Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.020
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Nucleus ; 15(1): 2351957, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38753956

RESUMO

Abnormal cell nuclear shapes are hallmarks of diseases, including progeria, muscular dystrophy, and many cancers. Experiments have shown that disruption of heterochromatin and increases in euchromatin lead to nuclear deformations, such as blebs and ruptures. However, the physical mechanisms through which chromatin governs nuclear shape are poorly understood. To investigate how heterochromatin and euchromatin might govern nuclear morphology, we studied chromatin microphase separation in a composite coarse-grained polymer and elastic shell simulation model. By varying chromatin density, heterochromatin composition, and heterochromatin-lamina interactions, we show how the chromatin phase organization may perturb nuclear shape. Increasing chromatin density stabilizes the lamina against large fluctuations. However, increasing heterochromatin levels or heterochromatin-lamina interactions enhances nuclear shape fluctuations by a "wetting"-like interaction. In contrast, fluctuations are insensitive to heterochromatin's internal structure. Our simulations suggest that peripheral heterochromatin accumulation could perturb nuclear morphology, while nuclear shape stabilization likely occurs through mechanisms other than chromatin microphase organization.


Assuntos
Núcleo Celular , Cromatina , Heterocromatina , Núcleo Celular/metabolismo , Heterocromatina/metabolismo , Heterocromatina/química , Cromatina/metabolismo , Cromatina/química , Polímeros/química , Polímeros/metabolismo , Eucromatina/metabolismo , Eucromatina/química , Humanos , Separação de Fases
2.
Nucleic Acids Res ; 52(9): e45, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38634798

RESUMO

Recent studies have combined DNA methyltransferase footprinting of genomic DNA in nuclei with long-read sequencing, resulting in detailed chromatin maps for multi-kilobase stretches of genomic DNA from one cell. Theoretically, nucleosome footprints and nucleosome-depleted regions can be identified using M.EcoGII, which methylates adenines in any sequence context, providing a high-resolution map of accessible regions in each DNA molecule. Here, we report PacBio long-read sequence data for budding yeast nuclei treated with M.EcoGII and a bioinformatic pipeline which corrects for three key challenges undermining this promising method. First, detection of m6A in individual DNA molecules by the PacBio software is inefficient, resulting in false footprints predicted by random gaps of seemingly unmethylated adenines. Second, there is a strong bias against m6A base calling as AT content increases. Third, occasional methylation occurs within nucleosomes, breaking up their footprints. After correcting for these issues, our pipeline calculates a correlation coefficient-based score indicating the extent of chromatin heterogeneity within the cell population for every gene. Although the population average is consistent with that derived using other techniques, we observe a wide range of heterogeneity in nucleosome positions at the single-molecule level, probably reflecting cellular chromatin dynamics.


Assuntos
Cromatina , Metilação de DNA , Nucleossomos , Análise de Sequência de DNA , Cromatina/metabolismo , Cromatina/genética , Cromatina/química , Nucleossomos/genética , Nucleossomos/metabolismo , Análise de Sequência de DNA/métodos , Software , Genoma Fúngico , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Saccharomycetales/genética , Saccharomycetales/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/genética
3.
Nucleic Acids Res ; 52(7): 3510-3521, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38452220

RESUMO

N6-Methyladenosine (m6A) is the most abundant chemical modification occurring on eukaryotic mRNAs, and has been reported to be involved in almost all stages of mRNA metabolism. The distribution of m6A sites is notably asymmetric along mRNAs, with a strong preference toward the 3' terminus of the transcript. How m6A regional preference is shaped remains incompletely understood. In this study, by performing m6A-seq on chromatin-associated RNAs, we found that m6A regional preference arises during transcription. Nucleosome occupancy is remarkedly increased in the region downstream of m6A sites, suggesting an intricate interplay between m6A methylation and nucleosome-mediated transcriptional dynamics. Notably, we found a remarkable slowdown of Pol-II movement around m6A sites. In addition, inhibiting Pol-II movement increases nearby m6A methylation levels. By analyzing massively parallel assays for m6A, we found that RNA secondary structures inhibit m6A methylation. Remarkably, the m6A sites associated with Pol-II pausing tend to be embedded within RNA secondary structures. These results suggest that Pol-II pausing could affect the accessibility of m6A motifs to the methyltransferase complex and subsequent m6A methylation by mediating RNA secondary structure. Overall, our study reveals a crucial role of transcriptional dynamics in the formation of m6A regional preference.


