Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 234(4): 3458-3468, 2019 04.
Article in English | MEDLINE | ID: mdl-30368811

ABSTRACT

Notochordal cells (NCs), characterized by their vacuolated morphology and coexpression of cytokeratin and vimentin intermediate filaments (IFs), form the immature nucleus pulposus (NP) of the intervertebral disc. As humans age, NCs give way to mature NP cells, which do not possess a vacuolated morphology and typically only express vimentin IFs. In light of their concomitant loss, we investigated the relationship between cytosolic vacuoles and cytokeratin IFs, specifically those containing cytokeratin-8 proteins, using a human chordoma cell line as a model for NCs. We demonstrate that the chemical disruption of IFs with acrylamide, F-actin with cytochalasin-D, and microtubules with nocodazole all result in a significant (p < 0.001) decrease in vacuolation. However, vacuole loss was the greatest in acrylamide-treated cells. Examination of the individual roles of vimentin and cytokeratin-8 IFs in the existence of vacuoles was accomplished using small interfering RNA-mediated RNA interference to knock down either vimentin or cytokeratin-8 expression. Reduction of cytokeratin-8 expression was associated with a less-vacuolated cell morphology. These data demonstrate that cytokeratin-8 IFs are involved in stabilizing vacuoles and that their diminished expression could play a role in the loss of vacuolation in NCs during aging. A better understanding of the NCs may assist in preservation of this cell type for NP maintenance and regeneration.


Subject(s)
Chordoma/metabolism , Intermediate Filaments/metabolism , Keratin-8/metabolism , Notochord/metabolism , Vacuoles/metabolism , Acrylamide/toxicity , Cell Line, Tumor , Chordoma/pathology , Cytochalasin D/toxicity , Humans , Intermediate Filaments/drug effects , Intermediate Filaments/genetics , Intermediate Filaments/pathology , Keratin-8/genetics , Nocodazole/toxicity , Notochord/drug effects , Notochord/pathology , Signal Transduction , Vacuoles/drug effects , Vacuoles/pathology
2.
Cell Physiol Biochem ; 38(1): 379-92, 2016.
Article in English | MEDLINE | ID: mdl-26824457

ABSTRACT

BACKGROUND: The microtubule assembly inhibitor nocodazole has been shown to trigger caspase-independent mitotic death and caspase dependent apoptosis. Similar to apoptosis of nucleated cells, erythrocytes may undergo eryptosis, the suicidal erythrocyte death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Stimulators of eryptosis include increase of cytosolic Ca2+ activity ([Ca2+]i), oxidative stress and ceramide. The present study explored, whether and how nocodazole induces eryptosis. METHODS: Flow cytometry was employed to determine phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, the abundance of reactive oxygen species (ROS) from 2',7'-dichlorodihydrofluorescein (DCF) diacetate dependent fluorescence as well as ceramide surface abundance utilizing specific antibodies. Tubulin abundance was quantified by TubulinTracker™ Green reagent and visualized by confocal microscopy. RESULTS: A 48 hours exposure of human erythrocytes to nocodazole (≥ 30 µg/ml) significantly increased the percentage of annexin-V-binding cells without significantly modifying average forward scatter. Nocodazole significantly increased Fluo3-fluorescence, significantly increased DCF fluorescence and significantly increased ceramide surface abundance. The effect of nocodazole on annexin-V-binding was significantly blunted, but not abolished by removal of extracellular Ca2+ and was not modified in the presence of Caspase 3 inhibitor zVAD (1 µM). Nocodazole treatment reduced the content of total tubulin. CONCLUSIONS: Nocodazole triggers cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect in part due to stimulation of Ca2+ entry, oxidative stress and ceramide.


Subject(s)
Erythrocytes/drug effects , Nocodazole/toxicity , Tubulin Modulators/toxicity , Aniline Compounds/chemistry , Apoptosis/drug effects , Calcium/metabolism , Ceramides/metabolism , Erythrocyte Membrane/drug effects , Erythrocytes/cytology , Erythrocytes/metabolism , Hemolysis/drug effects , Humans , Microscopy, Confocal , Phosphatidylserines/metabolism , Reactive Oxygen Species/metabolism , Tubulin/metabolism , Xanthenes/chemistry
3.
Cell Signal ; 27(3): 638-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25479591

ABSTRACT

Mitosis and epidermal growth factor (EGF) receptor (EGFR) are both targets for cancer therapy. The role of EGFR signaling in mitosis has been rarely studied and poorly understood. The limited studies indicate that the activation of EGFR and downstream signaling pathways is mostly inhibited during mitosis. However, we recently showed that EGFR is phosphorylated in response to EGF stimulation in mitosis. Here we studied EGF-induced EGFR activation and the activation of major signaling pathways downstream of EGFR during mitosis. We showed that EGFR was strongly activated by EGF during mitosis as all the five major tyrosine residues including Y992, Y1045, Y1068, Y1086, and Y1173 were phosphorylated to a level similar to that in the interphase. We further showed that the activated EGFR is able to selectively activate some downstream signaling pathways while avoiding others. Activated EGFR is able to activate PI3K and AKT2, but not AKT1, which may be responsible for the observed effects of EGF against nocodazole-induced cell death. Activated EGFR is also able to activate c-Src, c-Cbl and PLC-γ1 during mitosis. However, activated EGFR is unable to activate ERK1/2 and their downstream substrates RSK and Elk-1. While it activated Ras, EGFR failed to fully activate Raf-1 in mitosis due to the lack of phosphorylation at Y341 and the lack of dephosphorylation at pS259. We conclude that contrary to the dogma, EGFR is activated by EGF during mitosis. Moreover, EGFR-mediated cell signaling is regulated differently from the interphase to specifically serve the needs of the cell in mitosis.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Mitosis/physiology , Signal Transduction/drug effects , Apoptosis/drug effects , CSK Tyrosine-Protein Kinase , Cell Division , HeLa Cells , Humans , Interphase , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitosis/drug effects , Nocodazole/pharmacology , Nocodazole/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , src-Family Kinases/metabolism
4.
Toxicol In Vitro ; 28(3): 411-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24388784

ABSTRACT

The majority of environmental and commercial chemicals have not been evaluated for their potential to cause neurotoxicity. We have investigated if neuron specific anti-ßIII-tubulin antibodies are useful in a microplate assay of neurite outgrowth of retinoic acid-induced neurons from mouse P19 embryonal carcinoma cells. By incubating the P19-derived neurons with the primary anti-ßIII-tubulin antibody and a secondary Alexa Fluor 488-conjugated antibody, followed by measuring the fluorescence in a microplate reader, a time-dependent increase in anti-ßIII-tubulin immunofluorescence was observed. The relative fluorescence units increased by 4.3-fold from 2 to 10 days in culture. The results corresponded well with those obtained by semi-automatic tracing of neurites in fluorescence microscopy images of ßIII-tubulin-labeled neurons. The sensitivity of the neurite outgrowth assay using a microplate reader to detect neurotoxicity produced by nocodazole, methyl mercury chloride and okadaic acid was significantly higher than for a cell viability assay measuring intracellular fluorescence of calcein-AM. The microplate-based method to measure toxicity targeting neurites using anti-ßIII-tubulin antibodies is however less sensitive than the extracellular lactate dehydrogenase activity assay to detect general cytotoxicity produced by high concentrations of clomipramine, or glutamate-induced excitotoxicity. In conclusion, the fluorescence microplate assay for the detection of neurite outgrowth by measuring changes in ßIII-tubulin immunoreactivity is a rapid and sensitive method to assess chemical- or toxin-induced neurite toxicity.


Subject(s)
Antibodies/immunology , Neurites/metabolism , Neurotoxicity Syndromes/diagnosis , Tubulin/immunology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Fluoresceins/chemistry , Fluorescent Antibody Technique/methods , Methylmercury Compounds/toxicity , Mice , Microscopy, Fluorescence , Neurons/drug effects , Neurons/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Nocodazole/toxicity , Okadaic Acid/toxicity , Time Factors , Tretinoin/pharmacology
5.
Cell Death Dis ; 4: e468, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23348582

ABSTRACT

Mitotic cell death following prolonged arrest is an important death mechanism that is not completely understood. This study shows that Protein Tyrosine Phosphatase 1B (PTP1B) undergoes phosphorylation during mitotic arrest induced by microtubule-targeting agents (MTAs) in chronic myeloid leukaemia cells. Inhibition of cyclin-dependent kinase 1 (Cdk1) or polo-like kinase 1 (Plk1) during mitosis prevents PTP1B phosphorylation, implicating these kinases in PTP1B phosphorylation. In support of this, Cdk1 and Plk1 co-immunoprecipitate with endogenous PTP1B from mitotic cells. In addition, active recombinant Cdk1-cyclin B1 directly phosphorylates PTP1B at serine 386 in a kinase assay. Recombinant Plk1 phosphorylates PTP1B on serine 286 and 393 in vitro, however, it requires a priming phosphorylation by Cdk1 at serine 386 highlighting a novel co-operation between Cdk1 and Plk1 in the regulation of PTP1B. Furthermore, overexpression of wild-type PTP1B induced mitotic cell death, which is potentiated by MTAs. Moreover, mutation of serine 286 abrogates the cell death induced by PTP1B, whereas mutation of serine 393 does not, highlighting the importance of serine 286 phosphorylation in the execution of mitotic cell death. Finally, phosphorylation on serine 286 enhanced PTP1B phosphatase activity. Collectively, these data reveal that PTP1B activity promotes mitotic cell death and is regulated by the co-operative action of Cdk1 and Plk1 during mitotic arrest.


Subject(s)
Apoptosis/drug effects , CDC2 Protein Kinase/pharmacology , Cell Cycle Proteins/pharmacology , Protein Serine-Threonine Kinases/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins/pharmacology , Antineoplastic Agents/toxicity , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin B1/pharmacology , Humans , Immunoprecipitation , K562 Cells , Mitosis , Nocodazole/toxicity , Paclitaxel/toxicity , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Serine/chemistry , Polo-Like Kinase 1
6.
J Appl Toxicol ; 33(6): 426-33, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22081495

ABSTRACT

The mechanism of genotoxic potential of the cancer chemotherapeutic drugs amsacrine and nocodazole in mouse bone marrow was investigated using a micronucleus test complemented by fluorescence in situ hybridization assay with mouse centromeric and telomeric DNA probes. In animals treated with different doses of amsacrine (0.5-12 mg kg(-1) ), the frequencies of micronucleated polychromatic erythrocytes increased significantly after treatment with 9 and 12 mg kg(-1) . A statistically significant increase in micronuclei frequency was also detected for 75 mg kg(-1) nocodazole (two exposures, spaced 24 h apart). Both compounds caused significant suppressions of erythroblast proliferation at higher doses. Furthermore, the present study demonstrated for the first time that amsacrine has high incidences of clastogenicity and low incidences of aneugenicity whereas nocodazole has high incidences of aneugenicity and low incidences of clastogenicity during mitotic phases in vivo. The assay also showed that chromosomes can be enclosed in the micronuclei before and after centromere separation. Therefore, the clinical use of these genotoxic drugs must be weighed against the risks of the development of chromosomal aberrations in cancer patients and medical personnel exposed to drug regimens that include these chemicals.


Subject(s)
Amsacrine/toxicity , Antineoplastic Agents/toxicity , Bone Marrow Cells/drug effects , Mutagens , Nocodazole/toxicity , Animals , Antibiotics, Antineoplastic/toxicity , Centromere/drug effects , Colchicine/toxicity , Cytogenetics , DNA Probes , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Erythrocytes/ultrastructure , In Situ Hybridization , In Vitro Techniques , Male , Mice , Micronucleus Tests , Mitomycin/toxicity
7.
Cell Cycle ; 10(14): 2373-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21670593

ABSTRACT

Disruption of cell cycle checkpoints and interference with the normal cell cycle progression frequently result in cell death or malignant transformation. Hexavalent chromium [Cr(VI)] is a well-known carcinogen that has been implicated in the occurrence of many types of human malignancies, including lung cancer. However, the exact mechanism by which Cr(VI) causes malignant transformation in the lung remains unknown. We have demonstrated that chronic exposure to a non-cytotoxic concentration of Cr(VI) induced a variety of chromosomal abnormalities, including premature sister chromatid separation, chromosomal breakage and the presence of lagging/misaligned chromosomes. After treatment with nocodazole, both HeLa and normal lung bronchial epithelial cells were arrested at mitosis. However, Cr(VI) significantly compromised M-phase arrest induced by nocodazole. Cr(VI) suppressed BubR1 activation and reduced expression of Emi1, leading to an unscheduled activation of APC/C. Consistent with this observation, Cr(VI) treatment caused enhanced polyubiquitination of geminin during mitotic release, while it deregulated the activity of Cdt1, a DNA replication licensing factor. Combined, these results suggest that Cr(VI)-induced chromosomal instability is partly due to a perturbation of APC/C activities, leading to chromosomal instability.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Cycle Proteins/metabolism , Chromium/toxicity , Chromosomal Instability , F-Box Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Agents/toxicity , Cell Division , Geminin , HeLa Cells , Humans , Mitosis , Nocodazole/toxicity , Ubiquitination
8.
J Exp Biol ; 214(Pt 1): 80-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21147971

ABSTRACT

In the present study, we exposed the olfactory epithelia of crucian carp, Carassius carassius, and brown trout, Salmo trutta, to dextran coupled with Alexa dyes together with odorants. Dye uptake was severely reduced after pre-exposure to nocodazole, an inhibitor of microtubule polymerization that impairs endocytosis, supporting the hypothesis that odour-activated olfactory receptor molecules undergo endocytosis. Application of the bile acid taurolithocholate, a potent and specific odorant for fish, resulted in the labelling of a sparse (less than 3%) cell population with the typical morphology of ciliated sensory neurons (CSNs) - long dendrites and cell somata deep in the sensory epithelium. The dye was distributed throughout the sensory neuron, also revealing axons and target glomeruli. Stained axons redistribute at the entrance of the olfactory bulb and terminate in two small target areas, a dorsal and a medial one. These results are consistent with the notion that taurolithocholate is detected specifically by a few ciliated sensory neurons. Application of the olfactory epithelium of brown trout to bile acid stained cells with the appearance of CSNs. Application of an alarm agonist, hypxanthine-3-N-oxide, to crucian carp olfactory organ caused staining of another set of sensory neurons. Furthermore, our results show that odour-induced uptake of a dye can serve to identify the subtype of olfactory sensory neurons responding to a particular odorant, and to pinpoint the target regions of these neurons in the olfactory bulb as a first step to elucidating the neuronal network responding to a particular odour.


Subject(s)
Carps/metabolism , Nerve Net/cytology , Olfactory Receptor Neurons/drug effects , Taurolithocholic Acid/pharmacology , Trout/metabolism , Animals , Coloring Agents/pharmacokinetics , Dextrans , Endocytosis/drug effects , Image Processing, Computer-Assisted , Microscopy, Confocal , Microscopy, Electron, Scanning , Nerve Net/drug effects , Nocodazole/toxicity , Olfactory Receptor Neurons/metabolism , Staining and Labeling/methods
9.
Toxicol Appl Pharmacol ; 230(2): 235-46, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18485433

ABSTRACT

Arsenite, a known mitotic disruptor, causes cell cycle arrest and cell death at anaphase. The mechanism causing mitotic arrest is highly disputed. We compared arsenite to the spindle poisons nocodazole and paclitaxel. Immunofluorescence analysis of alpha-tubulin in interphase cells demonstrated that, while nocodazole and paclitaxel disrupt microtubule polymerization through destabilization and hyperpolymerization, respectively, microtubules in arsenite-treated cells remain comparable to untreated cells even at supra-therapeutic concentrations. Immunofluorescence analysis of alpha-tubulin in mitotic cells showed spindle formation in arsenite- and paclitaxel-treated cells but not in nocodazole-treated cells. Spindle formation in arsenite-treated cells appeared irregular and multi-polar. gamma-tubulin staining showed that cells treated with nocodazole and therapeutic concentrations of paclitaxel contained two centrosomes. In contrast, most arsenite-treated mitotic cells contained more than two centrosomes, similar to centrosome abnormalities induced by heat shock. Of the three drugs tested, only arsenite treatment increased expression of the inducible isoform of heat shock protein 70 (HSP70i). HSP70 and HSP90 proteins are intimately involved in centrosome regulation and mitotic spindle formation. HSP90 inhibitor 17-DMAG sensitized cells to arsenite treatment and increased arsenite-induced centrosome abnormalities. Combined treatment of 17-DMAG and arsenite resulted in a supra-additive effect on viability, mitotic arrest, and centrosome abnormalities. Thus, arsenite-induced abnormal centrosome amplification and subsequent mitotic arrest is independent of effects on tubulin polymerization and may be due to specific stresses that are protected against by HSP90 and HSP70.


Subject(s)
Arsenites/toxicity , Mitosis/drug effects , Stress, Physiological/pathology , Tubulin/biosynthesis , Antineoplastic Agents/toxicity , Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Blotting, Western , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Centrosome/drug effects , Centrosome/ultrastructure , Fever/pathology , Fluorescent Antibody Technique , HeLa Cells , Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Microtubules/drug effects , Microtubules/ultrastructure , Nocodazole/toxicity , Paclitaxel/toxicity , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure
10.
Oncogene ; 27(12): 1726-36, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-17891177

ABSTRACT

We used intrachromosomal substrates to directly monitor the effect of the cell cycle on the efficiency and the accuracy of nonhomologous end joining (NHEJ) in mammalian cells. We show that both KU and KU-independent (KU-alt) pathways are efficient when maintaining cells in G1/S, in G2/M or during dynamic progression through S phase. In addition, the accuracy of NHEJ is barely altered when the cells are blocked in G1/S or in G2/M. However, progression through S phase increases the frequency of deletions, which is a hallmark of the KU-alt pathway. Moreover, we show that the intermediates that are generated by the KU-dependent end joining of non-fully complementary ends, and which contain mismatches, nicks or gap intermediates, are less accurately processed when the cells progress through S phase. In conclusion, both KU and KU-alt processes are active throughout the cell cycle, but the repair is more error prone during S phase, both by increasing the mutagenic KU-alt pathway and decreasing the accuracy of the repair of the intermediates generated by the KU-dependent pathway.


Subject(s)
Antigens, Nuclear/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Mutagenesis/genetics , S Phase/genetics , Signal Transduction/genetics , Animals , Antigens, Nuclear/physiology , Antineoplastic Agents/toxicity , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA-Binding Proteins/physiology , Gene Deletion , Ku Autoantigen , Mimosine/toxicity , Molecular Sequence Data , Nocodazole/toxicity , Recombination, Genetic , S Phase/drug effects , Signal Transduction/drug effects
11.
Cell Cycle ; 6(12): 1521-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17507801

ABSTRACT

We recently reported that MEK1/2 plays an important role in microtubule organization and spindle pole tethering in mouse oocytes, but how the intracellular transport of this protein is regulated remains unknown. In the present study, we investigated the mechanisms of poleward MEK1/2 transport during the prometaphase I/metaphase I transition and MEK1/2 release from the spindle poles during the metaphase I/anaphase I transition in mouse oocytes. Firstly, we found that p-MEK1/2 was colocalized with dynactin at the spindle poles. Inhibition of the cytoplasmic dynein/dynactin complex by antibody microinjection blocked polar accumulation of p-MEK1/2 and caused obvious spindle abnormalities. Moreover, coimmunoprecipitation of p-MEK1/2 and dynein or dynactin from mouse oocyte extracts confirmed their association at metaphase I. Secondly, disruption of microtubules by nocodazole resulted in the failure of poleward p-MEK1/2 transport. Whereas, when the nocodazole-treated oocytes were recovered in fresh culture medium, the spindle reformed and p-MEK1/2 relocalized to the spindle poles. Finally, we examined the mechanism of p-MEK1/2 release from the spindle poles. In control oocytes, polar p-MEK1/2 was gradually released during metaphase I/anaphase I transition. By contrast, in the presence of nondegradable cyclin B (Delta90), p-MEK1/2 still remained at the spindle poles at anaphase I. Our results indicate that poleward MEK1/2 transport is a cytoplasmic dynein/dynactin-mediated and spindle microtubule-dependent intracellular movement, and that its subsequent anaphase release from spindle poles is dependent on cyclin B degradation.


Subject(s)
Cyclin B/metabolism , Cytoplasm/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Metaphase/physiology , Spindle Apparatus/metabolism , Animals , Dynactin Complex , Dyneins/metabolism , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Nocodazole/toxicity , Oocytes/cytology , Oocytes/physiology , Protein Transport/drug effects , Protein Transport/physiology , Spindle Apparatus/drug effects , Tubulin Modulators/toxicity
12.
J Neural Transm (Vienna) ; 114(4): 405-16, 2007.
Article in English | MEDLINE | ID: mdl-16906355

ABSTRACT

Li(+) exerts protective effect against several neurotoxins in neuronal cell preparations. Here we examined the antiapoptotic effects of GSK3beta in cerebellar granule neurons (CGNs) in the presence of several neurotoxins. Acute treatment with Li(+) protected neurons against nocodazole and serum/potassium (S/K) deprivation, but were ineffective against kainic acid and MPP(+). Li(+) 5 mM also decreased caspase-3 activation induced by nocodazole and S/K deprivation as measured by Ac-DEVD-p-nitroaniline and the breakdown of alpha-spectrin. All the neurotoxins used in the present study activated GSK3beta, evaluated with a specific antibody phospho-GSK-3beta (Ser9) by Western-blot and immunocytochemistry and were always inhibited by Li(+) 5 mM. Our results implicate Li(+) in the regulation of apoptosis mediated by caspase activation (Type I). Furthermore inhibition of GSK3beta by acute treatment with Li(+) 5 mM is not an indicator of neuroprotection. The acute antiapoptotic function of Li(+) is discussed in terms of its inhibition of Type I pathway, the intrinsic (mitochondrial) apoptotic pathway in cerebellar granule cells.


Subject(s)
Apoptosis/drug effects , Lithium Chloride/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Caspases/drug effects , Caspases/metabolism , Cells, Cultured , Dopamine Agents/toxicity , Enzyme Activation/drug effects , Excitatory Amino Acid Agonists/toxicity , Flow Cytometry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Kainic Acid/toxicity , Neurons/metabolism , Neurons/pathology , Nocodazole/toxicity , Rats , Rats, Sprague-Dawley
13.
J Alzheimers Dis ; 9(2): 195-205, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16873966

ABSTRACT

Synapse loss and neuronal death are key features of Alzheimer's disease pathology. Disrupted axonal transport of mitochondria is a potential mechanism that could contribute to both. As the major producer of ATP in the cell, transport of mitochondria to the synapse is required for synapse maintenance. However, mitochondria also play an important role in the regulation of apoptosis. Investigation of aluminum (Al) maltolate induced apoptosis in human NT2 cells led us to explore the relationship between apoptosis related changes and the disruption of mitochondrial transport. Similar to that observed with tau over expression, NT2 cells exhibit peri-nuclear clustering of mitochondria following treatment with Al maltolate. Neuritic processes largely lacked mitochondria, except in axonal swellings. Similar, but more rapid results were observed following staurosporine administration, indicating that the clustering effect was not specific to Al maltolate. Organelle clustering and transport disruption preceded apoptosis. Incubation with the caspase inhibitor zVAD-FMK effectively blocked apoptosis, however failed to prevent organelle clustering. Thus, transport disruption is associated with the initiation, but not necessarily the completion of apoptosis. These results, together with observed transport defects and apoptosis related changes in Alzheimer disease brain suggest that mitochondrial transport disruption may play a significant role in synapse loss and thus the pathogenesis or Alzheimer's disease.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Mitochondria/drug effects , Mitochondria/ultrastructure , Organometallic Compounds/toxicity , Pyrones/toxicity , Alzheimer Disease/pathology , Amino Acid Chloromethyl Ketones/toxicity , Animals , Antineoplastic Agents/toxicity , Cell Line , Cytochromes c/metabolism , Enzyme Inhibitors/toxicity , Humans , Hydrogen Peroxide/toxicity , Immunohistochemistry , In Situ Nick-End Labeling , Microtubules/drug effects , Neurites/ultrastructure , Neuroprotective Agents/toxicity , Nocodazole/toxicity , Organelles/drug effects , Organelles/ultrastructure , Rabbits , Staurosporine/toxicity
14.
J Exp Biol ; 208(Pt 22): 4305-15, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16272253

ABSTRACT

Birds are uricotelic, and because they excrete urate by renal tubular secretion, they provide a convenient model for examination of this process. Primary monolayer cultures of the isolated renal proximal tubule epithelium from the domestic chicken, Gallus gallus L., were mounted in Ussing chambers where several substrates/inhibitors of renal organic anion transporters were tested for the sidedness and specificity of their effects on transepithelial urate transport. Transepithelial electrical resistance, electrical potential and sodium-dependent glucose current were monitored to detect nonspecific effects. Under control short-circuited conditions the ratio of unidirectional fluxes of [(14)C]urate was found to be 3:1. Active net secretion was specifically inhibited by 1 mmol l(-1) probenecid and 10 mmol l(-1) para-aminohippuric acid (PAH). Bromocresol Green, cimetidine, nocodozole, cytochalasin D and ouabain also inhibited secretion but were toxic. Interstitial-side lithium (5 mmol l(-1)) and glutarate (1 mmol l(-1)) specifically blocked transport, but 10-100 micromol l(-1) glutarate had no effect. Interstitial estrone sulfate (ES) stimulated urate secretion at 10 micromol l(-1) but was inhibitory at 500 micromol l(-1). Active PAH secretion (5:1 flux ratio) was inhibited 34% by 330 micromol l(-1) urate. ES (500 micromol l(-1)) blocked the remainder. From the lumen side, glucose-free, Cl(-)-free and high K(+) (30 mmol l(-1)) solutions, or an alkaline pH of 7.7 had no effect on urate transport and neither did several compounds known to be uricosuric. Lumen-side methotrexate (500 micromol l(-1)) and MK571 (20 micromol l(-1)) strongly inhibited urate secretion. MK571 had no effect from the interstitial side. RT-PCR revealed mRNA for OAT1-, OAT3-, MRP2- and MRP4-like organic anion transporters in chicken proximal epithelium.


Subject(s)
Chickens/metabolism , Kidney Tubules, Proximal/metabolism , Organic Anion Transporters/metabolism , Uric Acid/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Bromcresol Green/toxicity , Carbon Radioisotopes/metabolism , Cimetidine/toxicity , Cytochalasins/toxicity , DNA Primers , Electric Impedance , Epithelium/metabolism , Estrone/analogs & derivatives , Estrone/toxicity , Glutarates/toxicity , Hydrogen-Ion Concentration , Lithium/toxicity , Membrane Potentials/drug effects , Nocodazole/toxicity , Organic Anion Transporters/genetics , Ouabain/toxicity , Probenecid/toxicity , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , p-Aminohippuric Acid/metabolism , p-Aminohippuric Acid/toxicity
15.
Mutat Res ; 588(2): 143-51, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16310403

ABSTRACT

Aneugenic compounds act on non-DNA targets to exert genotoxicity via an indirect mechanism. In contrast to DNA-binding agents, these compounds are expected to possess threshold levels of activity. Therefore, the risk for adverse effects following human exposure to an aneugen could be minimal, if the threshold of activity has been clearly determined in vivo and in vitro and providing the human exposure level is below this threshold. Thus, the development of a single-cell model to allow comparisons between in vitro and in vivo threshold values for aneugenic compounds is of importance. The in vivo micronucleus test is one of the main assays used in genetic toxicology, and is often performed in the mouse. Thus, an extensive database is available in the literature. However, there are only few data concerning the in vitro micronucleus assay using mouse cells, as the majority of in vitro micronucleus assays have been performed using human lymphocytes. In addition, there is a lack of data concerning thresholds for any compound using this model. First, we evaluated whether the use of mouse splenocytes would be an acceptable alternative to that of human lymphocytes to identify aneugens. To allow valid comparisons, the two protocols were first harmonized. Thus, phytohemagglutinin (PHA) and concanavalin A were used as specific mitogens for human lymphocytes and mouse splenocytes, respectively, in order to achieve similar cell-proliferation rates. To achieve similar and sufficient numbers of binucleated cells, cytochalasin B was added 44 and 56 h after culture initiation of the human and mouse cells, respectively. Second, we compared the sensitivity of the mouse protocol with that of the human protocol by exposing the cells to the aneugens nocodazole and paclitaxel. There was good reproducibility of the cytotoxic/genotoxic responses of the two cell models following exposure to the aneugens. The sensitivity of the mouse splenocytes to paclitaxel was higher than that of the human lymphocytes. The two cell types were equally sensitive to nocodazole.


Subject(s)
Aneugens/toxicity , Lymphocytes/drug effects , Nocodazole/toxicity , Paclitaxel/toxicity , Spleen/drug effects , Animals , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Cytochalasin B/pharmacology , Humans , Mice , Mitogens/pharmacology , Mutagenicity Tests , Phytohemagglutinins/pharmacology , Spindle Apparatus/drug effects , Spleen/cytology
16.
Mutagenesis ; 20(3): 173-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15817572

ABSTRACT

Aneugenic and clastogenic agents are good inducers of both micronuclei and apoptosis. In its turn, apoptosis may modify the threshold values for the induction of micronuclei. This is of major concern for accurate assessment of hazard related to exposure to mutagens. In the present work we studied the influence of caspases, the key regulators of the apoptotic process, on the induction of micronuclei in the cytokinesis block micronucleus assay. For this, we applied a combined approach in which both human peripheral blood mononucleated cells (PBMC) and the paired human breast carcinoma cell lines MCF-7, which is caspase-3 deficient, and the caspase-3 transfected MCF-7 (MCF-7casp-3) were used to study the influence of caspase activity on micronuclei. When nocodazole induced apoptosis was inhibited by the use of inhibitors of the two main apical caspases-8 and -9 in PBMC, the frequencies of micronucleated binucleates (MNCB) increased with inhibition of these caspases confirming that apoptosis can eliminate micronucleated cells. On the contrary when caspase-3 was inhibited, the frequencies of MNCB was lower, suggesting a role of caspase-3, also in micronuclei formation. To verify this hypothesis, we compared the induction of apoptosis and micronuclei by the aneugen nocodazole, the clastogen methyl methane sulfonate (MMS) and the non-mutagenic apoptogen staurosporin in MCF-7 and MCF-7casp-3 cells. The results showed that when caspase-3 activity was impaired, in the parental MCF-7 cell line or in the MCF-7casp-3 cells in the presence of the caspase-3 inhibitor, the frequencies of nocodazole or MMS induced micronuclei decreased. These results suggest that caspase-3, besides its function as an effector caspase in the apoptotic pathway, is also involved in the formation of micronuclei.


Subject(s)
Aneugens/toxicity , Apoptosis , Caspases/metabolism , Micronuclei, Chromosome-Defective , Mutagens/toxicity , Nocodazole/toxicity , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspase 3 , Caspase 7 , Caspase 8 , Caspase Inhibitors , Caspases/genetics , Cell Line, Tumor , Cysteine Proteinase Inhibitors/toxicity , Cytokinesis/drug effects , Humans , Methyl Methanesulfonate/toxicity , Micronucleus Tests , Monocytes/drug effects , Mutation , Staurosporine/toxicity , Transfection
17.
Reprod Toxicol ; 19(4): 459-71, 2005.
Article in English | MEDLINE | ID: mdl-15749259

ABSTRACT

Trisomies due to nondisjunction in oogenesis are still a major cause of genetic diseases in humans. In this study, we analysed spindle morphology of in vitro matured nocodazole-exposed mouse oocytes by novel non-invasive Polscope-microscopy, and compared images to those obtained by anti-tubulin immunofluorescence of fixed oocytes. Polscope revealed a reduction in the numbers of oocytes expressing a birefringent spindle, and alterations in spindle morphology at concentrations of nocodazole below those inducing detectable aberrations in immunofluorescence. Hyperploidy increased significantly at a concentration of 40 nM nocodazole in mouse metaphase II oocytes, similar to thresholds inducing nondisjunction in cultured human lymphocytes. In conclusion, Polscope represents a novel highly sensitive, non-invasive method to identify chemicals inducing severe spindle aberrations that predispose mammalian oocytes to nondisjunction. Polscope may provide information on the functionality of the spindle in experimental studies but is also compatible with clinical trials in human assisted reproduction due to its non-invasive nature.


Subject(s)
Aneuploidy , Mutagens/toxicity , Nocodazole/toxicity , Oocytes/drug effects , Spindle Apparatus/drug effects , Animals , Birefringence , Chromosome Segregation/drug effects , Chromosomes, Mammalian/drug effects , Chromosomes, Mammalian/ultrastructure , Female , In Vitro Techniques , Meiosis/drug effects , Metaphase/drug effects , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Microscopy, Polarization , Microtubules/drug effects , Microtubules/ultrastructure , Oocytes/ultrastructure , Spindle Apparatus/ultrastructure
18.
Exp Cell Res ; 303(1): 114-27, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15572032

ABSTRACT

Two-photon microscopy of rhodamine 123-labeled mitochondria revealed that mitochondria of neurons cultured from mouse respiratory center form functionally coupled, dynamically organized aggregates such as chains and clusters, while single mitochondria were rarely seen. Mitochondrial chain structures predominate in dendrites, while irregularly shaped mitochondrial clusters are mostly found in the soma. Both types of mitochondrial structures showed chaotic Brownian motions and the mitochondrial chains also revealed well-directed movements. The latter dislocations were arrested upon mitochondrial depolarization or blockade of mitochondrial ATP synthesis. Depolymerization of microtubules by colchicine or nocodazole or inhibition of protein phosphatases by calyculin A disrupted mitochondrial chains and the mitochondria accumulated in the soma. Forskolin and IBMX reversibly blocked directed movements of mitochondria, but did not affect their overall spatial distribution. Thus, protein phosphorylation seems to control both mitochondrial transport and organization. Protein phosphorylation downstream of enhanced cytosolic cAMP levels apparently regulates the transition from motile to non-motile mitochondria, while phosphorylation resulting from inhibition of types 1 and 2A protein phosphatases massively disturbs mitochondrial organization. The complex phosphorylation processes seem to control the close interaction of mitochondria and cytoskeleton which may guarantee that mitochondria are immobilized at energetic hot spots and rearranged in response to changes in local energy demands.


Subject(s)
Biological Transport/physiology , Cyclic AMP/metabolism , Microtubules/drug effects , Mitochondria/metabolism , Neurons/cytology , Animals , Cells, Cultured , Dendrites/drug effects , Enzyme Inhibitors/pharmacology , Marine Toxins , Mice , Microscopy, Fluorescence , Nocodazole/toxicity , Oxazoles/toxicity , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Respiratory Center/cytology
19.
Dev Cell ; 7(5): 637-51, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15525526

ABSTRACT

Cells that cannot satisfy the spindle assembly checkpoint (SAC) are delayed in mitosis (D-mitosis), a fact that has useful clinical ramifications. However, this delay is seldom permanent, and in the presence of an active SAC most cells ultimately escape mitosis and enter the next G1 as tetraploid cells. This review defines and discusses the various factors that determine how long a cell remains in mitosis when it cannot satisfy the SAC and also discusses the cell's subsequent fate.


Subject(s)
Mitosis/physiology , Spindle Apparatus/physiology , Animals , Apoptosis , Cellular Senescence , G1 Phase , Humans , Kinetochores/physiology , Microtubules/physiology , Models, Biological , Necrosis , Nocodazole/toxicity , Spindle Apparatus/drug effects , Time Factors
20.
Zygote ; 10(3): 245-51, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12214806

ABSTRACT

Incorporation of [3H]thymidine at different concentrations into mouse embryos at early developmental stages was determined by autoradiography. Methods to synchronise the G1-phase of mouse 2- and 4-cell embryos were also investigated. The results showed that the ability of embryos to incorporate [3H]thymidine increased with development. Embryos at the 4-cell stage were not labelled when the concentration of [3H]thymidine was lower than 5 microCi/ml, whereas the nuclei of embryos at morula and blastocyst stages began to show silver grains at a concentration of 0.1 microCi/ml of [3H]thymidine. After 2- and 4-cell mouse embryos were synchronised at the onset of G1-phase by treatment with low temperature or nocodazole, and DNA synthesis was detected by autoradiography, the duration of G1-phase was estimated. The result showed that 43% of the 2-cell embryos had a G1-phase of < or = 1 h, 22% had a G1-phase of < or = 2 h, 22% had a G1-phase of < or = 3 h and 13% had a G1-phase of < or = 4 h. The G1-phase in 85% of the 4-cell embryos was < or = 3 h, that in 8% of embryos was < or = 4 h and that in 7% of embryos was < or = 5 h. The toxicity of nocodazole on mouse embryo development was assessed based on both blastocyst formation and the number of blastomeres, and the results indicated that the effect of nocodazole on embryo development and cell cycle block was dose-dependent. The minimum concentration of nocodazole for metaphase block of mouse late 2-cell embryos was 0.05 microM, and the appropriate concentrations which did not impair development were 0.05-0.5 microM.


Subject(s)
Cleavage Stage, Ovum/cytology , G1 Phase , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/metabolism , Cleavage Stage, Ovum/drug effects , Cleavage Stage, Ovum/metabolism , Dose-Response Relationship, Drug , Embryonic and Fetal Development/drug effects , Female , G1 Phase/drug effects , In Vitro Techniques , Mice , Morula/cytology , Morula/drug effects , Morula/metabolism , Nocodazole/administration & dosage , Nocodazole/toxicity , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...