Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.356
Filtrar
1.
J Vis Exp ; (183)2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35604167

RESUMO

Flat cultures of mammalian cells are a widely used in vitro approach for understanding cell physiology, but this system is limited in modeling solid tissues due to unnaturally rapid cell replication. This is particularly challenging when modeling mature chromatin, as fast replicating cells are frequently involved in DNA replication and have a heterogeneous polyploid population. Presented below is a workflow for modeling, treating, and analyzing quiescent chromatin modifications using a three-dimensional (3D) cell culture system. Using this protocol, hepatocellular carcinoma cell lines are grown as reproducible 3D spheroids in an incubator providing active nutrient diffusion and low shearing forces. Treatment with sodium butyrate and sodium succinate induced an increase in histone acetylation and succinylation, respectively. Increases in levels of histone acetylation and succinylation are associated with a more open chromatin state. Spheroids are then collected for isolation of cell nuclei, from which histone proteins are extracted for the analysis of their post-translational modifications. Histone analysis is performed via liquid chromatography coupled online with tandem mass spectrometry, followed by an in-house computational pipeline. Finally, examples of data representation to investigate the frequency and occurrence of combinatorial histone marks are shown.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Histonas , Fígado , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Técnicas de Cultura de Células em Três Dimensões/métodos , Cromatina/fisiologia , Cromatografia Líquida , Histonas/análise , Histonas/metabolismo , Fígado/metabolismo , Mamíferos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Esferoides Celulares/metabolismo
2.
Genes Genet Syst ; 97(1): 3-14, 2022 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-35431282

RESUMO

Epigenetic marks including DNA methylation (DNAme) play a critical role in the transcriptional regulation of genes and retrotransposons. Defects in DNAme are detected in infertility, imprinting disorders and congenital diseases in humans, highlighting the broad importance of this epigenetic mark in both development and disease. While DNAme in terminally differentiated cells is stably propagated following cell division by the maintenance DNAme machinery, widespread erasure and subsequent de novo establishment of this epigenetic mark occur early in embryonic development as well as in germ cell development. Combined with deep sequencing, low-input methods that have been developed in the past several years have enabled high-resolution and genome-wide mapping of both DNAme and histone post-translational modifications (PTMs) in rare cell populations including developing germ cells. Epigenome studies using these novel methods reveal an unprecedented view of the dynamic chromatin landscape during germ cell development. Furthermore, integrative analysis of chromatin marks in normal germ cells and in those deficient in chromatin-modifying enzymes uncovers a critical interplay between histone PTMs and de novo DNAme in the germline. This review discusses work on mechanisms of the erasure and subsequent de novo DNAme in mouse germ cells as well as the outstanding questions relating to the regulation of the dynamic chromatin landscape in germ cells.


Assuntos
Cromatina , Metilação de DNA , Células Germinativas , Animais , Cromatina/genética , Cromatina/metabolismo , Cromatina/fisiologia , Metilação de DNA/fisiologia , Epigênese Genética , Feminino , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Células Germinativas/fisiologia , Histonas/genética , Histonas/metabolismo , Camundongos , Gravidez
3.
Life Sci Alliance ; 5(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35086934

RESUMO

The 3D conformation of the chromatin creates complex networks of noncoding regulatory regions (distal elements) and promoters impacting gene regulation. Despite the importance of the role of noncoding regions in complex diseases, little is known about their interplay within regulatory hubs and implication in multigenic diseases such as schizophrenia. Here we show that cis-regulatory hubs (CRHs) in neurons highlight functional interactions between distal elements and promoters, providing a model to explain epigenetic mechanisms involved in complex diseases. CRHs represent a new 3D model, where distal elements interact to create a complex network of active genes. In a disease context, CRHs highlighted strong enrichments in schizophrenia-associated genes, schizophrenia-associated SNPs, and schizophrenia heritability compared with equivalent structures. Finally, CRHs exhibit larger proportions of genes differentially expressed in schizophrenia compared with promoter-distal element pairs or TADs. CRHs thus capture causal regulatory processes improving the understanding of complex disease etiology such as schizophrenia. These multiple lines of genetic and statistical evidence support CRHs as 3D models to study dysregulation of gene expression in complex diseases more generally.


Assuntos
Biologia Computacional/métodos , Regulação da Expressão Gênica/genética , Herança Multifatorial/genética , Cromatina/genética , Cromatina/fisiologia , Elementos Facilitadores Genéticos/genética , Epigênese Genética/genética , Expressão Gênica/genética , Humanos , Modelos Genéticos , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Esquizofrenia/genética
4.
Cell Rep ; 37(9): 110064, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34852223

RESUMO

CD4+ T cells have a remarkable potential to differentiate into diverse effector lineages following activation. Here, we probe the heterogeneity present among naive CD4+ T cells before encountering their cognate antigen to ask whether their effector potential is modulated by pre-existing transcriptional and chromatin landscape differences. Single-cell RNA sequencing shows that key drivers of variability are genes involved in T cell receptor (TCR) signaling. Using CD5 expression as a readout of the strength of tonic TCR interactions with self-peptide MHC, and sorting on the ends of this self-reactivity spectrum, we find that pre-existing transcriptional differences among naive CD4+ T cells impact follicular helper T (TFH) cell versus non-TFH effector lineage choice. Moreover, our data implicate TCR signal strength during thymic development in establishing differences in naive CD4+ T cell chromatin landscapes that ultimately shape their effector potential.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Cromatina/fisiologia , Ativação Linfocitária/imunologia , Coriomeningite Linfocítica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Feminino , Perfilação da Expressão Gênica , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Masculino , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo
5.
Genes (Basel) ; 12(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34946946

RESUMO

Origins of DNA replication are specified by the ordered recruitment of replication factors in a cell-cycle-dependent manner. The assembly of the pre-replicative complex in G1 and the pre-initiation complex prior to activation in S phase are well characterized; however, the interplay between the assembly of these complexes and the local chromatin environment is less well understood. To investigate the dynamic changes in chromatin organization at and surrounding replication origins, we used micrococcal nuclease (MNase) to generate genome-wide chromatin occupancy profiles of nucleosomes, transcription factors, and replication proteins through consecutive cell cycles in Saccharomyces cerevisiae. During each G1 phase of two consecutive cell cycles, we observed the downstream repositioning of the origin-proximal +1 nucleosome and an increase in protected DNA fragments spanning the ARS consensus sequence (ACS) indicative of pre-RC assembly. We also found that the strongest correlation between chromatin occupancy at the ACS and origin efficiency occurred in early S phase, consistent with the rate-limiting formation of the Cdc45-Mcm2-7-GINS (CMG) complex being a determinant of origin activity. Finally, we observed nucleosome disruption and disorganization emanating from replication origins and traveling with the elongating replication forks across the genome in S phase, likely reflecting the disassembly and assembly of chromatin ahead of and behind the replication fork, respectively. These results provide insights into cell-cycle-regulated chromatin dynamics and how they relate to the regulation of origin activity.


Assuntos
Ciclo Celular/fisiologia , Cromatina/fisiologia , Origem de Replicação/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Divisão Celular , Cromatina/genética , Replicação do DNA/genética , Replicação do DNA/fisiologia , Fase G1 , Nucleossomos/metabolismo , Origem de Replicação/fisiologia , Fase S , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
6.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34819364

RESUMO

Mitotic errors can activate cyclic GMP-AMP synthase (cGAS) and induce type I interferon (IFN) signaling. Current models propose that chromosome segregation errors generate micronuclei whose rupture activates cGAS. We used a panel of antimitotic drugs to perturb mitosis in human fibroblasts and measured abnormal nuclear morphologies, cGAS localization, and IFN signaling in the subsequent interphase. Micronuclei consistently recruited cGAS without activating it. Instead, IFN signaling correlated with formation of cGAS-coated chromatin bridges that were selectively generated by microtubule stabilizers and MPS1 inhibitors. cGAS activation by chromatin bridges was suppressed by drugs that prevented cytokinesis. We confirmed cGAS activation by chromatin bridges in cancer lines that are unable to secrete IFN by measuring paracrine transfer of 2'3'-cGAMP to fibroblasts, and in mouse cells. We propose that cGAS is selectively activated by self-chromatin when it is stretched in chromatin bridges. Immunosurveillance of cells that fail mitosis, and antitumor actions of taxanes and MPS1 inhibitors, may depend on this effect.


