Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 79(2): 126, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35132494

ABSTRACT

B-type lamins are fundamental components of the nuclear lamina, a complex structure that acts as a scaffold for organization and function of the nucleus. Lamin B1 and B2, the most represented isoforms, are encoded by LMNB1 and LMNB2 gene, respectively. All B-type lamins are synthesized as precursors and undergo sequential post-translational modifications to generate the mature protein. B-type lamins are involved in a wide range of nuclear functions, including DNA replication and repair, regulation of chromatin and nuclear stiffness. Moreover, lamins B1 and B2 regulate several cellular processes, such as tissue development, cell cycle, cellular proliferation, senescence, and DNA damage response. During embryogenesis, B-type lamins are essential for organogenesis, in particular for brain development. As expected from the numerous and pivotal functions of B-type lamins, mutations in their genes or fluctuations in their expression levels are critical for the onset of several diseases. Indeed, a growing range of human disorders have been linked to lamin B1 or B2, increasing the complexity of the group of diseases collectively known as laminopathies. This review highlights the recent findings on the biological role of B-type lamins under physiological or pathological conditions, with a particular emphasis on brain disorders and cancer.


Subject(s)
Brain Diseases/metabolism , Lamin Type B/physiology , Laminopathies/metabolism , Neoplasms/metabolism , Animals , Humans
2.
Biosci Rep ; 41(1)2021 01 29.
Article in English | MEDLINE | ID: mdl-33416073

ABSTRACT

Triple negative breast cancer (TNBC) is a more common type of breast cancer with high distant metastasis and poor prognosis. The potential role of lamins in cancer progression has been widely revealed. However, the function of lamin B2 (LMNB2) in TNBC progression is still unclear. The present study aimed to investigate the role of LMNB2 in TNBC. The cancer genome atlas (TCGA) database analysis and immunohistochemistry (IHC) were performed to examine LMNB2 expression levels. LMNB2 short hairpin RNA plasmid or lentivirus was used to deplete the expression of LMNB2 in human TNBC cell lines including MDA-MB-468 and MDA-MB-231. Alterations in cell proliferation and apoptosis in vitro and the nude mouse tumorigenicity assay in vivo were subsequently analyzed. The human TNBC tissues shown high expression of LMNB2 according to the bioinformation analysis and IHC assays. LMNB2 expression was correlated with the clinical pathological features of TNBC patients, including pTNM stage and lymph node metastasis. Through in vitro and in vivo assays, we confirmed LMNB2 depletion suppressed the proliferation and induced the apoptosis of TNBC cells, and inhibited tumor growth of TNBC cells in mice, with the decrease in Ki67 expression or the increase in caspase-3 expression. In conclusion, LMNB2 may promote TNBC progression and could serve as a potential therapeutic target for TNBC treatment.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Lamin Type B/physiology , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Lamin Type B/genetics , Male , Middle Aged , RNA, Messenger/genetics
3.
Sheng Wu Gong Cheng Xue Bao ; 34(11): 1742-1749, 2018 Nov 25.
Article in Chinese | MEDLINE | ID: mdl-30499270

ABSTRACT

Lamin B1 is one of the essential members of the nuclear lamina protein family. Its main function is to maintain the integrity of nuclear skeleton, as well as to participate in the cell proliferation and aging by affecting the chromosome distribution. gene expression, and DNA damage repair. The abnormal expression of lamin B1 is related to certain diseases, including neurological diseases [e.g. neural tube defects (NDTs), adult-onset autosomal dominant leukodystrophy (ADLD)] and tumors (e.g. pancreatic cancer). It is also a potential tumor marker as well as drug target. Further research on lamin B1 will help people understand the molecular mechanism of the emergence and development of neural system diseases and tumors, and define a new future in drug target.


Subject(s)
Lamin Type B/physiology , Neoplasms , Nervous System Diseases , Cell Nucleus , Gene Expression , Humans
4.
Yakugaku Zasshi ; 137(11): 1315-1321, 2017.
Article in Japanese | MEDLINE | ID: mdl-29093367

ABSTRACT

Two types of cell death, necrosis and apoptosis, are defined in terms of cell death morphological features. We have been studying the mechanisms by which cell death processes are switched during the treatment of mouse tumor FM3A with anticancer, 5-fluoro-2'-deoxyuridine (FUdR): it induces original clone F28-7 to necrosis, but its sub-clone F28-7-A to apoptosis. We identified several such switch regulators of cell death: heat shock protein 90 (HSP90), lamin-B1, cytokeratin-19, and activating transcription factor 3 (ATF3), by using transcriptomic, proteomic analyses and siRNA screening. For example, the inhibition of HSP90 by its inhibitor geldanamycin in F28-7 caused a shift from necrosis to apoptosis. We also observed that the knockdown of lamin-B1, cytokeratin-19, or ATF3 expression in F28-7 resulted in a shift from necrosis to apoptosis. Recently, we used microRNA (miRNA, miR) microarray analyses to investigate the miRNA expression profiles in these sister cells. The miR-351 and miR-743a were expressed at higher levels in F28-7-A than in F28-7. Higher expression of miR-351 or miR-743a in F28-7, induced by transfecting the miR mimics, resulted in a switch of cell death mode: necrosis to apoptosis. Furthermore, transfection of an miR-351 inhibitor into F28-7-A resulted in morphological changes, and mode of cell death from apoptosis to necrosis. These findings suggest that the identified cell death regulators may have key roles in switching cell death mode. Possible mechanisms involving cell death regulators in the switch of necrosis or apoptosis are discussed. We propose a novel anticancer strategy targeting the switch regulators of necrosis or apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Deoxyuridine/analogs & derivatives , Necrosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Animals , Apoptosis/genetics , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Deoxyuridine/pharmacology , Deoxyuridine/therapeutic use , Gene Expression Profiling , HSP90 Heat-Shock Proteins/physiology , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Lamin Type B/genetics , Lamin Type B/physiology , Mice , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Necrosis/genetics
5.
Mol Biol Cell ; 28(23): 3333-3348, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28931598

ABSTRACT

Matrix stiffness that is sensed by a cell or measured by a purely physical probe reflects the intrinsic elasticity of the matrix and also how thick or thin the matrix is. Here, mesenchymal stem cells (MSCs) and their nuclei spread in response to thickness-corrected matrix microelasticity, with increases in nuclear tension and nuclear stiffness resulting from increases in myosin-II and lamin-A,C. Linearity between the widely varying projected area of a cell and its nucleus across many matrices, timescales, and myosin-II activity levels indicates a constant ratio of nucleus-to-cell volume, despite MSCs' lineage plasticity. Nuclear envelope fluctuations are suppressed on the stiffest matrices, and fluctuation spectra reveal a high nuclear tension that matches trends from traction force microscopy and from increased lamin-A,C. Transcriptomes of many diverse tissues and MSCs further show that lamin-A,C's increase with tissue or matrix stiffness anti-correlates with lamin-B receptor (LBR), which contributes to lipid/sterol biosynthesis. Adipogenesis (a soft lineage) indeed increases LBR:lamin-A,C protein stoichiometry in MSCs versus osteogenesis (stiff). The two factors compete for lamin-B in response to matrix elasticity, knockdown, myosin-II inhibition, and even constricted migration that disrupts and segregates lamins in situ. Matrix stiffness-driven contractility thus tenses the nucleus to favor lamin-A,C accumulation and suppress soft tissue phenotypes.


Subject(s)
Lamin Type A/metabolism , Lamin Type B/metabolism , Adipogenesis/physiology , Adipose Tissue/physiology , Animals , Cell Differentiation , Cell Nucleus/metabolism , Elasticity , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Lamin Type A/physiology , Lamin Type B/physiology , Lamins/metabolism , Mesenchymal Stem Cells/metabolism , Nuclear Envelope/metabolism , Osteogenesis/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Lamin B Receptor
6.
Mol Biol Cell ; 27(1): 35-47, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26510501

ABSTRACT

Lamin B1, a key component of the nuclear lamina, plays an important role in brain development and function. A duplication of the human lamin B1 (LMNB1) gene has been linked to adult-onset autosomal dominant leukodystrophy, and mouse and human loss-of-function mutations in lamin B1 are susceptibility factors for neural tube defects. In the mouse, experimental ablation of endogenous lamin B1 (Lmnb1) severely impairs embryonic corticogenesis. Here we report that in primary mouse cortical neurons, LMNB1 overexpression reduces axonal outgrowth, whereas deficiency of endogenous Lmnb1 results in aberrant dendritic development. In the absence of Lmnb1, both the length and complexity of dendrites are reduced, and their growth is unresponsive to KCl stimulation. This defective dendritic outgrowth stems from impaired ERK signaling. In Lmnb1-null neurons, ERK is correctly phosphorylated, but phospho-ERK fails to translocate to the nucleus, possibly due to delocalization of nuclear pore complexes (NPCs) at the nuclear envelope. Taken together, these data highlight a previously unrecognized role of lamin B1 in dendrite development of mouse cortical neurons through regulation of nuclear shuttling of specific signaling molecules and NPC distribution.


Subject(s)
Dendrites/physiology , Lamin Type B/physiology , Animals , Axons/metabolism , Dendrites/metabolism , Female , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Mice, Mutant Strains , Mutation , Neurogenesis/physiology , Neurons/metabolism , Nuclear Lamina/metabolism , Pregnancy
7.
Fly (Austin) ; 8(2): 63-7, 2014.
Article in English | MEDLINE | ID: mdl-25483250

ABSTRACT

Recent studies show that nuclear lamins, the type V intermediate filament proteins, are required for proper building of at least some organs. As the major structural components of the nuclear lamina found underneath the inner nuclear membranes, lamins are ubiquitously expressed in all animal cells. How the broadly expressed lamins support the building of specific tissues is not understood. By studying Drosophila testis, we have uncovered a mechanism by which lamin-B functions in the cyst stem cell (CySC) and its differentiated cyst cell, the cell types known to form the niche/microenvironment for the germline stem cells (GSC) and the developing germ line, to ensure testis organogenesis (1). In this extra view, we discuss some remaining questions and the implications of our findings in the understanding of how the ubiquitous nuclear lamina regulates tissue building in a context-dependent manner.


Subject(s)
Drosophila melanogaster/physiology , Lamin Type B/physiology , Nuclear Lamina/physiology , Stem Cells/physiology , Testis/physiology , Animals , Epidermal Growth Factor/metabolism , Female , Male , Morphogenesis , Nuclear Pore Complex Proteins/metabolism
8.
FASEB J ; 28(8): 3423-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24732130

ABSTRACT

Nuclear lamin B1 (LMNB1) constitutes one of the major structural proteins in the lamina mesh. We silenced the expression of LMNB1 by RNA interference in the colon cancer cell line DLD-1 and showed a dramatic redistribution of H3K27me3 from the periphery to a more homogeneous nuclear dispersion. In addition, we observed telomere attrition and an increased frequency of micronuclei and nuclear blebs. By 3D-FISH analyses, we demonstrated that the volume and surface of chromosome territories were significantly larger in LMNB1-depleted cells, suggesting that LMNB1 is required to maintain chromatin condensation in interphase nuclei. These changes led to a prolonged S phase due to activation of Chk1. Finally, silencing of LMNB1 resulted in extensive changes in alternative splicing of multiple genes and in a higher number of enlarged nuclear speckles. Taken together, our results suggest a mechanistic role of the nuclear lamina in the organization of chromosome territories, maintenance of genome integrity and proper gene splicing.


Subject(s)
Lamin Type B/physiology , S Phase/physiology , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Nucleus/ultrastructure , Chromosomes, Human, Pair 18/ultrastructure , Chromosomes, Human, Pair 19/ultrastructure , Colonic Neoplasms/pathology , Heterochromatin/physiology , Histones/metabolism , Humans , In Situ Hybridization, Fluorescence , Interphase , Lamin Type B/deficiency , Methylation , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , S Phase/drug effects , Telomere Shortening/drug effects
9.
J Cell Biol ; 204(5): 669-82, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24567359

ABSTRACT

Cell migration through solid tissue often involves large contortions of the nucleus, but biological significance is largely unclear. The nucleoskeletal protein lamin-A varies both within and between cell types and was shown here to contribute to cell sorting and survival in migration through constraining micropores. Lamin-A proved rate-limiting in 3D migration of diverse human cells that ranged from glioma and adenocarcinoma lines to primary mesenchymal stem cells (MSCs). Stoichiometry of A- to B-type lamins established an activation barrier, with high lamin-A:B producing extruded nuclear shapes after migration. Because the juxtaposed A and B polymer assemblies respectively conferred viscous and elastic stiffness to the nucleus, subpopulations with different A:B levels sorted in 3D migration. However, net migration was also biphasic in lamin-A, as wild-type lamin-A levels protected against stress-induced death, whereas deep knockdown caused broad defects in stress resistance. In vivo xenografts proved consistent with A:B-based cell sorting, and intermediate A:B-enhanced tumor growth. Lamins thus impede 3D migration but also promote survival against migration-induced stresses.


Subject(s)
Cell Movement/physiology , Lamin Type A/physiology , Lamin Type B/physiology , Apoptosis , Cell Line, Tumor , Cell Nucleus/ultrastructure , Cell Nucleus Shape , Cell Survival , Gene Knockdown Techniques , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type B/chemistry , Lamin Type B/genetics , Protein Structure, Tertiary
10.
Adv Exp Med Biol ; 773: 127-42, 2014.
Article in English | MEDLINE | ID: mdl-24563346

ABSTRACT

While speculation has centered on a role for nuclear lamins in tumor progression for many years, most of the diseases that have been linked to lamin mutation are dystrophic in nature, often limiting the proliferation potential of affected cells in vivo and in vitro. Nevertheless, these lamin mutations, particularly in the LMNA gene that encodes A-type lamins, have provided an interesting tool set to understand functions of nuclear intermediate filament proteins in cell cycle progress and various means of exit, including quiescence, senescence, and differentiation down various lineages. The picture that has emerged is complex with lamins controlling the activity of key cell cycle factors such as the retinoblastoma protein (RB) and interacting with several important signal transduction pathways. Here we describe the current state of knowledge and speculate that lamins may be intimately involved in the regulation of cell proliferation, acting at the interface between cancer and aging.


Subject(s)
Aging/physiology , Cell Cycle/physiology , Lamin Type A/physiology , Lamin Type B/physiology , Retinoblastoma Protein/physiology , Humans , Signal Transduction
11.
Science ; 334(6063): 1706-10, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22116031

ABSTRACT

B-type lamins, the major components of the nuclear lamina, are believed to be essential for cell proliferation and survival. We found that mouse embryonic stem cells (ESCs) do not need any lamins for self-renewal and pluripotency. Although genome-wide lamin-B binding profiles correlate with reduced gene expression, such binding is not directly required for gene silencing in ESCs or trophectoderm cells. However, B-type lamins are required for proper organogenesis. Defects in spindle orientation in neural progenitor cells and migration of neurons probably cause brain disorganizations found in lamin-B null mice. Thus, our studies not only disprove several prevailing views of lamin-Bs but also establish a foundation for redefining the function of the nuclear lamina in the context of tissue building and homeostasis.


Subject(s)
Embryonic Stem Cells/physiology , Lamin Type B/physiology , Organogenesis , Animals , Body Size , Brain/cytology , Brain/embryology , Cell Cycle , Cell Differentiation , Cell Movement , Cells, Cultured , Chromatin/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation, Developmental , Gene Silencing , Lamin Type B/genetics , Lamin Type B/metabolism , Male , Mice , Mice, Knockout , Neural Stem Cells/cytology , Neurons/cytology , Nuclear Lamina/physiology , Organ Size , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Promoter Regions, Genetic , Spindle Apparatus/physiology , Spindle Apparatus/ultrastructure , Transcription, Genetic , Trophoblasts/cytology
12.
Chromosome Res ; 18(5): 525-41, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20568006

ABSTRACT

Cell type and tissue architecture correlate with genome organization in higher eukaryotes, and structural nuclear landmarks are faithfully transmitted from one cell generation to the next. However, how nuclear components find their place in the nucleus after mitosis is still a matter of debate. As the major structural proteins within nuclei, the nuclear lamins are good candidates to re-establish nuclear compartments following mitosis. Human cells with reduced expression of the major B-type lamin protein, lamin B1, were generated using RNA interference. Mitotic and nuclear assembly phenotypes were then visualized in both fixed and living cells. Mitotic defects in lamin B1-depleted cells correlated with a general deterioration in nuclear compartmentalization and chromatin structure, frequent failure of chromosome segregation, and profound disorganization of centromeres. Examination of cells with normal lamin B1 expression indicated that small lamin B1 foci remain associated with major nuclear compartments--chromatin, nucleoli, and nuclear speckles--during an unperturbed mitosis. Our experiments show that normal lamin B1 expression is required for successful cell division and provide preliminary evidence that lamin B1-containing remnants of the interphase nucleoskeleton persist throughout mitosis. We suggest that these residual structures provide landmarks that are targeted during nuclear reassembly to allow key features of nuclear organization to be inherited from one cell cycle to the next.


Subject(s)
Cell Nucleus/metabolism , Lamin Type B/physiology , Mitosis/physiology , Cell Nucleolus/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Lamin Type B/biosynthesis , Nuclear Proteins/metabolism , RNA Interference , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors
13.
Nucleic Acids Symp Ser (Oxf) ; (53): 293-4, 2009.
Article in English | MEDLINE | ID: mdl-19749376

ABSTRACT

We report that anticancer 5-fluoro-2'-deoxyuridine (FUdR) shows cytotoxicity against mouse cancer cell line FM3A cells, using a progeny clone F28-7 and its variant F28-7-A. In this process, the cell-death morphology is different between F28-7 and F28-7-A cells, that is, necrosis in F28-7 but apoptosis in F28-7-A cells. Recently we have investigated the gene and protein expression profiles of necrosis and apoptosis induced by FUdR using transcriptomic and proteomic analyses. In the proteomic analysis of these cells before their exposure to FUdR, the nuclear inner-membrane protein lamin B1 is up-regulated in F28-7 but not in F28-7-A, suggesting that lamin B1 may possess a function to regulate the morphology of cell-death. A knockdown of lamin B1 expression in F28-7 cells has now been performed by use of the small interfering RNA technique, resulting in a decrease of the lamin B1-expression level down to the level in F28-7-A. Remarkably, the FUdR-induced death morphology of this knocked-down F28-7 was apoptosis, definitely different from the necrosis that occurs in the FUdR-treated original F28-7. Our present finding provides an interesting possibility that lamin-B1 may have an important role in regulating cell-death morphology.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Apoptosis , Floxuridine/toxicity , Lamin Type B/physiology , Animals , Cell Line, Tumor , Lamin Type B/antagonists & inhibitors , Lamin Type B/genetics , Mice , Necrosis , RNA Interference
14.
Dis Model Mech ; 2(3-4): 178-88, 2009.
Article in English | MEDLINE | ID: mdl-19259393

ABSTRACT

Duplication of the gene encoding lamin B1 (LMNB1) with increased mRNA and protein levels has been shown to cause severe myelin loss in the brains of adult-onset autosomal dominant leukodystrophy patients. Similar to many neurodegenerative disorders, patients with adult-onset autosomal dominant leukodystrophy are phenotypically normal until adulthood and the defect is specific to the central nervous system despite the ubiquitous expression pattern of lamin B1. We set out to dissect the molecular mechanisms underlying this demyelinating phenotype. Increased lamin B1 expression results in disturbances of inner nuclear membrane proteins, chromatin organization and nuclear pore transport in vitro. It also leads to premature arrest of oligodendrocyte differentiation, which might be caused by reduced transcription of myelin genes and by mislocalization of myelin proteins. We identified the microRNA miR-23 as a negative regulator of lamin B1 that can ameliorate the consequences of excessive lamin B1 at the cellular level. Our results indicate that regulation of lamin B1 is important for myelin maintenance and that miR-23 contributes to this process, at least in part, by downregulating lamin B1, therefore establishing novel functions of lamin B1 and miR-23 in the regulation of oligodendroglia development and myelin formation in vitro.


Subject(s)
Lamin Type B/genetics , Lamin Type B/physiology , MicroRNAs/genetics , Myelin Sheath/physiology , Oligodendroglia/physiology , Animals , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chromatin/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , Humans , Mice , Models, Biological , Myelin Sheath/metabolism , Phenotype
15.
J Cell Biol ; 184(1): 45-55, 2009 Jan 12.
Article in English | MEDLINE | ID: mdl-19139261

ABSTRACT

Interaction of lamins with chromatin and transcription factors regulate transcription. Oct-1 has previously been shown to colocalize partly with B-type lamins and is essential for transcriptional regulation of oxidative stress response genes. Using sequential extraction, co-immunoprecipitation (IP), fluorescence loss in photobleaching, and fluorescence resonance energy transfer, we confirm Oct-1-lamin B1 association at the nuclear periphery and show that this association is lost in Lmnb1(Delta/Delta) cells. We show that several Oct-1-dependent genes, including a subset involved in oxidative stress response, are dysregulated in Lmnb1(Delta/Delta) cells. Electrophoretic mobility shift assay and chromatin IP reveal that Oct-1 binds to the putative octamer-binding sequences of the dysregulated genes and that this activity is increased in cells lacking functional lamin B1. Like Oct1(-/-) cells, Lmnb1(Delta/Delta) cells have elevated levels of reactive oxygen species and are more susceptible to oxidative stress. Sequestration of Oct-1 at the nuclear periphery by lamin B1 may be a mechanism by which the nuclear envelope can regulate gene expression and contribute to the cellular response to stress, development, and aging.


Subject(s)
Lamin Type B/physiology , Octamer Transcription Factor-1/physiology , Oxidative Stress , Animals , Cells, Cultured , Cellular Senescence , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Nuclear Lamina/metabolism , Octamer Transcription Factor-1/metabolism , Reactive Oxygen Species/metabolism
17.
J Cell Sci ; 121(Pt 7): 1014-24, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18334554

ABSTRACT

Spatial organisation of nuclear compartments is an important regulator of chromatin function, yet the molecular principles that maintain nuclear architecture remain ill-defined. We have used RNA interference to deplete key structural nuclear proteins, the nuclear lamins. In HeLa cells, we show that reduced expression of lamin B1, but not lamin A/C, severely inhibits RNA synthesis--first by RNA polymerase II and later by RNA polymerase I. Declining levels of transcription correlate with different morphological changes in major nuclear compartments, nucleoli and nuclear speckles. Ultimately, nuclear changes linked to the loss of synthetic activity result in expansion of the inter-chromatin domain and corresponding changes in the structure and spatial organisation of chromosome territories, which relocate towards the nuclear periphery. These results show that a lamin B1-containing nucleoskeleton is required to maintain RNA synthesis and that ongoing synthesis is a fundamental determinant of global nuclear architecture in mammalian cells.


Subject(s)
Cell Nucleus/metabolism , Lamin Type B/metabolism , Nuclear Matrix/metabolism , RNA/biosynthesis , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Lamin Type A/genetics , Lamin Type A/metabolism , Lamin Type A/physiology , Lamin Type B/genetics , Lamin Type B/physiology , Microscopy, Fluorescence , RNA/genetics , RNA Interference , RNA Polymerase I/metabolism , RNA Polymerase II/metabolism
18.
J Cell Sci ; 121(Pt 2): 215-25, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18187453

ABSTRACT

Lamins are intermediate filament proteins and the major component of the nuclear lamina. Current views of the lamina are based on the remarkably regular arrangement of lamin LIII in amphibian oocyte nuclei. We have re-examined the LIII lamina and propose a new interpretation of its organization. Rather than consisting of two perpendicular arrays of parallel filaments, we suggest that the oocyte lamina consists of parallel filaments that are interconnected in register to give the impression of a second set of perpendicular filaments. We have also used the oocyte system to investigate the organization of somatic lamins. Currently, it is not feasible to examine the organization of somatic lamins in situ because of their tight association with chromatin. It is also difficult to assemble vertebrate lamin filaments in vitro. Therefore, we have used the oocyte system, where exogenously expressed somatic B-type and A-type lamins assemble into filaments. Expression of B-type lamins induces the formation of intranuclear membranes that are covered by single filament layers. LIII filaments appear identical to the endogenous lamina, whereas lamin B2 assembles into filaments that are organized less precisely. Lamin A induces sheets of thicker filaments on the endogenous lamina and significantly increases the rigidity of the nuclear envelope.


Subject(s)
Lamin Type A/physiology , Lamin Type B/physiology , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Cytoskeleton/metabolism , Female , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Models, Biological , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Xenopus laevis
19.
J Biol Chem ; 281(35): 25768-80, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16825190

ABSTRACT

Mutations in the nuclear envelope proteins lamins A and C cause a broad variety of human diseases, including Emery-Dreifuss muscular dystrophy, dilated cardiomyopathy, and Hutchinson-Gilford progeria syndrome. Cells lacking lamins A and C have reduced nuclear stiffness and increased nuclear fragility, leading to increased cell death under mechanical strain and suggesting a potential mechanism for disease. Here, we investigated the contribution of major lamin subtypes (lamins A, C, and B1) to nuclear mechanics by analyzing nuclear shape, nuclear dynamics over time, nuclear deformations under strain, and cell viability under prolonged mechanical stimulation in cells lacking both lamins A and C, cells lacking only lamin A (i.e. "lamin C-only" cells), cells lacking wild-type lamin B1, and wild-type cells. Lamin A/C-deficient cells exhibited increased numbers of misshapen nuclei and had severely reduced nuclear stiffness and decreased cell viability under strain. Lamin C-only cells had slightly abnormal nuclear shape and mildly reduced nuclear stiffness but no decrease in cell viability under strain. Interestingly, lamin B1-deficient cells exhibited normal nuclear mechanics despite having a significantly increased frequency of nuclear blebs. Our study indicates that lamins A and C are important contributors to the mechanical stiffness of nuclei, whereas lamin B1 contributes to nuclear integrity but not stiffness.


Subject(s)
Cell Nucleus/metabolism , Lamin Type A/chemistry , Lamin Type B/chemistry , Animals , Apoptosis , Cell Survival , Crosses, Genetic , Heterozygote , Lamin Type A/physiology , Lamin Type B/physiology , Mice , Mice, Transgenic , Nuclear Envelope/metabolism , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...