Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(12): 1733-1744, 2017 03 23.
Article in English | MEDLINE | ID: mdl-27641332

ABSTRACT

Long-term survival remains low for most patients with glioblastoma (GBM), which reveals the need for markers of disease outcome and novel therapeutic targets. We describe that ODZ1 (also known as TENM1), a type II transmembrane protein involved in fetal brain development, plays a crucial role in the invasion of GBM cells. Differentiation of glioblastoma stem-like cells drives the nuclear translocation of an intracellular fragment of ODZ1 through proteolytic cleavage by signal peptide peptidase-like 2a. The intracellular fragment of ODZ1 promotes cytoskeletal remodelling of GBM cells and invasion of the surrounding environment both in vitro and in vivo. Absence of ODZ1 by gene deletion or downregulation of ODZ1 by small interfering RNAs drastically reduces the invasive capacity of GBM cells. This activity is mediated by an ODZ1-triggered transcriptional pathway, through the E-box binding Myc protein, that promotes the expression and activation of Ras homolog family member A (RhoA) and subsequent activation of Rho-associated, coiled-coil containing protein kinase (ROCK). Overexpression of ODZ1 in GBM cells reduced survival of xenografted mice. Consistently, analysis of 122 GBM tumour samples revealed that the number of ODZ1-positive cells inversely correlated with overall and progression-free survival. Our findings establish a novel marker of invading GBM cells and consequently a potential marker of disease progression and a therapeutic target in GBM.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tenascin/genetics , Transcription, Genetic , rhoA GTP-Binding Protein/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Knockout Techniques , Glioblastoma/mortality , Glioblastoma/pathology , Heterografts , Humans , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Prognosis , Protein Transport , Proteolysis , Signal Transduction , Tenascin/deficiency , Tenascin/metabolism , Up-Regulation , rho-Associated Kinases/metabolism
2.
Oncogene ; 30(32): 3537-48, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21423202

ABSTRACT

Glioblastoma multiforme is one of the most devastating cancers and presents unique challenges to therapy because of its aggressive behavior. Cancer-initiating or progenitor cells have been described to be the only cell population with tumorigenic capacity in glioblastoma. Therefore, effective therapeutic strategies targeting these cells or the early precursors may be beneficial. We have established different cultures of glioblastoma-initiating cells (GICs) derived from surgical specimens and found that, after induction of differentiation, the NFκB transcriptional pathway was activated, as determined by analyzing key proteins such as p65 and IκB and the upregulation of a number of target genes. We also showed that blockade of nuclear factor (NF)κB signaling in differentiating GICs by different genetic strategies or treatment with small-molecule inhibitors, promoted replication arrest and senescence. This effect was partly mediated by reduced levels of the NFκB target gene cyclin D1, because its downregulation by RNA interference reproduced a similar phenotype. Furthermore, these results were confirmed in a xenograft model. Intravenous treatment of immunodeficient mice bearing human GIC-derived tumors with a novel small-molecule inhibitor of the NFκB pathway induced senescence of tumor cells but no ultrastructural alterations of the brain parenchyma were detected. These findings reveal that activation of NFκB may keep differentiating GICs from acquiring a mature postmitotic phenotype, thus allowing cell proliferation, and support the rationale for therapeutic strategies aimed to promote premature senescence of differentiating GICs by blocking key factors within the NFκB pathway.


Subject(s)
Cellular Senescence/genetics , Glioblastoma/genetics , NF-kappa B/genetics , Signal Transduction/genetics , Animals , Blotting, Western , Carbazoles/pharmacology , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/pathology , Glycosides/pharmacology , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sulfones/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Struct Biol ; 163(2): 137-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18571432

ABSTRACT

Cajal bodies (CBs) are nuclear organelles involved in the maturation of small nuclear ribonucleoproteins required for the processing of pre-mRNAs. They concentrate coilin, splicing factors and the survival of motor neuron protein (SMN). By using immunocytochemistry and transfection experiments with GFP-SUMO-1, DsRed1-Ubc9, GFP-coilin and GFP-SMN constructs we demonstrate the presence of SUMO-1 and the SUMO conjugating enzyme (Ubc9) in a subset of CBs in undifferentiated neuron-like UR61 cells. Furthermore, SUMO-1 is transiently localized into neuronal CBs from adult nervous tissue in response to osmotic stress or inhibition of methyltransferase activity. SUMO-1-positive CBs contain coilin, SMN and small nuclear ribonucleoproteins, suggesting that they are functional CBs involved in pre-mRNA processing. Since coilin and SMN have several putative motifs of SUMO-1 modification, we suggest that the sumoylation of coilin and/or SMN might play a role in the molecular reorganization of CBs during the neuronal differentiation or stress-response.


Subject(s)
Coiled Bodies/chemistry , SUMO-1 Protein/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Methyltransferases/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/chemistry , Nuclear Proteins/metabolism , Osmotic Pressure , PC12 Cells , RNA-Binding Proteins/metabolism , Rats , SMN Complex Proteins , SUMO-1 Protein/analysis , Survival of Motor Neuron 1 Protein , Ubiquitin-Conjugating Enzymes/analysis
4.
Neuroscience ; 140(2): 453-62, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16563640

ABSTRACT

In this study we have used the transcription assay with 5'-fluorouridine incorporation into nascent RNA to analyze the nuclear organization and dynamics of transcription sites in rat trigeminal ganglia neurons. The 5'-FU administrated by i.p. injection was successfully incorporated into nuclear domains containing actively transcribing genes of trigeminal neurons. 5'-Fluorouridine RNA-labeling was detected with immunocytochemistry at light and electron microscopy levels. The 5'-fluorouridine incorporation sites were detected in the nucleolus, particularly on the dense fibrillar component, and in numerous transcription foci spread throughout the euchromatin regions, without preferential positioning at the nuclear periphery or in the nuclear interior. Double labeling experiments to combine 5'-fluorouridine incorporation with molecular markers of nuclear compartments showed the absence of transcription sites in Cajal bodies and nuclear speckles of splicing factors. Similarly, no 5'-fluorouridine labeling was detected in well-characterized chromatin silencing domain, the telomeric heterochromatin. The specificity and sensitivity of the run-on transcription assay in trigeminal ganglia neurons was verified by the i.p. administration of the transcription inhibitor actinomycin D. The dramatic reduction in RNA synthesis upon actinomycin D treatment was associated with two important cellular events, heterochromatin silencing and formation of DNA damage/repair nuclear foci, demonstrated by the expression of tri-methylated histone H4 and phosphorylated H2AX, respectively. 5'-Fluorouridine incorporation in animal models provides a useful tool to investigate the organization of gene expression in mammalian neurons in both normal physiology and experimental pathology systems.


Subject(s)
Cell Nucleus/metabolism , Neurons, Afferent/metabolism , RNA, Messenger/biosynthesis , Transcription, Genetic/physiology , Trigeminal Ganglion/metabolism , Uridine/analogs & derivatives , Animals , Biological Assay/methods , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , DNA Repair/physiology , Dactinomycin/pharmacology , Euchromatin/genetics , Euchromatin/metabolism , Euchromatin/ultrastructure , Gene Expression/physiology , Gene Silencing/physiology , Histones/metabolism , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Neurons, Afferent/ultrastructure , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Staining and Labeling/methods , Transcriptional Activation/physiology , Trigeminal Ganglion/ultrastructure , Uridine/metabolism
5.
Apoptosis ; 11(5): 701-15, 2006 May.
Article in English | MEDLINE | ID: mdl-16532376

ABSTRACT

Here we show a detailed analysis of cellular and molecular events during in vivo apoptotic cell death in the INZs (interdigital necrotic zones) of the embryonic limb. As the apoptotic mechanisms proceed, the transcriptionally active chromatin and nuclear traffic of RNAs are disrupted, cytoskeletal components are disorganized and the adhesive properties of cells are compromised as Paxillin, a clue member of the focal adhesion complex, decreases in early apoptotic cells. Activation of effector caspases 3 and 7 follow nuclear degradation. In addition, active caspase2 is localized in the nuclei and cytoplasm of early apoptotic cells suggesting a major role in physiological conditions supported by its down-regulation in tissue survival experiments. However in caspase 2 siRNA assays we observed translocation of caspase 3 to the nuclei suggesting functional redundancy. We also observed release of cytochrome c and AIF from the mitochondria, and interestingly AIF becomes intranuclear in a caspase independent manner.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/physiology , Extremities/growth & development , Mitochondria/enzymology , Mitochondria/metabolism , Animals , Caspase 2 , Caspase 3 , Caspase 7 , Caspases/genetics , Caspases/metabolism , Cell Death , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Chick Embryo , Cytochromes c/metabolism , Cytoplasm/enzymology , Cytoplasm/metabolism , Down-Regulation , Enzyme Activation , Gene Expression Regulation, Developmental , Paxillin/metabolism , RNA, Small Interfering/analysis , RNA, Small Interfering/metabolism
6.
J Neurocytol ; 33(4): 393-405, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15520525

ABSTRACT

In this study we have taken advantage of the high nuclear responsiveness of type A sensory ganglia neurons to variations of cellular activity to investigate the reorganization and dynamics of nuclear compartments involved in transcription and RNA processing in response to neuronal injury. As experimental model we have used the inflammatory injury of the peripheral nerve endings induced by formalin injection in the areas of ophthalmic/maxillary nerve distribution. We have performed immunofluorescence and confocal laser microscopy analysis with specific antibodies for different nuclear compartments and ultrastructural analysis. The initial response to neuronal injury, within the 3 days post-injury, consisted of chromatin condensation, reduction in the expression level of acetylated histone H4, accumulation of perichromatin granules, reorganization of splicing factors in prominent nuclear speckles, reduction in the number of Cajal bodies and nucleolar alterations. These changes tended to revert by day 7 post-injury and are consistent with a transient inhibition of transcription and RNA processing. Moreover, we have observed an early and sustained expression of the transcription factor c-Jun. These results illustrate the transcription-dependent organization of nuclear compartments in type A trigeminal neurons and also support the importance of the nuclear response to axonal injury as a key component in the regenerative capacity of this neuronal population.


Subject(s)
Cell Nucleus/ultrastructure , Inflammation/pathology , Neurons/cytology , Peripheral Nerves/pathology , Trigeminal Ganglion/cytology , Animals , Cell Nucleus/metabolism , Male , Neurons/metabolism , Neurons/pathology , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Trigeminal Ganglion/pathology
8.
J Cell Biol ; 154(2): 293-307, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11470819

ABSTRACT

Cajal bodies (CBs) are nuclear suborganelles involved in the biogenesis of small nuclear ribonucleoproteins (snRNPs). In addition to snRNPs, they are highly enriched in basal transcription and cell cycle factors, the nucleolar proteins fibrillarin (Fb) and Nopp140 (Nopp), the survival motor neuron (SMN) protein complex, and the CB marker protein, p80 coilin. We report the generation of knockout mice lacking the COOH-terminal 487 amino acids of coilin. Northern and Western blot analyses demonstrate that we have successfully removed the full-length coilin protein from the knockout animals. Some homozygous mutant animals are viable, but their numbers are reduced significantly when crossed to inbred backgrounds. Analysis of tissues and cell lines from mutant animals reveals the presence of extranucleolar foci that contain Fb and Nopp but not other typical nucleolar markers. These so-called "residual" CBs neither condense Sm proteins nor recruit members of the SMN protein complex. Transient expression of wild-type mouse coilin in knockout cells results in formation of CBs and restores these missing epitopes. Our data demonstrate that full-length coilin is essential for proper formation and/or maintenance of CBs and that recruitment of snRNP and SMN complex proteins to these nuclear subdomains requires sequences within the coilin COOH terminus.


Subject(s)
Coiled Bodies/metabolism , Mice, Knockout/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/deficiency , Ribonucleoproteins, Small Nuclear/metabolism , Animals , Autoantigens/metabolism , Blotting, Northern , Cell Line , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Coiled Bodies/genetics , Coiled Bodies/ultrastructure , Cyclic AMP Response Element-Binding Protein , Fetal Viability/genetics , Gene Expression/drug effects , Gene Targeting , Green Fluorescent Proteins , Homozygote , Luminescent Proteins/genetics , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Phosphoproteins/metabolism , RNA Splicing , RNA, Messenger , RNA-Binding Proteins , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , SMN Complex Proteins , Survival Rate , snRNP Core Proteins
9.
J Cell Biol ; 154(2): 369-87, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11470825

ABSTRACT

The beta-catenin signaling pathway is deregulated in nearly all colon cancers. Nonhypercalcemic vitamin D3 (1alpha,25-dehydroxyvitamin D(3)) analogues are candidate drugs to treat this neoplasia. We show that these compounds promote the differentiation of human colon carcinoma SW480 cells expressing vitamin D receptors (VDRs) (SW480-ADH) but not that of a malignant subline (SW480-R) or metastasic derivative (SW620) cells lacking VDR. 1alpha,25(OH)2D(3) induced the expression of E-cadherin and other adhesion proteins (occludin, Zonula occludens [ZO]-1, ZO-2, vinculin) and promoted the translocation of beta-catenin, plakoglobin, and ZO-1 from the nucleus to the plasma membrane. Ligand-activated VDR competed with T cell transcription factor (TCF)-4 for beta-catenin binding. Accordingly, 1alpha,25(OH)2D(3) repressed beta-catenin-TCF-4 transcriptional activity. Moreover, VDR activity was enhanced by ectopic beta-catenin and reduced by TCF-4. Also, 1alpha,25(OH)2D(3) inhibited expression of beta-catenin-TCF-4-responsive genes, c-myc, peroxisome proliferator-activated receptor delta, Tcf-1, and CD44, whereas it induced expression of ZO-1. Our results show that 1alpha,25(OH)2D(3) induces E-cadherin and modulates beta-catenin-TCF-4 target genes in a manner opposite to that of beta-catenin, promoting the differentiation of colon carcinoma cells.


Subject(s)
Adenocarcinoma/metabolism , Cadherins/biosynthesis , Calcitriol/analogs & derivatives , Cell Differentiation/drug effects , Cholecalciferol/pharmacology , Colonic Neoplasms/metabolism , Cytoskeletal Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Trans-Activators , Active Transport, Cell Nucleus/drug effects , Adenocarcinoma/pathology , Antineoplastic Agents , Calcitriol/pharmacology , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Colonic Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Macromolecular Substances , Phenotype , Protein Binding/drug effects , RNA, Messenger/metabolism , Receptors, Calcitriol/metabolism , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , beta Catenin
10.
J Comp Neurol ; 430(2): 250-63, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11135260

ABSTRACT

Trigeminal ganglion neurons comprise three main cell body-size types. This cell size heterogeneity provides an excellent neuronal model to study the cell size-dependent organization and dynamics of the nucleoli, Cajal (coiled) bodies (CBs), and nuclear speckles of pre-mRNA splicing factors, nuclear structures that play a key role in the normal neuronal physiology. We have analyzed the number of nucleoli and CBs and the structural and molecular organization of CBs and nuclear speckles in the three neuronal types by using immunofluorescence with antibodies that recognize nucleoli (fibrillarin), CBs (coilin), and nuclear speckles (snRNPs), confocal microscopy, and electron microscopy. Whereas the mean number of nucleoli per neuron decreases as a function of cell size, the number of CBs per cell significantly increases in large neurons in comparison with the small ones. In addition, large neurons have a higher proportion of CBs associated with the nucleolus. In all neuronal types, CBs concentrate coilin, fibrillarin, snRNPs, and the survival motor neuron protein (SMN). Immunostaining for snRNPs shows small speckle domains and extensive areas of diffuse nucleoplasmic signal in large neurons, in contrast with the large nuclear speckles found in small neurons. Furthermore, flow cytometric analysis shows that all neurons are in the range of diploid cells. These findings indicate that the fusion behavior of nucleoli, the formation of CBs and their relationships with the nucleolus, as well as the compartmentalization of the pre-mRNA splicing machinery, is related to cell body size in the trigeminal ganglion neurons. Because transcriptional activity is a basic determinant mechanism of cell size in diploid cells, we suggest that our findings reflect a distinct transcription-dependent organization of the nucleolus and splicing machinery in the three cell types of trigeminal ganglion neurons.


Subject(s)
Cell Nucleolus/ultrastructure , Inclusion Bodies/ultrastructure , Neurons/cytology , Neurons/physiology , RNA Splicing/physiology , Rats/physiology , Trigeminal Ganglion/cytology , Trigeminal Ganglion/physiology , Animals , Cell Size , DNA/metabolism , Inclusion Bodies/metabolism , Male , Microscopy, Electron , Microscopy, Electron, Scanning , Neurology/methods , Organelles/ultrastructure , Rats, Sprague-Dawley , Ribonucleoproteins, Small Nuclear/physiology
11.
J Cell Biol ; 150(5): 1199-208, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10974006

ABSTRACT

The immunosuppressive and antiinflammatory actions of glucocorticoid hormones are mediated by their transrepression of activating protein-1 (AP-1) and nuclear factor-kappa B (NFkappaB) transcription factors. Inhibition of the c-Jun NH(2)-terminal kinase (JNK) signaling pathway, the main mediator of AP-1 activation, has been described in extracts of hormone-treated cells. Here, we show by confocal laser microscopy, enzymatic assays, and immunoblotting that the synthetic glucocorticoid dexamethasone inhibited tumor necrosis factor alpha (TNF-alpha)-induced phosphorylation and activation of JNK in the cytoplasm and nucleus of intact HeLa cells. As a result, c-Jun NH(2)-terminal domain phosphorylation and induction were impaired. Dexamethasone did not block the TNF-alpha-induced JNK nuclear translocation, but rather induced, per se, nuclear accumulation of the enzyme. Consistently with previous findings, a glucocorticoid receptor mutant (GRdim), which is deficient in dimerization, DNA binding, and transactivation, but retains AP-1 transrepressing activity, was as efficient as wild-type GR in mediating the same effects of dexamethasone on JNK in transfected Cos-7 cells. Our results show that glucocorticoids antagonize the TNF-alpha-induced activation of AP-1 by causing the accumulation of inactive JNK without affecting its subcellular distribution.


Subject(s)
Cell Nucleus/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/metabolism , Animals , COS Cells , Cell Fractionation , Cell Nucleus/drug effects , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Mitogen-Activated Protein Kinases/drug effects , Phosphorylation , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Transfection , Tumor Necrosis Factor-alpha/pharmacology
12.
Neurology ; 55(4): 552-9, 2000 Aug 22.
Article in English | MEDLINE | ID: mdl-10953191

ABSTRACT

OBJECTIVE: To assess if axonal damage in severe acute inflammatory demyelinating polyneuropathy (AIDP) correlates with the appearance of epiperineurium in nerve trunks. BACKGROUND: Increase of endoneurial fluid pressure in nerve trunks possessing epiperineurium may be an important mechanism of axonal damage in AIDP. METHODS: A 79-year-old man had a 2-day history of acroparesthesias and ascending paralysis culminating in quadriplegia, bilateral facial palsy, and mechanical ventilation. Five intravenous immunoglobulin cycles were given without response. He died on day 60. Electrophysiologic studies (days 4, 17, and 50) initially showed normal nerve conduction velocities with further slowing, progressive attenuation of compound muscle action potentials, and profuse denervation. The authors studied the preforaminal anterior and posterior L3 and L5 spinal roots, third and fifth lumbar nerves and their branches, and femoral and sural nerves. RESULTS: Density of myelinated fibers was preserved in L5 ventral and dorsal roots and reduced in sural nerve. Mild de-remyelination was observed in lumbar roots. In both lumbar nerves and their branches, there were extensive de-remyelination and centrofascicular or wedge-shaped areas with marked loss of large myelinated fibers. Axonal degeneration was the predominant lesion in sural nerve. CONCLUSION: The presence of epiperineurium correlates with a drastic change of pathology with superimposed ischemic lesions and distally accentuated axonal loss, suggesting that endoneurial fluid pressure increase could cause axonal damage in AIDP.


Subject(s)
Axons/pathology , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/physiopathology , Peripheral Nerves/physiopathology , Action Potentials , Aged , Axons/ultrastructure , Carcinoma, Transitional Cell/complications , Carcinoma, Transitional Cell/pathology , Electromyography , Fatal Outcome , Guillain-Barre Syndrome/complications , Guillain-Barre Syndrome/drug therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Lumbosacral Region , Male , Neural Conduction , Spinal Cord/pathology , Spinal Nerve Roots/physiopathology , Sural Nerve/physiopathology , Urinary Bladder Neoplasms/complications , Urinary Bladder Neoplasms/pathology
13.
Exp Cell Res ; 256(1): 179-91, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10739665

ABSTRACT

In response to cellular stress, the activation of the JNK cascade mediates phosphorylation of c-Jun that promotes its transactivation, which in turn activates the transcription of specific genes. In an experimental model of neuronal stress in vivo, by means of immunofluorescence and kinase assays we have found a reversible activation of JNK induced by the administration of the anti-cancer drug Adriamycin. In control neurons, a considerable basal level of the active, phosphorylated JNK was detected in neuronal nuclei, with a speckled distribution in addition to a diffuse nucleoplasmic signal. Adriamycin-induced neuronal stress was associated with a notable increase of this nuclear immunostaining, indicating activation of the JNK pathway which was confirmed by the increase of JNK enzymatic activity, while no changes in the total JNK were detected by Western blots. The JNK neuronal response to stress was also accompanied by an increase in the nuclear immunoreactivity for c-Jun and also by the de novo appearance of a strong nuclear phospho-c-Jun signal. These effects tend to revert to the control situation after 24 h of Adriamycin treatment. The nuclear compartmentalization of phospho-JNK and its substrate c-Jun was analyzed by confocal laser microscopy. Phospho-JNK strongly colocalizes with snRNPs in nuclear speckles, while the former was not concentrated in the coiled bodies. Upon stress induction, both c-Jun and phospho-c-Jun show a nucleoplasmic distribution in euchromatin domains, with the nucleoli free of immunolabeling. Furthermore, the nuclear speckles enriched in phospho-JNK exhibit a very low or undetectable signal with both c-Jun antibodies. Immunogold electron microscopy confirms the accumulation of phospho-JNK in interchromatin granule clusters (nuclear speckles), while in the nucleoplasm this kinase is mainly localized in perichromatin fibrils. Both c-Jun and phospho-c-Jun were also detected in perichromatin fibrils. Double labeling experiments show the colocalization of phospho-JNK and phospho-c-Jun in certain perichromatin fibrils. These results indicate that the neuronal response to the Adriamycin-induced stress is mediated by the activation of the JNK pathway. The accumulation of phospho-JNK in nuclear speckles raises the possibility that this kinase may be involved in the phosphorylation of an unknown splicing factor. Moreover, the colocalization of phospho-JNK and c-Jun in perichromatin fibrils, which are associated with sites of active transcription, suggests that these nuclear structures may be putative sites for the phosphorylation of JNK substrates.


Subject(s)
Cell Nucleus/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/physiology , Neurons/ultrastructure , Trigeminal Ganglion/cytology , Animals , Cell Nucleolus/physiology , Cell Nucleolus/ultrastructure , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Chromatin/physiology , Chromatin/ultrastructure , Doxorubicin/pharmacology , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/analysis , Neurons/drug effects , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Trigeminal Ganglion/physiology
14.
Glia ; 29(3): 246-59, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10642751

ABSTRACT

Administration of tellurium (Te) in weaning rats causes a well-established demyelinating neuropathy induced by the inhibition in myelinating Schwann cells (SC) of the synthesis of cholesterol, a major component of the myelin sheath, at the level of squalene epoxidase. We have used this experimental model of Te neuropathy to study the biogenesis and reorganization of the endomembranes of the nuclear envelope and endoplasmic reticulum (ER) in response to Te treatment by ultrastructural analysis and in situ hybridization for the detection of HMG CoA reductase and synthase mRNA, which encode key enzymes in cholesterol synthesis. The adaptive response of myelinating SC to cholesterol depletion includes cell hypertrophy, the formation of tubular invaginations of proliferating nuclear membranes giving rise to peculiar nuclear inclusions termed crystalloids, and, at the cytoplasmic level, the formation of lamellar bodies of rough ER, proliferation of the smooth ER, and overexpression of HMG CoA reductase and synthase mRNAs. The changes revert after withdrawal of Te treatment. Our results show that the biogenesis and structural organization of both endomembrane systems change dynamically upon Te-induced cholesterol depletion, indicating that this constituent plays a critical role in the organization of nuclear envelope and ER compartments in SC. The results also suggest that the HMG CoA reductase, an integral membrane protein of ER, provides the signal for the extensive membrane assembly. While the physiological meaning of crystalloid remains to be clarified, the hypertrophy of the smooth ER may represent a cytoprotective mechanism involved in detoxification of the neurotoxic agent or its metabolic derivates.


Subject(s)
Cell Nucleus/metabolism , Endoplasmic Reticulum, Smooth/physiology , Schwann Cells/drug effects , Schwann Cells/physiology , Tellurium/pharmacology , Animals , Cytoplasm/physiology , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl-CoA Synthase/genetics , Inclusion Bodies/ultrastructure , Lipid Metabolism , Male , Microspheres , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
15.
J Neuropathol Exp Neurol ; 58(12): 1234-43, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10604748

ABSTRACT

We present a cytological, immunocytochemical, and biochemical study of the cell death of mature myelinating Schwann cells (SCs) in the primary demyelinating neuropathy induced by tellurium (Te). Weaned rats were fed a diet containing 1.1% elemental Te. The animals were killed daily within the first week of Te diet. After 4 to 6 days of Te treatment some SCs underwent degeneration and necrosis. By electron microscopy analysis, degenerating SCs showed chromatin condensation, detachment from the nuclear envelope of condensed chromatin clumps, aggregation of interchromatin granule clusters, formation of intranuclear bundles of microfilaments, and cytoplasmic vesiculation. By confocal laser fluorescence microscopy, chromatin regions were stained with the TUNEL method for in situ labeling of DNA fragmentation and exhibited a progressive reduction of histone signal. In addition, splicing small nuclear ribonucleoprotein (snRNP) factors were redistributed in a few large nuclear domains and bright foci of intranuclear actin were observed. DNA electrophoresis revealed a smear pattern of DNA fragmentation in sciatic nerve samples from Te-treated animals. Upon Te treatment, no degradation of the caspase substrates poly (ADP-ribose) polymerase and lamin B was detected by Western blots or immunocytochemistry, respectively. The peculiar structural rearrangement of the transcription and splicing machinery as well as the vesicular degeneration of the cytoplasm in degenerating SCs support an autophagic cell death of the necrotic type. Unlike the apoptosis of pre-remyelinating SCs (11), this caspase independent cell death of necrotic type involves mature pre-demyelinating SCs and eliminates SCs injured by the neurotoxic effect of Te.


Subject(s)
Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Schwann Cells/pathology , Tellurium , Actins/physiology , Animals , Caspases/chemistry , Caspases/metabolism , Cell Nucleus/metabolism , DNA/metabolism , DNA Fragmentation , Demyelinating Diseases/genetics , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Male , Necrosis , RNA Splicing , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Schwann Cells/ultrastructure
16.
J Cell Biol ; 147(4): 715-28, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562276

ABSTRACT

The spliceosomal snRNAs U1, U2, U4, and U5 are synthesized in the nucleus, exported to the cytoplasm to assemble with Sm proteins, and reimported to the nucleus as ribonucleoprotein particles. Recently, two novel proteins involved in biogenesis of small nuclear ribonucleoproteins (snRNPs) were identified, the Spinal muscular atrophy disease gene product (SMN) and its associated protein SIP1. It was previously reported that in HeLa cells, SMN and SIP1 form discrete foci located next to Cajal (coiled) bodies, the so-called "gemini of coiled bodies" or "gems." An intriguing feature of gems is that they do not appear to contain snRNPs. Here we show that gems are present in a variable but small proportion of rapidly proliferating cells in culture. In the vast majority of cultured cells and in all primary neurons analyzed, SMN and SIP1 colocalize precisely with snRNPs in the Cajal body. The presence of SMN and SIP1 in Cajal bodies is confirmed by immunoelectron microscopy and by microinjection of antibodies that interfere with the integrity of the structure. The association of SMN with snRNPs and coilin persists during cell division, but at the end of mitosis there is a lag period between assembly of new Cajal bodies in the nucleus and detection of SMN in these structures, suggesting that SMN is targeted to preformed Cajal bodies. Finally, treatment of cells with leptomycin B (a drug that blocks export of U snRNAs to the cytoplasm and consequently import of new snRNPs into the nucleus) is shown to deplete snRNPs (but not SMN or SIP1) from the Cajal body. This suggests that snRNPs flow through the Cajal body during their biogenesis pathway.


Subject(s)
Autoantigens/metabolism , Muscular Atrophy, Spinal/genetics , Neurons/metabolism , Organelles/metabolism , Ribonucleoproteins, Small Nuclear/biosynthesis , Animals , Autoantigens/analysis , Autoantigens/genetics , Cell Line , HeLa Cells , Humans , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Male , Microscopy, Immunoelectron , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Organelles/ultrastructure , RNA-Binding Proteins , Rats , Rats, Sprague-Dawley , Ribonucleoproteins, Small Nuclear/analysis , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/ultrastructure , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/ultrastructure , Tumor Cells, Cultured , snRNP Core Proteins
17.
Acta Neuropathol ; 97(2): 143-55, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9928825

ABSTRACT

We have used an experimental model of tellurium (Te)-induced demyelinating neuropathy in the rat to study cellular mechanisms involved in the early response of myelinating Schwann cells (SCs) to injury, prior to demyelination. Starting at postnatal day 21, weaned rats were fed a diet containing 1.1% elemental Te. The animals were killed daily within the 1st week of Te diet and the sciatic nerves were processed for the ultrastructural and immunocytochemical studies. Immunohistochemistry revealed that Te induces an increased nuclear expression of c-Fos in SCs. By electron microscopy analysis, the early cytoplasmic alteration was a dramatic disorganization of the rough endoplasmic reticulum (ER) with cisternal dilations and redistribution and loss of membrane-bound ribosomes. This was followed by a prominent activation of the macroautophagy in SCs. This process involved the formation of autophagosomes containing well-preserved cell organelles, autolysosomes with cellular remnants in various phases of degeneration and lysosomes. Te treatment also induced the expression of free ubiquitin in the perikaryal region of the SC cytoplasm. Immunogold electron microscopy showed the subcellular distribution of ubiquitin in the cytosol, around of dilated ER cisterns and in the matrix of autolysosomes and residual bodies. At the nucleolar level, fibrillarin immunofluorescence revealed nucleolar segregation in SCs exposed to Te. The ultrastructural study confirmed the segregation of the nucleolar components with a peripheral distribution of the dense fibrillar component. These results support the hypothesis that the depletion of cholesterol induced by Te treatment triggers a stress response in myelinating SCs mediated by immediate early genes of the fos family. The cellular response includes a severe disruption of the protein synthesis machinery, namely the rough ER and nucleolus, with the subsequent activation of both ubiquitin and autophagic pathways of proteins and cell organelle degradation. This cytoplasmic remodeling may represent a cytoprotective mechanism in the response of SCs to a neurotoxic stress. Furthermore, it must be a prerequisite for the induction of phenotypic changes and cell repair mechanisms in SCs.


Subject(s)
Autophagy , Demyelinating Diseases/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Schwann Cells/metabolism , Schwann Cells/pathology , Ubiquitins/biosynthesis , Animals , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Nucleolus/pathology , Cell Nucleolus/ultrastructure , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoplasm/pathology , Cytoplasm/ultrastructure , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Endoplasmic Reticulum, Rough/drug effects , Endoplasmic Reticulum, Rough/pathology , Endoplasmic Reticulum, Rough/ultrastructure , Immunohistochemistry , Lysosomes/pathology , Lysosomes/ultrastructure , Male , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley , Ribosomes/drug effects , Ribosomes/pathology , Ribosomes/ultrastructure , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Tellurium
18.
Anat Embryol (Berl) ; 198(2): 111-22, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9725770

ABSTRACT

The morphology, organization and expression of proliferating cell nuclear antigen (PCNA) and the cytoskeletal proteins vimentin and GFAP in immature Bergmann glial cells were studied after a developmental injury induced by a single dose of the cytotoxic agent methylazoxymethanol (MAM) administered on postnatal day 5. This drug, which produces cell death of cerebellar granule cell precursors, did not induce apoptosis in Bergmann glial cells, which are in a proliferative stage. After MAM treatment, PCNA staining showed a severe depletion of PCNA-positive granule cell precursors, whereas PCNA-positive Bergmann glial nuclei in the Purkinje cell layer were preserved. Moreover, the quantitative analysis revealed an increase in the density of both Purkinje cells and PCNA-positive Bergmann glial cells per mm of Purkinje cell layer in MAM-treated rats relative to age-matched controls, but the numerical ratio between these two cell populations remains invariable after MAM treatment. Vimentin and GFAP immunocytochemistry revealed a reinforcement of the Bergmann glial palisade with overexpression of both proteins and thicker immunoreactive glial processes in MAM-treated rats. At the ultrastructural level, Bergmann glial processes closely associated with dying cells in different stages of apoptosis were observed. Frequently, these processes enclosed dying cells in extracellular compartments. Furthermore, phagosomes containing apoptotic bodies were found in Bergmann fibers of MAM-treated rats. These data indicate that the cell death of granule cell precursors triggers a reactive response in immature Bergmann glia. We suggest that this response reflects the plasticity of Bergmann glia to control the neuronal microenvironment in the maturing molecular layer, protecting healthy cells against the potentially harmful contents of dying cells. In situ labeling of cell death with the TUNEL method revealed that the cell death of granule cell precursors is of the apoptotic type. The participation of ameboid microglial cells in the phagocytosis of apoptotic cells was shown with tomato lectin histochemistry and ultrastructural analysis. Moreover, the presence of mitosis in this microglial population demonstrates its proliferative activity in regions of extensive cell death.


Subject(s)
Apoptosis , Cerebellar Cortex/pathology , Gliosis/pathology , Microglia/pathology , Neurons/pathology , Animals , Cerebellar Cortex/drug effects , Cerebellar Cortex/metabolism , Fluorescent Antibody Technique, Direct , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/metabolism , In Situ Nick-End Labeling , Lectins/metabolism , Male , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/toxicity , Microglia/metabolism , Mitosis , Neurons/drug effects , Neurons/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells , Vimentin/metabolism
19.
Acta Neuropathol ; 95(3): 269-79, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9542592

ABSTRACT

We have used an experimental model of tellurium(Te)-induced demyelinating neuropathy in the rat to study cellular mechanisms involved in regulating Schwann cell (SC) numbers during remyelination. Starting at postnatal day 21, weaned rats were fed a diet containing 1.1% elemental Te. Following 7 days of Te treatment and at several time points of post-tellurium treatment (PTe), the animals were processed for ultrastructural analysis, SC nuclei quantification and teased fibre preparations. It is well-established that Te induces a transient demyelinating/remyelinating sequence in sciatic nerves. The loss of the myelin sheath in this neuropathy produces active proliferation and overproduction of immature SCs. By electron microscopy analysis most mitotic SCs were located along demyelinated segments. Quantitative determination of SC nuclei per transverse section of sciatic nerve revealed a dramatic increase of SCs at 2 days PTe relative to control nerves. The number of SC nuclei then decreased progressively during the long-term period of recovery studied (330 days PTe). In Te-treated rats, SCs undergoing cell death were regularly found within the nerve fibre compartment, especially on demyelinated segments. Dying cells exhibited morphological features of apoptosis and appeared enclosed by lamellar processes of adjacent healthy SCs in extracellular compartments. Both healthy immature SCs and endoneurial macrophages were involved in the phagocytosis of apoptotic SCs. Particularly during remyelination, supernumerary endoneurial SCs were observed surrounding myelinated fibres. These cells progressively became atrophic with a morphological phenotype similar so that of "onion bulb" cells. On the other hand, teased fibre measurements revealed a remarkable permanent internodal shortening in remyelinated fibres from Te-treated sciatic nerves. These results indicate that a portion of redundant immature SCs are susceptible to elimination by apoptosis. However, other distinct biological mechanisms such as the persistence of supernumerary SCs in the endoneurium and the shortening of internodal lengths are also involved in regulating SC numbers during the remyelination stage.


Subject(s)
Apoptosis/drug effects , Demyelinating Diseases/pathology , Ranvier's Nodes/pathology , Schwann Cells/pathology , Animals , Cell Nucleus/pathology , Demyelinating Diseases/chemically induced , Disease Models, Animal , Male , Microscopy, Electron , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Ranvier's Nodes/ultrastructure , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Tellurium
20.
J Neurocytol ; 27(4): 205-17, 1998 Apr.
Article in English | MEDLINE | ID: mdl-10640180

ABSTRACT

Administration of hypertonic NaCl solutions by intraperitoneal injection evokes a transient expression of immediate-early genes in the hypothalamic magnocellular neurons of supraoptic nuclei (SON), which is followed by an upregulation of arginine vasopressin synthesis and a general increase in cellular metabolic activity. Here we have analysed the changes that occur in the nucleus of SON neurons during the period of transient Fos expression after injection of hypertonic saline. Within the first 30 minutes after injection, the nuclei become significantly smaller, contain more condensed chromatin and incorporate less 3H-uridine than the controls. By 12 hours these effects are reverting and at 24 hours the nuclei are already more active than the controls. Additionally, we observe an initial decrease in the number of coiled bodies per nucleus within the first 2 hours, followed by a 3-fold increase at 24 hours after injection. As coiled bodies are transcription-dependent subnuclear 'organelles', these results further support the view that injection of hypertonic saline causes a transient inhibition of nuclear activity. Our data show that SON neurons respond to acute osmotic/stress stimuli first with inhibition and then with activation of gene expression. Importantly, inhibition of transcriptional activity occurs simultaneously with maximal accumulation of Fos protein in the nucleus, raising the possibility that activation of c-fos expression may cause repression of target genes.


Subject(s)
Neurosecretory Systems/physiology , Supraoptic Nucleus/physiology , Transcription, Genetic/physiology , Water-Electrolyte Balance/physiology , Animals , Cell Nucleus/ultrastructure , Gene Expression/physiology , Hypertonic Solutions , Male , Microinjections , Microscopy, Immunoelectron , Neurons/physiology , Neurons/ultrastructure , Neurosecretory Systems/cytology , Osmotic Pressure , Rats , Rats, Sprague-Dawley , Ribonucleoproteins, Small Nuclear/genetics , Supraoptic Nucleus/cytology , Tritium , Uridine/metabolism , Uridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...