Assuntos
Adenosina , Adenosina/análogos & derivados , RNA Polimerase II , RNA Mensageiro , Transcrição Gênica , Adenosina/metabolismo , Metilação , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , RNA Polimerase II/metabolismo , Humanos , Conformação de Ácido Nucleico , Nucleossomos/metabolismo , Nucleossomos/genética , Metiltransferases/metabolismo , Metiltransferases/genética , Cromatina/metabolismo , Cromatina/genética , Cromatina/química
4.
Science ; 383(6688): 1215-1222, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38484065

RESUMO

DNA replication is initiated at multiple loci to ensure timely duplication of eukaryotic genomes. Sister replication forks progress bidirectionally, and replication terminates when two convergent forks encounter one another. To investigate the coordination of replication forks, we developed a replication-associated in situ HiC method to capture chromatin interactions involving nascent DNA. We identify more than 2000 fountain-like structures of chromatin contacts in human and mouse genomes, indicative of coupling of DNA replication forks. Replication fork interaction not only occurs between sister forks but also involves forks from two distinct origins to predetermine replication termination. Termination-associated chromatin fountains are sensitive to replication stress and lead to coupled forks-associated genomic deletions in cancers. These findings reveal the spatial organization of DNA replication forks within the chromatin context.


Assuntos
Cromatina , Replicação do DNA , DNA , Genoma Humano , Animais , Humanos , Camundongos , Cromatina/química , DNA/química , DNA/genética , Conformação Proteica , Sequenciamento de Nucleotídeos em Larga Escala
5.
Nature ; 627(8002): 196-203, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38355805

RESUMO

It is well established that neutrophils adopt malleable polymorphonuclear shapes to migrate through narrow interstitial tissue spaces1-3. However, how polymorphonuclear structures are assembled remains unknown4. Here we show that in neutrophil progenitors, halting loop extrusion-a motor-powered process that generates DNA loops by pulling in chromatin5-leads to the assembly of polymorphonuclear genomes. Specifically, we found that in mononuclear neutrophil progenitors, acute depletion of the loop-extrusion loading factor nipped-B-like protein (NIPBL) induced the assembly of horseshoe, banded, ringed and hypersegmented nuclear structures and led to a reduction in nuclear volume, mirroring what is observed during the differentiation of neutrophils. Depletion of NIPBL also induced cell-cycle arrest, activated a neutrophil-specific gene program and conditioned a loss of interactions across topologically associating domains to generate a chromatin architecture that resembled that of differentiated neutrophils. Removing NIPBL resulted in enrichment for mega-loops and interchromosomal hubs that contain genes associated with neutrophil-specific enhancer repertoires and an inflammatory gene program. On the basis of these observations, we propose that in neutrophil progenitors, loop-extrusion programs produce lineage-specific chromatin architectures that permit the packing of chromosomes into geometrically confined lobular structures. Our data also provide a blueprint for the assembly of polymorphonuclear structures, and point to the possibility of engineering de novo nuclear shapes to facilitate the migration of effector cells in densely populated tumorigenic environments.


Assuntos
Movimento Celular , Forma do Núcleo Celular , Neutrófilos , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatina/metabolismo , Cromossomos/química , Cromossomos/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Conformação de Ácido Nucleico , Diferenciação Celular/genética , Inflamação/genética , Elementos Facilitadores Genéticos , Linhagem da Célula/genética
6.
J Mol Biol ; 436(7): 168318, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37863247

RESUMO

Within the SET domain superfamily of lysine methyltransferases, there is a well-conserved subfamily, frequently referred to as the Set3 SET domain subfamily, which contain noncanonical SET domains carrying divergent amino acid sequences. These proteins are implicated in diverse biological processes including stress responses, cell differentiation, and development, and their disruption is linked to diseases including cancer and neurodevelopmental disorders. Interestingly, biochemical and structural analysis indicates that they do not possess catalytic methyltransferase activity. At the molecular level, Set3 SET domain proteins appear to play critical roles in the regulation of gene expression, particularly repression and heterochromatin maintenance, and in some cases, via scaffolding other histone modifying activities at chromatin. Here, we explore the common and unique functions among Set3 SET domain subfamily proteins and analyze what is known about the specific contribution of the conserved SET domain to functional roles of these proteins, as well as propose areas of investigation to improve understanding of this important, noncanonical subfamily of proteins.