Assuntos
Cromatina/fisiologia , Mitose/fisiologia , Nucleotidiltransferases/metabolismo , Linhagem Celular Tumoral , Cromatina/genética , Humanos , Interferon Tipo I/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Micronúcleo Germinativo/genética , Micronúcleo Germinativo/fisiologia , Mitose/efeitos dos fármacos , Mitose/genética , Neoplasias/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/fisiologia , Transdução de Sinais
7.
Biochem Soc Trans ; 49(6): 2727-2736, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34821364

RESUMO

DNA replication ensures the correct copying of the genome and the faithful transfer of the genetic information to the offspring. However, obstacles to replication fork (RF) progression cause RF stalling and compromise efficient genome duplication. Since replication uses the same DNA template as transcription, both transcription and replication must be coordinated to prevent Transcription-Replication Conflicts (TRCs) that could stall RF progression. Several factors contribute to limit the occurrence of such conflicts and their harmful impact on genome integrity. Increasing evidence indicates that chromatin homeostasis plays a key role in the cellular response to TRCs as well as in the preservation of genome integrity. Indeed, chromatin regulating enzymes are frequently mutated in cancer cells, a common characteristic of which is genome instability. Therefore, understanding the role of chromatin in TRC occurrence and resolution may help identify the molecular mechanism by which chromatin protects genome integrity, and the causes and physiological relevance of the high mutation rates of chromatin regulating factors in cancer. Here we review the current knowledge in the field, as well as the perspectives and future applications.


Assuntos
Cromatina/fisiologia , Genoma , Transcrição Gênica/fisiologia , Replicação do DNA
8.
Nat Commun ; 12(1): 6184, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702821

RESUMO

The spatiotemporal organization of chromatin influences many nuclear processes: from chromosome segregation to transcriptional regulation. To get a deeper understanding of these processes, it is essential to go beyond static viewpoints of chromosome structures, to accurately characterize chromatin's diffusion properties. We present GP-FBM: a computational framework based on Gaussian processes and fractional Brownian motion to extract diffusion properties from stochastic trajectories of labeled chromatin loci. GP-FBM uses higher-order temporal correlations present in the data, therefore, outperforming existing methods. Furthermore, GP-FBM allows to interpolate incomplete trajectories and account for substrate movement when two or more particles are present. Using our method, we show that average chromatin diffusion properties are surprisingly similar in interphase and mitosis in mouse embryonic stem cells. We observe surprising heterogeneity in local chromatin dynamics, correlating with potential regulatory activity. We also present GP-Tool, a user-friendly graphical interface to facilitate usage of GP-FBM by the research community.


Assuntos
Cromatina/fisiologia , Modelos Biológicos , Animais , Montagem e Desmontagem da Cromatina , Biologia Computacional , Proteínas de Homeodomínio/genética , Interfase , Camundongos , Mitose , Movimento (Física) , Células-Tronco Embrionárias Murinas , Distribuição Normal
9.
BMB Rep ; 54(10): 489-496, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34488934

RESUMO

Chromatin has highly organized structures in the nucleus, and these higher-order structures are proposed to regulate gene activities and cellular processes. Sequencing-based techniques, such as Hi-C, and fluorescent in situ hybridization (FISH) have revealed a spatial segregation of active and inactive compartments of chromatin, as well as the non-random positioning of chromosomes in the nucleus, respectively. However, regardless of their efficiency in capturing target genomic sites, these techniques are limited to fixed cells. Since chromatin has dynamic structures, live cell imaging techniques are highlighted for their ability to detect conformational changes in chromatin at a specific time point, or to track various arrangements of chromatin through long-term imaging. Given that the imaging approaches to study live cells are dramatically advanced, we recapitulate methods that are widely used to visualize the dynamics of higher-order chromatin structures. [BMB Reports 2021; 54(10): 489-496].


Assuntos
Cromatina/fisiologia , Imageamento Tridimensional/métodos , Imagem Óptica/métodos , Núcleo Celular/metabolismo , Cromatina/ultraestrutura , Cromossomos/metabolismo , Genoma/genética , Humanos , Relação Estrutura-Atividade , Ativação Transcricional/genética
10.
Genes (Basel) ; 12(9)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34573397

RESUMO

The eukaryotic nucleus is continuously being exposed to endogenous and exogenous sources that cause DNA breaks, whose faithful repair requires the activity of dedicated nuclear machineries. DNA is packaged into a variety of chromatin domains, each characterized by specific molecular properties that regulate gene expression and help maintain nuclear structure. These different chromatin environments each demand a tailored response to DNA damage. Silenced chromatin domains in particular present a major challenge to the cell's DNA repair machinery due to their specific biophysical properties and distinct, often repetitive, DNA content. To this end, we here discuss the interplay between silenced chromatin domains and DNA damage repair, specifically double-strand breaks, and how these processes help maintain genome stability.


Assuntos
Cromatina/fisiologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Inativação Gênica/fisiologia , Animais , Núcleo Celular/metabolismo , Cromatina/metabolismo , Reparo do DNA por Junção de Extremidades/genética , Heterocromatina/metabolismo , Humanos
11.
Clin Epigenetics ; 13(1): 175, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34535185

RESUMO

BACKGROUND: Organoids or spheroids have emerged as a physiologically relevant in vitro preclinical model to study patient-specific diseases. A recent study used spheroids of MCF10 cells to model breast cancer progression and identified targetable alterations more similar to those in vivo. Thus, it is practical and essential to explore and characterize the spheroids of the commonly used human breast cancer (BC) cells. METHODS: In this study, we conducted Hi-C analyses in three-dimensional (3D) spheroids of MCF10A, MCF7 and MCF7TR cells and compared TADs and looping genes with those in 2D monolayers. Furthermore, we performed in silico functional analysis on 3D-growth-specific looping genes and to compare patient outcomes with or without endocrinal therapy. Finally, we performed 3C/RT-qPCR validations in 3D spheroids and 3D-FISH confirmations in organoids of breast cancer patient tissues. RESULTS: We found that chromatin structures have experienced drastic changes during the 3D culture growth of BC cells although there is not much change in the quantity of chromatin domains. We also observed that the strengths of looping genes were statistically different between 2D monolayers and 3D spheroids. We further identified novel 3D growth-specific looping genes within Hippo relevant pathways, of which two genes showed potential prognostic values in measuring the outcome of the endocrine treatment. We finally confirmed a few selected genes in Hippo relevant pathways with enhanced looping in organoids of breast cancer patient tissues. CONCLUSIONS: Hence, our work has provided significant insights into our understanding of 3D-growth-specific chromatin architecture in tamoxifen-resistant breast cancer. Our analyses suggest that the strengthened looping-mediated Hippo relevant pathways may contribute to endocrine therapy resistance in breast cancer patients.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cromatina/metabolismo , Disruptores Endócrinos/farmacologia , Adulto , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Cromatina/fisiologia , Metilação de DNA , Disruptores Endócrinos/metabolismo , Disruptores Endócrinos/uso terapêutico , Feminino , Humanos , Pessoa de Meia-Idade , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
12.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-34299075

RESUMO

Organisms' survival is associated with the ability to respond to natural or anthropogenic environmental stressors. Frequently, these responses involve changes in gene regulation and expression, consequently altering physiology, development, or behavior. Here, we present modifications in response to heat exposure that mimics extreme summertime field conditions of lab-cultured and field-conditioned Nematostella vectensis. Using ATAC-seq and RNA-seq data, we found that field-conditioned animals had a more concentrated reaction to short-term thermal stress, expressed as enrichment of the DNA repair mechanism pathway. By contrast, lab animals had a more diffuse reaction that involved a larger number of differentially expressed genes and enriched pathways, including amino acid metabolism. Our results demonstrate that pre-conditioning affects the ability to respond efficiently to heat exposure in terms of both chromatin accessibility and gene expression and reinforces the importance of experimentally addressing ecological questions in the field.


Assuntos
Cromatina/fisiologia , Regulação da Expressão Gênica , Temperatura Alta , Laboratórios/estatística & dados numéricos , Anêmonas-do-Mar/genética , Transcriptoma , Animais , Monitoramento Ambiental , Perfilação da Expressão Gênica , Anêmonas-do-Mar/crescimento & desenvolvimento
13.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-34299232

RESUMO

The genetic architecture of complex traits is multifactorial. Genome-wide association studies (GWASs) have identified risk loci for complex traits and diseases that are disproportionately located at the non-coding regions of the genome. On the other hand, we have just begun to understand the regulatory roles of the non-coding genome, making it challenging to precisely interpret the functions of non-coding variants associated with complex diseases. Additionally, the epigenome plays an active role in mediating cellular responses to fluctuations of sensory or environmental stimuli. However, it remains unclear how exactly non-coding elements associate with epigenetic modifications to regulate gene expression changes and mediate phenotypic outcomes. Therefore, finer interrogations of the human epigenomic landscape in associating with non-coding variants are warranted. Recently, chromatin-profiling techniques have vastly improved our understanding of the numerous functions mediated by the epigenome and DNA structure. Here, we review various chromatin-profiling techniques, such as assays of chromatin accessibility, nucleosome distribution, histone modifications, and chromatin topology, and discuss their applications in unraveling the brain epigenome and etiology of complex traits at tissue homogenate and single-cell resolution. These techniques have elucidated compositional and structural organizing principles of the chromatin environment. Taken together, we believe that high-resolution epigenomic and DNA structure profiling will be one of the best ways to elucidate how non-coding genetic variations impact complex diseases, ultimately allowing us to pinpoint cell-type targets with therapeutic potential.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Cromatina/fisiologia , Sítios de Ligação/genética , Imunoprecipitação da Cromatina/métodos , Epigênese Genética/genética , Epigenoma/genética , Epigenômica/métodos , Regulação da Expressão Gênica/genética , Genoma , Estudo de Associação Genômica Ampla/métodos , Código das Histonas/genética , Humanos , Herança Multifatorial/genética , Nucleossomos/metabolismo , Nucleossomos/fisiologia , Polimorfismo de Nucleotídeo Único/genética , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
14.
J Virol ; 95(19): e0068621, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34232709

RESUMO

During persistent human papillomavirus infection, the viral genome replicates as an extrachromosomal plasmid that is efficiently partitioned to daughter cells during cell division. We have previously shown that an element which overlaps the human papillomavirus 18 (HPV18) transcriptional enhancer promotes stable DNA replication of replicons containing the viral replication origin. Here, we perform comprehensive analyses to elucidate the function of this maintenance element. We conclude that no unique element or binding site in this region is absolutely required for persistent replication and partitioning and instead propose that the overall chromatin architecture of this region is important to promote efficient use of the replication origin. These results have important implications for the genome partitioning mechanism of papillomaviruses. IMPORTANCE Persistent infection with oncogenic human papillomaviruses (HPVs) is responsible for ∼5% of human cancers. The viral DNA replicates as an extrachromosomal plasmid and is partitioned to daughter cells in dividing keratinocytes. Using a complementation assay that allows us to separate viral transcription and replication, we provide insight into viral sequences that are required for long-term replication and persistence in keratinocytes. Understanding how viral genomes replicate persistently for such long periods of time will guide the development of antiviral therapies.


