoma , Neoplasias/genética , Biologia de Sistemas/métodos , Células A549 , Linhagem Celular Tumoral , Mapeamento Cromossômico , DNA de Neoplasias/análise , DNA de Neoplasias/genética , Genes Neoplásicos , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Células K562 , Desequilíbrio de Ligação , Análise de Sequência de DNA/métodos , Integração de Sistemas
15.
Cell ; 174(1): 172-186.e21, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29958106

RESUMO

The fusion oncoprotein CBFß-SMMHC, expressed in leukemia cases with chromosome 16 inversion, drives leukemia development and maintenance by altering the activity of the transcription factor RUNX1. Here, we demonstrate that CBFß-SMMHC maintains cell viability by neutralizing RUNX1-mediated repression of MYC expression. Upon pharmacologic inhibition of the CBFß-SMMHC/RUNX1 interaction, RUNX1 shows increased binding at three MYC distal enhancers, where it represses MYC expression by mediating the replacement of the SWI/SNF complex component BRG1 with the polycomb-repressive complex component RING1B, leading to apoptosis. Combining the CBFß-SMMHC inhibitor with the BET inhibitor JQ1 eliminates inv(16) leukemia in human cells and a mouse model. Enhancer-interaction analysis indicated that the three enhancers are physically connected with the MYC promoter, and genome-editing analysis demonstrated that they are functionally implicated in deregulation of MYC expression. This study reveals a mechanism whereby CBFß-SMMHC drives leukemia maintenance and suggests that inhibitors targeting chromatin activity may prove effective in inv(16) leukemia therapy.


Assuntos
Apoptose , Cromatina/metabolismo , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Apoptose/efeitos dos fármacos , Azepinas/farmacologia , Azepinas/uso terapêutico , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/metabolismo , Inversão Cromossômica/efeitos dos fármacos , Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , DNA/química , DNA/metabolismo , DNA Helicases/metabolismo , Modelos Animais de Doenças , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Triazóis/uso terapêutico
16.
Nat Methods ; 15(6): 433-436, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29735996

RESUMO

Mapping proteomic composition at distinct genomic loci in living cells has been a long-standing challenge. Here we report that dCas9-APEX2 biotinylation at genomic elements by restricted spatial tagging (C-BERST) allows the rapid, unbiased mapping of proteomes near defined genomic loci, as demonstrated for telomeres and centromeres. C-BERST enables the high-throughput identification of proteins associated with specific sequences, thereby facilitating annotation of these factors and their roles.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Proteômica/métodos , Linhagem Celular Tumoral , Mapeamento Cromossômico , Endonucleases , Regulação da Expressão Gênica , Genoma , Genômica , Humanos , Enzimas Multifuncionais , Engenharia de Proteínas , Proteoma
17.
Cold Spring Harb Protoc ; 2018(4)2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29610357

RESUMO

Formaldehyde cross-linking of DNA to associated proteins is a relatively straightforward method, but it is also the most critical step in the chromatin immunoprecipitation (ChIP) and 3C analyses. Although formaldehyde is a highly permeable cross-linker, its maximum cross-linking efficiencies are estimated to be at ∼1% for mammalian cells because reactivity is limited to amines. Therefore, a relatively large number of cells are required for 3C and ChIP-based assays. Five hundred million cross-linked diploid cells are equivalent to ∼1.66 fmol of the genome. Thus, only ∼100 amol of genomic copies is analyzed for one ChIP assay. Because the quality of cross-linked chromatin can vary, even when generated under near-identical conditions, it is preferable to generate multiple large batches. This protocol describes growing and cross-linking IMR90 primary human fibroblast cells for ChIP analysis. For other cell types, some modification of the protocol is necessary.


Assuntos
Imunoprecipitação da Cromatina/métodos , Reagentes de Ligações Cruzadas/química , Formaldeído/química , Fibroblastos/citologia , Humanos , Temperatura , Fatores de Tempo
18.
Science ; 359(6376)2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29348367

RESUMO

Mitotic chromosomes fold as compact arrays of chromatin loops. To identify the pathway of mitotic chromosome formation, we combined imaging and Hi-C analysis of synchronous DT40 cell cultures with polymer simulations. Here we show that in prophase, the interphase organization is rapidly lost in a condensin-dependent manner, and arrays of consecutive 60-kilobase (kb) loops are formed. During prometaphase, ~80-kb inner loops are nested within ~400-kb outer loops. The loop array acquires a helical arrangement with consecutive loops emanating from a central "spiral staircase" condensin scaffold. The size of helical turns progressively increases to ~12 megabases during prometaphase. Acute depletion of condensin I or II shows that nested loops form by differential action of the two condensins, whereas condensin II is required for helical winding.


Assuntos
Cromossomos/química , Cromossomos/genética , Mitose , Adenosina Trifosfatases/metabolismo , Animais , Linhagem Celular , Biologia Computacional , Proteínas de Ligação a DNA/metabolismo , Genômica , Interfase , Complexos Multiproteicos/metabolismo , Prometáfase , Prófase , Xenopus laevis
19.
Nat Cell Biol ; 19(9): 1071-1080, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28825700

RESUMO

Structural maintenance of chromosomes (SMC) protein complexes are key determinants of chromosome conformation. Using Hi-C and polymer modelling, we study how cohesin and condensin, two deeply conserved SMC complexes, organize chromosomes in the budding yeast Saccharomyces cerevisiae. The canonical role of cohesin is to co-align sister chromatids, while condensin generally compacts mitotic chromosomes. We find strikingly different roles for the two complexes in budding yeast mitosis. First, cohesin is responsible for compacting mitotic chromosome arms, independently of sister chromatid cohesion. Polymer simulations demonstrate that this role can be fully accounted for through cis-looping of chromatin. Second, condensin is generally dispensable for compaction along chromosome arms. Instead, it plays a targeted role compacting the rDNA proximal regions and promoting resolution of peri-centromeric regions. Our results argue that the conserved mechanism of SMC complexes is to form chromatin loops and that distinct SMC-dependent looping activities are selectively deployed to appropriately compact chromosomes.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Estruturas Cromossômicas , Cromossomos Fúngicos/metabolismo , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mitose , Complexos Multiproteicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfatases/genética , Proteínas de Ciclo Celular/genética , Cromatina/química , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Cromossomos Fúngicos/química , Cromossomos Fúngicos/genética , Simulação por Computador , DNA Fúngico/química , DNA Fúngico/genética , DNA Ribossômico/química , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Proteínas de Ligação a DNA/genética , Modelos Genéticos , Modelos Moleculares , Complexos Multiproteicos/genética , Conformação de Ácido Nucleico , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Relação Estrutura-Atividade , Coesinas
20.
Biochim Biophys Acta ; 1859(11): 1389-1397, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27514584

RESUMO

RUNX1 is a transcription factor functioning both as an oncogene and a tumor suppressor in breast cancer. RUNX1 alters chromatin structure in cooperation with chromatin modifier and remodeling enzymes. In this study, we examined the relationship between RUNX1-mediated transcription and genome organization. We characterized genome-wide RUNX1 localization and performed RNA-seq and Hi-C in RUNX1-depleted and control MCF-7 breast cancer cells. RNA-seq analysis showed that RUNX1 depletion led to up-regulation of genes associated with chromatin structure and down-regulation of genes related to extracellular matrix biology, as well as NEAT1 and MALAT1 lncRNAs. Our ChIP-Seq analysis supports a prominent role for RUNX1 in transcriptional activation. About 30% of all RUNX1 binding sites were intergenic, indicating diverse roles in promoter and enhancer regulation and suggesting additional functions for RUNX1. Hi-C analysis of RUNX1-depleted cells demonstrated that overall three-dimensional genome organization is largely intact, but indicated enhanced association of RUNX1 near Topologically Associating Domain (TAD) boundaries and alterations in long-range interactions. These results suggest an architectural role for RUNX1 in fine-tuning local interactions rather than in global organization. Our results provide novel insight into RUNX1-mediated perturbations of higher-order genome organization that are functionally linked with RUNX1-dependent compromised gene expression in breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias da Mama/patologia , Imunoprecipitação da Cromatina , Matriz Extracelular/metabolismo , Feminino , Humanos , Células MCF-7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA