Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 587(7834): 377-386, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32894860

RESUMO

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Atenção à Saúde/métodos , Atenção à Saúde/tendências , Medicina/métodos , Medicina/tendências , Patologia , Análise de Célula Única , Inteligência Artificial , Atenção à Saúde/ética , Atenção à Saúde/normas , Diagnóstico Precoce , Educação Médica , Europa (Continente) , Feminino , Saúde , Humanos , Legislação Médica , Masculino , Medicina/normas
2.
Nat Methods ; 17(3): 279-282, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32066961

RESUMO

We introduce an engineered nanobody whose affinity to green fluorescent protein (GFP) can be switched on and off with small molecules. By controlling the cellular localization of GFP fusion proteins, the engineered nanobody allows interrogation of their roles in basic biological processes, an approach that should be applicable to numerous previously described GFP fusions. We also outline how the binding affinities of other nanobodies can be controlled by small molecules.


Assuntos
Proteínas de Fluorescência Verde/química , Fragmentos de Imunoglobulinas/química , Nanopartículas/química , Anticorpos de Domínio Único/química , Cristalografia por Raios X , DNA/química , Bases de Dados de Proteínas , Escherichia coli , Transferência Ressonante de Energia de Fluorescência , Produtos do Gene gag/química , Células HEK293 , HIV-1/química , Células HeLa , Humanos , Cinética , Ligantes , Microscopia de Fluorescência , Mitose , Domínios Proteicos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química
3.
Elife ; 82019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31204999

RESUMO

The organisation of mammalian genomes into loops and topologically associating domains (TADs) contributes to chromatin structure, gene expression and recombination. TADs and many loops are formed by cohesin and positioned by CTCF. In proliferating cells, cohesin also mediates sister chromatid cohesion, which is essential for chromosome segregation. Current models of chromatin folding and cohesion are based on assumptions of how many cohesin and CTCF molecules organise the genome. Here we have measured absolute copy numbers and dynamics of cohesin, CTCF, NIPBL, WAPL and sororin by mass spectrometry, fluorescence-correlation spectroscopy and fluorescence recovery after photobleaching in HeLa cells. In G1-phase, there are ~250,000 nuclear cohesin complexes, of which ~ 160,000 are chromatin-bound. Comparison with chromatin immunoprecipitation-sequencing data implies that some genomic cohesin and CTCF enrichment sites are unoccupied in single cells at any one time. We discuss the implications of these findings for how cohesin can contribute to genome organisation and cohesion.


Assuntos
Fator de Ligação a CCCTC/genética , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Dosagem de Genes , Expressão Gênica , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Fator de Ligação a CCCTC/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cromátides/genética , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos/genética , Recuperação de Fluorescência Após Fotodegradação/métodos , Fase G1/genética , Genoma Humano/genética , Células HeLa , Humanos , Espectrometria de Massas/métodos , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Coesinas
4.
J Cell Biol ; 217(7): 2309-2328, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29632028

RESUMO

The two Condensin complexes in human cells are essential for mitotic chromosome structure. We used homozygous genome editing to fluorescently tag Condensin I and II subunits and mapped their absolute abundance, spacing, and dynamic localization during mitosis by fluorescence correlation spectroscopy (FSC)-calibrated live-cell imaging and superresolution microscopy. Although ∼35,000 Condensin II complexes are stably bound to chromosomes throughout mitosis, ∼195,000 Condensin I complexes dynamically bind in two steps: prometaphase and early anaphase. The two Condensins rarely colocalize at the chromatid axis, where Condensin II is centrally confined, but Condensin I reaches ∼50% of the chromatid diameter from its center. Based on our comprehensive quantitative data, we propose a three-step hierarchical loop model of mitotic chromosome compaction: Condensin II initially fixes loops of a maximum size of ∼450 kb at the chromatid axis, whose size is then reduced by Condensin I binding to ∼90 kb in prometaphase and ∼70 kb in anaphase, achieving maximum chromosome compaction upon sister chromatid segregation.