Assuntos
Genoma Viral , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/fisiologia , Sequências Reguladoras de Ácido Nucleico , Replicon/fisiologia , Replicação Viral , Sítios de Ligação , Cromatina/fisiologia , Replicação do DNA , Elementos Facilitadores Genéticos , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiologia , Papillomavirus Humano 31/genética , Papillomavirus Humano 31/fisiologia , Queratinócitos/fisiologia , Queratinócitos/virologia , Plasmídeos , Regiões Promotoras Genéticas , Origem de Replicação , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica
15.
PLoS Genet ; 17(7): e1009691, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34292939

RESUMO

Mammalian genomes are partitioned into sub-megabase to megabase-sized units of preferential interactions called topologically associating domains or TADs, which are likely important for the proper implementation of gene regulatory processes. These domains provide structural scaffolds for distant cis regulatory elements to interact with their target genes within the three-dimensional nuclear space and architectural proteins such as CTCF as well as the cohesin complex participate in the formation of the boundaries between them. However, the importance of the genomic context in providing a given DNA sequence the capacity to act as a boundary element remains to be fully investigated. To address this question, we randomly relocated a topological boundary functionally associated with the mouse HoxD gene cluster and show that it can indeed act similarly outside its initial genomic context. In particular, the relocated DNA segment recruited the required architectural proteins and induced a significant depletion of contacts between genomic regions located across the integration site. The host chromatin landscape was re-organized, with the splitting of the TAD wherein the boundary had integrated. These results provide evidence that topological boundaries can function independently of their site of origin, under physiological conditions during mouse development.


Assuntos
Cromatina/fisiologia , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/fisiologia , Animais , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Proteínas de Ciclo Celular/genética , Cromatina/genética , Montagem e Desmontagem da Cromatina , DNA/genética , Elementos Facilitadores Genéticos/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Redes Reguladoras de Genes/genética , Genoma/genética , Genoma/fisiologia , Genômica/métodos , Camundongos , Camundongos Transgênicos
16.
Mol Cell ; 81(17): 3526-3541.e8, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34186021

RESUMO

BAP1 is mutated or deleted in many cancer types, including mesothelioma, uveal melanoma, and cholangiocarcinoma. It is the catalytic subunit of the PR-DUB complex, which removes PRC1-mediated H2AK119ub1, essential for maintaining transcriptional repression. However, the precise relationship between BAP1 and Polycombs remains elusive. Using embryonic stem cells, we show that BAP1 restricts H2AK119ub1 deposition to Polycomb target sites. This increases the stability of Polycomb with their targets and prevents diffuse accumulation of H2AK119ub1 and H3K27me3. Loss of BAP1 results in a broad increase in H2AK119ub1 levels that is primarily dependent on PCGF3/5-PRC1 complexes. This titrates PRC2 away from its targets and stimulates H3K27me3 accumulation across the genome, leading to a general chromatin compaction. This provides evidence for a unifying model that resolves the apparent contradiction between BAP1 catalytic activity and its role in vivo, uncovering molecular vulnerabilities that could be useful for BAP1-related pathologies.


Assuntos
Cromatina/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Animais , Linhagem Celular/metabolismo , Cromatina/genética , Cromatina/fisiologia , Células-Tronco Embrionárias/metabolismo , Heterocromatina , Histonas/metabolismo , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/fisiologia , Ubiquitinação
17.
Elife ; 102021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33942717

RESUMO

Intact-organism imaging of Drosophila larvae reveals and quantifies chromatin-aqueous phase separation. The chromatin can be organized near the lamina layer of the nuclear envelope, conventionally fill the nucleus, be organized centrally, or as a wetting droplet. These transitions are controlled by changes in nuclear volume and the interaction of chromatin with the lamina (part of the nuclear envelope) at the nuclear periphery. Using a simple polymeric model that includes the key features of chromatin self-attraction and its binding to the lamina, we demonstrate theoretically that it is the competition of these two effects that determines the mode of chromatin distribution. The qualitative trends as well as the composition profiles obtained in our simulations compare well with the observed intact-organism imaging and quantification. Since the simulations contain only a small number of physical variables we can identify the generic mechanisms underlying the changes in the observed phase separations.


Assuntos
Núcleo Celular/fisiologia , Cromatina/fisiologia , Simulação por Computador , Animais , Drosophila , Larva
18.
Circ Res ; 128(7): 887-907, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33793334

RESUMO

Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.


Assuntos
Hipertensão/etiologia , Rim/citologia , Renina/fisiologia , Animais , Arteríolas/embriologia , Pressão Sanguínea/fisiologia , Comunicação Celular , Diferenciação Celular , Plasticidade Celular , Cromatina/fisiologia , Montagem e Desmontagem da Cromatina/fisiologia , Conexinas/fisiologia , Homeostase , Humanos , Integrinas/fisiologia , Sistema Justaglomerular/citologia , Rim/irrigação sanguínea , Rim/embriologia , Glomérulos Renais/fisiologia , Camundongos , MicroRNAs/fisiologia , Fenótipo , Regeneração/fisiologia , Artéria Renal , Renina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Células-Tronco/fisiologia , Equilíbrio Hidroeletrolítico
19.
Dev Biol ; 476: 189-199, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33844976

RESUMO

The three-dimensional (3D) organization of the genome is highly dynamic, changing during development and varying across different tissues and cell types. Recent studies indicate that these changes alter regulatory interactions, leading to changes in gene expression. Despite its importance, the mechanisms that influence genomic organization remain poorly understood. We have previously identified a network of chromatin boundary elements, or insulators, in the Drosophila Antennapedia homeotic complex (ANT-C). These genomic elements interact with one another to tether chromatin loops that could block or promote enhancer-promoter interactions. To understand the function of these insulators, we assessed their interactions by measuring their 3D nuclear distance in developing animal tissues. Our data suggest that the ANT-C Hox complex might be in a folded or looped configuration rather than in a random or extended form. The architecture of the ANT-C complex, as read out by the pair-wise distance between insulators, undergoes a strong compression during late embryogenesis, coinciding with the reduction of cell and nuclear diameters due to continued cell divisions in post-cleavage cells. Our results suggest that genomic architecture and gene regulation may be influenced by cellular morphology and movement during development.


Assuntos
Mapeamento Cromossômico/métodos , Regulação da Expressão Gênica no Desenvolvimento/genética , Genoma/genética , Animais , Cromatina/genética , Cromatina/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Elementos Facilitadores Genéticos/genética , Feminino , Expressão Gênica/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo
20.
Nat Biotechnol ; 39(7): 819-824, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33846646

RESUMO

Methods for quantifying gene expression1 and chromatin accessibility2 in single cells are well established, but single-cell analysis of chromatin regions with specific histone modifications has been technically challenging. In this study, we adapted the CUT&Tag method3 to scalable nanowell and droplet-based single-cell platforms to profile chromatin landscapes in single cells (scCUT&Tag) from complex tissues and during the differentiation of human embryonic stem cells. We focused on profiling polycomb group (PcG) silenced regions marked by histone H3 Lys27 trimethylation (H3K27me3) in single cells as an orthogonal approach to chromatin accessibility for identifying cell states. We show that scCUT&Tag profiling of H3K27me3 distinguishes cell types in human blood and allows the generation of cell-type-specific PcG landscapes from heterogeneous tissues. Furthermore, we used scCUT&Tag to profile H3K27me3 in a patient with a brain tumor before and after treatment, identifying cell types in the tumor microenvironment and heterogeneity in PcG activity in the primary sample and after treatment.


Assuntos
Cromatina/fisiologia , Proteínas do Grupo Polycomb/metabolismo , Análise de Célula Única , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Diferenciação Celular , Cromatina/genética , Células-Tronco Embrionárias , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Células K562 , Proteínas do Grupo Polycomb/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...