Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 212(3): 281-8, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26811421

ABSTRACT

Topoisomerase IIß-binding protein 1 (TOPBP1) participates in DNA replication and DNA damage response; however, its role in DNA repair and relevance for human cancer remain unclear. Here, through an unbiased small interfering RNA screen, we identified and validated TOPBP1 as a novel determinant whose loss sensitized human cells to olaparib, an inhibitor of poly(ADP-ribose) polymerase. We show that TOPBP1 acts in homologous recombination (HR) repair, impacts olaparib response, and exhibits aberrant patterns in subsets of human ovarian carcinomas. TOPBP1 depletion abrogated RAD51 loading to chromatin and formation of RAD51 foci, but without affecting the upstream HR steps of DNA end resection and RPA loading. Furthermore, TOPBP1 BRCT domains 7/8 are essential for RAD51 foci formation. Mechanistically, TOPBP1 physically binds PLK1 and promotes PLK1 kinase-mediated phosphorylation of RAD51 at serine 14, a modification required for RAD51 recruitment to chromatin. Overall, our results provide mechanistic insights into TOPBP1's role in HR, with potential clinical implications for cancer treatment.


Subject(s)
Carrier Proteins/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Homologous Recombination , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Female , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA Interference , Rad51 Recombinase/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Polo-Like Kinase 1
2.
Cell Cycle ; 13(23): 3727-41, 2014.
Article in English | MEDLINE | ID: mdl-25483082

ABSTRACT

BRCA1 tumor suppressor regulates crucial cellular processes involved in DNA damage repair and cell cycle control. We showed that expression of BCR-ABL1 correlates with decreased level of BRCA1 protein, which promoted aberrant mitoses and aneuploidy as well as altered DNA damage response. Using polysome profiling and luciferase-BRCA1 3'UTR reporter system here we demonstrate that downregulation of BRCA1 protein in CML is caused by inhibition of BRCA1 mRNA translation, but not by increased protein degradation or reduction of mRNA level and half-life. We investigated 2 mRNA-binding proteins - HuR and TIAR showing specificity to AU-Rich Element (ARE) sites in 3'UTR of mRNA. BCR-ABL1 promoted cytosolic localization of TIAR and HuR, their binding to BRCA1 mRNA and formation of the TIAR-HuR complex. HuR protein positively regulated BRCA1 mRNA stability and translation, conversely TIAR negatively regulated BRCA1 translation and was found localized predominantly in the cytosolic stress granules in CML cells. TIAR-dependent downregulation of BRCA1 protein level was a result of ER stress, which is activated in BCR-ABL1 expressing cells, as we previously shown. Silencing of TIAR in CML cells strongly elevated BRCA1 level. Altogether, we determined that TIAR-mediated repression of BRCA1 mRNA translation is responsible for downregulation of BRCA1 protein level in BCR-ABL1 -positive leukemia cells. This mechanism may contribute to genomic instability and provide justification for targeting PARP1 and/or RAD52 to induce synthetic lethality in "BRCAness" CML and BCR-ABL1 -positive ALL cells.


Subject(s)
BRCA1 Protein/metabolism , Down-Regulation/physiology , Endoplasmic Reticulum Stress/physiology , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , RNA-Binding Proteins/metabolism , Animals , BRCA1 Protein/genetics , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
3.
Cell Cycle ; 11(21): 4069-78, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23095523

ABSTRACT

Activation of adaptive mechanisms plays a crucial role in cancer progression and drug resistance by allowing cell survival under stressful conditions. Therefore, inhibition of the adaptive response is considered as a prospective therapeutic strategy. The PERK-eIF2α phosphorylation pathway is an important arm of the unfolded protein response (UPR), which is induced under conditions of endoplasmic reticulum (ER) stress. Our previous work showed that ER stress is induced in chronic myeloid leukemia (CML) cells. Herein, we demonstrate that the PERK-eIF2α phosphorylation pathway is upregulated in CML cell lines and CD34+ cells from CML patients and is associated with CML progression and imatinib resistance. We also show that induction of apoptosis by imatinib results in the downregulation of the PERK-eIF2α phosphorylation arm. Furthermore, we demonstrate that inactivation of the PERK-eIF2α phosphorylation arm decreases the clonogenic and proliferative capacities of CML cells and sensitizes them to death by imatinib. These findings provide evidence for a pro-survival role of PERK-eIF2α phosphorylation arm that contributes to CML progression and development of imatinib resistance. Thus, the PERK-eIF2α phosphorylation arm may represent a suitable target for therapeutic intervention for CML disease.


Subject(s)
Antineoplastic Agents/toxicity , Cell Survival/drug effects , Eukaryotic Initiation Factor-2/metabolism , Fusion Proteins, bcr-abl/metabolism , Piperazines/toxicity , Pyrimidines/toxicity , eIF-2 Kinase/metabolism , Animals , Antigens, CD34/metabolism , Benzamides , Cell Line, Tumor , Endoplasmic Reticulum Stress , HL-60 Cells , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Phosphorylation , STAT5 Transcription Factor/metabolism , Signal Transduction
4.
Cell Cycle ; 11(20): 3837-50, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22983061

ABSTRACT

Impaired DNA damage response pathways may create vulnerabilities of cancer cells that can be exploited therapeutically. One such selective vulnerability is the sensitivity of BRCA1- or BRCA2-defective tumors (hence defective in DNA repair by homologous recombination, HR) to inhibitors of the poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme critical for repair pathways alternative to HR. While promising, treatment with PARP-1 inhibitors (PARP-1i) faces some hurdles, including (1) acquired resistance, (2) search for other sensitizing, non-BRCA1/2 cancer defects and (3) lack of biomarkers to predict response to PARP-1i. Here we addressed these issues using PARP-1i on 20 human cell lines from carcinomas of the breast, prostate, colon, pancreas and ovary. Aberrations of the Mre11-Rad50-Nbs1 (MRN) complex sensitized cancer cells to PARP-1i, while p53 status was less predictive, even in response to PARP-1i combinations with camptothecin or ionizing radiation. Furthermore, monitoring PARsylation and Rad51 foci formation as surrogate markers for PARP activity and HR, respectively, supported their candidacy for biomarkers of PARP-1i responses. As to resistance mechanisms, we confirmed the role of the multidrug resistance efflux transporters and its reversibility. More importantly, we demonstrated that shRNA lentivirus-mediated depletion of 53BP1 in human BRCA1-mutant breast cancer cells increased their resistance to PARP-1i. Given the preferential loss of 53BP1 in BRCA-defective and triple-negative breast carcinomas, our findings warrant assessment of 53BP1 among candidate predictive biomarkers of response to PARPi. Overall, this study helps characterize genetic and functional determinants of cellular responses to PARP-1i and contributes to the search for biomarkers to exploit PARP inhibitors in cancer therapy.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/drug therapy , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/genetics , Acid Anhydride Hydrolases , Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Biomarkers, Tumor/metabolism , Camptothecin/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Female , Gamma Rays/therapeutic use , Genes, MDR , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , MRE11 Homologue Protein , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , Tumor Suppressor p53-Binding Protein 1
5.
Apoptosis ; 17(9): 950-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696202

ABSTRACT

Chronic myeloid leukemia (CML) is a disorder of hematopoietic stem cells caused by the expression of BCR-ABL. Loss of p53 has not been implicated as important for the development of CML. Mutations in p53 protein are infrequent, however they correlate with the disease progression. The absence of p53 mutations does not exclude the possibility that other dysfunctions play an important role in CML pathology. Acetylation represents a very potent posttranslational mechanism regulating p53 stability, transcriptional activity and localization. In this study we have investigated whether the expression of BCR-ABL could influence the acetylation of p53, specifically at lysine 317/320 (K317/K320), which has been shown to regulate nuclear export and transcription-independent apoptotic activity of p53. We found that BCR-ABL expression increases K317 acetylation of p53 and is able to prevent a drop in acetylation observed upon DNA damage, followed by translocation of p53 to the cytoplasm and by Bax activation. We have shown that this site plays a crucial role in the regulation of p53 localization and p53-dependent, Bax-mediated apoptosis. Our study presents a novel BCR-ABL-dependent mechanism protecting from DNA-damage-induced cell death. It can, in addition to already known mechanisms, explain the resistance to p53-dependent apoptosis observed in CML cells expressing wt p53. We propose that the acetyltransferases regulating the p53 acetylation could be an interesting and potent target for therapeutic intervention.


Subject(s)
Apoptosis , DNA Damage , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Cell Line , Cell Survival , Cytoplasm/metabolism , Enzyme Activation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mitochondria/metabolism , Protein Transport , RNA Interference , RNA, Small Interfering , bcl-2-Associated X Protein/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
6.
Mol Cancer Ther ; 9(5): 1328-38, 2010 May.
Article in English | MEDLINE | ID: mdl-20442314

ABSTRACT

Recent findings showed that BRCA1, in addition to its role in DNA damage response, acts as an upstream regulator of genes involved in the mitotic checkpoint regulation, thus protecting against promotion of aberrant divisions and aneuploidy. Moreover, there is also an indication that the BRCA1 protein is downregulated in chronic myeloid leukemia (CML) patients. We have investigated a possible functional relationship between BRCA1 and mitotic checkpoint competence in cells with the same genetic background expressing different levels of Bcr-Abl, an oncogene responsible for CML. Herein, we show that Bcr-Abl strongly downregulates the BRCA1 protein level, which is partially reversed on treatment with imatinib, an inhibitor of Bcr-Abl tyrosine kinase. Bcr-Abl leads to decreased expression of genes involved in the mitotic checkpoint activation--Mad2, Bub1, Bub3, and BubR1, resulting in mitosis perturbances, weakened mitotic checkpoint function, and mitotic slippage after nocodazole treatment. Furthermore, high Bcr-Abl-expressing cells showed also postmitotic checkpoint dysfunctions and inability to effectively arrest in the 4NG1 phase of the cell cycle, which was associated with limited p21 induction. These observations had significant biological consequences, as we found a high level of improper divisions, chromosomal missegregation, and generation of polyploid cells on mitotic checkpoint prolonged activation. Additionally, Bcr-Abl-expressing cells showed resistance to death activated by spindle defects, reversed by imatinib. Our study presents new facts and supports the hypothesis concerning the mutator nature of Bcr-Abl itself. The functional interaction between Bcr-Abl and mitosis dysfunctions, due to compromised mitotic checkpoints, may have important implications for the generation of aneuploidy and CML progression.


Subject(s)
Cell Division/genetics , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Mitosis/genetics , Protein-Tyrosine Kinases/genetics , Tubulin Modulators/therapeutic use , Aneuploidy , Animals , Cells, Cultured , Disease Progression , Drug Delivery Systems , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Leukemic/physiology , Genes, cdc/physiology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Microtubules/drug effects , Protein-Tyrosine Kinases/physiology , Tubulin Modulators/administration & dosage , Tubulin Modulators/pharmacology
7.
Inflamm Bowel Dis ; 16(3): 379-89, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19774649

ABSTRACT

INTRODUCTION: Epidemiological data suggests that 5-aminosalicylic acid (5-ASA), a nonsteroidal antiinflammatory drug used in the treatment of inflammatory bowel diseases, prevents colorectal cancer development in these patients, although the mechanisms remain incompletely understood. METHODS AND RESULTS: Here we report that 5-ASA prevents growth of several colorectal cancer cell lines by interfering in the cell cycle, i.e., an S-phase and G2/M phase arrest, dependent on 5-ASA dosage and concentration, together with an increased mitotic index. In addition, prolonged cell cycle arrest by repeated 5-ASA treatment induced apoptosis and caused abnormal spindle organization leading to mitotic catastrophe, another form of cell death. CONCLUSION: These observations illustrate that 5-ASA has chemopreventive and chemotherapeutic properties.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Mesalamine/pharmacology , Caco-2 Cells , Cell Division/drug effects , Flow Cytometry , G2 Phase/drug effects , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Mitosis/drug effects , PPAR gamma/metabolism , S Phase/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/pathology , Survivin
8.
Mech Ageing Dev ; 130(1-2): 24-32, 2009.
Article in English | MEDLINE | ID: mdl-18538372

ABSTRACT

Induction of senescence has been proposed as a possible in vivo tumor response to anticancer treatment. Senescent cancer cells are often polyploid, however, their route to polyploidy is poorly recognized (endoreduplication versus aberrant mitoses). We showed that after treatment of HCT116 cells with a low dose of doxorubicin most of them stopped proliferation as documented by SA-beta-galactosidase activity and the lack of Ki67 expression. Increased expression of other common senescence markers, p53, p21 and cyclin D1, was also observed. The cells became giant, polyploid and polymorphic, with multinucleated cells comprising a substantial fraction. The vast majority of the doxorubicin-treated cells did not enter mitoses, as evidenced by mitotic index analysis, as well as by the predominantly cytoplasmic localization of cyclin B1 and a lack of separation of multiplied centrosomes. This allowed us to conclude that doxorubicin-treated HCT116 cells underwent endoreduplication. However, the rare events of aberrant mitoses of polyploid cells observed by us led to aneuploid progeny as was documented by cytogenetic analysis of survivors. Thus, a senescence-inducing treatment of HCT116 cancer cells had a dual effect-it stopped the proliferation of the majority of the cells, but also led to the appearance of proliferating aneuploid ones.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cellular Senescence/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Doxorubicin/pharmacology , Genomic Instability/drug effects , Apoptosis/drug effects , Colonic Neoplasms/pathology , DNA Replication/drug effects , Disease Progression , HCT116 Cells , Humans , Mitosis/drug effects , Polyploidy
9.
Postepy Biochem ; 53(1): 10-8, 2007.
Article in Polish | MEDLINE | ID: mdl-17718383

ABSTRACT

Human survivin is a member of the IAP (Inhibitor of Apoptosis) family. It was reported that survivin expression is associated with drug resistance, cancer progression and low patient survival rate in many cancers. Survivin is implicated in both: inhibition of apoptosis and regulation of cell division. As a member of Chromosomal Passenger Complex (CPC) it is involved in sister chromatids segregation during mitosis. On the other hand, survivin plays an important role in the surveillance mechanism called mitotic spindle assembly checkpoint (MSAC) which regulates metaphase to anaphase transition during mitosis. Additionally, survivin is necessary for cytokinesis progression. The present review is a summary of survivin's functions, focused on its role in cell division in normal and cancer cells, as well as introduction to discussion about anticancer therapies based on survivin depletion.


Subject(s)
Microtubule-Associated Proteins/metabolism , Mitosis/physiology , Neoplasm Proteins/metabolism , Anaphase , Aneuploidy , Apoptosis/physiology , Chromosomal Proteins, Non-Histone/metabolism , Cytokinesis/physiology , Humans , Inhibitor of Apoptosis Proteins/metabolism , Metaphase , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Polyploidy , Protein Binding , Protein Isoforms/genetics , Protein Structure, Tertiary , Spindle Apparatus/metabolism , Survivin
10.
Acta Biochim Pol ; 53(3): 485-95, 2006.
Article in English | MEDLINE | ID: mdl-16951738

ABSTRACT

The influence of mutations in the mitochondrial DNA (mtDNA) on the bioenergetic metabolism of the cell is still poorly understood. Many of the mutations in the mtDNA affect the expression of the mitochondrial genome. Investigations on cells from patients are not easy, especially as the mitochondrial DNA is heteroplasmic and this state is changed in culture. Moreover, the nuclear background and the mitochondrial haplotype may affect the behaviour of cells. Transfer of patient mitochondria to rho zero cell lines is also not optimal as these cells in general have many nuclear changes which may also affect cell behaviour. Thus, we decided to use inhibitors of mitochondrial genome expression, such as thiamphenicol, ethidium bromide and dideoxycytidine to investigate the bioenergetic metabolism of HeLa cells. We found that oxidative phosphorylation and glycolysis participate equally in ATP production in HeLa cells and that decreased activity of the respiratory chain leads to increased glycolysis and the reduction of cell growth. Insufficient ATP production in the oxidative phosphorylation process was not compensated by increased proliferation of the mitochondria. However, we were able to show that there are some mechanisms compensating limited expression of the mitochondrial genome within the mitochondria. Experiments with dideoxycytidine revealed that 10-fold decrease of the mtDNA copy number resulted in almost normal activity of cytochrome c oxidase. We found that mtDNA depletion is compensated mostly on the level of RNA metabolism in the mitochondria. Thus, our results are in agreement with the hypothesis that transcription initiation rather than mtDNA copy number is a rate limiting factor for expression of the mitochondrial genome.


Subject(s)
DNA, Mitochondrial/drug effects , Ethidium/pharmacology , Gene Expression/drug effects , Nuclear Proteins , Thiamphenicol/pharmacology , Zalcitabine/pharmacology , Animals , Antimetabolites/pharmacology , DNA, Mitochondrial/genetics , Electron Transport/genetics , Electron Transport Complex IV/genetics , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Nuclear Respiratory Factor 1/genetics , Oxidative Phosphorylation/drug effects , RNA/drug effects , RNA/genetics , RNA, Mitochondrial , Rats
11.
Mol Cancer Res ; 4(7): 457-69, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16849521

ABSTRACT

The Bcr-Abl oncoprotein plays a major role in the development and progression of chronic myeloid leukemia and is a determinant of chemotherapy resistance occurring during the blast crisis phase of the disease. The aim of this article was to investigate the possibility of combating the resistance to apoptosis caused by Bcr-Abl by inducing an alternative cell death process. As a model of chronic myeloid leukemia, we employed Bcr-Abl-transfected mouse progenitor 32D cells with low and high Bcr-Abl expression levels corresponding to drug-sensitive and drug-resistant cells, respectively. The drug curcumin (diferuloylmethane), a known potent inducer of cell death in many cancer cells, was investigated for efficacy with Bcr-Abl-expressing cells. Curcumin strongly inhibited cell proliferation and affected cell viability by inducing apoptotic symptoms in all tested cells; however, apoptosis was a relatively late event. G(2)-M cell cycle arrest, together with increased mitotic index and cellular and nuclear morphology resembling those described for mitotic catastrophe, was observed and preceded caspase-3 activation and DNA fragmentation. Mitosis-arrested cells displayed abnormal chromatin organization, multipolar chromosome segregation, aberrant cytokinesis, and multinucleated cells-morphologic changes typical of mitotic catastrophe. We found that the mitotic cell death symptoms correlated with attenuated expression of survivin, a member of the chromosomal passenger complex, and mislocalization of Aurora B, the partner of survivin in the chromosomal passenger complex. Inhibition of survivin expression with small interfering RNA exhibited similar mitotic disturbances, thus implicating survivin as a major, albeit not the only, target for curcumin action. This study shows that curcumin can overcome the broad resistance to cell death caused by expression of Bcr-Abl and suggests that curcumin may be a promising agent for new combination regimens for drug-resistant chronic myeloid leukemia.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mitosis/drug effects , Animals , Apoptosis/genetics , Aurora Kinase B , Aurora Kinases , Caspases/metabolism , Cell Growth Processes/drug effects , Chromosome Segregation , Down-Regulation/drug effects , Enzyme Activation , Fusion Proteins, bcr-abl/genetics , Genes, abl , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Inhibitor of Apoptosis Proteins , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis/genetics , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...