Assuntos
Histona-Lisina N-Metiltransferase , Domínios PR-SET , Sequência de Aminoácidos , Cromatina/química , Cromatina/metabolismo , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Animais
7.
Nucleic Acids Res ; 51(20): 11080-11103, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37823591

RESUMO

Chromatin association of the BRCA1-BARD1 heterodimer is critical to promote homologous recombination repair of DNA double-strand breaks (DSBs) in S/G2. How the BRCA1-BARD1 complex interacts with chromatin that contains both damage induced histone H2A ubiquitin and inhibitory H4K20 methylation is not fully understood. We characterised BRCA1-BARD1 binding and enzymatic activity to an array of mono- and di-nucleosome substrates using biochemical, structural and single molecule imaging approaches. We found that the BRCA1-BARD1 complex preferentially interacts and modifies di-nucleosomes over mono-nucleosomes, allowing integration of H2A Lys-15 ubiquitylation signals with other chromatin modifications and features. Using high speed- atomic force microscopy (HS-AFM) to monitor how the BRCA1-BARD1 complex recognises chromatin in real time, we saw a highly dynamic complex that bridges two nucleosomes and associates with the DNA linker region. Bridging is aided by multivalent cross-nucleosome interactions that enhance BRCA1-BARD1 E3 ubiquitin ligase catalytic activity. Multivalent interactions across nucleosomes explain how BRCA1-BARD1 can recognise chromatin that retains partial di-methylation at H4 Lys-20 (H4K20me2), a parental histone mark that blocks BRCA1-BARD1 interaction with nucleosomes, to promote its enzymatic and DNA repair activities.


Assuntos
Proteína BRCA1 , Cromatina , Nucleossomos , Ubiquitina-Proteína Ligases , Humanos , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Cromatina/química , Cromatina/metabolismo , Células HeLa , Histonas/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
8.
Nature ; 622(7981): 173-179, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37731000

RESUMO

Lysine residues in histones and other proteins can be modified by post-translational modifications that encode regulatory information1. Lysine acetylation and methylation are especially important for regulating chromatin and gene expression2-4. Pathways involving these post-translational modifications are targets for clinically approved therapeutics to treat human diseases. Lysine methylation and acetylation are generally assumed to be mutually exclusive at the same residue. Here we report cellular lysine residues that are both methylated and acetylated on the same side chain to form Nε-acetyl-Nε-methyllysine (Kacme). We show that Kacme is found on histone H4 (H4Kacme) across a range of species and across mammalian tissues. Kacme is associated with marks of active chromatin, increased transcriptional initiation and is regulated in response to biological signals. H4Kacme can be installed by enzymatic acetylation of monomethyllysine peptides and is resistant to deacetylation by some HDACs in vitro. Kacme can be bound by chromatin proteins that recognize modified lysine residues, as we demonstrate with the crystal structure of acetyllysine-binding protein BRD2 bound to a histone H4Kacme peptide. These results establish Kacme as a cellular post-translational modification with the potential to encode information distinct from methylation and acetylation alone and demonstrate that Kacme has all the hallmarks of a post-translational modification with fundamental importance to chromatin biology.


Assuntos
Acetilação , Cromatina , Lisina , Metilação , Processamento de Proteína Pós-Traducional , Sítio de Iniciação de Transcrição , Animais , Humanos , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Histona Desacetilases/metabolismo
9.
J Mol Biol ; 435(11): 168019, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37330288

RESUMO

All life forms sense and respond to mechanical stimuli. Throughout evolution, organisms develop diverse mechanosensing and mechanotransduction pathways, leading to fast and sustained mechanoresponses. Memory and plasticity characteristics of mechanoresponses are thought to be stored in the form of epigenetic modifications, including chromatin structure alterations. These mechanoresponses in the chromatin context share conserved principles across species, such as lateral inhibition during organogenesis and development. However, it remains unclear how mechanotransduction mechanisms alter chromatin structure for specific cellular functions, and if altered chromatin structure can mechanically affect the environment. In this review, we discuss how chromatin structure is altered by environmental forces via an outside-in pathway for cellular functions, and the emerging concept of how chromatin structure alterations can mechanically affect nuclear, cellular, and extracellular environments. This bidirectional mechanical feedback between chromatin of the cell and the environment can potentially have important physiological implications, such as in centromeric chromatin regulation of mechanobiology in mitosis, or in tumor-stroma interactions. Finally, we highlight the current challenges and open questions in the field and provide perspectives for future research.


Assuntos
Cromatina , Epigenoma , Mecanotransdução Celular , Mitose , Cromatina/química , Cromatina/genética , Mecanotransdução Celular/genética , Humanos , Neoplasias , Células Estromais
10.
Science ; 381(6655): 313-319, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37384673

RESUMO

Loss of H2A-H2B histone dimers is a hallmark of actively transcribed genes, but how the cellular machinery functions in the context of noncanonical nucleosomal particles remains largely elusive. In this work, we report the structural mechanism for adenosine 5'-triphosphate-dependent chromatin remodeling of hexasomes by the INO80 complex. We show how INO80 recognizes noncanonical DNA and histone features of hexasomes that emerge from the loss of H2A-H2B. A large structural rearrangement switches the catalytic core of INO80 into a distinct, spin-rotated mode of remodeling while its nuclear actin module remains tethered to long stretches of unwrapped linker DNA. Direct sensing of an exposed H3-H4 histone interface activates INO80, independently of the H2A-H2B acidic patch. Our findings reveal how the loss of H2A-H2B grants remodelers access to a different, yet unexplored layer of energy-driven chromatin regulation.


Assuntos
Chaetomium , Montagem e Desmontagem da Cromatina , Cromatina , Histonas , Nucleossomos , Cromatina/química , DNA/química , Histonas/química , Nucleossomos/química , Microscopia Crioeletrônica , Chaetomium/química , Chaetomium/ultraestrutura
11.
Nature ; 617(7962): 818-826, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37198486

RESUMO

Cancer cells rewire metabolism to favour the generation of specialized metabolites that support tumour growth and reshape the tumour microenvironment1,2. Lysine functions as a biosynthetic molecule, energy source and antioxidant3-5, but little is known about its pathological role in cancer. Here we show that glioblastoma stem cells (GSCs) reprogram lysine catabolism through the upregulation of lysine transporter SLC7A2 and crotonyl-coenzyme A (crotonyl-CoA)-producing enzyme glutaryl-CoA dehydrogenase (GCDH) with downregulation of the crotonyl-CoA hydratase enoyl-CoA hydratase short chain 1 (ECHS1), leading to accumulation of intracellular crotonyl-CoA and histone H4 lysine crotonylation. A reduction in histone lysine crotonylation by either genetic manipulation or lysine restriction impaired tumour growth. In the nucleus, GCDH interacts with the crotonyltransferase CBP to promote histone lysine crotonylation. Loss of histone lysine crotonylation promotes immunogenic cytosolic double-stranded RNA (dsRNA) and dsDNA generation through enhanced H3K27ac, which stimulates the RNA sensor MDA5 and DNA sensor cyclic GMP-AMP synthase (cGAS) to boost type I interferon signalling, leading to compromised GSC tumorigenic potential and elevated CD8+ T cell infiltration. A lysine-restricted diet synergized with MYC inhibition or anti-PD-1 therapy to slow tumour growth. Collectively, GSCs co-opt lysine uptake and degradation to shunt the production of crotonyl-CoA, remodelling the chromatin landscape to evade interferon-induced intrinsic effects on GSC maintenance and extrinsic effects on immune response.


Assuntos
Histonas , Lisina , Neoplasias , Processamento de Proteína Pós-Traducional , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Glutaril-CoA Desidrogenase/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/deficiência , Lisina/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , RNA de Cadeia Dupla/imunologia , Humanos , Animais , Camundongos , Interferon Tipo I/imunologia
12.
Nature ; 616(7957): 574-580, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37020029

RESUMO

Interactions between biomolecules underlie all cellular processes and ultimately control cell fate. Perturbation of native interactions through mutation, changes in expression levels or external stimuli leads to altered cellular physiology and can result in either disease or therapeutic effects1,2. Mapping these interactions and determining how they respond to stimulus is the genesis of many drug development efforts, leading to new therapeutic targets and improvements in human health1. However, in the complex environment of the nucleus, it is challenging to determine protein-protein interactions owing to low abundance, transient or multivalent binding and a lack of technologies that are able to interrogate these interactions without disrupting the protein-binding surface under study3. Here, we describe a method for the traceless incorporation of iridium-photosensitizers into the nuclear micro-environment using engineered split inteins. These Ir-catalysts can activate diazirine warheads through Dexter energy transfer to form reactive carbenes within an approximately 10 nm radius, cross-linking with proteins in the immediate micro-environment (a process termed µMap) for analysis using quantitative chemoproteomics4. We show that this nanoscale proximity-labelling method can reveal the critical changes in interactomes in the presence of cancer-associated mutations, as well as treatment with small-molecule inhibitors. µMap improves our fundamental understanding of nuclear protein-protein interactions and, in doing so, is expected to have a significant effect on the field of epigenetic drug discovery in both academia and industry.


Assuntos
Núcleo Celular , Cromatina , Reagentes de Ligações Cruzadas , Humanos , Núcleo Celular/química , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Reagentes de Ligações Cruzadas/análise , Reagentes de Ligações Cruzadas/química , Transferência de Energia , Epigenômica , Inteínas , Irídio , Mutação , Neoplasias/genética , Fármacos Fotossensibilizantes , Ligação Proteica , Mapas de Interação de Proteínas
13.
J Mol Biol ; 435(8): 168033, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-36858171

RESUMO

The nuclear protein 1 (NUPR1) is an intrinsically disordered protein involved in stress-mediated cellular conditions. Its paralogue nuclear protein 1-like (NUPR1L) is p53-regulated, and its expression down-regulates that of the NUPR1 gene. Peptidyl-arginine deiminase 4 (PADI4) is an isoform of a family of enzymes catalyzing arginine to citrulline conversion; it is also involved in stress-mediated cellular conditions. We characterized the interaction between NUPR1 and PADI4 in vitro, in silico, and in cellulo. The interaction of NUPR1 and PADI4 occurred with a dissociation constant of 18 ± 6 µM. The binding region of NUPR1, mapped by NMR, was a hydrophobic polypeptide patch surrounding the key residue Ala33, as pinpointed by: (i) computational results; and, (ii) site-directed mutagenesis of residues of NUPR1. The association between PADI4 and wild-type NUPR1 was also assessed in cellulo by using proximity ligation assays (PLAs) and immunofluorescence (IF), and it occurred mainly in the nucleus. Moreover, binding between NUPR1L and PADI4 also occurred in vitro with an affinity similar to that of NUPR1. Molecular modelling provided information on the binding hot spot for PADI4. This is an example of a disordered partner of PADI4, whereas its other known interacting proteins are well-folded. Altogether, our results suggest that the NUPR1/PADI4 complex could have crucial functions in modulating DNA-repair, favoring metastasis, or facilitating citrullination of other proteins.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Cromatina , Proteínas Intrinsicamente Desordenadas , Proteínas de Neoplasias , Proteínas Nucleares , Proteína-Arginina Desiminase do Tipo 4 , Sequência de Bases , Cromatina/química , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética
14.
J Mol Biol ; 435(16): 167889, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-36402224

RESUMO

p53 exerts its tumour suppressor activity by modulating hundreds of genes and it can also repress viral replication. Such is the case of human papillomavirus (HPV) through targeting the E2 master regulator, but the biochemical mechanism is not known. We show that the C-terminal DNA binding domain of HPV16 E2 protein (E2C) triggers heterotypic condensation with p53 at a precise 2/1 E2C/p53 stoichiometry at the onset for demixing, yielding large regular spherical droplets that increase in size with E2C concentration. Interestingly, transfection experiments show that E2 co-localizes with p53 in the nucleus with a grainy pattern, and recruits p53 to chromatin-associated foci, a function independent of the DNA binding capacity of p53 as judged by a DNA binding impaired mutant. Depending on the length, DNA can either completely dissolve or reshape heterotypic droplets into irregular condensates containing p53, E2C, and DNA, and reminiscent of that observed linked to chromatin. We propose that p53 is a scaffold for condensation in line with its structural and functional features, in particular as a promiscuous hub that binds multiple cellular proteins. E2 appears as both client and modulator, likely based on its homodimeric DNA binding nature. Our results, in line with the known role of condensation in eukaryotic gene enhancement and silencing, point at biomolecular condensation of E2 with p53 as a means to modulate HPV gene function, strictly dependent on host cell replication and transcription machinery.


Assuntos
Condensados Biomoleculares , Replicação do DNA , Proteínas de Ligação a DNA , Papillomavirus Humano 16 , Proteínas Oncogênicas Virais , Proteína Supressora de Tumor p53 , Replicação Viral , Humanos , Linhagem Celular Tumoral , Cromatina/química , Cromatina/metabolismo , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Papillomavirus Humano 16/fisiologia , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Infecções por Papillomavirus/virologia , Domínios Proteicos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Replicação Viral/fisiologia , Condensados Biomoleculares/metabolismo , Condensados Biomoleculares/virologia
15.
Elife ; 112022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-36196991

RESUMO

Chromosome segregation requires both the separation of sister chromatids and the sustained condensation of chromatids during anaphase. In yeast cells, cohesin is not only required for sister chromatid cohesion but also plays a major role determining the structure of individual chromatids in metaphase. Separase cleavage is thought to remove all cohesin complexes from chromosomes to initiate anaphase. It is thus not clear how the length and organisation of segregating chromatids is maintained during anaphase in the absence of cohesin. Here, we show that degradation of cohesin at the anaphase onset causes aberrant chromatid segregation. Hi-C analysis on segregating chromatids demonstrates that cohesin depletion causes loss of intrachromatid organisation. Surprisingly, tobacco etch virus (TEV)-mediated cleavage of cohesin does not dramatically disrupt chromatid organisation in anaphase, explaining why bulk segregation is achieved. In addition, we identified a small pool of cohesin complexes bound to telophase chromosomes in wild-type cells and show that they play a role in the organisation of centromeric regions. Our data demonstrates that in yeast cells cohesin function is not over in metaphase, but extends to the anaphase period when chromatids are segregating.


Assuntos
Proteínas de Ciclo Celular , Cromatina , Proteínas Cromossômicas não Histona , Saccharomyces cerevisiae , Anáfase/genética , Cromátides , Cromatina/química , Cromatina/metabolismo , Saccharomyces cerevisiae/genética , Separase/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Coesinas
16.
Nature ; 609(7929): 1048-1055, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36104563

RESUMO

Telomeres, the ends of eukaryotic chromosomes, play pivotal parts in ageing and cancer and are targets of DNA damage and the DNA damage response1-5. Little is known about the structure of telomeric chromatin at the molecular level. Here we used negative stain electron microscopy and single-molecule magnetic tweezers to characterize 3-kbp-long telomeric chromatin fibres. We also obtained the cryogenic electron microscopy structure of the condensed telomeric tetranucleosome and its dinucleosome unit. The structure displayed close stacking of nucleosomes with a columnar arrangement, and an unusually short nucleosome repeat  length that comprised about 132 bp DNA wound in a continuous superhelix around histone octamers. This columnar structure is primarily stabilized by the H2A carboxy-terminal and histone amino-terminal tails in a synergistic manner. The columnar conformation results in exposure of the DNA helix, which may make it susceptible to both DNA damage and the DNA damage response. The conformation also exists in an alternative open state, in which one nucleosome is unstacked and flipped out, which exposes the acidic patch of the histone surface. The structural features revealed in this work suggest mechanisms by which protein factors involved in telomere maintenance can access telomeric chromatin in its compact form.


Assuntos
Cromatina , DNA , Histonas , Conformação Molecular , Telômero , Cromatina/química , Cromatina/genética , Cromatina/ultraestrutura , DNA/química , DNA/metabolismo , DNA/ultraestrutura , Dano ao DNA , Histonas/química , Histonas/metabolismo , Histonas/ultraestrutura , Humanos , Microscopia Eletrônica , Nucleossomos/química , Nucleossomos/genética , Nucleossomos/ultraestrutura , Imagem Individual de Molécula , Telômero/química , Telômero/genética , Telômero/ultraestrutura
17.
Science ; 377(6609): 967-975, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-36007034

RESUMO

Chromosome engineering has been attempted successfully in yeast but remains challenging in higher eukaryotes, including mammals. Here, we report programmed chromosome ligation in mice that resulted in the creation of new karyotypes in the lab. Using haploid embryonic stem cells and gene editing, we fused the two largest mouse chromosomes, chromosomes 1 and 2, and two medium-size chromosomes, chromosomes 4 and 5. Chromatin conformation and stem cell differentiation were minimally affected. However, karyotypes carrying fused chromosomes 1 and 2 resulted in arrested mitosis, polyploidization, and embryonic lethality, whereas a smaller fused chromosome composed of chromosomes 4 and 5 was able to be passed on to homozygous offspring. Our results suggest the feasibility of chromosome-level engineering in mammals.


Assuntos
Fusão Gênica Artificial , Edição de Genes , Cariótipo , Translocação Genética , Animais , Fusão Gênica Artificial/métodos , Cromatina/química , Células-Tronco Embrionárias , Edição de Genes/métodos , Haploidia , Camundongos , Mitose
18.
J Mol Biol ; 434(19): 167756, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35870648

RESUMO

Histone chaperones, which constitute an interaction and functional network involved in all aspects of histone metabolism, have to date been identified only in eukaryotes. The Epstein-Barr virus tegument protein BKRF4 is a histone-binding protein that engages histones H2A-H2B and H3-H4, and cellular chromatin, inhibiting the host DNA damage response. Here, we identified BKRF4 as a bona fide viral histone chaperone whose histone-binding domain (HBD) forms a co-chaperone complex with the human histone chaperone ASF1 in vitro. We determined the crystal structures of the quaternary complex of the BKRF4 HBD with human H3-H4 dimer and the histone chaperone ASF1b and the ternary complex of the BKRF4 HBD with human H2A-H2B dimer. Through structural and biochemical studies, we elucidated the molecular basis for H3-H4 and H2A-H2B recognition by BKRF4. We also revealed two conserved motifs, D/EL and DEF/Y/W, within the BKRF4 HBD, which may represent common motifs through which histone chaperones target H3-H4 and H2A-H2B, respectively. In conclusion, our results identify BKRF4 as a histone chaperone encoded by the Epstein-Barr virus, representing a typical histone chaperone found in a non-eukaryote. We envision that more histone chaperones await identification and characterization in DNA viruses and even archaea.


Assuntos
Proteínas do Capsídeo , Proteínas de Ciclo Celular , Herpesvirus Humano 4 , Chaperonas de Histonas , Proteínas do Capsídeo/química , Proteínas de Ciclo Celular/química , Cromatina/química , Herpesvirus Humano 4/genética , Chaperonas de Histonas/química , Histonas/metabolismo , Humanos , Ligação Proteica , Conformação Proteica
19.
FEBS J ; 289(5): 1156-1159, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35233949

RESUMO

Over the past few decades, epigenetic regulators have emerged as major players in cellular processes that drive cancer initiation and progression, and subsequently modulate the responsiveness of cancers to therapeutic agents. This Special Issue of The FEBS Journal, Cancer Epigenetics, features an exciting collection of review articles that focus on the functions of a broad spectrum of epigenetic modulators in cancer. The diverse topics explored herein range from the roles of transposable elements and chromatin architecture in cancer and the most recent research advances on cancer-associated histone variants (oncohistones), to the effects of altered epigenetics on transcription and advanced cancer cell phenotypes. Moreover, the prospective key function of cancer metabolism in linking epigenetics and transcriptional regulation, and the potential of epigenetics for targeted cancer therapeutics is discussed. We hope that this collection of articles will give readers an enlightening overview of the most recent advances in the fast-moving field of cancer epigenetics.


Assuntos
Metilação de DNA , Epigênese Genética , Histonas/genética , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapêutico , Cromatina/química , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Elementos de DNA Transponíveis , Progressão da Doença , Histonas/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/diagnóstico , Neoplasias/patologia , Fenótipo , Transcrição Gênica , Resultado do Tratamento
20.
Nat Genet ; 54(2): 170-179, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115686

RESUMO

Inherited noncoding genetic variants confer significant disease susceptibility to childhood acute lymphoblastic leukemia (ALL) but the molecular processes linking germline polymorphisms with somatic lesions in this cancer are poorly understood. Through targeted sequencing in 5,008 patients, we identified a key regulatory germline variant in GATA3 associated with Philadelphia chromosome-like ALL (Ph-like ALL). Using CRISPR-Cas9 editing and samples from patients with Ph-like ALL, we showed that this variant activated a strong enhancer that upregulated GATA3 transcription. This, in turn, reshaped global chromatin accessibility and three-dimensional genome organization, including regions proximal to the ALL oncogene CRLF2. Finally, we showed that GATA3 directly regulated CRLF2 and potentiated the JAK-STAT oncogenic effects during leukemogenesis. Taken together, we provide evidence for a distinct mechanism by which a germline noncoding variant contributes to oncogene activation, epigenetic regulation and three-dimensional genome reprogramming.


Assuntos
Cromatina/química , Fator de Transcrição GATA3/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Criança , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Feminino , Fator de Transcrição GATA3/metabolismo , Genoma Humano , Humanos , Janus Quinases/metabolismo , Masculino , Oncogenes , Cromossomo Filadélfia , Ligação Proteica , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA