Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 228
Filtrer
1.
Cell Cycle ; 20(12): 1195-1208, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-34048314

RÉSUMÉ

ATP metabolism during mitosis needs to be coordinated with numerous energy-demanding activities, especially in cancer cells whose metabolic pathways are reprogramed to sustain rapid proliferation in a nutrient-deficient environment. Although strategies targeting the energy metabolic pathways have shown therapeutic efficacy in preclinical cancer models, how normal cells and cancer cells differentially respond to energy shortage is unclear. In this study, using time-lapse microscopy, we found that cancer cells displayed unique mitotic phenotypes in a dose-dependent manner upon decreasing ATP (i.e. energy) supply. When reduction in ATP concentration was moderate, chromosome movements in mitosis were barely affected, while the metaphase-anaphase transition was significantly prolonged due to reduced tension between the sister-kinetochores, which delayed the satisfaction of the spindle assembly checkpoint. Further reduction in ATP concentration led to a decreased level of Aurora-B at the centromere, resulting in increased chromosome mis-segregation after metaphase delay. In contrast to cancer cells, ATP restriction in non-transformed cells induced cell cycle arrest in interphase, rather than causing mitotic defects. In addition, data mining of cancer patient database showed a correlation between signatures of energy production and chromosomal instability possibly resulted from mitotic defects. Together, these results reveal that energy restriction induces differential responses in normal and cancer cells, with chromosome mis-segregation only observed in cancer cells. This points to targeting energy metabolism as a potentially cancer-selective therapeutic strategy.


Sujet(s)
Adénosine triphosphate/métabolisme , Adénosine triphosphate/pharmacologie , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Métaphase/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du col de l'utérus/métabolisme , Anaphase/effets des médicaments et des substances chimiques , Aurora kinase B/métabolisme , Femelle , Cellules HeLa , Humains , Interphase/effets des médicaments et des substances chimiques , Kinétochores/métabolisme , Microscopie/méthodes , NAD/pharmacologie , Appareil du fuseau/métabolisme , Imagerie accélérée/méthodes , Tumeurs du col de l'utérus/anatomopathologie
2.
Molecules ; 26(3)2021 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-33494466

RÉSUMÉ

Amsacrine, an anticancer drug first synthesised in 1970 by Professor Cain and colleagues, showed excellent preclinical activity and underwent clinical trial in 1978 under the auspices of the US National Cancer Institute, showing activity against acute lymphoblastic leukaemia. In 1984, the enzyme DNA topoisomerase II was identified as a molecular target for amsacrine, acting to poison this enzyme and to induce DNA double-strand breaks. One of the main challenges in the 1980s was to determine whether amsacrine analogues could be developed with activity against solid tumours. A multidisciplinary team was assembled in Auckland, and Professor Denny played a leading role in this approach. Among a large number of drugs developed in the programme, N-[2-(dimethylamino)-ethyl]-acridine-4-carboxamide (DACA), first synthesised by Professor Denny, showed excellent activity against a mouse lung adenocarcinoma. It underwent clinical trial, but dose escalation was prevented by ion channel toxicity. Subsequent work led to the DACA derivative SN 28049, which had increased potency and reduced ion channel toxicity. Mode of action studies suggested that both amsacrine and DACA target the enzyme DNA topoisomerase II but with a different balance of cellular consequences. As primarily a topoisomerase II poison, amsacrine acts to turn the enzyme into a DNA-damaging agent. As primarily topoisomerase II catalytic inhibitors, DACA and SN 28049 act to inhibit the segregation of daughter chromatids during anaphase. The balance between these two actions, one cell cycle phase specific and the other nonspecific, together with pharmacokinetic, cytokinetic and immunogenic considerations, provides links between the actions of acridine derivatives and anthracyclines such as doxorubicin. They also provide insights into the action of cytotoxic DNA-binding drugs.


Sujet(s)
Adénocarcinome pulmonaire/traitement médicamenteux , Antinéoplasiques , ADN tumoral/métabolisme , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de la topoisomérase-II , Adénocarcinome pulmonaire/histoire , Adénocarcinome pulmonaire/métabolisme , Amsacrine/composition chimique , Amsacrine/histoire , Amsacrine/pharmacocinétique , Amsacrine/usage thérapeutique , Anaphase/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/histoire , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/usage thérapeutique , Chromatides/métabolisme , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , ADN topoisomérases de type II/métabolisme , Histoire du 20ème siècle , Histoire du 21ème siècle , Humains , Tumeurs du poumon/histoire , Tumeurs du poumon/métabolisme , Souris , Naphtyridines/composition chimique , Naphtyridines/pharmacocinétique , Naphtyridines/usage thérapeutique , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Inhibiteurs de la topoisomérase-II/composition chimique , Inhibiteurs de la topoisomérase-II/pharmacocinétique , Inhibiteurs de la topoisomérase-II/usage thérapeutique
3.
J Cell Biol ; 220(1)2021 01 04.
Article de Anglais | MEDLINE | ID: mdl-33284320

RÉSUMÉ

During G1 in budding yeast, the Cdc42 GTPase establishes a polar front, along which actin is recruited to direct secretion for bud formation. Cdc42 localizes at the bud cortex and then redistributes between mother and daughter in anaphase. The molecular mechanisms that terminate Cdc42 bud-localized activity during mitosis are poorly understood. We demonstrate that the activity of the Cdc14 phosphatase, released through the mitotic exit network, is required for Cdc42 redistribution between mother and bud. Induced Cdc14 nucleolar release results in premature Cdc42 redistribution between mother and bud. Inhibition of Cdc14 causes persistence of Cdc42 bud localization, which perturbs normal cell size and spindle positioning. Bem3, a Cdc42 GAP, binds Cdc14 and is dephosphorylated at late anaphase in a Cdc14-dependent manner. We propose that Cdc14 dephosphorylates and activates Bem3 to allow Cdc42 inactivation and redistribution. Our results uncover a mechanism through which Cdc14 regulates the spatiotemporal activity of Cdc42 to maintain normal cell size at cytokinesis.


Sujet(s)
Mitose , Saccharomyces cerevisiae/cytologie , Saccharomyces cerevisiae/métabolisme , Protéine G cdc42/métabolisme , Anaphase/effets des médicaments et des substances chimiques , Composés hétérocycliques bicycliques/pharmacologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Taille de la cellule/effets des médicaments et des substances chimiques , Guanosine triphosphate/métabolisme , Métaphase/effets des médicaments et des substances chimiques , Mitose/effets des médicaments et des substances chimiques , Nocodazole/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Domaines protéiques , Protein Tyrosine Phosphatases/métabolisme , Saccharomyces cerevisiae/effets des médicaments et des substances chimiques , Protéines de Saccharomyces cerevisiae/métabolisme , Appareil du fuseau/effets des médicaments et des substances chimiques , Appareil du fuseau/métabolisme , Thiazolidines/pharmacologie , Facteurs temps
4.
Mol Cancer Ther ; 20(3): 477-489, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33277443

RÉSUMÉ

Cyclin-dependent kinase 2 (CDK2) antagonism inhibits clustering of excessive centrosomes at mitosis, causing multipolar cell division and apoptotic death. This is called anaphase catastrophe. To establish induced anaphase catastrophe as a clinically tractable antineoplastic mechanism, induced anaphase catastrophe was explored in different aneuploid cancers after treatment with CYC065 (Cyclacel), a CDK2/9 inhibitor. Antineoplastic activity was studied in preclinical models. CYC065 treatment augmented anaphase catastrophe in diverse cancers including lymphoma, lung, colon, and pancreatic cancers, despite KRAS oncoprotein expression. Anaphase catastrophe was a broadly active antineoplastic mechanism. Reverse phase protein arrays (RPPAs) revealed that along with known CDK2/9 targets, focal adhesion kinase and Src phosphorylation that regulate metastasis were each repressed by CYC065 treatment. Intriguingly, CYC065 treatment decreased lung cancer metastases in in vivo murine models. CYC065 treatment also significantly reduced the rate of lung cancer growth in syngeneic murine and patient-derived xenograft (PDX) models independent of KRAS oncoprotein expression. Immunohistochemistry analysis of CYC065-treated lung cancer PDX models confirmed repression of proteins highlighted by RPPAs, implicating them as indicators of CYC065 antitumor response. Phospho-histone H3 staining detected anaphase catastrophe in CYC065-treated PDXs. Thus, induced anaphase catastrophe after CYC065 treatment can combat aneuploid cancers despite KRAS oncoprotein expression. These findings should guide future trials of this novel CDK2/9 inhibitor in the cancer clinic.


Sujet(s)
Anaphase/effets des médicaments et des substances chimiques , Kinase-2 cycline-dépendante/antagonistes et inhibiteurs , Aneuploïdie , Animaux , Carcinogenèse , Prolifération cellulaire , Humains , Souris , Souris nude , Métastase tumorale , Transfection
5.
PLoS One ; 15(7): e0236373, 2020.
Article de Anglais | MEDLINE | ID: mdl-32702063

RÉSUMÉ

The diagnosis of patients with malignancies relies on the results of a clinical cytological examination. To enhance the diagnostic qualities of cytological examinations, it is important to have a detailed analysis of the cell's characteristics. There is, therefore, a need for developing a new auxiliary method for cytological diagnosis. In this study, we focused on studying the charge of the cell membrane surface of fixed cells, which is one of important cell's characteristics. Although fixed cells lose membrane potential which is observed in living cells owing to ion dynamics, we hypothesized that fixed cells still have a cell membrane surface charge due to cell membrane components and structure. We used 5 cell lines in this study (ARO, C32TG, RT4, TK, UM-UC-14). After fixation with CytoRich Red, we measured the cell membrane surface charge of fixed cells in solution using zeta potential measurements and fixed cells on glass slides, visualizing it using antibody-labeled beads and positively-charged beads. Furthermore, we measured the cell membrane surface charge of fixed cells under different conditions, such as different solution of fixative, ion concentration, pH, and pepsin treatments. The zeta potential measurements and visualization using the beads indicated that the cell membrane surface of fixed cells was negatively charged, and also that the charge varied among fixed cells. The charge state was affected by the different treatments. Moreover, the number of cell-bound beads was small in interphase, anaphase, and apoptotic cells. We concluded that the negative cell membrane surface charge was influenced by the three-dimensional structure of proteins as well as the different types of amino acids and lipids on the cell membrane. Thus, cell surface charge visualization can be applied as a new auxiliary method for clinical cytological diagnosis. This is the first systematic report of the cell membrane surface charge of fixed cells.


Sujet(s)
Lignée cellulaire/ultrastructure , Membrane cellulaire/ultrastructure , Cellules cultivées/ultrastructure , Cytodiagnostic , Anaphase/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Cycle cellulaire/effets des médicaments et des substances chimiques , Division cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées/effets des médicaments et des substances chimiques , Fixateurs/pharmacologie , Humains , Potentiels de membrane/effets des médicaments et des substances chimiques , Pepsine A/pharmacologie , Propriétés de surface
6.
Cell Cycle ; 19(15): 1899-1916, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32594833

RÉSUMÉ

The Ran GTPase plays critical roles in multiple cellular processes including interphase nucleocytoplasmic transport and mitotic spindle assembly. During mitosis in mammalian cells, GTP-bound Ran (Ran-GTP) is concentrated near mitotic chromatin while GDP-bound Ran (Ran-GDP) is more abundant distal to chromosomes. This pattern spatially controls spindle formation because Ran-GTP locally releases spindle assembly factors (SAFs), such as Hepatoma Up-Regulated Protein (HURP), from inhibitory interactions near chromosomes. Regulator of Chromatin Condensation 1 (RCC1) is Ran's chromatin-bound exchange factor, and RanBP1 is a conserved Ran-GTP-binding protein that has been implicated as a mitotic regulator of RCC1 in embryonic systems. Here, we show that RanBP1 controls mitotic RCC1 dynamics in human somatic tissue culture cells. In addition, we observed the re-localization of HURP in metaphase cells after RanBP1 degradation, consistent with the idea that altered RCC1 dynamics functionally modulate SAF activities. Together, our findings reveal an important mitotic role for RanBP1 in human somatic cells, controlling the spatial distribution and magnitude of mitotic Ran-GTP production and thereby ensuring the accurate execution of Ran-dependent mitotic events. ABBREVIATIONS: AID: Auxin-induced degron; FLIP: Fluorescence loss in photobleaching; FRAP: Fluorescence recovery after photobleaching; GDP: guanosine diphosphate; GTP: guanosine triphosphate; HURP: Hepatoma Up-Regulated Protein; NE: nuclear envelope; NEBD: Nuclear Envelope Breakdown; RanBP1: Ran-binding protein 1; RanGAP1: Ran GTPase-Activating Protein 1; RCC1: Regulator of Chromatin Condensation 1; RRR complex: RCC1/Ran/RanBP1 heterotrimeric complex; SAF: Spindle Assembly Factor; TIR1: Transport Inhibitor Response 1 protein; XEE: Xenopus egg extract.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Mammifères/métabolisme , Mitose , Protéines nucléaires/métabolisme , Transduction du signal , Protéine G ran/métabolisme , Anaphase/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chromatine/métabolisme , Chromosomes de mammifère/métabolisme , Acides indolacétiques/pharmacologie , Métaphase/effets des médicaments et des substances chimiques , Mitose/effets des médicaments et des substances chimiques , Modèles biologiques , Protéines tumorales/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Appareil du fuseau/effets des médicaments et des substances chimiques , Appareil du fuseau/métabolisme , Xenopus laevis
7.
Ecotoxicol Environ Saf ; 192: 110328, 2020 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-32078840

RÉSUMÉ

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.


Sujet(s)
Calixarènes/toxicité , Altération de l'ADN , Drosophila melanogaster/effets des médicaments et des substances chimiques , Oignons/effets des médicaments et des substances chimiques , Anaphase/effets des médicaments et des substances chimiques , Animaux , Calixarènes/composition chimique , Aberrations des chromosomes , Test des comètes , Cytotoxines/composition chimique , Cytotoxines/toxicité , Drosophila melanogaster/génétique , Hémocytes/effets des médicaments et des substances chimiques , Index mitotique , Oignons/génétique , Racines de plante/effets des médicaments et des substances chimiques , Quinoléines/synthèse chimique , Quinoléines/composition chimique , Quinoléines/toxicité , Télophase/effets des médicaments et des substances chimiques
8.
Nucleus ; 11(1): 19-31, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-31948316

RÉSUMÉ

Elastic tethers, connecting telomeres of all separating anaphase chromosome pairs, lose elasticity when they lengthen during anaphase. Treatment with phosphatase inhibitor CalyculinA causes anaphase chromosomes to move backwards after they reach the poles, suggesting that dephosphorylation causes loss of tether elasticity. We added 50nM CalyculinA to living anaphase crane-fly spermatocytes with different length tethers. When tethers were short, almost all partner chromosomes moved backwards after nearing the poles. When tethers were longer, fewer chromosomes moved backwards. With yet longer tethers none moved backward. This is consistent with tether elasticity being lost by dephosphorylation. 50nM CalyculinA blocks both PP1 and PP2A. To distinguish between PP1 and PP2A we treated cells with short tethers with 50nM okadaic acid which blocks solely PP2A, or with 1µM okadaic acid which blocks both PP1 and PP2A. Only 1µM okadaic acid caused chromosomes to move backward. Thus, tether elasticity is lost because of dephosphorylation by PP1.


Sujet(s)
Anaphase/physiologie , Chromosomes/métabolisme , Diptera/génétique , Élasticité , Télomère/métabolisme , Anaphase/effets des médicaments et des substances chimiques , Anaphase/génétique , Animaux , Chromosomes/effets des médicaments et des substances chimiques , Chromosomes/génétique , Diptera/cytologie , Diptera/effets des médicaments et des substances chimiques , Élasticité/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Toxines de la flore et de la faune marines/pharmacologie , Oxazoles/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/génétique , Télomère/effets des médicaments et des substances chimiques , Télomère/génétique
9.
Cell Cycle ; 18(20): 2770-2783, 2019 Oct.
Article de Anglais | MEDLINE | ID: mdl-31448675

RÉSUMÉ

Chromosomal instability (CIN) causes structural and numerical chromosome aberrations and represents a hallmark of cancer. Replication stress (RS) has emerged as a driver for structural chromosome aberrations while mitotic defects can cause whole chromosome missegregation and aneuploidy. Recently, first evidence indicated that RS can also influence chromosome segregation in cancer cells exhibiting CIN, but the underlying mechanisms remain unknown. Here, we show that chromosomally unstable cancer cells suffer from very mild RS, which allows efficient proliferation and which can be mimicked by treatment with very low concentrations of aphidicolin. Both, endogenous RS and aphidicolin-induced very mild RS cause chromosome missegregation during mitosis leading to the induction of aneuploidy. Moreover, RS triggers an increase in microtubule plus end growth rates in mitosis, an abnormality previously identified to cause chromosome missegregation in cancer cells. In fact, RS-induced chromosome missegregation is mediated by increased mitotic microtubule growth rates and is suppressed after restoration of proper microtubule growth rates and upon rescue of replication stress. Hence, very mild and cancer-relevant RS triggers aneuploidy by deregulating microtubule dynamics in mitosis.


Sujet(s)
Aneuploïdie , Prolifération cellulaire , Ségrégation des chromosomes , Microtubules/métabolisme , Mitose , Tumeurs/génétique , Anaphase/effets des médicaments et des substances chimiques , Aphidicoline/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Instabilité des chromosomes , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Réplication de l'ADN/effets des médicaments et des substances chimiques , Humains , Microtubules/effets des médicaments et des substances chimiques , Mitose/effets des médicaments et des substances chimiques , Mitose/génétique
10.
Mol Cancer Ther ; 18(10): 1775-1786, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31358662

RÉSUMÉ

The spindle assembly checkpoint maintains genomic integrity. A key component is tyrosine threonine kinase (TTK, also known as Mps1). TTK antagonism is hypothesized to cause genomic instability and cell death. Interrogating The Cancer Genome Atlas revealed high TTK expression in lung adenocarcinomas and squamous cell cancers versus the normal lung (P < 0.001). This correlated with an unfavorable prognosis in examined lung adenocarcinoma cases (P = 0.007). TTK expression profiles in lung tumors were independently assessed by RNA in situ hybridization. CFI-402257 is a highly selective TTK inhibitor. Its potent antineoplastic effects are reported here against a panel of well-characterized murine and human lung cancer cell lines. Significant antitumorigenic activity followed independent treatments of athymic mice bearing human lung cancer xenografts (6.5 mg/kg, P < 0.05; 8.5 mg/kg, P < 0.01) and immunocompetent mice with syngeneic lung cancers (P < 0.001). CFI-402257 antineoplastic mechanisms were explored. CFI-402257 triggered aneuploidy and apoptotic death of lung cancer cells without changing centrosome number. Reverse phase protein arrays (RPPA) of vehicle versus CFI-402257-treated lung cancers were examined using more than 300 critical growth-regulatory proteins. RPPA bioinformatic analyses discovered CFI-402257 enhanced MAPK signaling, implicating MAPK antagonism in augmenting TTK inhibitory effects. This was independently confirmed using genetic and pharmacologic repression of MAPK that promoted CFI-402257 anticancer actions. TTK antagonism exerted marked antineoplastic effects against lung cancers and MAPK inhibition cooperated. Future work should determine whether CFI-402257 treatment alone or with a MAPK inhibitor is active in the lung cancer clinic.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Tumeurs du poumon/enzymologie , Tumeurs du poumon/anatomopathologie , Polyploïdie , Inhibiteurs de protéines kinases/pharmacologie , Protein-tyrosine kinases/antagonistes et inhibiteurs , Anaphase/effets des médicaments et des substances chimiques , Animaux , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Centrosome/effets des médicaments et des substances chimiques , Centrosome/métabolisme , Humains , Souris , Protein-tyrosine kinases/métabolisme , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie
11.
J Cell Biol ; 218(8): 2492-2513, 2019 08 05.
Article de Anglais | MEDLINE | ID: mdl-31270138

RÉSUMÉ

The spatial organization of the genome is enigmatic. Direct evidence of physical contacts between chromosomes and their visualization at nanoscale resolution has been limited. We used superresolution microscopy to demonstrate that ribosomal DNA (rDNA) can form linkages between chromosomes. We observed rDNA linkages in many different human cell types and demonstrated their resolution in anaphase. rDNA linkages are coated by the transcription factor UBF and their formation depends on UBF, indicating that they regularly occur between transcriptionally active loci. Overexpression of c-Myc increases rDNA transcription and the frequency of rDNA linkages, further suggesting that their formation depends on active transcription. Linkages persist in the absence of cohesion, but inhibition of topoisomerase II prevents their resolution in anaphase. We propose that linkages are topological intertwines occurring between transcriptionally active rDNA loci spatially colocated in the same nucleolar compartment. Our findings suggest that active DNA loci engage in physical interchromosomal connections that are an integral and pervasive feature of genome organization.


Sujet(s)
Chromosomes humains/métabolisme , ADN ribosomique/métabolisme , Microscopie/méthodes , Anaphase/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire , Nucléole/effets des médicaments et des substances chimiques , Nucléole/métabolisme , ADN topoisomérases de type II/métabolisme , Humains , Cellules hybrides/effets des médicaments et des substances chimiques , Cellules hybrides/métabolisme , Cellules souches pluripotentes induites/métabolisme , Souris , Complexes protéiques d'initiation à la transcription pol1/métabolisme , Polyploïdie , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-myc/métabolisme , Telomerase/métabolisme , Inhibiteurs des topoisomérases/pharmacologie
12.
Elife ; 82019 02 13.
Article de Anglais | MEDLINE | ID: mdl-30758285

RÉSUMÉ

Microtubules (MTs) are essential for cleavage furrow positioning during cytokinesis, but the mechanisms by which MT-derived signals spatially define regions of cortical contractility are unresolved. In this study cytokinesis regulators visualized in Drosophila melanogaster (Dm) cells were found to localize to and track MT plus-ends during cytokinesis. The RhoA GEF Pebble (Dm ECT2) did not evidently tip-track, but rather localized rapidly to cortical sites contacted by MT plus-tips, resulting in RhoA activation and enrichment of myosin-regulatory light chain. The MT plus-end localization of centralspindlin was compromised following EB1 depletion, which resulted in a higher incidence of cytokinesis failure. Centralspindlin plus-tip localization depended on the C-terminus and a putative EB1-interaction motif (hxxPTxh) in RacGAP50C. We propose that MT plus-end-associated centralspindlin recruits a cortical pool of Dm ECT2 upon physical contact to activate RhoA and to trigger localized contractility.


Sujet(s)
Cytocinèse , Protéines de Drosophila/métabolisme , Drosophila melanogaster/cytologie , Drosophila melanogaster/métabolisme , Microtubules/métabolisme , Transduction du signal , Protéines G rho/métabolisme , Motifs d'acides aminés , Anaphase/effets des médicaments et des substances chimiques , Animaux , Concanavaline A/pharmacologie , Cytocinèse/effets des médicaments et des substances chimiques , Protéines à fluorescence verte/métabolisme , Microtubules/effets des médicaments et des substances chimiques , Myosines/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
13.
Environ Sci Pollut Res Int ; 25(36): 36117-36123, 2018 Dec.
Article de Anglais | MEDLINE | ID: mdl-30357725

RÉSUMÉ

2-Chlorophenol (2-CP), a class of chlorinated organic pollutants like other chlorophenols, is used as intermediate in the synthesis of the higher chlorinated congeners, certain dyes, preservatives, herbicides, fungicides, and plastics. In this study, cytotoxic and genotoxic effects of 2-CP were investigated on the root meristem cells of Allium cepa for its effects on root growth, mitotic index (MI), mitotic phases, chromosomal abnormalities (CAs), and DNA damage by using Allium anaphase-telophase and Comet assays. EC50 of 2-CP value was determined as approximately 25 mg/L by Allium root growth inhibition test. Three concentrations of 2-CP (12.5, 25, and 50 mg/L), distilled water (negative control), and methyl methane sulfonate (MMS, 10 mg/L, positive control) were applied to onion stem cells under different exposure periods (24, 48, 72, and 96 h). All the applied doses of 2-CP slightly decreased MIs. 2-CP induced total CAs such as disturbed anaphase-telophase, chromosome laggards, stickiness, and bridges and also DNA damage at significant levels. These results demonstrate that 2-CP has genotoxic effects in A. cepa root meristematic cells.


Sujet(s)
Chlorophénols/toxicité , Méristème/effets des médicaments et des substances chimiques , Oignons/effets des médicaments et des substances chimiques , Oignons/génétique , Racines de plante/effets des médicaments et des substances chimiques , Anaphase/effets des médicaments et des substances chimiques , Anaphase/génétique , Aberrations des chromosomes , Test des comètes , Altération de l'ADN/effets des médicaments et des substances chimiques , Méristème/génétique , Mitose/effets des médicaments et des substances chimiques , Index mitotique , Tests de mutagénicité/méthodes , Racines de plante/génétique , Polluants du sol/toxicité
14.
Nat Commun ; 9(1): 2042, 2018 05 23.
Article de Anglais | MEDLINE | ID: mdl-29795284

RÉSUMÉ

Kinetochores are multi-protein complexes that power chromosome movements by tracking microtubules plus-ends in the mitotic spindle. Human kinetochores bind up to 20 microtubules, even though single microtubules can generate sufficient force to move chromosomes. Here, we show that high microtubule occupancy at kinetochores ensures robust chromosome segregation by providing a strong mechanical force that favours segregation of merotelic attachments during anaphase. Using low doses of the microtubules-targeting agent BAL27862 we reduce microtubule occupancy and observe that spindle morphology is unaffected and bi-oriented kinetochores can still oscillate with normal intra-kinetochore distances. Inter-kinetochore stretching is, however, dramatically reduced. The reduction in microtubule occupancy and inter-kinetochore stretching does not delay satisfaction of the spindle assembly checkpoint or induce microtubule detachment via Aurora-B kinase, which was so far thought to release microtubules from kinetochores under low stretching. Rather, partial microtubule occupancy slows down anaphase A and increases incidences of lagging chromosomes due to merotelically attached kinetochores.


Sujet(s)
Aurora kinase B/métabolisme , Ségrégation des chromosomes/physiologie , Kinétochores/métabolisme , Microtubules/métabolisme , Appareil du fuseau/métabolisme , Anaphase/effets des médicaments et des substances chimiques , Anaphase/physiologie , Benzimidazoles/pharmacologie , Lignée cellulaire , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Humains , Microscopie intravitale , Kinétochores/ultrastructure , Microscopie électronique , Microtubules/ultrastructure , Oxadiazoles/pharmacologie , Appareil du fuseau/effets des médicaments et des substances chimiques
15.
Mol Cancer Ther ; 17(4): 724-731, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29559545

RÉSUMÉ

Cancer cells often have supernumerary centrosomes that promote genomic instability, a pathognomonic feature of cancer. During mitosis, cancer cells with supernumerary centrosomes undergo bipolar cell division by clustering centrosomes into two poles. When supernumerary centrosome clustering is antagonized, cancer cells are forced to undergo multipolar division leading to death of daughter cells. This proapoptotic pathway, called anaphase catastrophe, preferentially eliminates aneuploid cancer cells and malignant tumors in engineered mouse models. Anaphase catastrophe occurs through the loss or inhibition of the centrosomal protein CP110, a direct cyclin-dependent kinase 1 (CDK1) and CDK2 target. Intriguingly, CP110 is repressed by the KRAS oncoprotein. This sensitizes KRAS-driven lung cancers (an unmet medical need) to respond to CDK2 inhibitors. Anaphase catastrophe-inducing agents like CDK1 and CDK2 antagonists are lethal to cancer cells with supernumerary centrosomes, but can relatively spare normal cells with two centrosomes. This mechanism is proposed to provide a therapeutic window in the cancer clinic following treatment with a CDK1 or CDK2 inhibitor. Taken together, anaphase catastrophe is a clinically tractable mechanism that promotes death of neoplastic tumors with aneuploidy, a hallmark of cancer. Mol Cancer Ther; 17(4); 724-31. ©2018 AACR.


Sujet(s)
Anaphase/effets des médicaments et des substances chimiques , Aneuploïdie , Antinéoplasiques/pharmacologie , Tumeurs/prévention et contrôle , Animaux , Instabilité du génome , Humains , Tumeurs/génétique , Tumeurs/métabolisme
16.
Toxicol Ind Health ; 33(10): 802-809, 2017 Oct.
Article de Anglais | MEDLINE | ID: mdl-28893155

RÉSUMÉ

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.


Sujet(s)
Nanoparticules de magnétite/toxicité , Tests de mutagénicité/méthodes , Mutagènes/toxicité , Allium/effets des médicaments et des substances chimiques , Anaphase/effets des médicaments et des substances chimiques , Animaux , Test des comètes , Altération de l'ADN/effets des médicaments et des substances chimiques , Drosophila melanogaster/effets des médicaments et des substances chimiques , Larve/effets des médicaments et des substances chimiques , Nanoparticules de magnétite/composition chimique , Mutagènes/composition chimique , Taille de particule , Télophase/effets des médicaments et des substances chimiques , Ailes d'animaux/effets des médicaments et des substances chimiques
17.
Nat Commun ; 8: 15981, 2017 07 17.
Article de Anglais | MEDLINE | ID: mdl-28714471

RÉSUMÉ

Mutations in homologous recombination (HR) genes BRCA1 and BRCA2 predispose to tumorigenesis. HR-deficient cancers are hypersensitive to Poly (ADP ribose)-polymerase (PARP) inhibitors, but can acquire resistance and relapse. Mechanistic understanding how PARP inhibition induces cytotoxicity in HR-deficient cancer cells is incomplete. Here we find PARP inhibition to compromise replication fork stability in HR-deficient cancer cells, leading to mitotic DNA damage and consequent chromatin bridges and lagging chromosomes in anaphase, frequently leading to cytokinesis failure, multinucleation and cell death. PARP-inhibitor-induced multinucleated cells fail clonogenic outgrowth, and high percentages of multinucleated cells are found in vivo in remnants of PARP inhibitor-treated Brca2-/-;p53-/- and Brca1-/-;p53-/- mammary mouse tumours, suggesting that mitotic progression promotes PARP-inhibitor-induced cell death. Indeed, enforced mitotic bypass through EMI1 depletion abrogates PARP-inhibitor-induced cytotoxicity. These findings provide insight into the cytotoxic effects of PARP inhibition, and point at combination therapies to potentiate PARP inhibitor treatment of HR-deficient tumours.


Sujet(s)
Anaphase/effets des médicaments et des substances chimiques , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Tumeurs du sein/génétique , Cytocinèse/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Tumeurs mammaires de l'animal/génétique , Tumeurs expérimentales de la mamelle/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Animaux , Tumeurs du sein/traitement médicamenteux , Lignée cellulaire tumorale , Femelle , Cellules HeLa , Humains , Souris , Souris knockout , Mitose/effets des médicaments et des substances chimiques , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Réparation de l'ADN par recombinaison/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéines suppresseurs de tumeurs/génétique
18.
J Natl Cancer Inst ; 109(6)2017 06 01.
Article de Anglais | MEDLINE | ID: mdl-28376145

RÉSUMÉ

Background: The first generation CDK2/7/9 inhibitor seliciclib (CYC202) causes multipolar anaphase and apoptosis in lung cancer cells with supernumerary centrosomes (known as anaphase catastrophe). We investigated a new and potent CDK2/9 inhibitor, CCT68127 (Cyclacel). Methods: CCT68127 was studied in lung cancer cells (three murine and five human) and control murine pulmonary epithelial and human immortalized bronchial epithelial cells. Robotic CCT68127 cell-based proliferation screens were used. Cells undergoing multipolar anaphase and inhibited centrosome clustering were scored. Reverse phase protein arrays (RPPAs) assessed CCT68127 effects on signaling pathways. The function of PEA15, a growth regulator highlighted by RPPAs, was analyzed. Syngeneic murine lung cancer xenografts (n = 4/group) determined CCT68127 effects on tumorigenicity and circulating tumor cell levels. All statistical tests were two-sided. Results: CCT68127 inhibited growth up to 88.5% (SD = 6.4%, P < .003) at 1 µM, induced apoptosis up to 42.6% (SD = 5.5%, P < .001) at 2 µM, and caused G1 or G2/M arrest in lung cancer cells with minimal effects on control cells (growth inhibition at 1 µM: 10.6%, SD = 3.6%, P = .32; apoptosis at 2 µM: 8.2%, SD = 1.0%, P = .22). A robotic screen found that lung cancer cells with KRAS mutation were particularly sensitive to CCT68127 ( P = .02 for IC 50 ). CCT68127 inhibited supernumerary centrosome clustering and caused anaphase catastrophe by 14.1% (SD = 3.6%, P < .009 at 1 µM). CCT68127 reduced PEA15 phosphorylation by 70% (SD = 3.0%, P = .003). The gain of PEA15 expression antagonized and its loss enhanced CCT68127-mediated growth inhibition. CCT68127 reduced lung cancer growth in vivo ( P < .001) and circulating tumor cells ( P = .004). Findings were confirmed with another CDK2/9 inhibitor, CYC065. Conclusions: Next-generation CDK2/9 inhibition elicits marked antineoplastic effects in lung cancer via anaphase catastrophe and reduced PEA15 phosphorylation.


Sujet(s)
Adénosine/analogues et dérivés , Anaphase/effets des médicaments et des substances chimiques , Kinase-2 cycline-dépendante/antagonistes et inhibiteurs , Protéines et peptides de signalisation intracellulaire/génétique , Tumeurs du poumon/traitement médicamenteux , Phosphoprotéines/génétique , Inhibiteurs de protéines kinases/pharmacologie , Adénosine/pharmacologie , Adénosine/usage thérapeutique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéines régulatrices de l'apoptose , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Centrosome/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Tumeurs du poumon/génétique , Mâle , Souris , Phosphoprotéines/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes p21(ras)/génétique , Purines/pharmacologie , Roscovitine , Transduction du signal/effets des médicaments et des substances chimiques
19.
J Biol Chem ; 292(5): 1910-1924, 2017 02 03.
Article de Anglais | MEDLINE | ID: mdl-28028179

RÉSUMÉ

The suppression of mitotic Aurora kinases (AURKs) by AURK inhibitors frequently causes cytokinetic failure, leading to polyploidy or aneuploidy, indicating the critical role of AURK-mediated phosphorylation during cytokinesis. We demonstrate the deregulated expression of AKT3 in Aurora kinase inhibitor (AURKi)-resistant cells, which we established from human colorectal cancer HCT 116 cells. The AKT family, which includes AKT1, -2, and -3, plays multiple roles in antiapoptotic functions and drug resistance and is involved in cell growth and survival pathways. We found that an AKT inhibitor, AZD5363, showed synergistic effect with an AURKi, VX-680, on two AKT3-expressing AURKi-resistant cell lines, and AKT3 knockdown sensitized cells to VX-680. Consistent with these activities, AKT3 expression suppressed AURKi-induced apoptosis and conferred resistance to AURKi. Thus, AKT3 expression affects cell sensitivity to AURKi. Moreover, we found that AKT3 expression suppressed AURKi-induced aneuploidy, and inversely AKT3 knockdown enhanced it. In addition, partial co-localization of AKT3 with AURKB was observed during anaphase. Overall, this study suggests that AKT3 could repress the antiproliferative effects of AURKi, with a novel activity particularly suppressing the aneuploidy induction.


Sujet(s)
Anaphase/effets des médicaments et des substances chimiques , Aurora kinase B/antagonistes et inhibiteurs , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Protéines proto-oncogènes c-akt/biosynthèse , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Aneuploïdie , Aurora kinase B/génétique , Aurora kinase B/métabolisme , Cellules HEK293 , Cellules HeLa , Humains , Cellules MCF-7 , Protéines proto-oncogènes c-akt/génétique
20.
Cell Rep ; 17(7): 1728-1738, 2016 11 08.
Article de Anglais | MEDLINE | ID: mdl-27829144

RÉSUMÉ

To achieve chromosome segregation during mitosis, sister chromatids must undergo a dramatic change in their behavior to switch from balanced oscillations at the metaphase plate to directed poleward motion during anaphase. However, the factors that alter chromosome behavior at the metaphase-to-anaphase transition remain incompletely understood. Here, we perform time-lapse imaging to analyze anaphase chromosome dynamics in human cells. Using multiple directed biochemical, genetic, and physical perturbations, our results demonstrate that differences in the global phosphorylation states between metaphase and anaphase are the major determinant of chromosome motion dynamics. Indeed, causing a mitotic phosphorylation state to persist into anaphase produces dramatic metaphase-like oscillations. These induced oscillations depend on both kinetochore-derived and polar ejection forces that oppose poleward motion. Thus, our analysis of anaphase chromosome motion reveals that dephosphorylation of multiple mitotic substrates is required to suppress metaphase chromosome oscillatory motions and achieve directed poleward motion for successful chromosome segregation.


Sujet(s)
Anaphase , Chromosomes humains/métabolisme , Métaphase , Anaphase/effets des médicaments et des substances chimiques , Chromatides/métabolisme , Cellules HeLa , Humains , Kinétochores/effets des médicaments et des substances chimiques , Kinétochores/métabolisme , Métaphase/effets des médicaments et des substances chimiques , Modèles biologiques , Mouvement , Acide okadaïque/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE