Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Clin Pharmacol Ther ; 113(2): 349-359, 2023 02.
Article in English | MEDLINE | ID: mdl-36326573

ABSTRACT

Important discoveries by academic drug developers hold the promise of bringing innovative treatments that address unmet medical needs to the market. However, the drug development process has proved to be challenging and demanding for academic researchers, and regulatory challenges are an important barrier to implementing academic findings in clinical practice. European regulators offer varying degrees of support services to help drug developers meet regulatory standards and requirements. "Strengthening Training of Academia in Regulatory Sciences and Supporting Regulatory Scientific Advice" (STARS) is a European Commission-funded consortium aiming to strengthen the training of academics in regulatory science and requirements. Here, we report the results of four surveys that investigated the awareness and utilization of support tools offered by European regulators and identified the regulatory challenges and support needs of researchers. The surveys targeted four main European stakeholders in academic medicines research: academic research groups (706 respondents), academic research centers (99), funding organizations (49), and regulators (22). The results show that while European regulators provide various regulatory support tools, less than half of the responding academic researchers were aware of these tools and many experienced challenges in reaching a sufficient level of regulatory knowledge. There was a general lack of understanding of the regulatory environment that was aggravated by poor communication between stakeholders. The results of this study form a foundation for an improved European medicines regulatory network, in which regulatory challenges faced by academia are tackled.


Subject(s)
Drug Discovery , Drug and Narcotic Control , Humans , Europe , Surveys and Questionnaires
3.
Oncoscience ; 8: 134-153, 2021.
Article in English | MEDLINE | ID: mdl-34926718

ABSTRACT

Hec1 (Highly expressed in cancer 1) resides in the outer kinetochore where it works to facilitate proper kinetochore-microtubule interactions during mitosis. Hec1 is overexpressed in various cancers and its expression shows correlation with high tumour grade and poor patient prognosis. Chemical perturbation of Hec1 is anticipated to impair kinetochore-microtubule binding, activate the spindle assembly checkpoint (spindle checkpoint) and thereby suppress cell proliferation. In this study, we performed high-throughput screen to identify novel small molecules that target the Hec1 calponin homology domain (CHD), which is needed for normal microtubule attachments. 4 million compounds were first virtually fitted against the CHD, and the best hit molecules were evaluated in vitro. These approaches led to the identification of VTT-006, a 1,2-disubstituted-tetrahydro-beta-carboline derivative, which showed binding to recombinant Ndc80 complex and modulated Hec1 association with microtubules in vitro. VTT-006 treatment resulted in chromosome congression defects, reduced chromosome oscillations and induced loss of inter-kinetochore tension. Cells remained arrested in mitosis with an active spindle checkpoint for several hours before undergoing cell death. VTT-006 suppressed the growth of several cancer cell lines and enhanced the sensitivity of HeLa cells to Taxol. Our findings propose that VTT-006 is a potential anti-mitotic compound that disrupts M phase, impairs kinetochore-microtubule interactions, and activates the spindle checkpoint.

4.
Transl Oncol ; 12(1): 170-179, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30359947

ABSTRACT

Taxanes are chemotherapeutic agents used in the treatment of solid tumors, particularly of breast, ovarian, and lung origin. However, patients show divergent therapy responses, and the molecular determinants of taxane sensitivity have remained elusive. Especially the signaling pathways that promote death of the taxane-treated cells are poorly characterized. Here we describe a novel part of a signaling route in which c-Myc enhances paclitaxel sensitivity through upregulation of miR-203b-3p and miR-203a-3p; two clustered antiapoptosis protein Bcl-xL controlling microRNAs. In vitro, the miR-203b-3p decreases the expression of Bcl-xL by direct targeting of the gene's mRNA 3'UTR. Notably, overexpression of the miR-203b-3p changed the fate of paclitaxel-treated breast and ovarian cancer cells from mitotic slippage to cell death. In breast tumors, high expression of the miR-203b-3p and MYC was associated with better therapy response and patient survival. Interestingly, in the breast tumors, MYC expression correlated negatively with BCL2L1 expression but positively with miR-203b-3p and miR-203a-3p. Finally, silencing of MYC suppressed the transcription of both miRNAs in breast tumor cells. Pending further validation, these results may assist in patient stratification for taxane therapy.

5.
Drug Des Devel Ther ; 11: 1335-1351, 2017.
Article in English | MEDLINE | ID: mdl-28496304

ABSTRACT

PURPOSE: Overall, ~65% of patients diagnosed with advanced ovarian cancer (OC) will relapse after primary surgery and adjuvant first-line platinum- and taxane-based chemotherapy. Significant improvements in the treatment of OC are expected from the development of novel compounds having combined cytotoxic and antiangiogenic properties that make them effective on refractory tumors. METHODS: Permeability of NOV202 was determined with Caco-2 monolayer assay. The compound's pharmacokinetic profile and plasma:brain distribution were assessed in male C57Bl/6 mice. The compound's impacts on tubulin, microtubules and cell cycle were investigated by using in vitro tubulin polymerization assay, cell-based immunofluorescence and live cell microscopy. The IC50 concentrations of NOV202 were assessed in a panel of eight cancer cell lines. Impact of the compound on vascular tube formation was determined using the StemKit and Chick chorioallantoic membrane assays. The in vivo efficacy of the compound was analyzed with an OC xenograft mouse model. RESULTS: NOV202 was found to suppress cancer cell proliferation at low nanomolar concentrations (IC50 2.3-12.0 nM) and showed equal efficacy between OC cell line A2780 (IC50 2.4 nM) and its multidrug-resistant subline A2780/Adr (IC50 2.3 nM). Mechanistically, NOV202 targeted tubulin polymerization in vitro in a dose-dependent manner and in cells induced an M phase arrest. In vivo, NOV202 caused a dose-dependent reduction of tumor mass in an A2780 xenograft model, which at the highest dose (40 mg/kg) was comparable to the effect of paclitaxel (24 mg/kg). Interestingly, NOV202 exhibited vascular disrupting properties that were similar to the effects of Combretastatin A4. CONCLUSION: NOV202 is a novel tubulin and vascular targeting agent that shows strong anticancer efficacy in cells and OC xenograft models. The finding that the compound induced significantly more cell death in Pgp/MDR1 overexpressing OC cells compared to vincristine and paclitaxel warrants further development of the compound as a new therapy for OC patients with treatment refractory tumors and/or relapsing disease.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Br J Cancer ; 116(11): 1451-1461, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28449010

ABSTRACT

BACKGROUND: Several microRNA (miRNA) molecules have emerged as important post-transcriptional regulators of tumour suppressor and oncogene expression. Ras association domain family member 1 (RASSF1) is a critical tumour suppressor that controls multiple aspects of cell proliferation such as cell cycle, cell division and apoptosis. The expression of RASSF1 is lost in a variety of cancers due to the promoter hypermethylation. METHODS: miR-193a-3p was identified as a RASSF1-targeting miRNA by a dual screening approach. In cultured human cancer cells, immunoblotting, qRT-PCR, luciferase reporter assays, time-lapse microscopy and immunofluorescence methods were used to study the effects of excess miR-193a-3p on RASSF1 expression and cell division. RESULTS: Here, we report a new miRNA-mediated mechanism that regulates RASSF1 expression: miR-193a-3p binds directly to RASSF1-3'UTR and represses the mRNA and protein expression. In human cancer cells, excess of miR-193a-3p causes polyploidy through impairment of the Rassf1-Syntaxin 16 signalling pathway that is needed for completion of cytokinesis. In the next cell cycle the miR-193a-3p-overexpressing cells exhibit multipolar mitotic spindles, mitotic delay and elevated frequency of cell death. CONCLUSIONS: Our results suggest that besides epigenetic regulation, altered expression of specific miRNAs may contribute to the loss of Rassf1 in cancer cells and cause cell division errors.


Subject(s)
Cell Division/genetics , MicroRNAs/genetics , RNA, Messenger/metabolism , Tumor Suppressor Proteins/genetics , 3' Untranslated Regions , Cell Death/genetics , Cell Polarity/genetics , Cytokinesis/genetics , Down-Regulation , Gene Expression , Gene Expression Regulation, Neoplastic , HCT116 Cells , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/genetics , Signal Transduction/genetics , Syntaxin 16/metabolism , Transfection , Tumor Suppressor Proteins/metabolism
7.
Oncotarget ; 7(11): 12267-85, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26943585

ABSTRACT

The molecular pathways that contribute to the proliferation and drug response of cancer cells are highly complex and currently insufficiently characterized. We have identified a previously unknown microRNA-based mechanism that provides cancer cells means to stimulate tumorigenesis via increased genomic instability and, at the same time, evade the action of clinically utilized microtubule drugs. We demonstrate miR-493-3p to be a novel negative regulator of mitotic arrest deficient-2 (MAD2), an essential component of the spindle assembly checkpoint that monitors the fidelity of chromosome segregation. The microRNA targets the 3' UTR of Mad2 mRNA thereby preventing translation of the Mad2 protein. In cancer cells, overexpression of miR-493-3p induced a premature mitotic exit that led to increased frequency of aneuploidy and cellular senescence in the progeny cells. Importantly, excess of the miR-493-3p conferred resistance of cancer cells to microtubule drugs. In human neoplasms, miR-493-3p and Mad2 expression alterations correlated with advanced ovarian cancer forms and high miR-493-3p levels were associated with reduced survival of ovarian and breast cancer patients with aggressive tumors, especially in the paclitaxel therapy arm. Our results suggest that intratumoral profiling of miR-493-3p and Mad2 levels can have diagnostic value in predicting the efficacy of taxane chemotherapy.


Subject(s)
M Phase Cell Cycle Checkpoints/drug effects , Mad2 Proteins/metabolism , MicroRNAs/metabolism , Neoplasms/drug therapy , Paclitaxel/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chromosome Segregation , Female , HCT116 Cells , HeLa Cells , Humans , Mad2 Proteins/genetics , MicroRNAs/genetics , Mitosis/drug effects , Mitosis/physiology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Randomized Controlled Trials as Topic , Transfection
8.
J Cell Biol ; 206(6): 735-49, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25202032

ABSTRACT

Unless mitigated, external and physiological stresses are detrimental for cells, especially in mitosis, resulting in chromosomal missegregation, aneuploidy, or apoptosis. Heat shock proteins (Hsps) maintain protein homeostasis and promote cell survival. Hsps are transcriptionally regulated by heat shock factors (HSFs). Of these, HSF1 is the master regulator and HSF2 modulates Hsp expression by interacting with HSF1. Due to global inhibition of transcription in mitosis, including HSF1-mediated expression of Hsps, mitotic cells are highly vulnerable to stress. Here, we show that cells can counteract transcriptional silencing and protect themselves against proteotoxicity in mitosis. We found that the condensed chromatin of HSF2-deficient cells is accessible for HSF1 and RNA polymerase II, allowing stress-inducible Hsp expression. Consequently, HSF2-deficient cells exposed to acute stress display diminished mitotic errors and have a survival advantage. We also show that HSF2 expression declines during mitosis in several but not all human cell lines, which corresponds to the Hsp70 induction and protection against stress-induced mitotic abnormalities and apoptosis.


Subject(s)
DNA-Binding Proteins/genetics , Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Mitosis/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Survival , Chromatin/genetics , Gene Expression Regulation , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Proteins/biosynthesis , Humans , MCF-7 Cells , Mice , Mitotic Index , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Transcription Factors/biosynthesis , Transcription, Genetic
9.
Mol Cancer Ther ; 13(5): 1054-66, 2014 May.
Article in English | MEDLINE | ID: mdl-24748653

ABSTRACT

Mitosis is an attractive target for the development of new anticancer drugs. In a search for novel mitotic inhibitors, we virtually screened for low molecular weight compounds that would possess similar steric and electrostatic features, but different chemical structure than rigosertib (ON 01910.Na), a putative inhibitor of phosphoinositide 3-kinase (PI3K) and polo-like kinase 1 (Plk1) pathways. Highest scoring hit compounds were tested in cell-based assays for their ability to induce mitotic arrest. We identified a novel acridinyl-acetohydrazide, here named as Centmitor-1 (Cent-1), that possesses highly similar molecular interaction field as rigosertib. In cells, Cent-1 phenocopied the cellular effects of rigosertib and caused mitotic arrest characterized by chromosome alignment defects, multipolar spindles, centrosome fragmentation, and activated spindle assembly checkpoint. We compared the effects of Cent-1 and rigosertib on microtubules and found that both compounds modulated microtubule plus-ends and reduced microtubule dynamics. Also, mitotic spindle forces were affected by the compounds as tension across sister kinetochores was reduced in mitotic cells. Our results showed that both Cent-1 and rigosertib target processes that occur during mitosis as they had immediate antimitotic effects when added to cells during mitosis. Analysis of Plk1 activity in cells using a Förster resonance energy transfer (FRET)-based assay indicated that neither compound affected the activity of the kinase. Taken together, these findings suggest that Cent-1 and rigosertib elicit their antimitotic effects by targeting mitotic processes without impairment of Plk1 kinase activity.


Subject(s)
Acridones/pharmacology , Antimitotic Agents/pharmacology , Glycine/analogs & derivatives , Hydrazines/pharmacology , Sulfones/pharmacology , Acridones/chemistry , Antimitotic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Centrosome/metabolism , Drug Screening Assays, Antitumor , Glycine/chemistry , Glycine/pharmacology , HeLa Cells , High-Throughput Screening Assays , Humans , Hydrazines/chemistry , Microtubules/metabolism , Mitosis/drug effects , Molecular Structure , Molecular Weight , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Sulfones/chemistry , Polo-Like Kinase 1
10.
Chromosoma ; 122(5): 431-49, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23775312

ABSTRACT

Suppression of cell proliferation by targeting mitosis is one potential cancer intervention. A number of existing chemotherapy drugs disrupt mitosis by targeting microtubule dynamics. While efficacious, these drugs have limitations, i.e. neuropathy, unpredictability and development of resistance. In order to overcome these issues, a great deal of effort has been spent exploring novel mitotic targets including Polo-like kinase 1, Aurora kinases, Mps1, Cenp-E and KSP/Eg5. Here we summarize the latest developments in the discovery and clinical evaluation of new mitotic drug targets.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Mitosis/genetics , Molecular Targeted Therapy , Aurora Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Humans , Kinesins/metabolism , Mitosis/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
11.
Exp Cell Res ; 318(5): 578-92, 2012 Mar 10.
Article in English | MEDLINE | ID: mdl-22227008

ABSTRACT

The spindle assembly checkpoint (SAC) is a conserved mechanism that ensures the fidelity of chromosome distribution in mitosis by preventing anaphase onset until the correct bipolar microtubule-kinetochore attachments are formed. Errors in SAC function may contribute to tumorigenesis by inducing numerical chromosome anomalies (aneuploidy). On the other hand, total disruption of SAC can lead to massive genomic imbalance followed by cell death, a phenomena that has therapeutic potency. We performed a cell-based high-throughput screen with a compound library of 2000 bioactives for novel SAC inhibitors and discovered a plant-derived phenolic compound eupatorin (3',5-dihydroxy-4',6,7-trimethoxyflavone) as an anti-mitotic flavonoid. The premature override of the microtubule drug-imposed mitotic arrest by eupatorin is dependent on microtubule-kinetochore attachments but not interkinetochore tension. Aurora B kinase activity, which is essential for maintenance of normal SAC signaling, is diminished by eupatorin in cells and in vitro providing a mechanistic explanation for the observed forced mitotic exit. Eupatorin likely has additional targets since eupatorin treatment of pre-mitotic cells causes spindle anomalies triggering a transient M phase delay followed by impaired cytokinesis and polyploidy. Finally, eupatorin potently induces apoptosis in multiple cancer cell lines and suppresses cancer cell proliferation in organotypic 3D cell culture model.


Subject(s)
Antimitotic Agents/pharmacology , Apoptosis/drug effects , Flavonoids/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Polyploidy , Aurora Kinase B , Aurora Kinases , Cell Culture Techniques , Cell Proliferation/drug effects , Centrosome/metabolism , HeLa Cells , High-Throughput Screening Assays , Humans , Leupeptins/pharmacology , Male , Microscopy, Fluorescence , Nocodazole/pharmacology , Prostatic Neoplasms , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Thiones/pharmacology , Time-Lapse Imaging
12.
Mol Oncol ; 5(1): 105-11, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21051298

ABSTRACT

Cell division is orchestrated by a complex protein network that aims to maintenance of genomic stability. Visualisation of mitotic protein-protein associations in space and time has been limited due to the lack of proper biochemical and easy-to-use imaging tools. Here we report adaptation of the in situ proximity ligation assay (is-PLA) to study mitotic protein interactions with spatio-temporal resolution. We examined the composition of the Chromosomal Passenger Complex (CPC) at various mitotic phases and after chemical treatments using is-PLA with antibodies against the core CPC subunits Aurora B, INCENP, Survivin and Borealin. Our results support the notion that the core CPC functions as a single structural unit at centromeres in early mitosis and at central spindle after the onset of anaphase. Treatment of cells with the Aurora B inhibitor ZM447439 diminished the is-PLA signals at centromeres suggesting that Aurora B activity contributes to structural maintenance and/or proper subcellular localization of the core CPC. Is-PLA-based analysis of interaction between INCENP and Polo-like kinase 1 (Plk1) proposes that the kinase co-travels with CPC during late mitosis. The data illustrates both the strengths and limitations of the is-PLA in the analysis of mitotic macromolecule associations at sub-organelle level.


Subject(s)
Chromosomes, Human , Humans
13.
J Cell Sci ; 124(Pt 2): 216-27, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21172807

ABSTRACT

The p38 mitogen-activated protein kinase (p38 MAPK) family, which is comprised of four protein isoforms, p38α, p38ß, p38γ and p38δ, forms one of the key MAPK pathways. The p38 MAPKs are implicated in many cellular processes including inflammation, differentiation, cell growth, cell cycle and cell death. The function of p38 MAPKs in mitotic entry has been well established, but their role in mitotic progression has remained controversial. We identify p38γ MAPK as a modulator of mitotic progression and mitotic cell death. In HeLa cells, loss of p38γ results in multipolar spindle formation and chromosome misalignment, which induce a transient M phase arrest. The majority of p38γ-depleted cells die at mitotic arrest or soon after abnormal exit from M-phase. We show that p38 MAPKs are activated at the kinetochores and spindle poles throughout mitosis by kinase(s) that are stably bound to these structures. Finally, p38γ is required for the normal kinetochore localization of polo-like kinase 1 (Plk1), and this contributes to the activity of the p38 MAPK pathway. Our data suggest a link between mitotic regulation and the p38 MAPK pathway, in which p38γ prevents chromosomal instability and supports mitotic cell viability.


Subject(s)
Cells/cytology , Cells/enzymology , Mitogen-Activated Protein Kinase 12/deficiency , Mitosis , Cell Death , Cell Line , Cell Survival , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 12/genetics , Spindle Apparatus/enzymology
14.
Carcinogenesis ; 32(3): 305-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163887

ABSTRACT

Frameshift mutations frequently accumulate in microsatellite-unstable colorectal cancers (MSI CRCs) typically leading to downregulation of the target genes due to nonsense-mediated messenger RNA decay. However, frameshift mutations that occur in the 3' end of the coding regions can escape decay, which has largely been ignored in previous works. In this study, we characterized nonsense-mediated decay-escaping frameshift mutations in MSI CRC in an unbiased, genome wide manner. Combining bioinformatic search with expression profiling, we identified genes that were predicted to escape decay after a deletion in a microsatellite repeat. These repeats, located in 258 genes, were initially sequenced in 30 MSI CRC samples. The mitotic checkpoint kinase TTK was found to harbor decay-escaping heterozygous mutations in exon 22 in 59% (105/179) of MSI CRCs, which is notably more than previously reported. Additional novel deletions were found in exon 5, raising the mutation frequency to 66%. The exon 22 of TTK contains an A(9)-G(4)-A(7) locus, in which the most common mutation was a mononucleotide deletion in the A(9) (c.2560delA). When compared with identical non-coding repeats, TTK was found to be mutated significantly more often than expected without selective advantage. Since TTK inhibition is known to induce override of the mitotic spindle assembly checkpoint (SAC), we challenged mutated cancer cells with the microtubule-stabilizing drug paclitaxel. No evidence of checkpoint weakening was observed. As a conclusion, heterozygous TTK mutations occur at a high frequency in MSI CRCs. Unexpectedly, the plausible selective advantage in tumourigenesis does not appear to be related to SAC.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Frameshift Mutation/genetics , Microsatellite Instability , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus , Adenocarcinoma/pathology , Aged , Blotting, Western , Colorectal Neoplasms/pathology , Computational Biology , DNA, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Male , Microsatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein-Tyrosine Kinases , Tumor Cells, Cultured
15.
Carcinogenesis ; 30(6): 1032-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19395653

ABSTRACT

Fisetin is a natural flavonol present in edible vegetables, fruits and wine at 2-160 microg/g concentrations and an ingredient in nutritional supplements with much higher concentrations. The compound has been reported to exert anticarcinogenic effects as well as antioxidant and anti-inflammatory activity via its ability to act as an inhibitor of cell proliferation and free radical scavenger, respectively. Our cell-based high-throughput screen for small molecules that override chemically induced mitotic arrest identified fisetin as an antimitotic compound. Fisetin rapidly compromised microtubule drug-induced mitotic block in a proteasome-dependent manner in several human cell lines. Moreover, in unperturbed human cancer cells fisetin caused premature initiation of chromosome segregation and exit from mitosis without normal cytokinesis. To understand the molecular mechanism behind these mitotic errors, we analyzed the consequences of fisetin treatment on the localization and phoshorylation of several mitotic proteins. Aurora B, Bub1, BubR1 and Cenp-F rapidly lost their kinetochore/centromere localization and others became dephosphorylated upon addition of fisetin to the culture medium. Finally, we identified Aurora B kinase as a novel direct target of fisetin. The activity of Aurora B was significantly reduced by fisetin in vitro and in cells, an effect that can explain the observed forced mitotic exit, failure of cytokinesis and decreased cell viability. In conclusion, our data propose that fisetin perturbs spindle checkpoint signaling, which may contribute to the antiproliferative effects of the compound.


Subject(s)
Flavonoids/pharmacology , Mitosis/drug effects , Spindle Apparatus/metabolism , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Enzyme Activation , Flavonols , Humans , Kinetochores/drug effects , Kinetochores/physiology , Microfilament Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/drug effects
16.
Mol Oncol ; 3(3): 262-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19393581

ABSTRACT

Genomic stability depends on the normal function of the kinetochore, a multi-protein assemblage, which consists of over 80 molecules including both constitutive and transiently binding components. Information regarding the spatial-temporal assembly of kinetochore subcomplexes is often limited by technical difficulties in their isolation. To study kinetochore subcomplex formation, we targeted separately Hec1 and Spc24, two subunits of the Ndc80 kinetochore compilation, to the plasma membrane by fusing them with the amino-terminal palmitoylation and myristoylation (pm) sequence of the receptor tyrosine kinase Fyn. We found that in early mitotic cells, pm-GFP-Hec1 and pm-GFP-Spc24 fusion proteins localised to the plasma membrane and were able to recruit all subunits of the Ndc80 complex (Ndc80/Hec1, Nuf2, Spc24 and Spc25) to these foci. In interphase cells, only Hec1-Nuf2 and Spc24-Spc25 heterodimers accumulated to the plasma membrane foci. The results propose that the assembly of Ndc80 tetramer can take place outside of the kinetochore but requires co-factors that are only present in mitotic cells. These findings provide the first experimental evidence on the successful employment of the plasma membrane targeting technique in the study of kinetochore biochemistry.


Subject(s)
Cell Membrane/metabolism , Gene Expression , Kinetochores/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/biosynthesis , Protein Multimerization , Cell Membrane/genetics , Cytoskeletal Proteins , Genomic Instability/genetics , HeLa Cells , Humans , Lipoylation/genetics , Mitosis/genetics , Multiprotein Complexes/genetics , Nuclear Proteins/genetics
17.
Chromosoma ; 118(1): 71-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18784935

ABSTRACT

Incenp is an essential mitotic protein that, together with Aurora B, Survivin, and Borealin, forms the core of the chromosomal passenger protein complex (CPC). The CPC regulates various mitotic processes and functions to maintain genomic stability. The proper subcellular localization of the CPC and its full catalytic activity require the presence of each core subunit in the complex. We have investigated the mitotic tasks of the CPC using a function blocking antibody against Incenp microinjected into cells at different mitotic phases. This method allowed temporal analysis of CPC functions without perturbation of complex assembly or activity prior to injection. We have also studied the dynamic properties of Incenp and Aurora B using fusion protein photobleaching. We found that in early mitotic cells, Incenp and Aurora B exhibit dynamic turnover at centromeres, which is prevented by the anti-Incenp antibody. In these cells, the loss of centromeric CPC turnover is accompanied by forced mitotic exit without the execution of cytokinesis. Introduction of anti-Incenp antibody into early anaphase cells causes abnormalities in sister chromatid separation through defects in anaphase spindle functions. In summary, our data uncovers new mitotic roles for the CPC in anaphase and proposes that CPC turnover at centromeres modulates spindle assembly checkpoint signaling.


Subject(s)
Anaphase/physiology , Centromere/metabolism , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Xenopus Proteins/metabolism , Animals , Aurora Kinase B , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Plasmids , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Synaptonemal Complex/metabolism , Xenopus , Xenopus Proteins/genetics
18.
Chromosoma ; 117(2): 169-79, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18057949

ABSTRACT

Kinetochores bind microtubules laterally in a transient fashion and stably, by insertion of plus ends. These pathways may exist to carry out distinct tasks during different stages of mitosis and likely depend on distinct molecular mechanisms. On isolated chromosomes, we found microtubule nucleation/binding depended additively on both dynein/dynactin and on the Ndc80/Hec1 complex. Studying chromosome movement in living Xenopus cells within the simplified geometry of monopolar spindles, we quantified the relative contributions of dynein/dynactin and the Ndc80/Hec1 complex. Inhibition of dynein/dynactin alone had minor effects but did suppress transient, rapid, poleward movements. In contrast, inhibition of the Ndc80 complex blocked normal end-on attachments of microtubules to kinetochores resulting in persistent rapid poleward movements that required dynein/dynactin. In normal cells with bipolar spindles, dynein/dynactin activity on its own allowed attachment and rapid movement of chromosomes on prometaphase spindles but failed to support metaphase alignment and chromatid movement in anaphase. Thus, in prometaphase, dynein/dynactin likely mediates early transient, lateral interactions of kinetochores and microtubules. However, mature attachment via the Ndc80 complex is essential for metaphase alignment and anaphase A.


Subject(s)
Anaphase , Chromosomes/ultrastructure , Nuclear Proteins/physiology , Animals , Cell Cycle , Cell Cycle Proteins , Chromosomes/metabolism , Cytoskeletal Proteins , Dynactin Complex , Dyneins/metabolism , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , Models, Biological , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Spindle Apparatus , Transfection , Tubulin/metabolism , Xenopus laevis
19.
Cell Cycle ; 6(13): 1579-85, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17617734

ABSTRACT

Physical connection between the sister chromatids is mediated by the cohesin protein complex. During prophase, cohesin is removed from the chromosome arms while the centromeres remain united. Shugoshin1 (Sgo1) is required for maintenance of centromeric cohesion from prophase to the metaphase-anaphase transition. Furthermore, Sgo1 has been proposed to regulate kinetochore microtubule stability and sense interkinetochore tension, two tasks which are tightly coupled with the function of the Chromosomal Passenger Complex (CPC) and Polo-like kinase 1 (Plk1). Here we show that depletion or chemical inhibition of Aurora B kinase (AurB), the catalytic subunit of the CPC, disrupts accumulation of Sgo1 on the kinetochores in HeLa cells and causes Sgo1 to localize on the chromosome arms. RNAi assays show that depletion of Sgo1 did not affect AurB localization but diminished Plk1 kinetochore binding. Furthermore, we demonstrate that vertebrate Sgo1 is phosphorylated by both AurB and Plk1 in vitro. The data presented here includes an extensive analysis of kinetochore targeting interdependencies of mitotic proteins that propose a novel branch in kinetochore assembly where Sgo1 and Plk1 have central roles. Furthermore our studies implicate Sgo1 in the tension sensing mechanism of the spindle checkpoint by regulating Plk1 kinetochore affinity.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Kinetochores/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Aurora Kinase B , Aurora Kinases , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Humans , Microfilament Proteins/metabolism , Models, Biological , Phosphorylation , Protein Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Protein Transport , Spindle Apparatus/metabolism , Polo-Like Kinase 1
20.
Exp Cell Res ; 312(18): 3459-70, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16962097

ABSTRACT

In somatic cells, integrity of cell division is safeguarded by the spindle checkpoint, a signaling cascade that delays the separation of sister chromatids in the presence of misaligned chromosomes. Aurora kinases play important roles in this process by promoting centrosome maturation, chromosome bi-orientation, spindle checkpoint signaling, and cytokinesis. To investigate the functions of Aurora kinases in male meiosis, we applied a small molecule Aurora inhibitor, ZM447439, to seminiferous tubules in vitro. Primary and secondary spermatocytes exposed to ZM447439 exhibit defects in the spindle morphology and fail to align their chromosomes at the metaphase plate. Moreover, the treated spermatocytes undergo a forced exit from the meiotic M-phase without cytokinesis. These results suggest that the activities of Aurora kinases are required for normal spindle assembly as well as for establishment and maintenance of proper microtubule-kinetochore attachments and spindle checkpoint signaling in male mammalian meiosis.


Subject(s)
Benzamides/pharmacology , Chromosomes/metabolism , Meiosis/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Signal Transduction/physiology , Spermatocytes , Spindle Apparatus/metabolism , Animals , Apoptosis/physiology , Aurora Kinases , Benzamides/metabolism , Cell Shape , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Genes, cdc , Leupeptins/pharmacology , Male , Protein Serine-Threonine Kinases/metabolism , Quinazolines/metabolism , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/cytology , Spermatocytes/cytology , Spermatocytes/drug effects , Spermatocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...