Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 597(22): 2782-2790, 2023 11.
Article in English | MEDLINE | ID: mdl-37339933

ABSTRACT

The nuclear envelope plays an essential role in organizing the genome inside of the nucleus. The inner nuclear membrane is coated with a meshwork of filamentous lamin proteins that provide a surface to organize a variety of cellular processes. A subset of nuclear lamina- and membrane-associated proteins functions as anchors to hold transcriptionally silent heterochromatin at the nuclear periphery. While most chromatin tethers are integral membrane proteins, a limited number are lamina-bound. One example is the mammalian proline-rich 14 (PRR14) protein. PRR14 is a recently characterized protein with unique function that is different from other known chromatin tethers. Here, we review our current understanding of PRR14 structure and function in organizing heterochromatin at the nuclear periphery.


Subject(s)
Chromatin , Heterochromatin , Animals , Chromatin/genetics , Chromatin/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Cell Nucleus/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Nuclear Envelope , Mammals/genetics
2.
Genome Biol ; 23(1): 185, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36050765

ABSTRACT

BACKGROUND: Lamina-associated domains (LADs) are large genomic regions that are positioned at the nuclear lamina. It has remained largely unclear what drives the positioning and demarcation of LADs. Because the insulator protein CTCF is enriched at LAD borders, it was postulated that CTCF binding could position some LAD boundaries, possibly through its function in stalling cohesin and hence preventing cohesin invading into the LAD. To test this, we mapped genome-nuclear lamina interactions in mouse embryonic stem cells after rapid depletion of CTCF and other perturbations of cohesin dynamics. RESULTS: CTCF and cohesin contribute to a sharp transition in lamina interactions at LAD borders, while LADs are maintained after depletion of these proteins, also at borders marked by CTCF. CTCF and cohesin may thus reinforce LAD borders, but do not position these. CTCF binding sites within LADs are locally detached from the lamina and enriched for accessible DNA and active histone modifications. Remarkably, despite lamina positioning being strongly correlated with genome inactivity, this DNA remains accessible after the local detachment is lost following CTCF depletion. At a chromosomal scale, cohesin depletion and cohesin stabilization by depletion of the unloading factor WAPL quantitatively affect lamina interactions, indicative of perturbed chromosomal positioning in the nucleus. Finally, while H3K27me3 is locally enriched at CTCF-marked LAD borders, we find no evidence for an interplay between CTCF and H3K27me3 on lamina interactions. CONCLUSIONS: These findings illustrate that CTCF and cohesin are not primary determinants of LAD patterns. Rather, these proteins locally modulate NL interactions.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histones , Nuclear Lamina , Animals , Cell Cycle Proteins/genetics , Chromatin/metabolism , DNA/metabolism , Histones/metabolism , Mice , Nuclear Lamina/chemistry , Cohesins
3.
Nucleic Acids Res ; 50(8): 4258-4271, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35420130

ABSTRACT

The link between genomic structure and biological function is yet to be consolidated, it is, however, clear that physical manipulation of the genome, driven by the activity of a variety of proteins, is a crucial step. To understand the consequences of the physical forces underlying genome organization, we build a coarse-grained polymer model of the genome, featuring three fundamentally distinct classes of interactions: lengthwise compaction, i.e., compaction of chromosomes along its contour, self-adhesion among epigenetically similar genomic segments, and adhesion of chromosome segments to the nuclear envelope or lamina. We postulate that these three types of interactions sufficiently represent the concerted action of the different proteins organizing the genome architecture and show that an interplay among these interactions can recapitulate the architectural variants observed across the tree of life. The model elucidates how an interplay of forces arising from the three classes of genomic interactions can drive drastic, yet predictable, changes in the global genome architecture, and makes testable predictions. We posit that precise control over these interactions in vivo is key to the regulation of genome architecture.


Subject(s)
Chromosomes , Genome , Nuclear Lamina , Chromosomes/chemistry , Chromosomes/metabolism , Genomics , Nuclear Envelope , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism
4.
Chromosome Res ; 30(1): 123-136, 2022 03.
Article in English | MEDLINE | ID: mdl-35239049

ABSTRACT

More than one third of the mammalian genome is in a close association with the nuclear lamina, thus these genomic regions were termed lamina-associated domains (LADs). This association is fundamental for many aspects of chromatin biology including transcription, replication, and DNA damage repair. LADs association with the nuclear envelope is thought to be dependent on two major mechanisms: The first mechanism is the interaction between nuclear membrane proteins such as LBR with heterochromatin modifications that are enriched in LADs chromatin. The second mechanism is based on proteins that bind the borders of the LADs and support the association of the LADs with the nuclear envelope. Two factors were suggested to support the second mechanism: CCCTC-binding factor (CTCF) and YY1 based on their enriched binding to LADs borders. However, this mechanism has not been proven yet at a whole genome level. Here, to test if CTCF supports the LADs landscape, we generated melanoma cells with a partial loss of function (pLoF) of CTCF by the CRISPR-Cas9 system and determined the LADs landscape by lamin B ChIP-seq analysis. We found that under regular growth conditions, CTCF pLoF led to modest changes in the LADs landscape that included an increase in the signal of 2% of the LADs and a decrease in the signal of 8% of the LADs. However, CTCF importance for the LADs landscape was much higher upon induction of a chromatin stress. We induced chromatin stress by inhibiting RNA polymerase II, an intervention that is known to alter chromatin compaction and supercoiling. Notably, only in CTCF pLoF cells, the chromatin stress led to the dissociation of 7% of the LADs from the lamina. The CTCF-dependent LADs had almost three times shorter median length than the non-affected LADs, were enriched in CTCF binding at their borders, and were higher in their facultative-status (cell-type specific). Thus, it appears that CTCF is a key factor in facilitating the association of short facultative LADs with the nuclear lamina upon chromatin stress.


Subject(s)
Chromatin , Nuclear Lamina , Animals , Chromatin/genetics , Chromatin/metabolism , Genome , Genomics , Heterochromatin/metabolism , Mammals/genetics , Nuclear Lamina/chemistry , Nuclear Lamina/genetics , Nuclear Lamina/metabolism
5.
Methods Mol Biol ; 2157: 159-172, 2021.
Article in English | MEDLINE | ID: mdl-32820403

ABSTRACT

The organization of DNA within the eukaryotic nucleus is important for cellular processes such as regulation of gene expression and repair of DNA damage. To comprehend cell-to-cell variation within a complex system, systematic analysis of individual cells is necessary. While many tools exist to capture DNA conformation and chromatin context, these methods generally require large populations of cells for sufficient output. Here we describe single-cell DamID, a technique to capture contacts between DNA and a given protein of interest. By fusing the bacterial methyltransferase Dam to nuclear lamina protein lamin B1, genomic regions in contact with the nuclear periphery can be mapped. Single-cell DamID generates contact maps with sufficient throughput and resolution to reliably identify patterns of similarity as well as variation in nuclear organization of interphase chromosomes.


Subject(s)
Chromatin/metabolism , Genomics/methods , Nuclear Lamina/metabolism , Animals , Chromatin/chemistry , DNA/chemistry , DNA/metabolism , Humans , Lamin Type B/chemistry , Lamin Type B/metabolism , Nuclear Lamina/chemistry
6.
Genome Biol ; 21(1): 85, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32241294

ABSTRACT

At the nuclear periphery, associations of chromatin with the nuclear lamina through lamina-associated domains (LADs) aid functional organization of the genome. We review the organization of LADs and provide evidence of LAD heterogeneity from cell ensemble and single-cell data. LADs are typically repressive environments in the genome; nonetheless, we discuss findings of lamin interactions with regulatory elements of active genes, and the role lamins may play in genome regulation. We address the relationship between LADs and other genome organizers, and the involvement of LADs in laminopathies. The current data lay the basis for future studies on the significance of lamin-chromatin interactions in health and disease.


Subject(s)
Nuclear Lamina/physiology , Chromatin/metabolism , Euchromatin/metabolism , Genome , Heterochromatin/metabolism , Humans , Intermediate Filaments/metabolism , Laminopathies/genetics , Lamins/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Single-Cell Analysis
7.
Elife ; 92020 01 28.
Article in English | MEDLINE | ID: mdl-31989921

ABSTRACT

The nucleus of oocytes (germinal vesicle) is unusually large and its nuclear envelope (NE) is densely packed with nuclear pore complexes (NPCs) that are stockpiled for embryonic development. We showed that breakdown of this specialized NE is mediated by an Arp2/3-nucleated F-actin 'shell' in starfish oocytes, in contrast to microtubule-driven tearing in mammalian fibroblasts. Here, we address the mechanism of F-actin-driven NE rupture by correlated live-cell, super-resolution and electron microscopy. We show that actin is nucleated within the lamina, sprouting filopodia-like spikes towards the nuclear membranes. These F-actin spikes protrude pore-free nuclear membranes, whereas the adjoining stretches of membrane accumulate NPCs that are associated with the still-intact lamina. Packed NPCs sort into a distinct membrane network, while breaks appear in ER-like, pore-free regions. We reveal a new function for actin-mediated membrane shaping in nuclear rupture that is likely to have implications in other contexts, such as nuclear rupture observed in cancer cells.


Subject(s)
Actins , Nuclear Envelope , Oocytes/cytology , Actins/chemistry , Actins/metabolism , Animals , Microscopy, Electron , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Starfish
8.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118530, 2019 12.
Article in English | MEDLINE | ID: mdl-31415840

ABSTRACT

Phasor-assisted Metal Induced Energy Transfer-Fluorescence Lifetime Imaging Microscopy (MIET-FLIM) nanoscopy is introduced as a powerful tool for functional cell biology research. Thin metal substrates can be used to obtain axial super-resolution via nanoscale distance-dependent MIET from fluorescent dyes towards a nearby metal layer, thereby creating fluorescence lifetime contrast between dyes located at different nanoscale distance from the metal. Such data can be used to achieve axially super-resolved microscopy images, a process known as MIET-FLIM nanoscopy. Suitability of the phasor approach in MIET-FLIM nanoscopy is first demonstrated using nanopatterned substrates, and furthermore applied to characterize the distance distribution of the epithelial basal membrane of a biological cell from the gold substrate. The phasor plot of an entire cell can be used to characterize the full Förster resonance energy transfer (FRET) trajectory as a large distance heterogeneity within the sensing range of about 100 nm from the metal surface is present due to the extended shape of cell with curvatures. In contrast, the different proteins of nuclear lamina show strong confinement close to the nuclear envelope in nanoscale. We find the lamin B layer resides in average at shorter distances from the gold surface compared to the lamin A/C layer located in more extended ranges. This and the observed heterogeneity of the protein layer thicknesses suggests that A- and B-type lamins form distinct networks in the nuclear lamina. Our results provide detailed insights for the study of the different roles of lamin proteins in chromatin tethering and nuclear mechanics.


Subject(s)
Fluorescence Resonance Energy Transfer , Nanotechnology , Nuclear Lamina/chemistry , Nuclear Proteins/metabolism , A549 Cells , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Microscopy, Fluorescence , Nuclear Lamina/metabolism , Nuclear Proteins/chemistry , Optical Imaging
9.
Cells ; 8(4)2019 04 18.
Article in English | MEDLINE | ID: mdl-31003483

ABSTRACT

The nuclear lamina consists of a dense fibrous meshwork of nuclear lamins, Type V intermediate filaments, and is ~14 nm thick according to recent cryo-electron tomography studies. Recent advances in light microscopy have extended the resolution to a scale allowing for the fine structure of the lamina to be imaged in the context of the whole nucleus. We review quantitative approaches to analyze the imaging data of the nuclear lamina as acquired by structured illumination microscopy (SIM) and single molecule localization microscopy (SMLM), as well as the requisite cell preparation techniques. In particular, we discuss the application of steerable filters and graph-based methods to segment the structure of the four mammalian lamin isoforms (A, C, B1, and B2) and extract quantitative information.


Subject(s)
Nuclear Lamina/chemistry , Nuclear Lamina/ultrastructure , Animals , Cell Nucleus/metabolism , Electron Microscope Tomography/methods , Humans , Intermediate Filament Proteins/analysis , Intermediate Filaments/chemistry , Intermediate Filaments/physiology , Lamin Type A/analysis , Lamin Type B/analysis , Lamins/chemistry , Lamins/physiology , Nuclear Lamina/physiology , Protein Isoforms/analysis
10.
Nucleus ; 9(1): 258-275, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29637811

ABSTRACT

Mammalian nuclei are equipped with a framework of intermediate filaments that function as a karyoskeleton. This nuclear scaffold, formed primarily by lamins (A-type and B-type), maintains the spatial and functional organization of the genome and of sub-nuclear compartments. Over the past decade, a body of evidence has highlighted the significance of these structural nuclear proteins in the maintenance of nuclear architecture and mechanical stability, as well as genome function and integrity. The importance of these structures is now unquestioned given the wide range of degenerative diseases that stem from LMNA gene mutations, including muscular dystrophy disorders, peripheral neuropathies, lipodystrophies, and premature aging syndromes. Here, we review our knowledge about how alterations in nuclear lamins, either by mutation or reduced expression, impact cellular mechanisms that maintain genome integrity. Despite the fact that DNA replication is the major source of DNA damage and genomic instability in dividing cells, how alterations in lamins function impact replication remains minimally explored. We summarize recent studies showing that lamins play a role in DNA replication, and that the DNA damage that accumulates upon lamins dysfunction is elicited in part by deprotection of replication forks. We also discuss the emerging model that DNA damage and replication stress are "sensed" at the cytoplasm by proteins that normally survey this space in search of foreign nucleic acids. In turn, these cytosolic sensors activate innate immune responses, which are materializing as important players in aging and cancer, as well as in the response to cancer immunotherapy.


Subject(s)
DNA/genetics , Genomic Instability , Interferons/genetics , Lamins/genetics , Animals , DNA Damage , DNA Replication , Humans , Interferons/metabolism , Lamins/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism
11.
Nucleus ; 9(1): 216-226, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29517398

ABSTRACT

The nuclear lamina contributes to the regulation of gene expression and to chromatin organization. Mutations in A-type nuclear lamins cause laminopathies, some of which are associated with a loss of heterochromatin at the nuclear periphery. Until recently however, little if any information has been provided on where and how lamin A interacts with the genome and on how disease-causing lamin A mutations may rearrange genome conformation. Here, we review aspects of nuclear lamin association with the genome. We highlight recent evidence of reorganization of lamin A-chromatin interactions in cellular models of laminopathies, and implications on the 3-dimensional rearrangement of chromatin in these models, including patient cells. We discuss how a hot-spot lipodystrophic lamin A mutation alters chromatin conformation and epigenetic patterns at an anti-adipogenic locus, and conclude with remarks on links between lamin A, Polycomb and the pathophysiology of laminopathies. The recent findings presented here collectively argue towards a deregulation of large-scale and local spatial genome organization by a subset of lamin A mutations causing laminopathies.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Lamin Type A/genetics , Musculoskeletal Diseases/genetics , Mutation , Nuclear Lamina/chemistry , Nuclear Lamina/genetics , Chromatin/metabolism , Humans , Lamin Type A/metabolism , Musculoskeletal Diseases/metabolism , Nuclear Lamina/metabolism
12.
Curr Opin Cell Biol ; 48: 63-71, 2017 10.
Article in English | MEDLINE | ID: mdl-28641117

ABSTRACT

The nucleus is a mechanosensitive and load-bearing structure. Structural components of the nucleus interact to maintain nuclear integrity and have become subjects of exciting research that is relevant to cell and developmental biology. Here we outline the boundaries of what is known about key architectural elements within the nucleus and highlight their potential structural and transcriptional regulatory functions.


Subject(s)
Cell Nucleus/metabolism , Animals , Cell Nucleus/chemistry , Cell Nucleus/genetics , Chromatin/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Mechanotransduction, Cellular , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism
13.
Cell ; 169(5): 780-791, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525751

ABSTRACT

In metazoan cell nuclei, hundreds of large chromatin domains are in close contact with the nuclear lamina. Such lamina-associated domains (LADs) are thought to help organize chromosomes inside the nucleus and have been associated with gene repression. Here, we discuss the properties of LADs, the molecular mechanisms that determine their association with the nuclear lamina, their dynamic links with other nuclear compartments, and their proposed roles in gene regulation.


Subject(s)
Cell Nucleus/chemistry , Chromatin/chemistry , Animals , Cell Nucleus/metabolism , Gene Expression Regulation , Heterochromatin , Humans , Lamins/metabolism , Nuclear Lamina/chemistry , Nuclear Pore/metabolism
14.
Nature ; 543(7644): 261-264, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28241138

ABSTRACT

The nuclear lamina is a fundamental constituent of metazoan nuclei. It is composed mainly of lamins, which are intermediate filament proteins that assemble into a filamentous meshwork, bridging the nuclear envelope and chromatin. Besides providing structural stability to the nucleus, the lamina is involved in many nuclear activities, including chromatin organization, transcription and replication. However, the structural organization of the nuclear lamina is poorly understood. Here we use cryo-electron tomography to obtain a detailed view of the organization of the lamin meshwork within the lamina. Data analysis of individual lamin filaments resolves a globular-decorated fibre appearance and shows that A- and B-type lamins assemble into tetrameric filaments of 3.5 nm thickness. Thus, lamins exhibit a structure that is remarkably different from the other canonical cytoskeletal elements. Our findings define the architecture of the nuclear lamin meshworks at molecular resolution, providing insights into their role in scaffolding the nuclear lamina.


Subject(s)
Lamins/chemistry , Lamins/ultrastructure , Nuclear Lamina/chemistry , Nuclear Lamina/ultrastructure , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cryoelectron Microscopy , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Humans , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/ultrastructure , Lamins/metabolism , Mice , Nuclear Lamina/metabolism , Tomography
15.
J Mech Behav Biomed Mater ; 63: 35-43, 2016 10.
Article in English | MEDLINE | ID: mdl-27341289

ABSTRACT

Intermediate filament (IF) proteins are known mainly by their propensity to form viscoelastic filamentous networks within cells. In addition, IF-proteins are essential parts of various biological materials, such as horn and hagfish slime threads, which exhibit a range of mechanical properties from hard to elastic. These properties and their self-assembly nature made IF-proteins attractive building blocks for biomimetic and biological materials in diverse applications. Here we show that a type V IF-protein, the Caenorhabditis elegans nuclear lamin (Ce-lamin), is a promising building block for protein-based fibers. Electron cryo-tomography of vitrified sections enabled us to depict the higher ordered assembly of the Ce-lamin into macroscopic fibers through the creation of paracrystalline fibers, which are prominent in vitro structures of lamins. The lamin fibers respond to tensile force as other IF-protein-based fibers, i.e., hagfish slime threads, and possess unique mechanical properties that may potentially be used in certain applications. The self-assembly nature of lamin proteins into a filamentous structure, which is further assembled into a complex network, can be easily modulated. This knowledge may lead to a better understanding of the relationship in IF-proteins-based fibers and materials, between their hierarchical structures and their mechanical properties.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans , Lamins/chemistry , Nuclear Lamina/chemistry , Animals
16.
Histochem Cell Biol ; 145(4): 419-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883443

ABSTRACT

The nuclear lamina represents a multifunctional platform involved in such diverse yet interconnected processes as spatial organization of the genome, maintenance of mechanical stability of the nucleus, regulation of transcription and replication. Most of lamina activities are exerted through tethering of lamina-associated chromatin domains (LADs) to the nuclear periphery. Yet, the lamina is a dynamic structure demonstrating considerable expansion during the cell cycle to accommodate increased number of LADs formed during DNA replication. We analyzed dynamics of nuclear growth during interphase and changes in lamina structure as a function of cell cycle progression. The nuclear lamina demonstrates steady growth from G1 till G2, while quantitative analysis of lamina meshwork by super-resolution microscopy revealed that microdomain organization of the lamina is maintained, with lamin A and lamin B microdomain periodicity and interdomain gap sizes unchanged. FRAP analysis, in contrast, demonstrated differences in lamin A and B1 exchange rates; the latter showing higher recovery rate in S-phase cells. In order to further analyze the mechanism of lamina growth in interphase, we generated a lamina-free nuclear envelope in living interphase cells by reversible hypotonic shock. The nuclear envelope in nuclear buds formed after such a treatment initially lacked lamins, and analysis of lamina formation revealed striking difference in lamin A and B1 assembly: lamin A reassembled within 30 min post-treatment, whereas lamin B1 did not incorporate into the newly formed lamina at all. We suggest that in somatic cells lamin B1 meshwork growth is coordinated with replication of LADs, and lamin A meshwork assembly seems to be chromatin-independent process.


Subject(s)
Interphase , Nuclear Lamina/metabolism , Animals , Cells, Cultured , Cricetulus , Humans , Mice , Nuclear Lamina/chemistry , Swine
17.
Sci Rep ; 5: 17186, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26603343

ABSTRACT

In the interphase nucleus, chromatin is organized into three-dimensional conformation to coordinate genome functions. The lamina-chromatin association is important to facilitate higher-order chromatin in mammalian cells, but its biological significances and molecular mechanisms remain poorly understood. One obstacle is that the list of lamina-associated proteins remains limited, presumably due to the inherent insolubility of lamina proteins. In this report, we identified 182 proteins associated with lamin B1 (a constitutive component of lamina) in mouse hepatocytes, by adopting virus-based proximity-dependent biotin identification. These proteins are functionally related to biological processes such as chromatin organization. As an example, we validated the association between lamin B1 and core histone macroH2A1, a histone associated with repressive chromatin. Furthermore, we mapped Lamina-associated domains (LADs) in mouse liver cells and found that boundaries of LADs are enriched for macroH2A. More interestingly, knocking-down of macroH2A1 resulted in the release of heterochromatin foci marked by histone lysine 9 trimethylation (H3K9me3) and the decondensation of global chromatin structure. However, down-regulation of lamin B1 led to redistribution of macroH2A1. Taken together, our data indicated that macroH2A1 is associated with lamina and is required to maintain chromatin architecture in mouse liver cells.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Nuclear Lamina/metabolism , Animals , Chromatin/chemistry , Chromatography, High Pressure Liquid , Down-Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Heterochromatin/metabolism , Histones/antagonists & inhibitors , Histones/genetics , Lamin Type B/antagonists & inhibitors , Lamin Type B/genetics , Lamin Type B/metabolism , Methylation , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Lamina/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Tandem Mass Spectrometry
18.
Nat Mater ; 14(12): 1252-1261, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26301768

ABSTRACT

Nuclear lamins play central roles at the intersection between cytoplasmic signalling and nuclear events. Here, we show that at least two N- and C-terminal lamin epitopes are not accessible at the basal side of the nuclear envelope under environmental conditions known to upregulate cell contractility. The conformational epitope on the Ig-domain of A-type lamins is more buried in the basal than apical nuclear envelope of human mesenchymal stem cells undergoing osteogenesis (but not adipogenesis), and in fibroblasts adhering to rigid (but not soft) polyacrylamide hydrogels. This structural polarization of the lamina is promoted by compressive forces, emerges during cell spreading, and requires lamin A/C multimerization, intact nucleoskeleton-cytoskeleton linkages (LINC), and apical-actin stress-fibre assembly. Notably, the identified Ig-epitope overlaps with emerin, DNA and histone binding sites, and comprises various laminopathy mutation sites. Our findings should help decipher how the physical properties of cellular microenvironments regulate nuclear events.


Subject(s)
Cytoskeleton/metabolism , Lamin Type A/metabolism , Nuclear Lamina/metabolism , Biopolymers/chemistry , Biopolymers/metabolism , Humans , Lamin Type A/chemistry , Nuclear Lamina/chemistry , Protein Conformation
19.
Mol Biol Cell ; 26(22): 4075-86, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26310440

ABSTRACT

The nuclear lamina is a key structural element of the metazoan nucleus. However, the structural organization of the major proteins composing the lamina is poorly defined. Using three-dimensional structured illumination microscopy and computational image analysis, we characterized the supramolecular structures of lamin A, C, B1, and B2 in mouse embryo fibroblast nuclei. Each isoform forms a distinct fiber meshwork, with comparable physical characteristics with respect to mesh edge length, mesh face area and shape, and edge connectivity to form faces. Some differences were found in face areas among isoforms due to variation in the edge lengths and number of edges per face, suggesting that each meshwork has somewhat unique assembly characteristics. In fibroblasts null for the expression of either lamins A/C or lamin B1, the remaining lamin meshworks are altered compared with the lamin meshworks in wild-type nuclei or nuclei lacking lamin B2. Nuclei lacking LA/C exhibit slightly enlarged meshwork faces and some shape changes, whereas LB1-deficient nuclei exhibit primarily a substantial increase in face area. These studies demonstrate that individual lamin isoforms assemble into complex networks within the nuclear lamina and that A- and B-type lamins have distinct roles in maintaining the organization of the nuclear lamina.


Subject(s)
Nuclear Lamina/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Fibroblasts/metabolism , HeLa Cells , Humans , Imaging, Three-Dimensional/methods , Intermediate Filaments/metabolism , Mice , Microscopy/methods , Morphogenesis , Nuclear Lamina/chemistry , Protein Isoforms
20.
Nucleus ; 6(3): 236-46, 2015.
Article in English | MEDLINE | ID: mdl-25996284

ABSTRACT

The cell nucleus is structurally and functionally organized by lamins, intermediate filament proteins that form the nuclear lamina. Point mutations in genes that encode a specific subset of lamins, the A-type lamins, cause a spectrum of diseases termed laminopathies. Recent evidence points to a role for A-type lamins in intracellular redox homeostasis. To determine whether lamin A/C depletion and prelamin A accumulation differentially induce oxidative stress, we have performed a quantitative microscopy-based analysis of reactive oxygen species (ROS) levels and mitochondrial membrane potential (Δψm) in human fibroblasts subjected to sustained siRNA-mediated knockdown of LMNA and ZMPSTE24, respectively. We measured a highly significant increase in basal ROS levels and an even more prominent rise of induced ROS levels in lamin A/C depleted cells, eventually resulting in Δψm hyperpolarization and apoptosis. Depletion of ZMPSTE24 on the other hand, triggered a senescence pathway that was associated with moderately increased ROS levels and a transient Δψm depolarization. Both knockdowns were accompanied by an upregulation of several ROS detoxifying enzymes. Taken together, our data suggest that both persistent prelamin A accumulation and lamin A/C depletion elevate ROS levels, but to a different extent and with different effects on cell fate. This may contribute to the variety of disease phenotypes witnessed in laminopathies.


Subject(s)
Fibroblasts/metabolism , Lamin Type A/metabolism , Mitochondria/metabolism , Nuclear Lamina/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Fibroblasts/cytology , Gene Expression Regulation , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Membrane Potential, Mitochondrial , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/pathology , Nuclear Lamina/chemistry , Oxidative Stress , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL