Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cell ; 161(7): 1502-4, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091034

ABSTRACT

Human cancer cells bear complex chromosome rearrangements that can be potential drivers of cancer development. However, the molecular mechanisms underlying these rearrangements have been unclear. Zhang et al. use a new technique combining live-cell imaging and single-cell sequencing to demonstrate that chromosomes mis-segregated to micronuclei frequently undergo chromothripsis-like rearrangements in the subsequent cell cycle.


Subject(s)
Chromosome Breakage , DNA Damage , Micronuclei, Chromosome-Defective , Humans
2.
Cell ; 156(5): 868-9, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581486

ABSTRACT

The spindle assembly checkpoint prevents separation of sister chromatids until each kinetochore is attached to the mitotic spindle. Rodriguez-Bravo et al. report that the nuclear pore complex scaffolds spindle assembly checkpoint signaling in interphase, providing a store of inhibitory signals that limits the speed of the subsequent mitosis.


Subject(s)
Anaphase , Cell Cycle Proteins/metabolism , M Phase Cell Cycle Checkpoints , Mad2 Proteins/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Humans
3.
Cell ; 154(1): 47-60, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827674

ABSTRACT

During mitotic exit, missegregated chromosomes can recruit their own nuclear envelope (NE) to form micronuclei (MN). MN have reduced functioning compared to primary nuclei in the same cell, although the two compartments appear to be structurally comparable. Here we show that over 60% of MN undergo an irreversible loss of compartmentalization during interphase due to NE collapse. This disruption of the MN, which is induced by defects in nuclear lamina assembly, drastically reduces nuclear functions and can trigger massive DNA damage. MN disruption is associated with chromatin compaction and invasion of endoplasmic reticulum (ER) tubules into the chromatin. We identified disrupted MN in both major subtypes of human non-small-cell lung cancer, suggesting that disrupted MN could be a useful objective biomarker for genomic instability in solid tumors. Our study shows that NE collapse is a key event underlying MN dysfunction and establishes a link between aberrant NE organization and aneuploidy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Genomic Instability , Lung Neoplasms/pathology , Micronuclei, Chromosome-Defective , Nuclear Envelope/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Damage , Humans , Interphase , Lamins/metabolism , Lung Neoplasms/genetics
4.
Cell ; 154(5): 971-982, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23993091

ABSTRACT

Intracellular proteins with long lifespans have recently been linked to age-dependent defects, ranging from decreased fertility to the functional decline of neurons. Why long-lived proteins exist in metabolically active cellular environments and how they are maintained over time remains poorly understood. Here, we provide a system-wide identification of proteins with exceptional lifespans in the rat brain. These proteins are inefficiently replenished despite being translated robustly throughout adulthood. Using nucleoporins as a paradigm for long-term protein persistence, we found that nuclear pore complexes (NPCs) are maintained over a cell's life through slow but finite exchange of even its most stable subcomplexes. This maintenance is limited, however, as some nucleoporin levels decrease during aging, providing a rationale for the previously observed age-dependent deterioration of NPC function. Our identification of a long-lived proteome reveals cellular components that are at increased risk for damage accumulation, linking long-term protein persistence to the cellular aging process. PAPERCLIP:


Subject(s)
Brain/cytology , Cellular Senescence , Nuclear Pore Complex Proteins/metabolism , Proteome/metabolism , Animals , Brain/metabolism , Neuroglia/metabolism , Neurons/metabolism , Nuclear Pore/metabolism , Protein Biosynthesis , Rats
5.
Genes Dev ; 34(13-14): 913-930, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32499403

ABSTRACT

During mitosis, transcription of genomic DNA is dramatically reduced, before it is reactivated during nuclear reformation in anaphase/telophase. Many aspects of the underlying principles that mediate transcriptional memory and reactivation in the daughter cells remain unclear. Here, we used ChIP-seq on synchronized cells at different stages after mitosis to generate genome-wide maps of histone modifications. Combined with EU-RNA-seq and Hi-C analyses, we found that during prometaphase, promoters, enhancers, and insulators retain H3K4me3 and H3K4me1, while losing H3K27ac. Enhancers globally retaining mitotic H3K4me1 or locally retaining mitotic H3K27ac are associated with cell type-specific genes and their transcription factors for rapid transcriptional activation. As cells exit mitosis, promoters regain H3K27ac, which correlates with transcriptional reactivation. Insulators also gain H3K27ac and CCCTC-binding factor (CTCF) in anaphase/telophase. This increase of H3K27ac in anaphase/telophase is required for posttranscriptional activation and may play a role in the establishment of topologically associating domains (TADs). Together, our results suggest that the genome is reorganized in a sequential order, in which histone methylations occur first in prometaphase, histone acetylation, and CTCF in anaphase/telophase, transcription in cytokinesis, and long-range chromatin interactions in early G1. We thus provide insights into the histone modification landscape that allows faithful reestablishment of the transcriptional program and TADs during cell division.


Subject(s)
Chromatin/metabolism , Histone Code/genetics , Histones/metabolism , Mitosis/genetics , Protein Processing, Post-Translational/genetics , Transcriptional Activation/genetics , Animals , Cell Cycle Checkpoints/genetics , Chromosomes/genetics , Chromosomes/metabolism , Enhancer Elements, Genetic , Genome/genetics , Humans , Promoter Regions, Genetic , Protein Binding , Time Factors
6.
Cell ; 149(4): 733-5, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22579277

ABSTRACT

Nuclear export of mRNAs is thought to occur exclusively through nuclear pore complexes. In this issue of Cell, Speese et al. identify an alternate pathway for mRNA export in muscle cells where ribonucleoprotein complexes involved in forming neuromuscular junctions transit the nuclear envelope by fusing with and budding through the nuclear membrane.

7.
Genes Dev ; 32(19-20): 1321-1331, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30228202

ABSTRACT

The total number of nuclear pore complexes (NPCs) per nucleus varies greatly between different cell types and is known to change during cell differentiation and cell transformation. However, the underlying mechanisms that control how many nuclear transport channels are assembled into a given nuclear envelope remain unclear. Here, we report that depletion of the NPC basket protein Tpr, but not Nup153, dramatically increases the total NPC number in various cell types. This negative regulation of Tpr occurs via a phosphorylation cascade of extracellular signal-regulated kinase (ERK), the central kinase of the mitogen-activated protein kinase (MAPK) pathway. Tpr serves as a scaffold for ERK to phosphorylate the nucleoporin (Nup) Nup153, which is critical for early stages of NPC biogenesis. Our results reveal a critical role of the Nup Tpr in coordinating signal transduction pathways during cell proliferation and the dynamic organization of the nucleus.


Subject(s)
Nuclear Pore Complex Proteins/physiology , Nuclear Pore/physiology , Proto-Oncogene Proteins/physiology , Animals , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interphase , Mice , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/metabolism , Proto-Oncogene Proteins/metabolism
8.
Nat Rev Genet ; 20(1): 39-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30356165

ABSTRACT

The genome is packaged and organized nonrandomly within the 3D space of the nucleus to promote efficient gene expression and to faithfully maintain silencing of heterochromatin. The genome is enclosed within the nucleus by the nuclear envelope membrane, which contains a set of proteins that actively participate in chromatin organization and gene regulation. Technological advances are providing views of genome organization at unprecedented resolution and are beginning to reveal the ways that cells co-opt the structures of the nuclear periphery for nuclear organization and gene regulation. These genome regulatory roles of proteins of the nuclear periphery have important influences on development, disease and ageing.


Subject(s)
Gene Expression Regulation/physiology , Genome, Human/physiology , Heterochromatin/metabolism , Nuclear Envelope/metabolism , Animals , Heterochromatin/genetics , Humans , Nuclear Envelope/genetics
9.
Nat Rev Mol Cell Biol ; 14(1): 55-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23258296

ABSTRACT

Protein turnover is an effective way of maintaining a functional proteome, as old and potentially damaged polypeptides are destroyed and replaced by newly synthesized copies. An increasing number of intracellular proteins, however, have been identified that evade this turnover process and instead are maintained over a cell's lifetime. This diverse group of long-lived proteins might be particularly prone to accumulation of damage and thus have a crucial role in the functional deterioration of key regulatory processes during ageing.


Subject(s)
Cellular Senescence , Proteins/metabolism , Proteome/metabolism , Homeostasis , Humans , Protein Biosynthesis
10.
Cell ; 141(6): 1030-41, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20550937

ABSTRACT

In metazoa, nuclear pore complexes (NPCs) assemble from disassembled precursors into a reforming nuclear envelope (NE) at the end of mitosis and into growing intact NEs during interphase. Here, we show via RNAi-mediated knockdown that ELYS, a nucleoporin critical for the recruitment of the essential Nup107/160 complex to chromatin, is required for NPC assembly at the end of mitosis but not during interphase. Conversely, the transmembrane nucleoporin POM121 is critical for the incorporation of the Nup107/160 complex into new assembly sites specifically during interphase. Strikingly, recruitment of the Nup107/160 complex to an intact NE involves a membrane curvature-sensing domain of its constituent Nup133, which is not required for postmitotic NPC formation. Our results suggest that in organisms with open mitosis, NPCs assemble via two distinct mechanisms to accommodate cell cycle-dependent differences in NE topology.


Subject(s)
Cell Cycle , Eukaryotic Cells/metabolism , Nuclear Pore/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Minor Histocompatibility Antigens , Nuclear Pore Complex Proteins/metabolism , Protein Multimerization , Transcription Factors/metabolism , Xenopus
11.
Cell ; 140(3): 372-83, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20144761

ABSTRACT

Nuclear pore complexes have recently been shown to play roles in gene activation; however their potential involvement in metazoan transcription remains unclear. Here we show that the nucleoporins Sec13, Nup98, and Nup88, as well as a group of FG-repeat nucleoporins, bind to the Drosophila genome at functionally distinct loci that often do not represent nuclear envelope contact sites. Whereas Nup88 localizes to silent loci, Sec13, Nup98, and a subset of FG-repeat nucleoporins bind to developmentally regulated genes undergoing transcription induction. Strikingly, RNAi-mediated knockdown of intranuclear Sec13 and Nup98 specifically inhibits transcription of their target genes and prevents efficient reactivation of transcription after heat shock, suggesting an essential role of NPC components in regulating complex gene expression programs of multicellular organisms.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Nuclear Pore Complex Proteins/metabolism , Animals , Drosophila melanogaster/genetics , Heat-Shock Response , RNA Polymerase II/metabolism , Transcription, Genetic
12.
Genes Dev ; 31(22): 2222-2234, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29269482

ABSTRACT

Recent studies have shown that a subset of nucleoporins (Nups) can detach from the nuclear pore complex and move into the nuclear interior to regulate transcription. One such dynamic Nup, called Nup98, has been implicated in gene activation in healthy cells and has been shown to drive leukemogenesis when mutated in patients with acute myeloid leukemia (AML). Here we show that in hematopoietic cells, Nup98 binds predominantly to transcription start sites to recruit the Wdr82-Set1A/COMPASS (complex of proteins associated with Set1) complex, which is required for deposition of the histone 3 Lys4 trimethyl (H3K4me3)-activating mark. Depletion of Nup98 or Wdr82 abolishes Set1A recruitment to chromatin and subsequently ablates H3K4me3 at adjacent promoters. Furthermore, expression of a Nup98 fusion protein implicated in aggressive AML causes mislocalization of H3K4me3 at abnormal regions and up-regulation of associated genes. Our findings establish a function of Nup98 in hematopoietic gene activation and provide mechanistic insight into which Nup98 leukemic fusion proteins promote AML.


Subject(s)
Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nuclear Pore Complex Proteins/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cells, Cultured , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Developmental , Humans , Methylation , Mice
13.
Cell ; 136(2): 284-95, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19167330

ABSTRACT

In dividing cells, nuclear pore complexes (NPCs) disassemble during mitosis and reassemble into the newly forming nuclei. However, the fate of nuclear pores in postmitotic cells is unknown. Here, we show that NPCs, unlike other nuclear structures, do not turn over in differentiated cells. While a subset of NPC components, like Nup153 and Nup50, are continuously exchanged, scaffold nucleoporins, like the Nup107/160 complex, are extremely long-lived and remain incorporated in the nuclear membrane during the entire cellular life span. Besides the lack of nucleoporin expression and NPC turnover, we discovered an age-related deterioration of NPCs, leading to an increase in nuclear permeability and the leaking of cytoplasmic proteins into the nucleus. Our finding that nuclear "leakiness" is dramatically accelerated during aging and that a subset of nucleoporins is oxidatively damaged in old cells suggests that the accumulation of damage at the NPC might be a crucial aging event.


Subject(s)
Cell Nucleus/physiology , Mitosis , Nuclear Pore/physiology , Animals , Caenorhabditis elegans , Down-Regulation , Mice , Nuclear Pore Complex Proteins/physiology , Rats
14.
Genes Dev ; 30(20): 2253-2258, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27807035

ABSTRACT

The organization of the genome in the three-dimensional space of the nucleus is coupled with cell type-specific gene expression. However, how nuclear architecture influences transcription that governs cell identity remains unknown. Here, we show that nuclear pore complex (NPC) components Nup93 and Nup153 bind superenhancers (SE), regulatory structures that drive the expression of key genes that specify cell identity. We found that nucleoporin-associated SEs localize preferentially to the nuclear periphery, and absence of Nup153 and Nup93 results in dramatic transcriptional changes of SE-associated genes. Our results reveal a crucial role of NPC components in the regulation of cell type-specifying genes and highlight nuclear architecture as a regulatory layer of genome functions in cell fate.


Subject(s)
Cell Differentiation/genetics , Cell Nucleus/metabolism , Gene Expression Regulation/genetics , Nuclear Pore Complex Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Chromatin/metabolism , Enhancer Elements, Genetic/physiology , Genome/genetics , Humans , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/genetics , Protein Binding , Protein Transport
15.
Genes Dev ; 30(10): 1155-71, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198230

ABSTRACT

Nuclear pore complexes (NPCs) emerged as nuclear transport channels in eukaryotic cells ∼1.5 billion years ago. While the primary role of NPCs is to regulate nucleo-cytoplasmic transport, recent research suggests that certain NPC proteins have additionally acquired the role of affecting gene expression at the nuclear periphery and in the nucleoplasm in metazoans. Here we identify a widely expressed variant of the transmembrane nucleoporin (Nup) Pom121 (named sPom121, for "soluble Pom121") that arose by genomic rearrangement before the divergence of hominoids. sPom121 lacks the nuclear membrane-anchoring domain and thus does not localize to the NPC. Instead, sPom121 colocalizes and interacts with nucleoplasmic Nup98, a previously identified transcriptional regulator, at gene promoters to control transcription of its target genes in human cells. Interestingly, sPom121 transcripts appear independently in several mammalian species, suggesting convergent innovation of Nup-mediated transcription regulation during mammalian evolution. Our findings implicate alternate transcription initiation as a mechanism to increase the functional diversity of NPC components.


Subject(s)
Evolution, Molecular , Gene Expression Regulation , Membrane Glycoproteins/metabolism , Mutant Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , 5' Untranslated Regions/genetics , Cell Membrane/metabolism , Cell Nucleus/metabolism , Exons/genetics , HeLa Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Nuclear Localization Signals , Nuclear Pore Complex Proteins/chemistry , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , Solubility , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Initiation Site
16.
Genes Dev ; 29(4): 337-49, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691464

ABSTRACT

Nuclear pore complexes (NPCs) are composed of several copies of ∼30 different proteins called nucleoporins (Nups). NPCs penetrate the nuclear envelope (NE) and regulate the nucleocytoplasmic trafficking of macromolecules. Beyond this vital role, NPC components influence genome functions in a transport-independent manner. Nups play an evolutionarily conserved role in gene expression regulation that, in metazoans, extends into the nuclear interior. Additionally, in proliferative cells, Nups play a crucial role in genome integrity maintenance and mitotic progression. Here we discuss genome-related functions of Nups and their impact on essential DNA metabolism processes such as transcription, chromosome duplication, and segregation.


Subject(s)
Genome , Nuclear Pore Complex Proteins/metabolism , Animals , Chromosome Duplication/genetics , Chromosome Segregation/genetics , Gene Expression Regulation , Genomic Instability , Humans , Nuclear Pore Complex Proteins/genetics
17.
Genes Dev ; 29(12): 1224-38, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26080816

ABSTRACT

Nucleoporins (Nups) are a family of proteins best known as the constituent building blocks of nuclear pore complexes (NPCs), membrane-embedded channels that mediate nuclear transport across the nuclear envelope. Recent evidence suggests that several Nups have additional roles in controlling the activation and silencing of developmental genes; however, the mechanistic details of these functions remain poorly understood. Here, we show that depletion of Nup153 in mouse embryonic stem cells (mESCs) causes the derepression of developmental genes and induction of early differentiation. This loss of stem cell identity is not associated with defects in the nuclear import of key pluripotency factors. Rather, Nup153 binds around the transcriptional start site (TSS) of developmental genes and mediates the recruitment of the polycomb-repressive complex 1 (PRC1) to a subset of its target loci. Our results demonstrate a chromatin-associated role of Nup153 in maintaining stem cell pluripotency by functioning in mammalian epigenetic gene silencing.


Subject(s)
Embryonic Stem Cells/physiology , Gene Silencing , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Pluripotent Stem Cells/physiology , Animals , Binding Sites , Cell Differentiation , Cell Nucleus/metabolism , Chromatin/metabolism , Chromosome Mapping , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 1/metabolism , Protein Binding
19.
PLoS Genet ; 9(2): e1003308, 2013.
Article in English | MEDLINE | ID: mdl-23468646

ABSTRACT

Faithful execution of developmental gene expression programs occurs at multiple levels and involves many different components such as transcription factors, histone-modification enzymes, and mRNA processing proteins. Recent evidence suggests that nucleoporins, well known components that control nucleo-cytoplasmic trafficking, have wide-ranging functions in developmental gene regulation that potentially extend beyond their role in nuclear transport. Whether the unexpected role of nuclear pore proteins in transcription regulation, which initially has been described in fungi and flies, also applies to human cells is unknown. Here we show at a genome-wide level that the nuclear pore protein NUP98 associates with developmentally regulated genes active during human embryonic stem cell differentiation. Overexpression of a dominant negative fragment of NUP98 levels decreases expression levels of NUP98-bound genes. In addition, we identify two modes of developmental gene regulation by NUP98 that are differentiated by the spatial localization of NUP98 target genes. Genes in the initial stage of developmental induction can associate with NUP98 that is embedded in the nuclear pores at the nuclear periphery. Alternatively, genes that are highly induced can interact with NUP98 in the nuclear interior, away from the nuclear pores. This work demonstrates for the first time that NUP98 dynamically associates with the human genome during differentiation, revealing a role of a nuclear pore protein in regulating developmental gene expression programs.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Nuclear Pore Complex Proteins , Transcription, Genetic , Active Transport, Cell Nucleus/genetics , Cytoplasm/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome, Human , Humans , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism
20.
Curr Opin Cell Biol ; 20(4): 386-92, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495454

ABSTRACT

The nuclear envelope is a double-layered membrane that encloses the nuclear genome and transcriptional machinery. In dividing cells of metazoa, the nucleus completely disassembles during mitosis, creating the need to re-establish the nuclear compartment at the end of each cell division. Given the crucial role of the nuclear envelope in gene regulation and cellular organization, it is not surprising that its biogenesis and organization have become active research areas. We will review recent insights into nuclear membrane dynamics during the cell cycle.


Subject(s)
Life Cycle Stages/physiology , Nuclear Envelope/physiology , Vertebrates/physiology , Animals , Cell Cycle/physiology
SELECTION OF CITATIONS
SEARCH DETAIL