Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Ecotoxicol Environ Saf ; 192: 110328, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32078840

ABSTRACT

In this study pillar[5]arene (P5) and a quinoline-functionalized pillar[5]arene (P5-6Q) which is used for detecting radioactive element, gas adsorption and toxic ions were synthesized. These materials were characterized by Nuclear Magnetic Resonance (NMR), Fourier Transform Infrared (FTIR), elemental analysis, melting point, Mass Spectroscopy, Scanning Electron Microscopy (SEM) and Zeta Potential. The cytotoxic and genotoxic potential of P5 and P5-6Q at distinct concentrations of 12.5, 25, 50, and 100 µg/mL were also investigated by Allium ana-telophase and comet assays on Allium cepa roots and Drosophila melanogaster haemocytes. P5 and P5-6Q showed dose dependent cytotoxic effect by decreasing mitotic index (MI) and genotoxic effect by increasing chromosomal aberrations (CAs such as disturbed anaphase-telophase, polyploidy, stickiness, chromosome laggards and bridges) and DNA damage at the exposed concentrations. These changes in P5-6Q were lower than P5. Further research is necessary to clarify the cytotoxic and genotoxic action mechanisms of P5 and P5-6Q at molecular levels.


Subject(s)
Calixarenes/toxicity , DNA Damage , Drosophila melanogaster/drug effects , Onions/drug effects , Anaphase/drug effects , Animals , Calixarenes/chemistry , Chromosome Aberrations , Comet Assay , Cytotoxins/chemistry , Cytotoxins/toxicity , Drosophila melanogaster/genetics , Hemocytes/drug effects , Mitotic Index , Onions/genetics , Plant Roots/drug effects , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/toxicity , Telophase/drug effects
2.
Toxicol Ind Health ; 33(10): 802-809, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28893155

ABSTRACT

In this study, the genotoxic potential of <50 nm, <100 nm iron oxide (Fe2O3) nanoparticles (IONPs) and ionic form were investigated using the wing somatic mutation and recombination test (SMART) and Allium and comet assays. In the SMART assay, different concentrations (1, 2, 5 and 10 mM) of NPs and ionic forms were fed to transheterozygous larvae of Drosophila melanogaster. No significant genotoxic effect was observed in <100 nm NPs and ionic form, while <50 nm IONPs showed genotoxicity at 1 and 10 mM concentrations. Allium cepa root meristems were exposed to five concentrations (0.001, 0.01, 0.1, 1 and 10 mM) of <50 nm and ionic forms for 4 h and three concentrations (2.5, 5 and 10 mM) for <100 nm of IONPs for 24 and 96 h. There was a statistically significant effect at 96 h at all concentrations of <100 nm IONPs. Similarly, <50 nm of IONPs and ionic forms also showed a statistically significant effect on mitotic index frequencies for all concentrations at 4 h. There was a dose-dependent increase in chromosomal abnormalities for IONPs and ionic form. Comet assay results showed time- and concentration-dependent increases in <100 nm NPs. There was a concentration-dependent increase in <50 nm NPs and ionic form ( p < 0.05). Consequently, the <50 nm of Fe2O3 was found toxic compared to 100 nm Fe2O3 and ionic form.


Subject(s)
Magnetite Nanoparticles/toxicity , Mutagenicity Tests/methods , Mutagens/toxicity , Allium/drug effects , Anaphase/drug effects , Animals , Comet Assay , DNA Damage/drug effects , Drosophila melanogaster/drug effects , Larva/drug effects , Magnetite Nanoparticles/chemistry , Mutagens/chemistry , Particle Size , Telophase/drug effects , Wings, Animal/drug effects
3.
Genes Cells ; 21(9): 978-93, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27458047

ABSTRACT

ICRF-193 [meso-4,4-(2,3-butanediyl)-bis(2,6-piperazinedione)] is a complex-stabilizing inhibitor of DNA topoisomerase II (topo II) that is used as an effective anticancer drug. ICRF-193 inhibits topo II catalytic activity in vitro and blocks nuclear division in vivo. Here, we examined the effects of ICRF-193 treatment on chromatin behavior and spindle dynamics using detailed live mitotic cell analysis in the fission yeast, Schizosaccharomyces pombe. Time-lapse movie analysis showed that ICRF-193 treatment leads to an elongation of presumed chromatin fibers connected to kinetochores during mid-mitosis. Anaphase spindles begin to arch, and eventually spindle poles come together abruptly, as if the spindle snapped at the point of spindle microtubule overlap in telophase. Segregating chromosomes appeared as elastic clumps and subsequently pulled back and merged. The snapped spindle phenotype was abolished by microtubule destabilization after thiabendazole treatment, accompanied by unequal chromosome segregation or severe defects in spindle extension. Thus, we conclude that ICRF-193-treated, unseparated sister chromatids pulling toward opposite spindle poles produce the arched and snapped telophase spindle. ICRF-193 treatment increased DNA content, suggesting that the failure of sister chromatids to separate properly in anaphase, causes the spindle to break in telophase, resulting in polyploidization.


Subject(s)
Piperazines/pharmacology , Schizosaccharomyces/drug effects , Spindle Apparatus/drug effects , Telophase/drug effects , Anaphase/drug effects , Anaphase/physiology , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/genetics , Cell Nucleus Division , Chromatids/drug effects , Chromatids/genetics , Chromatids/metabolism , Chromosome Segregation , Diketopiperazines , Kinetochores/metabolism , Microtubules/drug effects , Mitosis , Ploidies , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/physiology , Telophase/physiology , Topoisomerase II Inhibitors/pharmacology
4.
New Phytol ; 207(4): 1061-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26061286

ABSTRACT

Stress-activated plant mitogen-activated protein (MAP) kinase pathways play roles in growth adaptation to the environment by modulating cell division through cytoskeletal regulation, but the mechanisms are poorly understood. We performed protein interaction and phosphorylation experiments with cytoskeletal proteins, mass spectrometric identification of MPK6 complexes and immunofluorescence analyses of the microtubular cytoskeleton of mitotic cells using wild-type, mpk6-2 mutant and plants overexpressing the MAP kinase-inactivating phosphatase, AP2C3. We showed that MPK6 interacted with γ-tubulin and co-sedimented with plant microtubules polymerized in vitro. It was the active form of MAP kinase that was enriched with microtubules and followed similar dynamics to γ-tubulin, moving from poles to midzone during the anaphase-to-telophase transition. We found a novel substrate for MPK6, the microtubule plus end protein, EB1c. The mpk6-2 mutant was sensitive to 3-nitro-l-tyrosine (NO2 -Tyr) treatment with respect to mitotic abnormalities, and root cells overexpressing AP2C3 showed defects in chromosome segregation and spindle orientation. Our data suggest that the active form of MAP kinase interacts with γ-tubulin on specific subsets of mitotic microtubules during late mitosis. MPK6 phosphorylates EB1c, but not EB1a, and has a role in maintaining regular planes of cell division under stress conditions.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitogen-Activated Protein Kinases/metabolism , Spindle Apparatus/metabolism , Stress, Physiological , Tubulin/metabolism , Anaphase/drug effects , Arabidopsis/cytology , Arabidopsis/drug effects , Butadienes/pharmacology , Cell Proliferation/drug effects , Chromosome Segregation/drug effects , Cytokinesis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Kinetochores/drug effects , Kinetochores/metabolism , Meristem/cytology , Meristem/drug effects , Meristem/metabolism , Microtubules/drug effects , Nitriles/pharmacology , Nitrosation/drug effects , Phosphorylation/drug effects , Plant Cells/drug effects , Plant Cells/metabolism , Spindle Apparatus/drug effects , Stress, Physiological/drug effects , Telophase/drug effects , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
5.
J Pharmacol Exp Ther ; 352(1): 175-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25369797

ABSTRACT

To date, many anticancer drugs have been developed by directly or indirectly targeting microtubules, which are involved in cell division. Although this approach has yielded many anticancer drugs, these drugs produce undesirable side effects. An alternative strategy is needed, and targeting mitotic exit may be one alternative approach. Localization of phosphorylated barrier-to-autointegration factor (BAF) to the chromosomal core region is essential for nuclear envelope compartment relocalization. In this study, we isolated brazilin from Caesalpinia sappan Leguminosae and demonstrated that it inhibited BAF phosphorylation in vitro and in vivo. Moreover, we demonstrated direct binding between brazilin and BAF. The inhibition of BAF phosphorylation induced abnormal nuclear envelope reassembly and cell death, indicating that perturbation of nuclear envelope reassembly could be a novel approach to anticancer therapy. We propose that brazilin isolated from C. sappan may be a new anticancer drug candidate that induces cell death by inhibiting vaccinia-related kinase 1-mediated BAF phosphorylation.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Benzopyrans/isolation & purification , Benzopyrans/pharmacology , Caesalpinia/chemistry , DNA-Binding Proteins/metabolism , Nuclear Envelope/drug effects , Nuclear Proteins/metabolism , Animals , Antineoplastic Agents/metabolism , Benzopyrans/metabolism , Cell Death/drug effects , Drug Evaluation, Preclinical , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Nuclear Envelope/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Telophase/drug effects
6.
Cell Cycle ; 12(4): 663-73, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23370391

ABSTRACT

Aurora B kinase is an integral regulator of cytokinesis as it stabilizes the intercellular canal within the midbody to ensure proper chromosomal segregation during cell division. Here we identified an E3 ligase subunit, F box protein FBXL2, that by recognizing a calmodulin binding signature within Aurora B, ubiquitinates and removes the kinase from the midbody. Calmodulin, by competing with the F box protein for access to the calmodulin binding signature, protected Aurora B from FBXL2. Calmodulin co-localized with Aurora B on the midbody, preserved Aurora B levels in cells, and stabilized intercellular canals during delayed abscission. Genetic or pharmaceutical depletion of endogenous calmodulin significantly reduced Aurora B protein levels at the midbody resulting in tetraploidy and multi-spindle formation. The calmodulin inhibitor, calmidazolium, reduced Aurora B protein levels resulting in tetraploidy, mitotic arrest, and apoptosis of tumorigenic cells and profoundly inhibiting tumor formation in athymic nude mice. These observations indicate molecular interplay between Aurora B and calmodulin in telophase and suggest that calmodulin acts as a checkpoint sensor for chromosomal segregation errors during mitosis.


Subject(s)
Calmodulin/genetics , Cytokinesis/genetics , F-Box Proteins/genetics , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/genetics , Telophase/genetics , Animals , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Binding Sites , Calmodulin/metabolism , Cell Line, Tumor , Chromosome Segregation/drug effects , Cytokinesis/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , F-Box Proteins/metabolism , Humans , Imidazoles/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Telophase/drug effects , Tumor Burden/drug effects
7.
Cell Cycle ; 11(22): 4211-21, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23095638

ABSTRACT

Energy- and nutrient-sensing proteins such as AMPK, mTOR and S6K1 are now recognized as novel regulators of mitotic completion in proliferating cells. We investigated the cellular distribution of the Ser2481 autophosphorylation of mTOR, which directly monitors mTORC-specific catalytic activity, during mammalian cell mitosis and cytokinesis. Automated immunofluorescence experiments in human carcinoma cell lines revealed that phospho-mTOR (Ser2481) exhibited profound spatial and temporal dynamics during cell division. Phospho-mTOR (Ser2481) was strikingly enriched in mitotic cells, and in prophase, bright phospho-mTOR (Ser2481) staining could be clearly observed among condensed chromosomes. Phospho-mTOR (Ser2481) then redistributes from diffuse cytosolic staining that partially colocalizes with the mitotic spindle during the early phases of mitosis to the furrow at the onset of cytokinesis. Like the bona fide chromosomal passenger proteins (CPPs) INCENP and Aurora B, phospho-mTOR (Ser2481) displayed noteworthy accumulation in the central spindle midzone and the midbody regions, which persisted during the furrowing process. Accordingly, double-staining experiments confirmed that phospho-mTOR (Ser2481) largely colocalized with CCPs in the midbodies. The CPP-like mitotic localization of phospho-mTOR (Ser2481) was fully prevented by the microtubule-depolymerizing drug nocodazole; mitotic traveling of phospho-mTOR (Ser2481) to the midbody during telophase and cytokinesis, where it appears to be integrated into the CPP-driven cytokinetic machinery, may therefore require dynamic microtubules. Although the Ser2448-phosphorylated form of mTOR was also found at high levels during M-phase in human cancer cells, we failed to observe a significant association of phospho-mTOR (Ser2448) with CCP-positive mitotic and cytokinetic structures. Our findings add phospho-mTOR (Ser2481) to the growing list of phospho-active forms of proteins belonging to the AMPK/mTOR/S6K1 signaling axis that reside at the mitotic and cytokinetic apparatus. Future studies should elucidate how the specific ability of phospho-mTOR (Ser2481) to spatially and temporally couple to the cleavage furrow and midbody region as a CPP-like protein can signal to or from adjacent signaling complexes and/or with the basic machinery of cell abscission.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , Cell Division , Cell Line, Tumor , Cytokinesis/drug effects , HeLa Cells , Humans , MCF-7 Cells , Microtubules/chemistry , Microtubules/metabolism , Nocodazole/pharmacology , Phosphorylation/drug effects , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Telophase/drug effects
8.
Zh Evol Biokhim Fiziol ; 48(1): 17-21, 2012.
Article in Russian | MEDLINE | ID: mdl-22567971

ABSTRACT

Evolutionary conservative chemosignal 2,5-dimethylpyrazin that is pheromone in female mice has been shown to increase frequency of mitotic aberrations analyzed with aid of metaphasic and ana-telophasic analysis in bone marrow cells. Replacement of one of methyl radicals in the pheromone molecule by the carboxyl radical reveals specificity of action of the used derivative: the frequency of disturbances revealed only by the ana-telophasic analysis increases, whereas by the metaphasic analysis, no induction of disturbance is detected. In the sperm head abnormality test there is shown a rise of the anomalies by both compounds. Possible mechanisms of specific action of the tested substances on stability of genetic apparatus of the bone marrow dividing cells in the house mouse are discussed.


Subject(s)
Bone Marrow Cells/metabolism , Pyrazines/adverse effects , Pyrazines/chemistry , Signal Transduction/drug effects , Sperm Head/metabolism , Testis/metabolism , Animals , Bone Marrow Cells/pathology , Female , Male , Mice , Pyrazines/pharmacology , Sex Attractants/adverse effects , Sex Attractants/chemistry , Sex Attractants/pharmacology , Sperm Head/pathology , Telophase/drug effects , Testis/pathology
9.
Biol Reprod ; 87(1): 11, 1-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539682

ABSTRACT

Meiosis in mammalian females is marked by two arrest points, at prophase I and metaphase II, which must be tightly regulated in order to produce a haploid gamete at the time of fertilization. The transition metal zinc has emerged as a necessary and dynamic regulator of the establishment, maintenance, and exit from metaphase II arrest, but the roles of zinc during prophase I arrest are largely unknown. In this study, we investigate the mechanisms of zinc regulation during the first meiotic arrest. Disrupting zinc availability in the prophase I arrested oocyte by treatment with the heavy metal chelator N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylenediamine (TPEN) causes meiotic resumption even in the presence of pharmacological inhibitors of meiosis. We further show that the MOS-MAPK pathway mediates zinc-dependent prophase I arrest, as the pathway prematurely activates during TPEN-induced meiotic resumption. Conversely, inhibition of the MOS-MAPK pathway maintains prophase I arrest. While prolonged zinc insufficiency ultimately results in telophase I arrest, early and transient exposure of oocytes to TPEN is sufficient to induce meiotic resumption and bypass the telophase I block, allowing the formation of developmentally competent eggs upon parthenogenetic activation. These results establish zinc as a crucial regulator of meiosis throughout the entirety of oocyte maturation, including the maintenance of and release from the first and second meiotic arrest points.


Subject(s)
Meiotic Prophase I/physiology , Oocytes/cytology , Oocytes/metabolism , Zinc/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , Female , In Vitro Techniques , MAP Kinase Signaling System , Meiotic Prophase I/drug effects , Mice , Oocytes/drug effects , Oogenesis/drug effects , Oogenesis/physiology , Parthenogenesis , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-mos/metabolism , Telophase/drug effects , Telophase/physiology , Zinc/deficiency
10.
Eur J Cell Biol ; 91(5): 413-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22365812

ABSTRACT

Mitosis is a continuous process to separate replicated chromosomes into two daughter cells through prophase, metaphase, anaphase, and telophase. Although a number of methods have been established to synchronize cells at different phases of the cell cycle, it is difficult to synchronize cells at the specific phases, anaphase and telophase, during mitosis because of the short duration of anaphase. Here, we show that HeLa S3 cells in anaphase and in telophase are successfully enriched by treatment with a combination of low concentrations of the microtubule-depolymerizing agent nocodazole and the myosin II inhibitor blebbistatin. After 9-h release from thymidine block at G1/S phase, addition of nocodazole at 20 ng/ml but not 40 ng/ml ensures rapid release from the nocodazole arrest. Subsequently, the cells are cultured in the presence of 50 µM blebbistatin for 20 and 50 min to enrich cells in anaphase and telophase, respectively. Western blot analysis verifies down-regulation of phospho-histone H3-Ser10, phospho-Aurora A/B/C, and cyclin B1 during M-phase progression. Furthermore, we show how the electrophoretic mobility shifts of the Src-family kinases c-Yes and c-Src can change in each phase of mitosis. These results provide a useful synchronization method for biochemically examining protein dynamics during M-phase progression.


Subject(s)
Anaphase/drug effects , Cell Cycle Checkpoints/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Metaphase/drug effects , Nocodazole/pharmacology , Telophase/drug effects , Tubulin Modulators/pharmacology , Anaphase/physiology , Cell Culture Techniques/methods , Cell Cycle Checkpoints/physiology , HeLa Cells , Humans , Metaphase/physiology , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Telophase/physiology
11.
Mol Hum Reprod ; 17(5): 296-304, 2011 May.
Article in English | MEDLINE | ID: mdl-21266449

ABSTRACT

JMY is a transcriptional co-factor of p53. Latest work has revealed that JMY is also an actin nucleation factor that regulates new filament assembly and activates Arp2/3 complex in somatic cells; however, roles of JMY in mouse oocyte are unknown. Here we showed the expression and functions of JMY during mouse oocyte meiotic maturation. JMY mRNA is expressed largely from germinal vesicle to metaphase I stage, and gradually decreased during anaphase I, telophase I (TI) and metaphase II (MII) stages. Immunostaining results showed that JMY localized at the spindle and cytoplasm of oocytes. Depletion of JMY by RNAi resulted in symmetric division, failure of spindle migration and cytokinesis during oocyte meiotic maturation, showing a 2-cell-like MII oocyte and TI stage arrest. Actin cap and cortical granules-free domain formation were also disrupted after JMY RNAi, indicating the failure of spindle migration. JMY antibody injection results were consistent with those of JMY RNAi, further confirming the involvement of JMY in oocyte polarity. Our data indicate that JMY is required for spindle migration, asymmetric division and cytokinesis during mouse oocyte maturation.


Subject(s)
Asymmetric Cell Division/genetics , Cytokinesis/genetics , Nuclear Proteins/genetics , Oocytes/metabolism , Trans-Activators/genetics , Anaphase/drug effects , Animals , Antibodies/pharmacology , Asymmetric Cell Division/drug effects , Cell Cycle Proteins , Cell Polarity/drug effects , Cell Polarity/genetics , Cytokinesis/drug effects , Cytoplasm/drug effects , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoskeleton/drug effects , Cytoskeleton/genetics , Cytoskeleton/metabolism , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Meiosis/genetics , Metaphase/drug effects , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oocytes/cytology , Oocytes/drug effects , RNA, Small Interfering/genetics , Telophase/drug effects , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
12.
Anticancer Res ; 30(12): 4937-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21187473

ABSTRACT

BACKGROUND: The rate-limiting enzyme of the mevalonate pathway, 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase, provides essential intermediates for the prenylation or dolichylation of growth-related proteins. d-δ-tocotrienol, a post-transcriptional down-regulator of HMG CoA reductase, suppresses the proliferation of murine B16 melanoma cells. Dietary d-δ-tocotrienol suppresses the growth of implanted B16 melanomas without toxicity to host mice. MATERIALS AND METHODS: The proliferation of human A2058 and A375 melanoma cells following a 72 h incubation in 96-well plates was measured by CellTiter 96® Aqueous One Solution. Cell cycle distribution was determined by flow cytometry. Fluorescence microscopy following acridine orange and ethidium bromide dual staining and procaspase-3 cleavage were used to detect apoptosis. Western-blot was employed to measure protein expression. RESULTS: d-δ-Tocotrienol induced dose-dependent suppression of cell proliferation with 50% inhibitory concentrations (IC(50)) of 37.5 ± 1.4 (A2058) and 22.3 ± 1.8 (A375) µmol/l, respectively (data are reported as mean ± standard deviation). d-δ-Tocotrienol-mediated cell cycle arrest at the G(1) phase was accompanied by reduced expression of cyclin-dependent kinase 4. Concomitantly, d-δ-tocotrienol induced caspase-3 activation and apoptosis. The impact of d-δ-tocotrienol on A2058 cell proliferation was potentiated by lovastatin (IC(50)=3.1 ± 0.5 µmol/l), a competitive inhibitor of HMG CoA reductase. CONCLUSION: d-δ-Tocotrienol may have potential application in melanoma chemoprevention and/or therapy.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Vitamin E/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , G1 Phase/drug effects , Humans , Lovastatin/administration & dosage , Lovastatin/pharmacology , Telophase/drug effects , Vitamin E/administration & dosage , Vitamin E/pharmacology
13.
Nat Chem Biol ; 6(9): 674-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20693991

ABSTRACT

Cellular metal ion fluxes are known in alkali and alkaline earth metals but are not well documented in transition metals. Here we describe major changes in the zinc physiology of the mammalian oocyte as it matures and initiates embryonic development. Single-cell elemental analysis of mouse oocytes by synchrotron-based X-ray fluorescence microscopy (XFM) revealed a 50% increase in total zinc content within the 12-14-h period of meiotic maturation. Perturbation of zinc homeostasis with a cell-permeable small-molecule chelator blocked meiotic progression past telophase I. Zinc supplementation rescued this phenotype when administered before this meiotic block. However, after telophase arrest, zinc triggered parthenogenesis, suggesting that exit from this meiotic step is tightly regulated by the availability of a zinc-dependent signal. These results implicate the zinc bolus acquired during meiotic maturation as an important part of the maternal legacy to the embryo.


Subject(s)
Mammals/embryology , Meiosis/physiology , Oocytes/cytology , Oocytes/metabolism , Zinc/metabolism , Animals , Chelating Agents/pharmacology , Embryonic Development/drug effects , Embryonic Development/physiology , Female , Meiosis/drug effects , Mice , Microscopy, Fluorescence , Oocytes/drug effects , Oocytes/growth & development , Parthenogenesis/drug effects , Parthenogenesis/physiology , Pregnancy , Telophase/drug effects , Telophase/physiology , Zinc/antagonists & inhibitors
14.
Chemosphere ; 75(7): 934-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19201446

ABSTRACT

We used the anaphase-telophase chromosome aberration and comet (Single Cell Gel Electrophoresis, SCGE) assays to evaluate the genotoxic effects of copper sulphate (CS) and cobalt chloride (CC) chemicals prepared in two concentrations (EC(50), 2xEC(50)), using methyl methanesulfonate (MMS) as a positive control and untreated cells as a negative control. In Allium root growth inhibition test, EC(50) values for CS and CC are 1.5 and 5.5 ppm, respectively. Mitotic index (MI) decreased in all concentrations tested of CS and CC compared to the control at each exposure time. The bridge, stickiness, vagrant chromosomes, fragments, c-anaphase and multipolarity chromosome aberrations were observed in anaphase-telophase cells. The total chromosome aberrations were more frequent with an increasing in the exposure time and the concentrations of both chemicals. The genotoxicity of CS and CC in Allium cepa root cells was analyzed using a mild alkaline comet assay at pH 12.3, which allows the detection of single strand breaks. In all the concentrations, CS and CC induced a significant increase (P<0.05) in DNA damage. No significant difference was found between positive control (300+/-5.81) and 3 ppm CS (280+/-4.61). The methods used are applicable for biological monitoring of environmental pollutants.


Subject(s)
Chromosome Aberrations , Cobalt/toxicity , Copper Sulfate/toxicity , Environmental Pollutants/toxicity , Onions/drug effects , Anaphase/drug effects , Cobalt/pharmacology , Comet Assay , Copper Sulfate/pharmacology , DNA Damage , Environmental Pollutants/pharmacology , Mitosis , Onions/genetics , Onions/growth & development , Plant Roots/drug effects , Plant Roots/genetics , Plant Roots/growth & development , Telophase/drug effects
15.
J Reprod Dev ; 54(4): 244-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18490860

ABSTRACT

Sperm-specific phospholipase C, PLCzeta, is a candidate for the Ca(2+) oscillation-inducing factor that is introduced into the ooplasm upon sperm-egg fusion. In addition to the 647-residue full-length PLCzeta, s-PLCzeta lacking the N-terminal 110 amino acids is known to be present in the mouse testis. In this study, we attempted to obtain full-term offspring from s-PLCzeta-activated eggs by round spermatid injection. Metaphase II-arrested eggs injected with a high RNA concentration of s-PLCzeta RNA normally developed to blastocysts. When the round spermatid nucleus was injected into telophase II-stage eggs previously activated by s-PLCzeta RNA, three live offspring were successfully obtained by transfer of the developed 4-cell embryos to pseudopregnant mice. These three offspring all grew to be normal adults and reproduced healthy second-generation mice.


Subject(s)
EF Hand Motifs/physiology , Embryonic Development/drug effects , Oocytes/drug effects , Parturition/drug effects , Phosphoinositide Phospholipase C/pharmacology , Type C Phospholipases/pharmacology , Animals , Animals, Newborn , Embryo Culture Techniques , Embryo, Mammalian , Female , Isoenzymes/chemistry , Isoenzymes/pharmacology , Mice , Mice, Inbred C57BL , Oogenesis/drug effects , Parthenogenesis/drug effects , Phosphoinositide Phospholipase C/chemistry , Pregnancy , Telophase/drug effects , Type C Phospholipases/chemistry
16.
Mol Biol Cell ; 19(5): 2328-38, 2008 May.
Article in English | MEDLINE | ID: mdl-18287523

ABSTRACT

mDia proteins are mammalian homologues of Drosophila diaphanous and belong to the formin family proteins that catalyze actin nucleation and polymerization. Although formin family proteins of nonmammalian species such as Drosophila diaphanous are essential in cytokinesis, whether and how mDia proteins function in cytokinesis remain unknown. Here we depleted each of the three mDia isoforms in NIH 3T3 cells by RNA interference and examined this issue. Depletion of mDia2 selectively increased the number of binucleate cells, which was corrected by coexpression of RNAi-resistant full-length mDia2. mDia2 accumulates in the cleavage furrow during anaphase to telophase, and concentrates in the midbody at the end of cytokinesis. Depletion of mDia2 induced contraction at aberrant sites of dividing cells, where contractile ring components such as RhoA, myosin, anillin, and phosphorylated ERM accumulated. Treatment with blebbistatin suppressed abnormal contraction, corrected localization of the above components, and revealed that the amount of F-actin at the equatorial region during anaphase/telophase was significantly decreased with mDia2 RNAi. These results demonstrate that mDia2 is essential in mammalian cell cytokinesis and that mDia2-induced F-actin forms a scaffold for the contractile ring and maintains its position in the middle of a dividing cell.


Subject(s)
Actins/metabolism , Cytokinesis , Fibroblasts/cytology , Microtubule-Associated Proteins/metabolism , NADPH Dehydrogenase/metabolism , Anaphase/drug effects , Animals , Cytokinesis/drug effects , Fibroblasts/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Mice , Microtubule-Associated Proteins/deficiency , NADPH Dehydrogenase/deficiency , NIH 3T3 Cells , Protein Isoforms/metabolism , Protein Transport/drug effects , RNA Interference/drug effects , Telophase/drug effects , rho GTP-Binding Proteins/metabolism
17.
Mol Reprod Dev ; 75(5): 801-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18157849

ABSTRACT

The present study was designed to investigate the effects of nicotine on development of bovine embryos derived from parthenogenetic activation (PA) and in vitro fertilization (IVF). Nicotine caused disfigured secondary meiotic spindle structures and affected embryonic development in a dose-dependent manner. Concentrations at 0.01-0.5 mM resulted in cleavage and blastocyst rates similar to the controls for both PA and IVF embryos. Nicotine at 2.0 and 4.0 mM significantly decreased the cleavage rates and none of the embryos developed beyond the 16-cell stage. Nicotine might disrupt the polymerization of microfilaments leading to impaired chromosome alignment or segregation, and induce the formation of polynuclei with a variety of abnormal nuclear structures such as 2-6 nuclei, 2-4 metaphase plates, 2-4 sets of anaphase/telophase plates, and the co-existence of polynuclei and 2-4 sets of anaphase/telophase plates. Nicotine adversely affected blastocyst chromosomal composition. Fifty-six to 70% of the IVF blastocysts and 71-88% of the PA blastocysts were polyploid and/or mixoploid after culture in 0.2-1.0 mM nicotine-containing media, which were higher (P < 0.05 or P < 0.01) than the controls. Cell numbers of the nicotine-cultured blastocysts were significantly lower than the control. In conclusion, nicotine induced disfigured spindles and irregular chromosome alignment and possibly impaired cytokinesis, which lead to decreased quality of the yielded blastocysts.


Subject(s)
Blastocyst/pathology , Cell Nucleus/pathology , Chromosome Aberrations/chemically induced , Embryonic Development/drug effects , Giant Cells/pathology , Nicotine/toxicity , Nicotinic Agonists/toxicity , Anaphase/drug effects , Animals , Blastocyst/metabolism , Cattle , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Embryo Culture Techniques , Female , Fertilization in Vitro , Giant Cells/metabolism , Parthenogenesis , Telophase/drug effects
18.
Curr Biol ; 17(23): 2081-6, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18060787

ABSTRACT

For mammalian somatic cells, the importance of microtubule cytoskeleton integrity during interphase cell-cycle progression is uncertain. The loss, suppression, or stabilization of the microtubule cytoskeleton has been widely reported to cause a G1 arrest in a variable, and often high, proportion of cell populations, suggesting the existence of a "microtubule damage," "microtubule integrity," or "postmitotic" checkpoint in G1 or G2. We found that when normal human cells (hTERT RPE1 and primary fibroblasts) are continuously exposed to nocodazole, they remain in mitosis for 10-48 hr before they slip out of mitosis and arrest in G1; this finding is consistent with previous reports. To eliminate the persistent effects of prolonged mitosis, we isolated anaphase-telophase cells that were just finishing a mitosis of normal duration, then we rapidly and completely disassembled microtubules by chilling the preparations to 0 degrees C for 10 minutes in the continuous presence of nocodazole or colcemid treatment to ensure that the cells entered G1 without a microtubule cytoskeleton. Without microtubules, cells progressed from anaphase to a subsequent mitosis with essentially normal kinetics. Similar results were obtained for cells in which the microtubule cytoskeleton was partially diminished by lower nocodazole doses or augmented and stabilized with taxol. Thus, after a preceding mitosis of normal duration, the integrity of the microtubule cytoskeleton is not subject to checkpoint surveillance, nor is it required for the normal human cell to progress through G1 and the remainder of interphase.


Subject(s)
Anaphase/drug effects , Anaphase/physiology , Cell Cycle/drug effects , Cytoskeleton/drug effects , Microtubules/drug effects , Cell Cycle/physiology , Cell Line , Cytoskeleton/metabolism , Fibroblasts , Humans , Image Processing, Computer-Assisted , Microscopy, Video , Microtubules/metabolism , Mitosis/drug effects , Mitosis/physiology , Nocodazole/pharmacology , Paclitaxel/pharmacology , Telophase/drug effects , Telophase/physiology
19.
Methods Enzymol ; 428: 63-76, 2007.
Article in English | MEDLINE | ID: mdl-17875412

ABSTRACT

To maximize the probability of survival and proliferation, cells coordinate various intracellular activities in response to changes in the extracellular environment. Eukaryotic cells transduce diverse cellular stimuli by multiple mitogen-activated protein kinase (MAPK) cascades. Exposure of cells to stress results in rapid activation of a highly conserved family of MAPKs, known as stress-activated protein kinases (SAPKs). Activation of SAPKs results in the generation of a set of adaptive responses that leads to the modulation of several aspects of cell physiology essential for cell survival, such as gene expression, translation, and morphogenesis. This chapter proposes that regulation of cell cycle progression is another general stress response critical for cell survival. Studies from yeast, both Schizosaccharomyces pombe and Saccharomyces cerevisiae, have served to start understanding how SAPKs control cell cycle progression in response to stress.


Subject(s)
Cell Cycle/physiology , Osmotic Pressure , Saccharomyces cerevisiae/physiology , Schizosaccharomyces/physiology , Anaphase/drug effects , Cyclin-Dependent Kinases/physiology , G1 Phase/physiology , G2 Phase/physiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Kinase Kinases/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/physiology , Saccharomyces cerevisiae Proteins/physiology , Telophase/drug effects
20.
Dev Cell ; 12(1): 17-30, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17199038

ABSTRACT

The centromere is the chromosomal site that joins to microtubules during mitosis for proper segregation. Determining the location of a centromere-specific histone H3 called CENP-A at the centromere is vital for understanding centromere structure and function. Here, we report the identification of three human proteins essential for centromere/kinetochore structure and function, hMis18alpha, hMis18beta, and M18BP1, the complex of which is accumulated specifically at the telophase-G1 centromere. We provide evidence that such centromeric localization of hMis18 is essential for the subsequent recruitment of de novo-synthesized CENP-A. If any of the three is knocked down by RNAi, centromere recruitment of newly synthesized CENP-A is rapidly abolished, followed by defects such as misaligned chromosomes, anaphase missegregation, and interphase micronuclei. Tricostatin A, an inhibitor to histone deacetylase, suppresses the loss of CENP-A recruitment to centromeres in hMis18alpha RNAi cells. Telophase centromere chromatin may be primed or licensed by the hMis18 complex and RbAp46/48 to recruit CENP-A through regulating the acetylation status in the centromere.


Subject(s)
Autoantigens/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Cycle Proteins , Centromere/drug effects , Centromere Protein A , Chromosomal Proteins, Non-Histone/chemistry , Chromosome Segregation/drug effects , Consensus Sequence , Genome, Human/drug effects , HeLa Cells , Humans , Hydroxamic Acids/pharmacology , Metaphase/drug effects , Molecular Sequence Data , Mutation/genetics , Phylogeny , Protein Binding/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-myb/metabolism , RNA Interference , Recombinant Fusion Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Telophase/drug effects , Vertebrates
SELECTION OF CITATIONS
SEARCH DETAIL
...