Assuntos
Adenosina Trifosfatases/genética , Segregação de Cromossomos/genética , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Mitose/genética , Complexos Multiproteicos/genética , Anáfase/genética , Cromátides/genética , Edição de Genes , Humanos
5.
EMBO J ; 36(24): 3573-3599, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29217591

RESUMO

Mammalian genomes are spatially organized into compartments, topologically associating domains (TADs), and loops to facilitate gene regulation and other chromosomal functions. How compartments, TADs, and loops are generated is unknown. It has been proposed that cohesin forms TADs and loops by extruding chromatin loops until it encounters CTCF, but direct evidence for this hypothesis is missing. Here, we show that cohesin suppresses compartments but is required for TADs and loops, that CTCF defines their boundaries, and that the cohesin unloading factor WAPL and its PDS5 binding partners control the length of loops. In the absence of WAPL and PDS5 proteins, cohesin forms extended loops, presumably by passing CTCF sites, accumulates in axial chromosomal positions (vermicelli), and condenses chromosomes. Unexpectedly, PDS5 proteins are also required for boundary function. These results show that cohesin has an essential genome-wide function in mediating long-range chromatin interactions and support the hypothesis that cohesin creates these by loop extrusion, until it is delayed by CTCF in a manner dependent on PDS5 proteins, or until it is released from DNA by WAPL.


Assuntos
Fator de Ligação a CCCTC/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Fator de Ligação a CCCTC/genética , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Genoma Humano/genética , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Coesinas
6.
Biophys J ; 113(7): 1383-1394, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28978433

RESUMO

Genome dynamics are intimately linked to the regulation of gene expression, the most fundamental mechanism in biology, yet we still do not know whether the very process of transcription drives spatial organization at specific gene loci. Here, we have optimized the ANCHOR/ParB DNA-labeling system for real-time imaging of a single-copy, estrogen-inducible transgene in human cells. Motion of an ANCHOR3-tagged DNA locus was recorded in the same cell before and during the appearance of nascent MS2-labeled mRNA. We found that transcription initiation by RNA polymerase 2 resulted in confinement of the mRNA-producing gene domain within minutes. Transcription-induced confinement occurred in each single cell independently of initial, highly heterogeneous mobility. Constrained mobility was maintained even when inhibiting polymerase elongation. Chromatin motion at constant step size within a largely confined area hence leads to increased collisions that are compatible with the formation of gene-specific chromatin domains, and reflect the assembly of functional protein hubs and DNA processing during the rate-limiting steps of transcription.


Assuntos
Ciclina D1/biossíntese , Transcrição Gênica , Linhagem Celular Tumoral , Cromatina/metabolismo , Ciclina D1/genética , Recuperação de Fluorescência Após Fotodegradação , Loci Gênicos , Humanos , Microscopia de Fluorescência , Imagem Molecular , Movimento (Física) , RNA Polimerase II/metabolismo , RNA Mensageiro/biossíntese , Espectrometria de Fluorescência , Transfecção , Transgenes
7.
Nat Commun ; 7: 13887, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27976684

RESUMO

Genome integrity relies on precise coordination between DNA replication and chromosome segregation. Whereas replication stress attracted much attention, the consequences of mitotic perturbations for genome integrity are less understood. Here, we knockdown 47 validated mitotic regulators to show that a broad spectrum of mitotic errors correlates with increased DNA breakage in daughter cells. Unexpectedly, we find that only a subset of these correlations are functionally linked. We identify the genuine mitosis-born DNA damage events and sub-classify them according to penetrance of the observed phenotypes. To demonstrate the potential of this resource, we show that DNA breakage after cytokinesis failure is preceded by replication stress, which mounts during consecutive cell cycles and coincides with decreased proliferation. Together, our results provide a resource to gauge the magnitude and dynamics of DNA breakage associated with mitotic aberrations and suggest that replication stress might limit propagation of cells with abnormal karyotypes.


Assuntos
Ciclo Celular , Proliferação de Células , Dano ao DNA/genética , Mitose/genética , Linhagem Celular Tumoral , Citocinese/genética , Quebras de DNA , Técnicas de Silenciamento de Genes , Humanos , Processamento de Imagem Assistida por Computador , Microscopia Confocal , Fenótipo , Imagem com Lapso de Tempo
8.
Nat Cell Biol ; 18(6): 692-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27136266

RESUMO

The formation of mitotic chromosomes requires both compaction of chromatin and the resolution of replicated sister chromatids. Compaction occurs during mitotic prophase and prometaphase, and in prophase relies on the activity of condensin II complexes. Exactly when and how sister chromatid resolution occurs has been largely unknown, as has its molecular requirements. Here, we established a method to visualize sister resolution by sequential replication labelling with two distinct nucleotide derivatives. Quantitative three-dimensional imaging then allowed us to measure the resolution of sister chromatids throughout mitosis by calculating their non-overlapping volume within the whole chromosome. Unexpectedly, we found that sister chromatid resolution starts already at the beginning of prophase, proceeds concomitantly with chromatin compaction and is largely completed by the end of prophase. Sister chromatid resolution was abolished by inhibition of topoisomerase IIα and by depleting or preventing mitotic activation of condensin II, whereas blocking cohesin dissociation from chromosomes had little effect. Mitotic sister chromatid resolution is thus an intrinsic part of mitotic chromosome formation in prophase that relies largely on DNA decatenation and shares the molecular requirement for condensin II with prophase compaction.


Assuntos
Cromátides/metabolismo , Mitose/fisiologia , Prometáfase/fisiologia , Prófase/fisiologia , Adenosina Trifosfatases/metabolismo , Antígenos de Neoplasias/metabolismo , Linhagem Celular , Replicação do DNA/fisiologia , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Imageamento Tridimensional/métodos , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo
9.
J Cell Biol ; 212(6): 647-59, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26953350

RESUMO

To prevent genome instability, mitotic exit is delayed until all chromosomes are properly attached to the mitotic spindle by the spindle assembly checkpoint (SAC). In this study, we characterized the function of ARHGEF17, identified in a genome-wide RNA interference screen for human mitosis genes. Through a series of quantitative imaging, biochemical, and biophysical experiments, we showed that ARHGEF17 is essential for SAC activity, because it is the major targeting factor that controls localization of the checkpoint kinase Mps1 to the kinetochore. This mitotic function is mediated by direct interaction of the central domain of ARHGEF17 with Mps1, which is autoregulated by the activity of Mps1 kinase, for which ARHGEF17 is a substrate. This mitosis-specific role is independent of ARHGEF17's RhoGEF activity in interphase. Our study thus assigns a new mitotic function to ARHGEF17 and reveals the molecular mechanism for a key step in SAC establishment.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Cinetocoros/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Fuso Acromático/metabolismo , Linhagem Celular Tumoral , Células HeLa , Humanos , Mitose/fisiologia , Transdução de Sinais/fisiologia , Fuso Acromático/fisiologia
10.
Mol Syst Biol ; 11(9): 828, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26415501

RESUMO

A remarkable observation emerging from recent cancer genome analyses is the identification of chromothripsis as a one-off genomic catastrophe, resulting in massive somatic DNA structural rearrangements (SRs). Largely due to lack of suitable model systems, the mechanistic basis of chromothripsis has remained elusive. We developed an integrative method termed "complex alterations after selection and transformation (CAST)," enabling efficient in vitro generation of complex DNA rearrangements including chromothripsis, using cell perturbations coupled with a strong selection barrier followed by massively parallel sequencing. We employed this methodology to characterize catastrophic SR formation processes, their temporal sequence, and their impact on gene expression and cell division. Our in vitro system uncovered a propensity of chromothripsis to occur in cells with damaged telomeres, and in particular in hyperploid cells. Analysis of primary medulloblastoma cancer genomes verified the link between hyperploidy and chromothripsis in vivo. CAST provides the foundation for mechanistic dissection of complex DNA rearrangement processes.


Assuntos
Cromossomos Humanos/genética , Rearranjo Gênico , Genoma Humano/genética , Instabilidade Genômica/genética , Neoplasias/genética , Aneuploidia , Divisão Celular , Linhagem Celular , Aberrações Cromossômicas , Variações do Número de Cópias de DNA/genética , Humanos , Meduloblastoma/genética , Poliploidia , Telômero/genética , Telômero/patologia , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
11.
Cell Rep ; 12(9): 1519-30, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26299969

RESUMO

Many cellular processes involve the recruitment of proteins to specific membranes, which are decorated with distinctive lipids that act as docking sites. The phosphoinositides form signaling hubs, and we examine mechanisms underlying recruitment. We applied a physiological, quantitative, liposome microarray-based assay to measure the membrane-binding properties of 91 pleckstrin homology (PH) domains, the most common phosphoinositide-binding target. 10,514 experiments quantified the role of phosphoinositides in membrane recruitment. For most domains examined, the observed binding specificity implied cooperativity with additional signaling lipids. Analyses of PH domains with similar lipid-binding profiles identified a conserved motif, mutations in which-including some found in human cancers-induced discrete changes in binding affinities in vitro and protein mislocalization in vivo. The data set reveals cooperativity as a key mechanism for membrane recruitment and, by enabling the interpretation of disease-associated mutations, suggests avenues for the design of small molecules targeting PH domains.


