Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Gynecol Oncol ; 128(3): 461-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23146687

ABSTRACT

OBJECTIVE: Endometrial tumors with non-functional p53, such as serous uterine endometrial carcinomas, are aggressive malignancies with a poor outcome, yet they have an Achilles' heel: due to loss of p53 function, these tumors may be sensitive to treatments which abrogate the G2/M checkpoint. Our objective was to exploit this weakness to induce mitotic cell death using two strategies: (1) EGFR inhibitor gefitinib combined with paclitaxel to arrest cells at mitosis, or (2) BI2536, an inhibitor of polo-like kinase 1 (PLK1), to block PLK1 activity. METHODS: We examined the impact of combining gefitinib and paclitaxel or PLK1 inhibitor on expression of G2/M checkpoint controllers, cell viability, and cell cycle progression in endometrial cancer cells with mutant p53. RESULTS: In cells lacking normal p53 activity, each treatment activated CDC25C and inactivated Wee1, which in turn activated cdc2 and sent cells rapidly through the G2/M checkpoint and into mitosis. Live cell imaging demonstrated irreversible mitotic arrest and eventual cell death. Combinatorial therapy with paclitaxel and gefitinib was highly synergistic and resulted in a 10-fold reduction in the IC50 for paclitaxel, from 14nM as a single agent to 1.3nM in the presence of gefitinib. However, BI2536 alone at low concentrations (5nM) was the most effective treatment and resulted in massive mitotic cell death. In a xenograft mouse model with p53-deficient cells, low dose BI2536 significantly inhibited tumor growth. CONCLUSIONS: These findings reveal induction of mitotic cell death as a therapeutic strategy for endometrial tumors lacking functional p53.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Checkpoints/drug effects , Endometrial Neoplasms/drug therapy , Mitosis/drug effects , Paclitaxel/pharmacology , Pteridines/pharmacology , Quinazolines/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gefitinib , Humans , M Phase Cell Cycle Checkpoints/drug effects , Mice , Mice, Nude , Mitosis/genetics , Paclitaxel/administration & dosage , Pteridines/administration & dosage , Quinazolines/administration & dosage , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
2.
Cell Biol Int ; 35(7): 687-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21250945

ABSTRACT

'Neosis' describes the process whereby p53 function-deficient tumour cells undergo self-renewal after genotoxic damage apparently via senescing ETCs (endopolyploid tumour cells). We previously reported that autophagic digestion and extrusion of DNA occurs in ETC and subsequently revealed that self-renewal transcription factors are also activated under these conditions. Here, we further studied this phenomenon in a range of cell lines after genotoxic damage induced by gamma irradiation, ETO (etoposide) or PXT (paclitaxel) treatment. These experiments revealed that chromatin degradation by autophagy was compatible with continuing mitotic activity in ETC. While the actively polyploidizing primary ETC produced early after genotoxic insult activated self-renewal factors throughout the polygenome, the secondary ETC restored after failed multipolar mitosis underwent subnuclei differentiation. As such, only a subset of subnuclei continued to express OCT4 and NANOG, while those lacking these factors stopped DNA replication and underwent degradation and elimination through autophagy. The surviving subnuclei sequestered nascent cytoplasm to form subcells, while being retained within the confines of the old ETC. Finally, the preformed paradiploid subcells became released from their linking chromosome bridges through autophagy and subsequently began cell divisions. These data show that 'neotic' ETC resulting from genotoxically damaged p53 function-deficient tumour cells develop through a heteronuclear system differentiating the polyploid genome into rejuvenated 'viable' subcells (which provide mitotically propagating paradiploid descendents) and subnuclei, which become degraded and eliminated by autophagy. The whole process reduces aneuploidy in descendants of ETC.


Subject(s)
Autophagy , Chromatin/metabolism , DNA Fragmentation , DNA Replication , Neoplasms/genetics , Ploidies , Tumor Suppressor Protein p53/deficiency , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , DNA Replication/drug effects , DNA Replication/radiation effects , Etoposide/pharmacology , Gamma Rays/adverse effects , Genome, Human , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mitosis , Nanog Homeobox Protein , Neoplasms/metabolism , Neoplasms/pathology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/genetics
3.
Cancer Res ; 69(6): 2296-304, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19258501

ABSTRACT

Cancer is frequently characterized histologically by the appearance of large cells that are either aneuploid or polyploid. Aneuploidy and polyploidy are hallmarks of radiation-induced mitotic catastrophe (MC), a common phenomenon occurring in tumor cells with impaired p53 function following exposure to various cytotoxic and genotoxic agents. MC is characterized by altered expression of mitotic regulators, untimely and abnormal cell division, delayed DNA damage, and changes in morphology. We report here that cells undergoing radiation-induced MC are more plastic with regards to ploidy and that this plasticity allows them to reorganize their genetic material through reduction division to produce smaller cells which are morphologically indistinguishable from control cells. Experiments conducted with the large-scale digital cell analysis system are discussed and show that a small fraction of polyploid cancer cells formed via radiation-induced MC can survive and start a process of depolyploidization that yields various outcomes. Although most multipolar divisions failed and cell fusion occurred, some of these divisions were successful and originated a variety of cell progeny characterized by different ploidy. Among these ploidy phenotypes, a progeny of small mononucleated cells, indistinguishable from the untreated control cells, is often seen. We report here evidence that meiosis-specific genes are expressed in the polyploid cells during depolyploidization. Tumor cells might take advantage of the temporary change from a promitotic to a promeiotic division regimen to facilitate depolyploidization and restore the proliferative state of the tumor cell population. These events might be mechanisms by which tumor progression and resistance to treatment occur in vivo.


Subject(s)
Gene Expression Regulation, Neoplastic/radiation effects , Meiosis/genetics , Mitosis/radiation effects , Neoplasms/genetics , Polyploidy , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Nucleus/radiation effects , Cell Nucleus/ultrastructure , Chromosome Segregation/genetics , Chromosome Segregation/radiation effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , HCT116 Cells , HeLa Cells , Humans , Metaphase/genetics , Metaphase/radiation effects , Mitosis/genetics , Neoplasms/pathology , Neoplasms/radiotherapy
4.
Cell Biol Int ; 32(9): 1044-56, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18602486

ABSTRACT

Recent findings including computerised live imaging suggest that polyploidy cells transiently emerging after severe genotoxic stress (and named 'endopolyploid cells') may have a role in tumour regrowth after anti-cancer treatment. Until now, mostly the factors enabling metaphase were studied in them. Here we investigate the mitotic activities and the role of Aurora-B, in view of potential depolyploidisation of these cells, because Aurora-B kinase is responsible for coordination and completion of mitosis. We observed that endopolyploid giant cells are formed via different means in irradiated p53 tumours, by: (1) division/fusion of daughter cells creating early multi-nucleated cells; (2) asynchronous division/fusion of sub-nuclei of these multi-nucleated cells; (3) a series of polyploidising mitoses reverting replicative interphase from aborted metaphase and forming giant cells with a single nucleus; (4) micronucleation of arrested metaphases enclosing genome fragments; or (5) incomplete division in the multi-polar mitoses forming late multi-nucleated giant cells. We also observed that these activities can release para-diploid cells, although infrequently. While apoptosis typically occurs after a substantial delay in these cells, we also found that approximately 2% of the endopolyploid cells evade apoptosis and senescence arrest and continue some form of mitotic activity. We describe here that catalytically active Aurora-B kinase is expressed in the nuclei of many endopolyploid cells in interphase, as well as being present at the centromeres, mitotic spindle and cleavage furrow during their attempted mitotes. The totally micronucleated giant cells (containing sub-genomic fragments in multiple micronuclei) represented only the minor fraction which failed to undergo mitosis, and Aurora-B was absent from it. These observations suggest that most endopolyploid tumour cells are not reproductively inert and that Aurora-B may contribute to the establishment of resistant tumours post-irradiation.


Subject(s)
Cell Division/radiation effects , Giant Cells/enzymology , Giant Cells/pathology , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/deficiency , Anaphase/radiation effects , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/radiation effects , Cell Survival/radiation effects , Chromosomes, Human/metabolism , Chromosomes, Human/radiation effects , DNA, Neoplasm/metabolism , Giant Cells/radiation effects , Humans , In Situ Hybridization, Fluorescence , Time Factors , Tubulin/metabolism , X-Rays
5.
J Microsc ; 228(Pt 3): 296-308, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045324

ABSTRACT

The Large-Scale Digital Cell Analysis System (LSDCAS) was designed to provide a highly extensible open source live cell imaging system. Analysis of cell growth data has demonstrated a lack of perturbation in cells imaged using LSDCAS, through reference to cell growth data from cells growing in CO(2) incubators. LSDCAS consists of data acquisition, data management and data analysis software, and is currently a Core research facility at the Holden Comprehensive Cancer Center at the University of Iowa. Using LSDCAS analysis software, this report and others show that although phase-contrast imaging has no apparent effect on cell growth kinetics and viability, fluorescent image acquisition in the cell lines tested caused a measurable level of growth perturbation using LSDCAS. This report describes the current design of the system, reasons for the implemented design, and details its basic functionality. The LSDCAS software runs on the GNU/Linux operating system, and provides easy to use, graphical programs for data acquisition and quantitative analysis of cells imaged with phase-contrast or fluorescence microscopy (alone or in combination), and complete source code is freely available under the terms of the GNU Public Software License at the project website (http://lsdcas.engineering.uiowa.edu).


Subject(s)
Cell Physiological Phenomena , Cells/cytology , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Microscopy, Phase-Contrast/methods , Cell Line, Tumor , Cell Proliferation , Humans , Time Factors
6.
Radiat Res ; 168(2): 183-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17638400

ABSTRACT

Human gliomas are among the most aggressive tumors, and they respond poorly to treatment. The efficacy of surgical, radiation and chemotherapy treatment of these tumors is limited by the development of resistance. Interventions aimed at altering the response of these tumors to radiation or chemotherapy treatments are needed to improve survival rate and prognosis. Glioblastomas are generally p53 (TP53) functional tumors; however, DNA repair pathways are activated in these tumors instead of the pathways to apoptosis. Thus resistance to treatment is seen in the ability of these tumors to overcome cell death. We present data that demonstrate that U87MG glioblastoma cells transduced with a dominant-negative p53 adenovirus construct become sensitized to radiation-induced mitotic catastrophe through abrogation of G(2)/M checkpoint control and overaccumulation of cyclin B1. These findings suggest that interventions abrogating the G(2)/M checkpoint sensitize these cells to radiation-induced mitotic catastrophe and may represent a novel mechanism to increase the efficacy of radiation in wild-type p53 gliomas that are resistant to apoptosis.


Subject(s)
Glioblastoma/radiotherapy , Mitosis/radiation effects , Tumor Suppressor Protein p53/physiology , Adenoviridae/genetics , CDC2 Protein Kinase/physiology , Cell Line, Tumor , Cyclin B/physiology , Cyclin B1 , DNA Damage , DNA Repair , G1 Phase , Glioblastoma/pathology , Humans , Transduction, Genetic
7.
Cancer Cell Int ; 6: 11, 2006 Apr 26.
Article in English | MEDLINE | ID: mdl-16640786

ABSTRACT

BACKGROUND: We have demonstrated that in some human cancer cells both chronic mild heat and ionizing radiation exposures induce a transient block in S and G2 phases of the cell cycle. During this delay, cyclin B1 protein accumulates to supranormal levels, cyclin B1-dependent kinase is activated, and abrogation of the G2/M checkpoint control occurs resulting in mitotic catastrophe (MC). RESULTS: Using syngenic mouse embryonic fibroblasts (MEF) with wild-type or mutant p53, we now show that, while both cell lines exhibit delays in S/G2 phase post-irradiation, the mutant p53 cells show elevated levels of cyclin B1 followed by MC, while the wild-type p53 cells present both a lower accumulation of cyclin B1 and a lower frequency of MC. CONCLUSION: These results are in line with studies reporting the role of p53 as a post-transcriptional regulator of cyclin B1 protein and confirm that dysregulation of cyclin B1 promote radiation-induced MC. These findings might be exploited to design strategies to augment the yield of MC in tumor cells that are resistant to radiation-induced apoptosis.

8.
Article in English | MEDLINE | ID: mdl-16685859

ABSTRACT

The Large Scale Digital Cell Analysis System (LSDCAS) developed at the University of Iowa provides capabilities for extended-time live cell image acquisition. This paper presents a new approach to quantitative analysis of live cell image data. By using time as an extra dimension, level set methods are employed to determine cell trajectories from 2D + time data sets. When identifying the cell trajectories, cell cluster separation and mitotic cell detection steps are performed. Each of the trajectories corresponds to the motion pattern of an individual cell in the data set. At each time frame, number of cells, cell locations, cell borders, cell areas, and cell states are determined and recorded. The proposed method can help solving cell analysis problems of general importance including cell pedigree analysis and cell tracking. The developed method was tested on cancer cell image sequences and its performance compared with manually-defined ground truth. The similarity Kappa Index is 0.84 for segmentation area and the signed border positioning segmentation error is 1.6 +/- 2.1 microm.


Subject(s)
Cell Nucleus/ultrastructure , Glioma/ultrastructure , Image Cytometry/methods , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Mitosis , Pattern Recognition, Automated/methods , Algorithms , Artificial Intelligence , Cell Count/methods , Cell Line, Tumor , Cell Movement , Humans , Reproducibility of Results , Sensitivity and Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...