Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38477372

ABSTRACT

Biogenesis of inclusion bodies (IBs) facilitates protein quality control (PQC). Canonical aggresomes execute degradation of misfolded proteins while non-degradable amyloids sequester into insoluble protein deposits. Lewy bodies (LBs) are filamentous amyloid inclusions of α-synuclein, but PQC benefits and drawbacks associated with LB-like IBs remain underexplored. Here, we report that crosstalk between filamentous LB-like IBs and aggresome-like IBs of α-synuclein (Syn-aggresomes) buffer the load, aggregation state, and turnover of the amyloidogenic protein in mouse primary neurons and HEK293T cells. Filamentous LB-like IBs possess unorthodox PQC capacities of self-quarantining α-synuclein amyloids and being degradable upon receding fresh amyloidogenesis. Syn-aggresomes equilibrate biogenesis of filamentous LB-like IBs by facilitating spontaneous degradation of α-synuclein and conditional turnover of disintegrated α-synuclein amyloids. Thus, both types of IB primarily contribute to PQC. Incidentally, the overgrown perinuclear LB-like IBs become degenerative once these are misidentified by BICD2, a cargo-adapter for the cytosolic motor-protein dynein. Microscopy indicates that microtubules surrounding the perinuclear filamentous inclusions are also distorted, misbalancing the cytoskeleton-nucleoskeleton tension leading to widespread lamina injuries. Together, nucleocytoplasmic mixing, DNA damage, and deregulated transcription of stress chaperones defeat the proteostatic purposes of the filamentous amyloids of α-synuclein.


Subject(s)
Nuclear Lamina , alpha-Synuclein , Animals , Humans , Mice , alpha-Synuclein/metabolism , Amyloid/metabolism , Amyloidogenic Proteins/metabolism , HEK293 Cells , Inclusion Bodies/metabolism , Nuclear Lamina/metabolism , Nuclear Lamina/pathology
2.
Curr Opin Cell Biol ; 85: 102257, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806292

ABSTRACT

The role of lamin B1 in human health and aging has attracted increasing attention as mounting evidence reveals its significance in diverse cellular processes. Both upregulation and downregulation of lamin B1 have been implicated in age-associated organ dysfunctions and various human diseases, including central nervous system disorders. Additionally, lamin B1 levels undergo alterations in cancer cells, and a tumor-specific association exists between lamin B1 abundance and cancer aggressiveness. Investigating the connectivity between lamin B1 abundance and human health is of utmost importance for further research. This review presents recent advancements in understanding lamin B1's role in nuclear lamina function and its implications for human health.


Subject(s)
Aging , Nuclear Lamina , Humans , Aging/pathology , Lamin Type B/genetics , Neoplasms/pathology , Nuclear Lamina/pathology
3.
Pathol Oncol Res ; 28: 1610684, 2022.
Article in English | MEDLINE | ID: mdl-36561231

ABSTRACT

Background: The nuclear laminar protein Lamin A and inner nuclear membrane protein Emerin plays important role in sustaining nuclear structure. However, They have not investigated the significance of these proteins for development of pancreatic intraductal papillary mucinous neoplasm (IPMN). Methods: We examined pancreatic IPMN specimens for nuclear morphology and nuclear protein expression pattern of Lamin A and Emerin. Forty-two IPMN specimens were included, with 30 classified as intraductal papillary mucinous adenoma (IPMA) and 12 as intraductal papillary mucinous carcinoma (IPMC). Results: Classification according to histological subtype revealed that 26 specimens were of the gastric subtype (1 IPMC case), 8 were pancreatobiliary (6 IPMC cases), 6 were intestinal (3 IPMC cases), and 2 were oncocytic (all cases were IPMC). The frequency of IPMN subtypes in this study seemed to agree with those in previous reports. We analyzed Feulgen staining sections for nuclear morphological analysis using computer-assisted image analysis. Nuclear area and perimeter were significantly larger in IPMC than in IPMA. Finally, we examined the positive ratios of Lamin A and Emerin in immunohistochemical staining sections by image analysis. We found a negative correlation between the nuclear size and Lamin A-positive ratio, which was significantly lower in IPMC than that in IPMA. However, no significant correlation was observed between nuclear size and Emerin expression was observed, and no differences were found in the Emerin-positive ratio between IPMA and IPMC. Conclusion: Our results suggest that a decreased Lamin A positive ratio induces nuclear enlargement in adenomas, which thereby induce promotion to carcinomas. Furthermore, Lamin A expression can be a reliable biomarker for distinguishing between IPMC and IPMA.


Subject(s)
Adenocarcinoma, Mucinous , Adenocarcinoma, Papillary , Carcinoma, Pancreatic Ductal , Pancreatic Intraductal Neoplasms , Pancreatic Neoplasms , Humans , Lamin Type A , Carcinoma, Pancreatic Ductal/pathology , Nuclear Lamina/metabolism , Nuclear Lamina/pathology , Adenocarcinoma, Mucinous/pathology , Pancreatic Neoplasms/pathology , Adenocarcinoma, Papillary/pathology
4.
Hum Cell ; 35(2): 583-598, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34984662

ABSTRACT

Breast cancer is life threatening among women because its migration by hematogenous metastasis, where, besides biochemical cues, breast circulating tumor cells (CTCs) expose to suspension state and shear stress. However, the combined effects of these mechanical factors on CTCs migration were unclear. Here, suspension state and shear stress were loaded to breast tumor cells (BTCs) to mimic two mechanical cues in the mechanical environment of breast CTCs and the mechanobiological mechanism of suspension state and shear stress regulating the migration of (BTCs) was investigated. The migration and nuclear lamina protein A/C (Lamin A/C) accumulation were enhanced in MDA-MB-231 and SK-BR-3 BTCs exposed to shear stress though lower than that of suspended cells with different yes-associated protein (YAP) subcellular localization. Knockdown of LMNA downregulated and upregulated YAP targets in suspended BTCs and BTCs exposed to shear stress, respectively, which inhibited MDA-MB-231 BTCs migration in vitro and in vivo. Large tumor suppressor (LATS) responded to suspension state and shear stress, knockdown of which decreased the migration of MDA-MB-231 BTCs. These findings uncover the mechanobiological mechanism that suspension state and shear stress antagonistically promote BTCs migration by Lamin A/C and LATS through YAP and the potential for targeting YAP in CTCs prognosis. Shear stress regulates suspended breast cancer cells migration by Lamin A/C and LATS through YAP.


Subject(s)
Breast Neoplasms , Neoplastic Cells, Circulating , Nuclear Lamina , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Humans , Neoplastic Cells, Circulating/pathology , Nuclear Lamina/metabolism , Nuclear Lamina/pathology , YAP-Signaling Proteins
5.
Sci Rep ; 11(1): 13195, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162976

ABSTRACT

Chromatin organization within the nuclear volume is essential to regulate many aspects of its function and to safeguard its integrity. A key player in this spatial scattering of chromosomes is the nuclear envelope (NE). The NE tethers large chromatin domains through interaction with the nuclear lamina and other associated proteins. This organization is perturbed in cells from Hutchinson-Gilford progeria syndrome (HGPS), a genetic disorder characterized by premature aging features. Here, we show that HGPS-related lamina defects trigger an altered 3D telomere organization with increased contact sites between telomeres and the nuclear lamina, and an altered telomeric chromatin state. The genome-wide replication timing signature of these cells is perturbed, with a shift to earlier replication for regions that normally replicate late. As a consequence, we detected a higher density of replication forks traveling simultaneously on DNA fibers, which relies on limiting cellular dNTP pools to support processive DNA synthesis. Remarkably, increasing dNTP levels in HGPS cells rescued fragile telomeres, and improved the replicative capacity of the cells. Our work highlights a functional connection between NE dysfunction and telomere homeostasis in the context of premature aging.


Subject(s)
Chromatin/ultrastructure , Deoxyribonucleotides/metabolism , Lamin Type A/physiology , Nuclear Lamina/pathology , Progeria/genetics , Telomere Homeostasis/genetics , Telomere/pathology , Adult , Animals , Cells, Cultured , Cellular Senescence/genetics , DNA Damage , DNA Replication , Fibroblasts , Genes, Reporter , Green Fluorescent Proteins , Histone Code , Humans , Infant, Newborn , Lamin Type A/analysis , Lamin Type A/deficiency , Lamin Type A/genetics , Lamin Type B/analysis , Mice , Mice, Knockout , Progeria/pathology , Recombinant Fusion Proteins/metabolism , Skin/pathology
6.
J Virol ; 95(17): e0087321, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34133898

ABSTRACT

Nuclear envelope budding in herpesvirus nuclear egress may be negatively regulated, since the pUL31/pUL34 nuclear egress complex heterodimer can induce membrane budding without capsids when expressed ectopically or on artificial membranes in vitro, but not in the infected cell. We have previously described a pUL34 mutant that contained alanine substitutions at R158 and R161 and that showed impaired growth, impaired pUL31/pUL34 interaction, and unregulated budding. Here, we determine the phenotypic contributions of the individual substitutions to these phenotypes. Neither substitution alone was able to reproduce the impaired growth or nuclear egress complex (NEC) interaction phenotypes. Either substitution, however, could fully reproduce the unregulated budding phenotype, suggesting that misregulated budding may not substantially impair virus replication. In addition, the R158A substitution caused relocalization of the NEC to intranuclear punctate structures and recruited lamin A/C to these structures, suggesting that this residue might be important for recruitment of kinases for dispersal of nuclear lamins. IMPORTANCE Herpesvirus nuclear egress is a complex, regulated process coordinated by two virus proteins that are conserved among the herpesviruses that form a heterodimeric nuclear egress complex (NEC). The NEC drives budding of capsids at the inner nuclear membrane and recruits other viral and host cell proteins for disruption of the nuclear lamina, membrane scission, and fusion. The structural basis of individual activities of the NEC, apart from membrane budding, are not clear, nor is the basis of the regulation of membrane budding. Here, we explore the properties of NEC mutants that have an unregulated budding phenotype, determine the significance of that regulation for virus replication, and also characterize a structural requirement for nuclear lamina disruption.


Subject(s)
Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Mutation , Nuclear Lamina/metabolism , Viral Proteins/metabolism , Virus Replication , Active Transport, Cell Nucleus , Animals , Chlorocebus aethiops , Herpes Simplex/genetics , Herpes Simplex/metabolism , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Nuclear Envelope/virology , Nuclear Lamina/pathology , Nuclear Lamina/virology , Vero Cells , Viral Proteins/genetics , Virus Release
7.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33758005

ABSTRACT

The nuclear lamina is a proteinaceous network of filaments that provide both structural and gene regulatory functions by tethering proteins and large domains of DNA, the so-called lamina-associated domains (LADs), to the periphery of the nucleus. LADs are a large fraction of the mammalian genome that are repressed, in part, by their association to the nuclear periphery. The genesis and maintenance of LADs is poorly understood as are the proteins that participate in these functions. In an effort to identify proteins that reside at the nuclear periphery and potentially interact with LADs, we have taken a two-pronged approach. First, we have undertaken an interactome analysis of the inner nuclear membrane bound LAP2ß to further characterize the nuclear lamina proteome. To accomplish this, we have leveraged the BioID system, which previously has been successfully used to characterize the nuclear lamina proteome. Second, we have established a system to identify proteins that bind to LADs by developing a chromatin-directed BioID system. We combined the BioID system with the m6A-tracer system which binds to LADs in live cells to identify both LAD proximal and nuclear lamina proteins. In combining these datasets, we have further characterized the protein network at the nuclear lamina, identified putative LAD proximal proteins and found several proteins that appear to interface with both micro-proteomes. Importantly, several proteins essential for LAD function, including heterochromatin regulating proteins related to H3K9 methylation, were identified in this study.


Subject(s)
Nuclear Lamina/metabolism , Proteome/metabolism , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Genome , Heterochromatin/metabolism , Humans , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , NIH 3T3 Cells , Nuclear Lamina/genetics , Nuclear Lamina/pathology , Nuclear Proteins/genetics , Protein Binding/physiology , Protein Domains/physiology , Proteome/genetics , Proteomics/methods
8.
Acta Neuropathol Commun ; 9(1): 45, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741069

ABSTRACT

The most common genetic cause of familial and sporadic amyotrophic lateral sclerosis (ALS) is a GGGGCC hexanucleotide repeat expansion (HRE) in the C9orf72 gene. While direct molecular hallmarks of the C9orf72 HRE (repeat RNA foci, dipeptide repeat protein pathology) are well characterized, the mechanisms by which the C9orf72 HRE causes ALS and the related neurodegenerative disease frontotemporal dementia (FTD) remain poorly understood. Recently, alterations to the nuclear pore complex and nucleocytoplasmic transport have been accepted as a prominent pathomechanism underlying C9orf72 ALS/FTD. However, global disruptions to nuclear morphology and the nuclear lamina itself remain controversial. Here, we use a large number of induced pluripotent stem cell derived spinal neurons and postmortem human motor cortex sections to thoroughly examine nuclear morphology and nuclear lamina disruptions with light microscopy. In contrast to previous studies in artificial overexpression model systems, endogenous levels of the C9orf72 HRE do not increase the frequency of nuclear lamina invaginations. In addition, the C9orf72 HRE has no impact on overall nuclear shape and size. Notably, the frequency of nuclear Lamin B1 invaginations increases with cellular aging, independent of the C9orf72 HRE. Together, our data suggest that nuclear morphology is unaltered in C9orf72 ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , Frontotemporal Dementia/pathology , Nuclear Lamina/pathology , Nuclear Lamina/ultrastructure , Aged , Amyotrophic Lateral Sclerosis/genetics , Autopsy , Cellular Senescence/physiology , DNA Repeat Expansion , Female , Frontotemporal Dementia/genetics , Humans , Lamin Type B/metabolism , Male , Middle Aged , Neurons/pathology , Neurons/ultrastructure , Nuclear Lamina/genetics
9.
Nat Cell Biol ; 23(2): 116-126, 2021 02.
Article in English | MEDLINE | ID: mdl-33558730

ABSTRACT

Alterations in nuclear shape are present in human diseases and ageing. A compromised nuclear lamina is molecularly interlinked to altered chromatin functions and genomic instability. Whether these alterations are a cause or a consequence of the pathological state are important questions in biology. Here, we summarize the roles of nuclear envelope components in chromatin organization, phase separation and transcriptional and epigenetic regulation. Examining these functions in healthy backgrounds will guide us towards a better understanding of pathological alterations.


Subject(s)
Nuclear Lamina/pathology , Aging/pathology , Animals , Disease , Epigenesis, Genetic , Humans , Lamins/metabolism , Nuclear Lamina/genetics , Protein Processing, Post-Translational
10.
Nucleus ; 11(1): 299-314, 2020 12.
Article in English | MEDLINE | ID: mdl-33030403

ABSTRACT

Decades of studies have established that nuclear lamin polymers form the nuclear lamina, a protein meshwork that supports the nuclear envelope structure and tethers heterochromatin to the nuclear periphery. Much less is known about unpolymerized nuclear lamins in the nuclear interior, some of which are now known to undergo specific phosphorylation. A recent finding that phosphorylated lamins bind gene enhancer regions offers a new hypothesis that lamin phosphorylation may influence transcriptional regulation in the nuclear interior. In this review, we discuss the regulation, localization, and functions of phosphorylated lamins. We summarize kinases that phosphorylate lamins in a variety of biological contexts. Our discussion extends to laminopathies, a spectrum of degenerative disorders caused by lamin gene mutations, such as cardiomyopathies and progeria. We compare the prevailing hypothesis for laminopathy pathogenesis based on lamins' function at the nuclear lamina with an emerging hypothesis based on phosphorylated lamins' function in the nuclear interior.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Lamins/metabolism , Nuclear Lamina/metabolism , Progeria/metabolism , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Humans , Lamins/genetics , Nuclear Lamina/genetics , Nuclear Lamina/pathology , Phosphorylation , Progeria/genetics , Progeria/pathology
11.
Nucleus ; 11(1): 237-249, 2020 12.
Article in English | MEDLINE | ID: mdl-32910721

ABSTRACT

The nuclear membranes function as a barrier to separate the cell nucleus from the cytoplasm, but this barrier can be compromised by nuclear membrane ruptures, leading to intermixing of nuclear and cytoplasmic contents. Spontaneous nuclear membrane ruptures (i.e., ruptures occurring in the absence of mechanical stress) have been observed in cultured cells, but they are more frequent in the setting of defects or deficiencies in nuclear lamins and when cells are subjected to mechanical stress. Nuclear membrane ruptures in cultured cells have been linked to DNA damage, but the relevance of ruptures to developmental or physiologic processes in vivo has received little attention. Recently, we addressed that issue by examining neuronal migration in the cerebral cortex, a developmental process that subjects the cell nucleus to mechanical stress. In the setting of lamin B1 deficiency, we observed frequent nuclear membrane ruptures in migrating neurons in the developing cerebral cortex and showed that those ruptures are likely the cause of observed DNA damage, neuronal cell death, and profound neuropathology. In this review, we discuss the physiologic relevance of nuclear membrane ruptures, with a focus on migrating neurons in cell culture and in the cerebral cortex of genetically modified mice.


Subject(s)
Cerebral Cortex/metabolism , DNA Damage , Lamin Type B/deficiency , Neurons/metabolism , Nuclear Lamina/metabolism , Polyneuropathies/metabolism , Animals , Cell Movement/genetics , Cerebral Cortex/pathology , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Mice, Transgenic , Neurons/pathology , Nuclear Lamina/genetics , Nuclear Lamina/pathology , Polyneuropathies/genetics , Polyneuropathies/pathology
12.
Am J Hum Genet ; 107(4): 753-762, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32910914

ABSTRACT

Lamin B1 plays an important role in the nuclear envelope stability, the regulation of gene expression, and neural development. Duplication of LMNB1, or missense mutations increasing LMNB1 expression, are associated with autosomal-dominant leukodystrophy. On the basis of its role in neurogenesis, it has been postulated that LMNB1 variants could cause microcephaly. Here, we confirm this hypothesis with the identification of de novo mutations in LMNB1 in seven individuals with pronounced primary microcephaly (ranging from -3.6 to -12 SD) associated with relative short stature and variable degree of intellectual disability and neurological features as the core symptoms. Simplified gyral pattern of the cortex and abnormal corpus callosum were noted on MRI of three individuals, and these individuals also presented with a more severe phenotype. Functional analysis of the three missense mutations showed impaired formation of the LMNB1 nuclear lamina. The two variants located within the head group of LMNB1 result in a decrease in the nuclear localization of the protein and an increase in misshapen nuclei. We further demonstrate that another mutation, located in the coil region, leads to increased frequency of condensed nuclei and lower steady-state levels of lamin B1 in proband lymphoblasts. Our findings collectively indicate that de novo mutations in LMNB1 result in a dominant and damaging effect on nuclear envelope formation that correlates with microcephaly in humans. This adds LMNB1 to the growing list of genes implicated in severe autosomal-dominant microcephaly and broadens the phenotypic spectrum of the laminopathies.


Subject(s)
Dwarfism/genetics , Intellectual Disability/genetics , Lamin Type B/genetics , Microcephaly/genetics , Mutation , Nuclear Lamina/genetics , Amino Acid Sequence , Base Sequence , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Child, Preschool , Corpus Callosum/diagnostic imaging , Corpus Callosum/metabolism , Corpus Callosum/pathology , Dwarfism/diagnostic imaging , Dwarfism/metabolism , Dwarfism/pathology , Female , Gene Expression , Humans , Infant , Intellectual Disability/diagnostic imaging , Intellectual Disability/metabolism , Intellectual Disability/pathology , Lamin Type B/metabolism , Lymphocytes/metabolism , Lymphocytes/pathology , Magnetic Resonance Imaging , Male , Microcephaly/diagnostic imaging , Microcephaly/metabolism , Microcephaly/pathology , Nuclear Lamina/metabolism , Nuclear Lamina/pathology
13.
Cells ; 8(7)2019 07 01.
Article in English | MEDLINE | ID: mdl-31266244

ABSTRACT

The eukaryotic nucleus controls most cellular processes. It is isolated from the cytoplasm by the nuclear envelope, which plays a prominent role in the structural organization of the cell, including nucleocytoplasmic communication, chromatin positioning, and gene expression. Alterations in nuclear composition and function are eminently pronounced upon stress and during premature and physiological aging. These alterations are often accompanied by epigenetic changes in histone modifications. We review, here, the role of nuclear envelope proteins and histone modifiers in the 3-dimensional organization of the genome and the implications for gene expression. In particular, we focus on the nuclear lamins and the chromatin-associated protein BAF, which are linked to Hutchinson-Gilford and Nestor-Guillermo progeria syndromes, respectively. We also discuss alterations in nuclear organization and the epigenetic landscapes during normal aging and various stress conditions, ranging from yeast to humans.


Subject(s)
Aging/physiology , Epigenesis, Genetic/physiology , Nuclear Lamina/metabolism , Stress, Physiological , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Code/genetics , Histones/metabolism , Humans , Lamins/genetics , Lamins/metabolism , Nuclear Lamina/pathology , Progeria/genetics , Progeria/pathology
14.
Cells ; 8(3)2019 03 11.
Article in English | MEDLINE | ID: mdl-30862117

ABSTRACT

Extracellular mechanical stimuli are translated into biochemical signals inside the cell via mechanotransduction. The nucleus plays a critical role in mechanoregulation, which encompasses mechanosensing and mechanotransduction. The nuclear lamina underlying the inner nuclear membrane not only maintains the structural integrity, but also connects the cytoskeleton to the nuclear envelope. Lamin mutations, therefore, dysregulate the nuclear response, resulting in abnormal mechanoregulations, and ultimately, disease progression. Impaired mechanoregulations even induce malfunction in nuclear positioning, cell migration, mechanosensation, as well as differentiation. To know how to overcome laminopathies, we need to understand the mechanisms of laminopathies in a mechanobiological way. Recently, emerging studies have demonstrated the varying defects from lamin mutation in cellular homeostasis within mechanical surroundings. Therefore, this review summarizes recent findings highlighting the role of lamins, the architecture of nuclear lamina, and their disease relevance in the context of nuclear mechanobiology. We will also provide an overview of the differentiation of cellular mechanics in laminopathy.


Subject(s)
Biophysical Phenomena , Nuclear Lamina/pathology , Animals , Cytoskeleton/metabolism , Humans , Lamins/metabolism , Mechanotransduction, Cellular , Models, Biological
15.
Cells ; 8(3)2019 03 13.
Article in English | MEDLINE | ID: mdl-30871242

ABSTRACT

Emerin is an essential LEM (LAP2, Emerin, MAN1) domain protein in metazoans and an integral membrane protein associated with inner and outer nuclear membranes. Mutations in the human EMD gene coding for emerin result in the rare genetic disorder: Emery⁻Dreifuss muscular dystrophy type 1 (EDMD1). This disease belongs to a broader group called laminopathies-a heterogeneous group of rare genetic disorders affecting tissues of mesodermal origin. EDMD1 phenotype is characterized by progressive muscle wasting, contractures of the elbow and Achilles tendons, and cardiac conduction defects. Emerin is involved in many cellular and intranuclear processes through interactions with several partners: lamins; barrier-to-autointegration factor (BAF), ß-catenin, actin, and tubulin. Our study demonstrates the presence of the emerin fraction which associates with mitotic spindle microtubules and centrosomes during mitosis and colocalizes during early mitosis with lamin A/C, BAF, and membranes at the mitotic spindle. Transfection studies with cells expressing EGFP-emerin protein demonstrate that the emerin fusion protein fraction also localizes to centrosomes and mitotic spindle microtubules during mitosis. Transient expression of emerin deletion mutants revealed that the resulting phenotypes vary and are mutant dependent. The most frequent phenotypes include aberrant nuclear shape, tubulin network mislocalization, aberrant mitosis, and mislocalization of centrosomes. Emerin deletion mutants demonstrated different chromatin binding capacities in an in vitro nuclear assembly assay and chromatin-binding properties correlated with the strength of phenotypic alteration in transfected cells. Aberrant tubulin staining and microtubule network phenotype appearance depended on the presence of the tubulin binding region in the expressed deletion mutants. We believe that the association with tubulin might help to "deliver" emerin and associated membranes to decondensing chromatin. Preliminary analyses of cells from Polish patients with EDMD1 revealed that for several mutations thought to be null for emerin protein, a truncated emerin protein was present. We infer that the EDMD1 phenotype may be strengthened by the toxicity of truncated emerin expressed in patients with certain nonsense mutations in EMD.


Subject(s)
Membrane Proteins/metabolism , Mitosis , Muscular Dystrophy, Emery-Dreifuss/pathology , Nuclear Lamina/pathology , Nuclear Proteins/metabolism , Antibodies/metabolism , Cell Cycle , Centrosome/metabolism , DNA-Binding Proteins/metabolism , Epitopes/metabolism , Gene Deletion , HeLa Cells , Humans , Lamin Type A/metabolism , Membrane Proteins/deficiency , Microtubules/metabolism , Nuclear Proteins/deficiency , Phenotype , Protein Binding , Spindle Apparatus/metabolism , Tubulin/metabolism
16.
Science ; 363(6428)2019 02 15.
Article in English | MEDLINE | ID: mdl-30765536

ABSTRACT

How hexanucleotide GGGGCC (G4C2) repeat expansions in C9orf72 cause frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) is not understood. We developed a mouse model engineered to express poly(PR), a proline-arginine (PR) dipeptide repeat protein synthesized from expanded G4C2 repeats. The expression of green fluorescent protein-conjugated (PR)50 (a 50-repeat PR protein) throughout the mouse brain yielded progressive brain atrophy, neuron loss, loss of poly(PR)-positive cells, and gliosis, culminating in motor and memory impairments. We found that poly(PR) bound DNA, localized to heterochromatin, and caused heterochromatin protein 1α (HP1α) liquid-phase disruptions, decreases in HP1α expression, abnormal histone methylation, and nuclear lamina invaginations. These aberrations of histone methylation, lamins, and HP1α, which regulate heterochromatin structure and gene expression, were accompanied by repetitive element expression and double-stranded RNA accumulation. Thus, we uncovered mechanisms by which poly(PR) may contribute to the pathogenesis of C9orf72-associated FTD and ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Dipeptides/metabolism , Heterochromatin/pathology , RNA, Double-Stranded/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , C9orf72 Protein/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Dipeptides/genetics , Disease Models, Animal , Green Fluorescent Proteins , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nuclear Lamina/pathology , Repetitive Sequences, Nucleic Acid
17.
Free Radic Biol Med ; 134: 567-580, 2019 04.
Article in English | MEDLINE | ID: mdl-30769159

ABSTRACT

Recent reports in pathophysiology of neurodegenerative diseases (ND) have linked nuclear lamina degradation/deficits to neuronal cell death. Lamin-B1 damage is specifically involved in this process leading to nuclear envelope invagination and heterochromatin rearrangement. The underlying mechanisms involved in these events are not yet defined. In this study, while examining the effect of Thioredoxin-1(Trx1) inhibition on cell death in a model of oxidative stress, we noted robust nuclear invagination in SH-SY5Y cells. Evaluation of nuclear lamina proteins revealed lamin-B1 cleavage that was prevented by caspase-6 (CASP6) inhibitor and exacerbated after pharmacologic/genetic inhibition of Trx1 system, but not after glutathione depletion. Activation of CASP6 was upstream of CASP3/7 activation and its inhibition was sufficient to prevent cell death in our system. The effect of Trx1 redox status on CASP6 activation was assessed by administration of reduced/oxidized forms in cell-free nuclei preparation and purified enzymatic assays. Although reduced Trx1 decreased CASP6 enzymatic activity and lamin-B1 cleavage, the fully oxidized Trx1 showed opposite effects. The enhanced CASP6 activation was also associated with lower levels of DJ-1, a neuroprotective and master regulator of cellular antioxidants. The implication of our findings in ND pathophysiology was strengthened with detection of lower Trx1 levels in the hippocampi tissue of a mouse model of Alzheimer's disease. This coincided with higher CASP6 activation resulting in increased lamin-B1 and DJ-1 depletion. This study provides a first mechanistic explanation for the key regulatory role of Trx1 as a gatekeeper in activation of CASP6 and induction of nuclear invagination, an important player in ND pathophysiology.


Subject(s)
Alzheimer Disease/pathology , Antioxidants/metabolism , Caspase 6/metabolism , Neuroblastoma/pathology , Nuclear Lamina/pathology , Oxidative Stress , Thioredoxins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Caspase 6/genetics , Female , Glutathione , Humans , Male , Mice , Mice, Transgenic , Neuroblastoma/genetics , Neuroblastoma/metabolism , Nuclear Lamina/metabolism , Oxidation-Reduction , Thioredoxins/genetics , Tumor Cells, Cultured
18.
Dev Cell ; 45(2): 212-225.e7, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29689196

ABSTRACT

Meiotic chromosome movement is important for the pairwise alignment of homologous chromosomes, which is required for correct chromosome segregation. Movement is driven by cytoplasmic forces, transmitted to chromosome ends by nuclear membrane-spanning proteins. In animal cells, lamins form a prominent scaffold at the nuclear periphery, yet the role lamins play in meiotic chromosome movement is unclear. We show that chromosome movement correlates with reduced lamin association with the nuclear rim, which requires lamin phosphorylation at sites analogous to those that open lamina network crosslinks in mitosis. Failure to remodel the lamina results in delayed meiotic entry, altered chromatin organization, unpaired or interlocked chromosomes, and slowed chromosome movement. The remodeling kinases are delivered to lamins via chromosome ends coupled to the nuclear envelope, potentially enabling crosstalk between the lamina and chromosomal events. Thus, opening the lamina network plays a role in modulating contacts between chromosomes and the nuclear periphery during meiosis.


Subject(s)
Animals, Genetically Modified/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Chromosome Segregation , Chromosomes/genetics , Meiotic Prophase I/genetics , Nuclear Lamina/pathology , Animals , Animals, Genetically Modified/growth & development , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/pathology , Chromosome Pairing , Cytoplasm , Gene Expression Regulation , Nuclear Envelope/genetics , Nuclear Envelope/pathology , Nuclear Lamina/genetics , Phosphorylation
19.
Nucleus ; 9(1): 249-260, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29557732

ABSTRACT

The nuclear lamina is a complex reticular structure that covers the inner face of the nucleus membrane in metazoan cells. It is mainly formed by intermediate filaments called lamins, and exerts essential functions to maintain the cellular viability. Lamin A/C provides mechanical steadiness to the nucleus and regulates genetic machinery. Laminopathies are tissue-specific or systemic disorders caused by variants in LMNA gene (primary laminopathies) or in other genes encoding proteins which are playing some role in prelamin A maturation or in lamin A/C function (secondary laminopathies). Those disorders in which adipose tissue is affected are called laminopathic lipodystrophies and include type 2 familial partial lipodystrophy and certain premature aging syndromes. This work summarizes the main clinical features of these syndromes, their associated comorbidities and the clues for the differential diagnosis with other lipodystrophic disorders.


Subject(s)
Lipodystrophy/diagnosis , Nuclear Lamina/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cell Nucleus/metabolism , Cell Nucleus/pathology , Humans , Lipodystrophy/metabolism , Nuclear Lamina/metabolism
20.
Neuron ; 93(2): 331-347, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-28065649

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by a limited expansion of CGG repeats in the 5' UTR of FMR1. Two mechanisms are proposed to cause FXTAS: RNA gain-of-function, where CGG RNA sequesters specific proteins, and translation of CGG repeats into a polyglycine-containing protein, FMRpolyG. Here we developed transgenic mice expressing CGG repeat RNA with or without FMRpolyG. Expression of FMRpolyG is pathogenic, while the sole expression of CGG RNA is not. FMRpolyG interacts with the nuclear lamina protein LAP2ß and disorganizes the nuclear lamina architecture in neurons differentiated from FXTAS iPS cells. Finally, expression of LAP2ß rescues neuronal death induced by FMRpolyG. Overall, these results suggest that translation of expanded CGG repeats into FMRpolyG alters nuclear lamina architecture and drives pathogenesis in FXTAS.


Subject(s)
Ataxia/genetics , DNA-Binding Proteins/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Membrane Proteins/metabolism , Nuclear Lamina/metabolism , Peptides/genetics , Protein Biosynthesis , RNA, Messenger/metabolism , Tremor/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Ataxia/metabolism , Brain/metabolism , Brain/pathology , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Humans , Male , Mice , Mice, Transgenic , Nuclear Lamina/pathology , Peptides/metabolism , Real-Time Polymerase Chain Reaction , Tremor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...