Assuntos
Membrana Celular/metabolismo , Proteínas Fúngicas/metabolismo , Fosfatidilinositóis/metabolismo , Chaetomium/metabolismo , Proteínas Fúngicas/química , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Saccharomyces cerevisiae/metabolismo
12.
J Cell Biol ; 209(5): 705-20, 2015 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-26056140

RESUMO

Targeting of inner nuclear membrane (INM) proteins is essential for nuclear architecture and function, yet its mechanism remains poorly understood. Here, we established a new reporter that allows real-time imaging of membrane protein transport from the ER to the INM using Lamin B receptor and Lap2ß as model INM proteins. These reporters allowed us to characterize the kinetics of INM targeting and establish a mathematical model of this process and enabled us to probe its molecular requirements in an RNA interference screen of 96 candidate genes. Modeling of the phenotypes of genes involved in transport of these INM proteins predicted that it critically depended on the number and permeability of nuclear pores and the availability of nuclear binding sites, but was unaffected by depletion of most transport receptors. These predictions were confirmed with targeted validation experiments on the functional requirements of nucleoporins and nuclear lamins. Collectively, our data support a diffusion retention model of INM protein transport in mammalian cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Imagem Molecular , Membrana Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático/genética , Células HeLa , Humanos , Proteínas de Membrana/genética , Membrana Nuclear/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptor de Lamina B
13.
PLoS One ; 10(2): e0116783, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25658810

RESUMO

Polo-like kinase 1 (PLK1) orchestrates multiple events of cell division. Although PLK1 function has been intensively studied in centriole-containing and rapidly cycling somatic cells, much less is known about its function in the meiotic divisions of mammalian oocytes, which arrest for a long period of time in prophase before meiotic resumption and lack centrioles for spindle assembly. Here, using specific small molecule inhibition combined with live mouse oocyte imaging, we comprehensively characterize meiotic PLK1's functions. We show that PLK1 becomes activated at meiotic resumption on microtubule organizing centers (MTOCs) and later at kinetochores. PLK1 is required for efficient meiotic resumption by promoting nuclear envelope breakdown. PLK1 is also needed to recruit centrosomal proteins to acentriolar MTOCs to promote normal spindle formation, as well as for stable kinetochore-microtubule attachment. Consequently, PLK1 inhibition leads to metaphase I arrest with misaligned chromosomes activating the spindle assembly checkpoint (SAC). Unlike in mitosis, the metaphase I arrest is not bypassed by the inactivation of the SAC. We show that PLK1 is required for the full activation of the anaphase promoting complex/cyclosome (APC/C) by promoting the degradation of the APC/C inhibitor EMI1 and is therefore essential for entry into anaphase I. Moreover, our data suggest that PLK1 is required for proper chromosome segregation and the maintenance of chromosome condensation during the meiosis I-II transition, independently of the APC/C. Thus, our results define the meiotic roles of PLK1 in oocytes and reveal interesting differential requirements of PLK1 between mitosis and oocyte meiosis in mammals.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Meiose/fisiologia , Oócitos/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Animais , Western Blotting , Segregação de Cromossomos/fisiologia , Feminino , Processamento de Imagem Assistida por Computador , Cinetocoros/metabolismo , Camundongos , Microscopia Confocal , Centro Organizador dos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Quinase 1 Polo-Like
14.
Curr Biol ; 24(21): 2598-605, 2014 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-25447992

RESUMO

During animal cell division, an actin-based ring cleaves the cell into two. Problems with this process can cause chromosome missegregation and defects in cytoplasmic inheritance and the partitioning of organelles, which in turn are associated with human diseases. Although much is known about how chromosome segregation is coupled to cell division, the way organelles coordinate their inheritance during partitioning to daughter cells is less well understood. Here, using a high-content live-imaging small interfering RNA screen, we identify Myosin-XIX (Myo19) as a novel regulator of cell division. Previously, this actin-based motor was shown to control the interphase movement of mitochondria. Our analysis shows that Myo19 is indeed localized to mitochondria and that its silencing leads to defects in the distribution of mitochondria within cells and in mitochondrial partitioning at division. Furthermore, many Myo19 RNAi cells undergo stochastic division failure--a phenotype that can be mimicked using a treatment that blocks mitochondrial fission and rescued by decreasing mitochondrial fusion, implying that mitochondria can physically interfere with cytokinesis. Strikingly, using live imaging we also observe the inappropriate movement of mitochondria to the poles of spindles in cells depleted for Myo19 as they enter anaphase. Since this phenocopies the results of an acute loss of actin filaments in anaphase, these data support a model whereby the Myo19 actin-based motor helps to control mitochondrial movement to ensure their faithful segregation during division. The presence of DNA within mitochondria makes their inheritance an especially important aspect of symmetrical cell division.


Assuntos
Divisão Celular/fisiologia , Mitocôndrias/metabolismo , Miosinas/fisiologia , Actinas/metabolismo , Citocinese , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Mitocôndrias/ultraestrutura , Dinâmica Mitocondrial , Miosinas/genética , Miosinas/metabolismo
15.
EMBO J ; 33(22): 2643-58, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25257309

RESUMO

Although splicing is essential for the expression of most eukaryotic genes, inactivation of splicing factors causes specific defects in mitosis. The molecular cause of this defect is unknown. Here, we show that the spliceosome subunits SNW1 and PRPF8 are essential for sister chromatid cohesion in human cells. A transcriptome-wide analysis revealed that SNW1 or PRPF8 depletion affects the splicing of specific introns in a subset of pre-mRNAs, including pre-mRNAs encoding the cohesion protein sororin and the APC/C subunit APC2. SNW1 depletion causes cohesion defects predominantly by reducing sororin levels, which causes destabilisation of cohesin on DNA. SNW1 depletion also reduces APC/C activity and contributes to cohesion defects indirectly by delaying mitosis and causing "cohesion fatigue". Simultaneous expression of sororin and APC2 from intron-less cDNAs restores cohesion in SNW1-depleted cells. These results indicate that the spliceosome is required for mitosis because it enables expression of genes essential for cohesion. Our transcriptome-wide identification of retained introns in SNW1- and PRPF8-depleted cells may help to understand the aetiology of diseases associated with splicing defects, such as retinosa pigmentosum and cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Proteínas do Citoesqueleto/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Precursores de RNA/metabolismo , Splicing de RNA/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas do Citoesqueleto/genética , Deleção de Genes , Células HeLa , Humanos , Coativadores de Receptor Nuclear/genética , Precursores de RNA/genética , Transcriptoma/fisiologia
16.
Methods Cell Biol ; 122: 219-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24857732

RESUMO

The nuclear pore complex (NPC) mediates selective transport across the nuclear envelope (NE) and plays crucial roles in several additional cellular functions. In higher eukaryotes, the NPC and the NE disassemble and reassemble during cell division and live-cell imaging has been a powerful tool to analyze these dynamic processes. Here, we present a method for the kinetic analysis of postmitotic NPC assembly and reestablishment of transport competence in intact cells by multicolor 4D imaging and photoswitching. By applying the methods we have established previously using normal rat kidney to HeLa cells, we demonstrate the conservation of NPC assembly in different mammalian cells. We recently showed that the molecular organization of the NPC can be studied by combining stochastic super-resolution microscopy with single-particle averaging and present this method here in detail.


Assuntos
Microscopia/métodos , Complexos Multiproteicos/análise , Complexo de Proteínas Formadoras de Poros Nucleares/análise , Poro Nuclear/metabolismo , Animais , Transporte Biológico , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Mitose , Ratos , Pontos de Checagem da Fase S do Ciclo Celular
17.
J Cell Biol ; 204(5): 697-712, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24590172

RESUMO

SAS-6 proteins are thought to impart the ninefold symmetry of centrioles, but the mechanisms by which their assembly occurs within cells remain elusive. In this paper, we provide evidence that the N-terminal, coiled-coil, and C-terminal domains of HsSAS-6 are each required for procentriole formation in human cells. Moreover, the coiled coil is necessary and sufficient to mediate HsSAS-6 centrosomal targeting. High-resolution imaging reveals that GFP-tagged HsSAS-6 variants localize in a torus around the base of the parental centriole before S phase, perhaps indicative of an initial loading platform. Moreover, fluorescence recovery after photobleaching analysis demonstrates that HsSAS-6 is immobilized progressively at centrosomes during cell cycle progression. Using fluorescence correlation spectroscopy and three-dimensional stochastic optical reconstruction microscopy, we uncover that HsSAS-6 is present in the cytoplasm primarily as a homodimer and that its oligomerization into a ninefold symmetrical ring occurs at centrioles. Together, our findings lead us to propose a mechanism whereby HsSAS-6 homodimers are targeted to centrosomes where the local environment and high concentration of HsSAS-6 promote oligomerization, thus initiating procentriole formation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Centríolos/ultraestrutura , Dimerização , Recuperação de Fluorescência Após Fotodegradação , Humanos , Modelos Biológicos , Transporte Proteico
18.
J Struct Biol ; 184(3): 445-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24145303

RESUMO

In contrast to cytoplasmic organelles, which are usually separated from the rest of the cell by phospholipid membranes, nuclear compartments are readily accessible to diffusing proteins and must rely on different mechanisms to maintain their integrity. Specific interactions between scaffolding proteins are known to have important roles for the formation and maintenance of nuclear structures. General physical mechanisms such as molecular crowding, phase separation or colloidal behavior have also been suggested, but their physiological significance remains uncertain. For macromolecular crowding, a role in the maintenance of nucleoli and promyelocytic leukemia (PML) nuclear bodies has been shown. Here, we tested whether a modulation of the compaction state of chromatin, which directly influences the local crowding state, has an impact on the formation and maintenance of densely packed heterochromatin. By osmotic perturbations, we could modify the packing state of chromatin in a controlled manner and show that chromatin compaction, which is associated with increased crowding conditions, is not, per se, sufficient to initiate the formation of new bona fide heterochromatin structures nor is it necessary to maintain already established heterochromatin domains. In consequence, if an increase in crowding induced by chromatin compaction maybe an early step in heterochromatin formation, specific protein-protein interactions are nevertheless required to make heterochromatin long lasting and independent of the crowding state.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/química , Cromatina/metabolismo , Heterocromatina/metabolismo , Animais , Eucromatina/química , Eucromatina/metabolismo , Heterocromatina/química , Histonas/análise , Histonas/genética , Histonas/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Células NIH 3T3 , Pressão Osmótica , Fotodegradação , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Espectrometria de Fluorescência
19.
Nature ; 501(7468): 564-8, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23975099

RESUMO

Mammalian genomes contain several billion base pairs of DNA that are packaged in chromatin fibres. At selected gene loci, cohesin complexes have been proposed to arrange these fibres into higher-order structures, but how important this function is for determining overall chromosome architecture and how the process is regulated are not well understood. Using conditional mutagenesis in the mouse, here we show that depletion of the cohesin-associated protein Wapl stably locks cohesin on DNA, leads to clustering of cohesin in axial structures, and causes chromatin condensation in interphase chromosomes. These findings reveal that the stability of cohesin-DNA interactions is an important determinant of chromatin structure, and indicate that cohesin has an architectural role in interphase chromosome territories. Furthermore, we show that regulation of cohesin-DNA interactions by Wapl is important for embryonic development, expression of genes such as c-myc (also known as Myc), and cell cycle progression. In mitosis, Wapl-mediated release of cohesin from DNA is essential for proper chromosome segregation and protects cohesin from cleavage by the protease separase, thus enabling mitotic exit in the presence of functional cohesin complexes.


Assuntos
Cromatina/química , Cromatina/metabolismo , Segregação de Cromossomos , Proteínas/metabolismo , Animais , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromátides/genética , Cromátides/metabolismo , Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos/genética , Cromossomos de Mamíferos/química , Cromossomos de Mamíferos/genética , Cromossomos de Mamíferos/metabolismo , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário/genética , Endopeptidases/metabolismo , Regulação da Expressão Gênica/genética , Genes myc/genética , Interfase , Camundongos , Mitose , Prófase , Proteínas/genética , Separase , Coesinas
20.
Science ; 341(6146): 655-8, 2013 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-23845946

RESUMO

Much of life's essential molecular machinery consists of large protein assemblies that currently pose challenges for structure determination. A prominent example is the nuclear pore complex (NPC), for which the organization of its individual components remains unknown. By combining stochastic super-resolution microscopy, to directly resolve the ringlike structure of the NPC, with single particle averaging, to use information from thousands of pores, we determined the average positions of fluorescent molecular labels in the NPC with a precision well below 1 nanometer. Applying this approach systematically to the largest building block of the NPC, the Nup107-160 subcomplex, we assessed the structure of the NPC scaffold. Thus, light microscopy can be used to study the molecular organization of large protein complexes in situ in whole cells.


Assuntos
Microscopia/métodos , Matriz Nuclear/ultraestrutura , Complexo de Proteínas Formadoras de Poros Nucleares/química , Poro Nuclear/ultraestrutura , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Humanos , Microscopia Confocal/métodos , Nanopartículas/química , Complexo de Proteínas Formadoras de Poros Nucleares/imunologia , Tamanho da Partícula , Anticorpos de Domínio Único/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA