Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Publication year range
1.
Mol Cell ; 79(3): 443-458.e7, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32649883

ABSTRACT

Despite the prominent role of TDP-43 in neurodegeneration, its physiological and pathological functions are not fully understood. Here, we report an unexpected role of TDP-43 in the formation of dynamic, reversible, liquid droplet-like nuclear bodies (NBs) in response to stress. Formation of NBs alleviates TDP-43-mediated cytotoxicity in mammalian cells and fly neurons. Super-resolution microscopy reveals distinct functions of the two RRMs in TDP-43 NB formation. TDP-43 NBs are partially colocalized with nuclear paraspeckles, whose scaffolding lncRNA NEAT1 is dramatically upregulated in stressed neurons. Moreover, increase of NEAT1 promotes TDP-43 liquid-liquid phase separation (LLPS) in vitro. Finally, we discover that the ALS-associated mutation D169G impairs the NEAT1-mediated TDP-43 LLPS and NB assembly, causing excessive cytoplasmic translocation of TDP-43 to form stress granules, which become phosphorylated TDP-43 cytoplasmic foci upon prolonged stress. Together, our findings suggest a stress-mitigating role and mechanism of TDP-43 NBs, whose dysfunction may be involved in ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Intranuclear Inclusion Bodies/metabolism , Neurons/metabolism , RNA, Long Noncoding/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Arsenites/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila melanogaster , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/ultrastructure , Mice , Mutation , Neurons/drug effects , Neurons/ultrastructure , Primary Cell Culture , Protein Transport/drug effects , RNA, Long Noncoding/metabolism , Signal Transduction , Stress, Physiological
2.
Pathol Int ; 73(8): 341-350, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37154498

ABSTRACT

Nuclear morphology of carcinoma cells is critical for the pathological diagnosis of papillary thyroid carcinoma (PTC). However, three-dimensional architecture of PTC nuclei is still elusive. In this study, we analyzed the three-dimensional ultrastructure of PTC nuclei using serial block-face scanning electron microscopy which takes advantage of the high-throughput acquisition of serial electron microscopic images and three-dimensional reconstruction of subcellular structures. En bloc-stained and resin-embedded specimens were prepared from surgically removed PTCs and normal thyroid tissues. We acquired two-dimensional images from serial block-face scanning electron microscopy and reconstructed three-dimensional nuclear structures. Quantitative comparisons showed that the nuclei of carcinoma cells were larger and more complex than those of normal follicular cells. The three-dimensional reconstruction of carcinoma nuclei divided intranuclear cytoplasmic inclusions into "open intranuclear cytoplasmic inclusions" connecting to cytoplasm outside the nucleus and "closed intranuclear cytoplasmic inclusions" without that connection. Cytoplasm with abundant organelles was observed in open inclusions, but closed inclusions contained fewer organelles with or without degeneration. Granules with a dense core were only observed in closed inclusions. Our observations suggested that open inclusions originate from nuclear invaginations, and disconnection from cytoplasm leads to closed inclusions.


Subject(s)
Carcinoma , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/diagnosis , Volume Electron Microscopy , Intranuclear Inclusion Bodies/pathology , Intranuclear Inclusion Bodies/ultrastructure , Carcinoma/pathology , Thyroid Neoplasms/pathology , Microscopy, Electron, Scanning
3.
Nucleic Acids Res ; 48(21): 11890-11912, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33068409

ABSTRACT

Eukaryotic cells compartmentalize their internal milieu in order to achieve specific reactions in time and space. This organization in distinct compartments is essential to allow subcellular processing of regulatory signals and generate specific cellular responses. In the nucleus, genetic information is packaged in the form of chromatin, an organized and repeated nucleoprotein structure that is a source of epigenetic information. In addition, cells organize the distribution of macromolecules via various membrane-less nuclear organelles, which have gathered considerable attention in the last few years. The macromolecular multiprotein complexes known as Promyelocytic Leukemia Nuclear Bodies (PML NBs) are an archetype for nuclear membrane-less organelles. Chromatin interactions with nuclear bodies are important to regulate genome function. In this review, we will focus on the dynamic interplay between PML NBs and chromatin. We report how the structure and formation of PML NBs, which may involve phase separation mechanisms, might impact their functions in the regulation of chromatin dynamics. In particular, we will discuss how PML NBs participate in the chromatinization of viral genomes, as well as in the control of specific cellular chromatin assembly pathways which govern physiological mechanisms such as senescence or telomere maintenance.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Genome, Viral , Intranuclear Inclusion Bodies/metabolism , Promyelocytic Leukemia Protein/genetics , Protein Processing, Post-Translational , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Cellular Senescence , Chromatin/chemistry , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , Genome, Human , Histones/genetics , Histones/metabolism , Host-Pathogen Interactions/genetics , Humans , Intranuclear Inclusion Bodies/chemistry , Intranuclear Inclusion Bodies/ultrastructure , Promyelocytic Leukemia Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Telomere Homeostasis , Viruses/genetics , Viruses/metabolism
4.
Brain ; 143(1): 222-233, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31819945

ABSTRACT

Essential tremor is one of the most common movement disorders. Despite its high prevalence and heritability, the genetic aetiology of essential tremor remains elusive. Up to now, only a few genes/loci have been identified, but these genes have not been replicated in other essential tremor families or cohorts. Here we report a genetic study in a cohort of 197 Chinese pedigrees clinically diagnosed with essential tremor. Using a comprehensive strategy combining linkage analysis, whole-exome sequencing, long-read whole-genome sequencing, repeat-primed polymerase chain reaction and GC-rich polymerase chain reaction, we identified an abnormal GGC repeat expansion in the 5' region of the NOTCH2NLC gene that co-segregated with disease in 11 essential tremor families (5.58%) from our cohort. Clinically, probands that had an abnormal GGC repeat expansion were found to have more severe tremor phenotypes, lower activities of daily living ability. Obvious genetic anticipation was also detected in these 11 essential tremor-positive families. These results indicate that abnormal GGC repeat expansion in the 5' region of NOTCH2NLC gene is associated with essential tremor, and provide strong evidence that essential tremor is a family of diseases with high clinical and genetic heterogeneities.


Subject(s)
Asian People/genetics , Essential Tremor/genetics , Trinucleotide Repeat Expansion/genetics , Adult , Aged , Female , GC Rich Sequence , Genetic Linkage , Humans , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/ultrastructure , Male , Microscopy, Electron , Middle Aged , Neurodegenerative Diseases/genetics , Pedigree , Polymerase Chain Reaction , Skin/ultrastructure , Exome Sequencing , Whole Genome Sequencing
5.
Vet Pathol ; 56(6): 907-914, 2019 11.
Article in English | MEDLINE | ID: mdl-31331256

ABSTRACT

Five chimney swift fledglings died following a progressive loss of appetite and condition while being cared for by an experienced wildlife rehabilitator. All animals had severe necrotizing and heterophilic ventriculitis, with myriad epithelial cells characterized by karyomegaly with intranuclear inclusion bodies. Transmission electron microscopy showed distention of epithelial cell nuclei and chromatin peripheralization by nonenveloped, icosahedral, 75- to 85-nm-diameter virions. Degenerate nested PCR for a highly conserved region of the adenovirus DNA polymerase gene was positive. BLAST analysis of the amplicon sequence indicated the presence of a novel adenovirus, with 74% homology to Antarctic penguin adenoviruses and 72% homology to a bat adenovirus, at low query coverages of only 65% and 63%, respectively. BLAST analysis of the predicted amino acid sequence generated the highest scores for squamate adenoviruses at 100% query coverage. Based on phylogenetic analysis of the partial amino acid sequence of the DNA polymerase, the chimney swift virus was a novel adenovirus most closely related to the Atadenovirus genus. Using a probe based on the novel viral sequence, DNA in situ hybridization identified viral nucleic acid in the nucleus. While the tentatively named chimney swift adenovirus-1 (CsAdV-1) is so far classified with the Atadenoviruses, it is relatively divergent from other members of that genus and may represent the first identified member of a new genus of Adenoviruses.


Subject(s)
Adenoviridae Infections/veterinary , Adenoviridae/classification , Bird Diseases/virology , Cerebral Ventriculitis/veterinary , Adenoviridae/genetics , Adenoviridae Infections/diagnostic imaging , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Amino Acid Sequence , Animals , Bird Diseases/diagnostic imaging , Bird Diseases/pathology , Birds , Cerebral Ventriculitis/diagnostic imaging , Cerebral Ventriculitis/pathology , Cerebral Ventriculitis/virology , In Situ Hybridization/veterinary , Intranuclear Inclusion Bodies/ultrastructure , Maine , Microscopy, Electron, Transmission/veterinary , Phylogeny , Polymerase Chain Reaction/veterinary , Virion
6.
Vet Pathol ; 54(3): 520-526, 2017 05.
Article in English | MEDLINE | ID: mdl-28113038

ABSTRACT

Nuclear glycogen inclusions occur infrequently in pathologic conditions but also in normal human and animal tissues. Their function or significance is unclear. To the best of the authors' knowledge, no reports of nuclear glycogen inclusions in canine parietal cells exist. After initial observations of nuclear inclusions/pseudoinclusions during routine histopathology, the authors retrospectively examined samples of gastric mucosa from dogs presenting with gastrointestinal signs for the presence of intranuclear inclusions/pseudoinclusions and determined their composition using histologic and electron-microscopic methods. In 24 of 108 cases (22%), the authors observed various numbers of intranuclear inclusions/pseudoinclusions within scattered parietal cells. Nuclei were characterized by marked karyomegaly and chromatin margination around a central optically empty or slightly eosinophilic area. The intranuclear inclusions/pseudoinclusions stained positive with periodic acid-Schiff (PAS) and were diastase sensitive, consistent with glycogen. Several PAS-positive/diastase-sensitive sections were further examined by transmission electron microscopy, also using periodic acid-thiocarbohydrazide-silver proteinate (PA-TCH-SP) staining to identify polysaccharides. Ultrastructurally, the nuclear inclusions were composed of electron-dense particles that were not membrane bound, without evidence of nuclear membrane invaginations or cytoplasmic organelles in the nuclei, and positive staining with PA-TCH-SP, confirming a glycogen composition. No cytoplasmic glycogen deposits were observed, suggesting that the intranuclear glycogen inclusions were probably synthesized in loco. Nuclear glycogen inclusions were not associated with gastritis or colonization by Helicobacter-like organisms ( P > .05). Our findings suggest that nuclear glycogen inclusions in canine parietal cells could be an incidental finding. Nevertheless, since nuclear glycogen is present in several pathologic conditions, further investigations could be warranted to determine their true significance.


Subject(s)
Dog Diseases/pathology , Glycogen/metabolism , Intranuclear Inclusion Bodies/pathology , Parietal Cells, Gastric/pathology , Animals , Dogs , Female , Intranuclear Inclusion Bodies/ultrastructure , Male , Microscopy, Electron, Transmission/veterinary , Parietal Cells, Gastric/ultrastructure , Retrospective Studies , Stomach Diseases/pathology , Stomach Diseases/veterinary
8.
Biofizika ; 60(5): 941-6, 2015.
Article in Russian | MEDLINE | ID: mdl-26591606

ABSTRACT

The ultrastructure of the retinal pigment epithelium of a diurnal rodent (Brandt's vole) was described taking into account 1) the functions of the pigment epithelium as a participant in the renewal of photoreceptor outer segment and. 2) digestion of outer segment membranes into phagosomes of the retinal pigment epithelium. The myeloid bodies were observed after exposure of the pigment epithelium to light (200 lux, 4 hours) and darkness (0,1 lux, 1,5-hour). In the cytoplasm of the pigment epithelium of the vole no myeloid bodies were observed. Instead of it small lamellar bodies, which have the spiral form and size (from - 200 to 400 nm) were found. The structure of these lamellar bodies was described. Furthermore, the structures, which were presumably responsible for the transport of the digested material, were revealed. The evidence of it is the presence of 1) dense precipitate in the apical domain of the pigment epithelium and 2) microtubules which participate in transport of this precipitate.


Subject(s)
Intranuclear Inclusion Bodies/ultrastructure , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Pigment Epithelium/ultrastructure , Animals , Arvicolinae , Biological Transport , Intranuclear Inclusion Bodies/radiation effects , Light , Photoreceptor Cells, Vertebrate/radiation effects , Retinal Pigment Epithelium/radiation effects
9.
Hum Mol Genet ; 21(3): 485-94, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22010050

ABSTRACT

α-Synuclein and mutant huntingtin are the major constituents of the intracellular aggregates that characterize the pathology of Parkinson's disease (PD) and Huntington's disease (HD), respectively. α-Synuclein is likely to be a major contributor to PD, since overexpression of this protein resulting from genetic triplication is sufficient to cause human forms of PD. We have previously demonstrated that wild-type α-synuclein overexpression impairs macroautophagy in mammalian cells and in transgenic mice. Overexpression of human wild-type α-synuclein in cells and Drosophila models of HD worsens the disease phenotype. Here, we examined whether α-synuclein overexpression also worsens the HD phenotype in a mammalian system using two widely used N-terminal HD mouse models (R6/1 and N171-82Q). We also tested the effects of α-synuclein deletion in the same N-terminal HD mouse models, as well as assessed the effects of α-synuclein deletion on macroautophagy in mouse brains. We show that overexpression of wild-type α-synuclein in both mouse models of HD enhances the onset of tremors and has some influence on the rate of weight loss. On the other hand, α-synuclein deletion in both HD models increases autophagosome numbers and this is associated with a delayed onset of tremors and weight loss, two of the most prominent endophenotypes of the HD-like disease in mice. We have therefore established a functional link between these two aggregate-prone proteins in mammals and provide further support for the model that wild-type α-synuclein negatively regulates autophagy even at physiological levels.


Subject(s)
Huntington Disease/metabolism , alpha-Synuclein/metabolism , Age of Onset , Animals , Brain/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Intranuclear Inclusion Bodies/ultrastructure , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Tremor/epidemiology , Tremor/metabolism , Weight Loss , alpha-Synuclein/deficiency , alpha-Synuclein/genetics
10.
J Virol ; 87(15): 8441-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698311

ABSTRACT

ORF78 (ac78) of Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is a baculovirus core gene of unknown function. To determine the role of ac78 in the baculovirus life cycle, an AcMNPV mutant with ac78 deleted, Ac78KO, was constructed. Quantitative PCR analysis revealed that ac78 is a late gene in the viral life cycle. After transfection into Spodoptera frugiperda cells, Ac78KO produced a single-cell infection phenotype, indicating that no infectious budded viruses (BVs) were produced. The defect in BV production was also confirmed by both viral titration and Western blotting. However, viral DNA replication was unaffected, and occlusion bodies were formed. An analysis of BVs and occlusion-derived viruses (ODVs) revealed that AC78 is associated with both forms of the virions and is an envelope structural protein. Electron microscopy revealed that AC78 also plays an important role in the embedding of ODV into the occlusion body. The results of this study demonstrate that AC78 is a late virion-associated protein and is essential for the viral life cycle.


Subject(s)
Nucleopolyhedroviruses/physiology , Viral Proteins/metabolism , Virus Release , Animals , Cell Nucleus/ultrastructure , Cell Nucleus/virology , DNA, Viral/chemistry , DNA, Viral/genetics , Gene Deletion , Gene Expression Profiling , Genes, Essential , Intranuclear Inclusion Bodies/metabolism , Intranuclear Inclusion Bodies/ultrastructure , Microscopy, Electron, Transmission , Molecular Sequence Data , Nucleopolyhedroviruses/genetics , Nucleopolyhedroviruses/ultrastructure , Sequence Analysis, DNA , Sf9 Cells , Spodoptera , Viral Proteins/genetics , Virus Replication
11.
Am J Pathol ; 180(3): 1095-1106, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266251

ABSTRACT

Progressive multifocal leukoencephalopathy is a fatal demyelinating disorder caused by JC virus infection. JC virus was recently found to target promyelocytic leukemia nuclear bodies (PML-NBs), punctuate domains in the nuclei. Thus, the virus progenies cluster in dots as intranuclear inclusions (ie, as dot-shaped inclusions). In the present study, both the viral major and minor capsid proteins were expressed from polycistronic expression vectors with a powerful promoter, and formation into virus-like particles (VLPs) was examined by electron microscopy. When the upstream regulatory sequence including the agnogene (nt 275 to 490) was present, capsid protein expression was suppressed, but numerous VLPs were efficiently formed with restricted accumulation to PML-NBs. VLPs were uniform, and the cells were severely degraded. In contrast, when the 5' terminus of the agnogene (nt 275 to 409; 135 bp) was deleted, capsid protein expression was markedly enhanced, but VLPs were more randomly produced in the nucleus outside of PML-NBs. VLPs were pleomorphic, and cell degradation was minimal. JC virus association with PML-NBs was confirmed in human brain tissues by structured illumination microscopy. PML-NBs were shaped in spherical shells, with viral capsid proteins circumscribing the surface. These findings indicate that PML-NBs are intranuclear locations for pathogenic JC virus proliferation. Either the agnogene or its product likely supports efficient progeny production at PML-NBs, leading to subsequent degeneration of host glial cells.


Subject(s)
Intranuclear Inclusion Bodies/ultrastructure , JC Virus/ultrastructure , Leukemia, Promyelocytic, Acute/pathology , Leukoencephalopathy, Progressive Multifocal/pathology , Capsid Proteins/ultrastructure , Cells, Cultured , Humans , Leukemia, Promyelocytic, Acute/virology , Microscopy , Microscopy, Electron , Transfection , Virion/ultrastructure
12.
Biol Cell ; 104(7): 418-32, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22443097

ABSTRACT

BACKGROUND INFORMATION: Promyelocytic leukaemia (PML) bodies are specific nuclear structures with functional significance for acute promyelocytic leukaemia. In this study, we analysed the trajectories of PML bodies using single-particle tracking. RESULTS: We observed that the recovery of PML protein after photobleaching was ATP dependent in both wild-type (wt) and A-type lamin-deficient cells. The movement of PML bodies was faster and the nuclear area occupied by particular PML bodies was larger in A-type lamin-deficient fibroblasts compared with their wt counterparts. Moreover, dysfunction of the LMNA gene increased the frequency of mutual interactions between individual PML bodies and influenced the morphology of these domains at the ultrastructural level. As a consequence of A-type lamin deficiency, PML protein accumulated in nuclear blebs and frequently appeared at the nuclear periphery. CONCLUSIONS: We suggest that the physiological function of lamin A proteins is important for events that occur in the compartment of PML bodies. This observation was confirmed in other experimental models characterised by lamin changes, including apoptosis or the differentiation of mouse embryonic stem cells.


Subject(s)
Intranuclear Inclusion Bodies/metabolism , Lamin Type A/deficiency , Leukemia, Promyelocytic, Acute/metabolism , Animals , Apoptosis , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , Intranuclear Inclusion Bodies/ultrastructure , Kinetics , Lamin Type A/metabolism , Mice , Reproducibility of Results
13.
Cell Mol Life Sci ; 69(18): 3027-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22476590

ABSTRACT

The nucleus of higher eukaryotes, such as humans and mice, is compartmentalized into multiple nuclear bodies, an organization that allows for the regulation of complex gene expression pathways that are characteristic of these organisms. Paraspeckles are recently discovered, mammalian-specific nuclear bodies built on a long, non-protein-coding RNA, NEAT1 (nuclear-enriched abundant transcript 1), which assembles various protein components including RNA-binding proteins of the DBHS (Drosophila behavior and human splicing) family. Paraspeckles have been proposed to control several biological processes, such as stress responses and cellular differentiation, but their function at the whole animal level remains unclear. In this review, we summarize a series of studies on paraspeckles that have been carried out in the decade since their discovery and discuss their physiological function and molecular mechanism.


Subject(s)
Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/metabolism , Animals , Humans , Intranuclear Inclusion Bodies/ultrastructure , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Editing , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
14.
Microsc Microanal ; 19(2): 360-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23410959

ABSTRACT

We used hybrid detectors (HyDs) to monitor the trajectories and interactions of promyelocytic leukemia (GFP-PML) nuclear bodies (NBs) and mCherry-53BP1-positive DNA lesions. 53BP1 protein accumulates in NBs that occur spontaneously in the genome or in γ-irradiation-induced foci. When we induced local DNA damage by ultraviolet irradiation, we also observed accumulation of 53BP1 proteins into discrete bodies, instead of the expected dispersed pattern. In comparison with photomultiplier tubes, which are used for standard analysis by confocal laser scanning microscopy, HyDs significantly eliminated photobleaching of GFP and mCherry fluorochromes during image acquisition. The low laser intensities used for HyD-based confocal analysis enabled us to observe NBs for the longer time periods, necessary for studies of the trajectories and interactions of PML and 53BP1 NBs. To further characterize protein interactions, we used resonance scanning and a novel bioinformatics approach to register and analyze the movements of individual PML and 53BP1 NBs. The combination of improved HyD-based confocal microscopy with a tailored bioinformatics approach enabled us to reveal damage-specific properties of PML and 53BP1 NBs.


Subject(s)
DNA/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/pathology , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Cell Line, Tumor , DNA/metabolism , DNA Damage , Humans , Intranuclear Inclusion Bodies/ultrastructure , Leukemia, Promyelocytic, Acute/metabolism , Time-Lapse Imaging , Tumor Suppressor p53-Binding Protein 1
15.
J Cell Sci ; 123(Pt 3): 392-400, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20130140

ABSTRACT

Promyelocytic leukemia nuclear bodies (PML-NBs) are mobile subnuclear organelles formed by PML and Sp100 protein. They have been reported to have a role in transcription, DNA replication and repair, telomere lengthening, cell cycle control and tumor suppression. We have conducted high-resolution 4Pi fluorescence laser-scanning microscopy studies complemented with correlative electron microscopy and investigations of the accessibility of the PML-NB subcompartment. During interphase PML-NBs adopt a spherical organization characterized by the assembly of PML and Sp100 proteins into patches within a 50- to 100-nm-thick shell. This spherical shell of PML and Sp100 imposes little constraint to the exchange of components between the PML-NB interior and the nucleoplasm. Post-translational SUMO modifications, telomere repeats and heterochromatin protein 1 were found to localize in characteristic patterns with respect to PML and Sp100. From our findings, we derived a model that explains how the three-dimensional organization of PML-NBs serves to concentrate different biological activities while allowing for an efficient exchange of components.


Subject(s)
Intranuclear Inclusion Bodies/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antigens, Nuclear/metabolism , Antigens, Nuclear/ultrastructure , Autoantigens/metabolism , Autoantigens/ultrastructure , Cell Line, Tumor , HeLa Cells , Humans , Intranuclear Inclusion Bodies/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Models, Biological , Nuclear Proteins/ultrastructure , Promyelocytic Leukemia Protein , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/ultrastructure , Tumor Suppressor Proteins/ultrastructure , Ubiquitins/metabolism
16.
Am J Pathol ; 178(2): 803-16, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281813

ABSTRACT

Many models of human tauopathies have been generated in mice by expression of a human mutant tau with maintained expression of mouse endogenous tau. Because murine tau might interfere with the toxic effects of human mutant tau, we generated a model in which a pathogenic human tau protein is expressed in the absence of wild-type tau protein, with the aim of facilitating the study of the pathogenic role of the mutant tau and to reproduce more faithfully a human tauopathy. The Tg30 line is a tau transgenic mouse model overexpressing human 1N4R double-mutant tau (P301S and G272V) that develops Alzheimer's disease-like neurofibrillary tangles in an age-dependent manner. By crossing Tg30 mice with mice invalidated for their endogenous tau gene, we obtained Tg30xtau(-/-) mice that express only exogenous human double-mutant 1N4R tau. Although Tg30xtau(-/-) mice express less tau protein compared with Tg30, they exhibit signs of decreased survival, increased proportion of sarkosyl-insoluble tau in the brain and in the spinal cord, increased number of Gallyas-positive neurofibrillary tangles in the hippocampus, increased number of inclusions in the spinal cord, and a more severe motor phenotype. Deletion of murine tau accelerated tau aggregation during aging of this mutant tau transgenic model, suggesting that murine tau could interfere with the development of tau pathology in transgenic models of human tauopathies.


Subject(s)
Gene Knockout Techniques , Microtubule-Associated Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , tau Proteins/metabolism , Animals , Cell Count , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/pathology , Intranuclear Inclusion Bodies/ultrastructure , Memory, Short-Term/drug effects , Mice , Mice, Transgenic , Models, Animal , Motor Activity/drug effects , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/ultrastructure , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Solubility/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Survival Analysis , tau Proteins/chemistry
17.
Neuropathology ; 31(2): 188-93, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20667015

ABSTRACT

We report an incipient case of intranuclear inclusion body disease (INIBD) in a 78-year-old woman. No apparent neurological symptoms were noticed during the clinical course. Post mortem examination revealed widespread occurrence of eosinophilic intranuclear inclusions in neuronal and glial cells of the central and peripheral nervous systems, as well as in parenchymal cells of the visceral organs. The inclusions were observed more frequently in glial cells than in neuronal cells. Ultrastructurally, the inclusions consisted of granular and filamentous material. Immunohistochemically, the inclusions were positive for ubiquitin, ubiquitin-related proteins (NEDD8 ultimate buster 1, small ubiquitin modifier-1, small ubiquitin modifier-2 and p62), promyelocytic leukemia protein and abnormally expanded polyglutamine. Consistent with previous studies, the vast majority of inclusion-bearing glial cells were astrocytes. Furthermore, p25α-positive oligodendrocytes rarely contained intranuclear inclusions. These findings suggest that INIBD may occur in non-demented elderly individuals and that oligodendrocyte is also involved in the disease process of INIBD.


Subject(s)
Brain Diseases/pathology , Intranuclear Inclusion Bodies/ultrastructure , Neurodegenerative Diseases/pathology , Aged , Astrocytes/pathology , Diabetes Mellitus , Female , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Oligodendroglia/pathology
18.
Neurobiol Dis ; 33(3): 331-41, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19130884

ABSTRACT

Huntington's disease (HD) is caused by an expanded CAG repeat in the HD gene. The pathological threshold for expansion in HD is around 36 CAG repeats, although 'super-long' expansions are found in brains of HD patients. We examined the effect of varying the CAG repeat length (from 170 to 450) on behavior and neuropathology of R6/2 mice. Unexpectedly, we found that increasing the repeat length delayed onset of disease and prolonged survival, from around 4 months to over 18 months in mice with the longest repeats. The delay in onset correlated with a delayed appearance of neuronal intranuclear inclusions (NIIs). However, super-long CAG repeats are not neuroprotective. Mice carrying 2 copies of the mutant transgene die earlier than those carrying a single copy. Furthermore, neurodegeneration is present in super-long repeat length mice at mid-stage disease, whereas little neurodegeneration is seen in mice with shorter CAG repeats until end stage. Expanding the CAG repeat beyond the range where NII formation is the dominant pathology has unmasked a slowly progressing neurological phenotype in R6/2 mice with brain pathology, including the identification of a novel form of inclusion, that more closely resembles that seen in adult onset cases of HD. This mouse may represent a better model for adult-onset HD than R6/2 mice with shorter repeats.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Trinucleotide Repeat Expansion , Age of Onset , Aging , Animals , Body Weight , Brain/pathology , Brain/ultrastructure , Disease Progression , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/physiopathology , Intranuclear Inclusion Bodies/ultrastructure , Mice , Mice, Transgenic , Motor Activity , Nerve Degeneration , Neurons/physiology , Neurons/ultrastructure , Phenotype , Postural Balance , Survival Rate
20.
Neurol Neurochir Pol ; 43(2): 113-20, 2009.
Article in English | MEDLINE | ID: mdl-19484687

ABSTRACT

BACKGROUND AND PURPOSE: Oculopharyngeal muscular dystrophy (OPMD) is mostly an autosomal dominant myopathic disorder, characterized by progressive bilateral ptosis, dysphagia and proximal muscle weakness, appearing usually in the fifth to sixth decade of life. The underlying cause of OPMD is an expanded GCG repeat in the first exon of the gene encoding poly (A)-binding protein nuclear 1 (PABPN1) localized on chromosome 14.q11.2-q13. The number of GCG expansion ranges from 8 to 13 repeats. PABPN1 is a nuclear multifunctional protein which is involved in transcription regulation and post-transcriptional processes. MATERIAL AND METHODS: We report on clinical characteristics in 9 Polish patients with genetically confirmed OPMD. RESULTS: The expanded repeat ranged from (GCG)8 to (GCG)11. Ptosis and dysphagia were present in all examined cases. In 4 patients weakness of extraocular muscle was found and two of them experienced transient diplopia. Mild limb-girdle weakness was observed in 6 patients. Muscle biopsy performed in all cases showed myopathic changes with rare rimmed vacuoles. Strikingly, despite thorough examination on electron microscopy, intranuclear inclusions typical for OPMD were found only in one patient. CONCLUSIONS: Genetic testing is necessary to confirm the diagnosis of OPMD, especially in cases with ptosis and external ophthalmoparesis, which may be initially diagnosed as mitochondrial myopathy.


Subject(s)
Muscular Dystrophy, Oculopharyngeal/diagnosis , Muscular Dystrophy, Oculopharyngeal/genetics , Adult , Aged , Biopsy , Cell Nucleus/ultrastructure , Diagnosis, Differential , Electromyography , Female , Genotype , Humans , Intranuclear Inclusion Bodies/ultrastructure , Male , Middle Aged , Mitochondrial Myopathies/diagnosis , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Mutation , Pedigree , Phenotype , Poly(A)-Binding Protein I/genetics
SELECTION OF CITATIONS
SEARCH DETAIL