Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(8): 6189-6206, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38577779

ABSTRACT

Identification of intracellular targets of anticancer drug candidates provides key information on their mechanism of action. Exploiting the ability of the anticancer (C∧N)-chelated half-sandwich iridium(III) complexes to covalently bind proteins, click chemistry with a bioorthogonal azido probe was used to localize a phenyloxazoline-chelated iridium complex within cells and profile its interactome at the proteome-wide scale. Proteins involved in protein folding and actin cytoskeleton regulation were identified as high-affinity targets. Upon iridium complex treatment, the folding activity of Heat Shock Protein HSP90 was inhibited in vitro and major cytoskeleton disorganization was observed. A wide array of imaging and biochemical methods validated selected targets and provided a multiscale overview of the effects of this complex on live human cells. We demonstrate that it behaves as a dual agent, inducing both electrophilic and oxidative stresses in cells that account for its cytotoxicity. The proposed methodological workflow can open innovative avenues in metallodrug discovery.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Iridium , Oxidative Stress , Humans , Iridium/chemistry , Iridium/pharmacology , Oxidative Stress/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Click Chemistry
2.
J Med Chem ; 64(22): 16675-16686, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34761949

ABSTRACT

Half-sandwich complexes of iridium(III) are currently being developed as anticancer drug candidates. In this context, we introduce IrBDP for which the C^N chelating phenyloxazoline ligand carries a fluorescent and lipophilic BODIPY reporter group, designed for intracellular tracking and hydrophobic compartment tropism. High-resolution analysis of cells cultured with IrBDP showed that it quickly permeates the plasma membrane and accumulates in the mitochondria and endoplasmic reticulum (ER), generating ER stress, dispersal of the Golgi apparatus, cell proliferation arrest and apoptotic cell death. Moreover, IrBDP forms fluorescent adducts with a subset of amino acids, namely histidine and cysteine, via coordination of N or S donor atoms of their side chains. Consistently, in vivo formation of covalent adducts with specific proteins is demonstrated, providing a molecular basis for the observed cytotoxicity and cellular response. Collectively, these results provide a new entry to the development of half-sandwich iridium-based anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Boron Compounds/chemistry , Endoplasmic Reticulum Stress , Iridium/chemistry , Proteins/chemistry , HeLa Cells , Humans
3.
Dalton Trans ; 49(48): 17635-17641, 2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33226042

ABSTRACT

Transition metal-based anticancer compounds, as an alternative to platinum derivatives, are raising scientific interest as they may present distinct although poorly understood mechanisms of action. We used a structure-activity relationship-based methodology to investigate the chemical and biological features of a series of ten (C^N)-chelated half-sandwich iridiumIII complexes of the general formula [IrCp*(phox)Cl], where (phox) is a 2-phenyloxazoline ligand forming a 5-membered metallacycle. This series of compounds undergoes a fast exchange of their chlorido ligand once solubilised in DMSO. They were cytotoxic to HeLa cells with IC50 values in the micromolar range and induced a rapid activation of caspase-3, an apoptosis marker. In vitro, the oxidative power of all the complexes towards NADH was highlighted but only the complexes bearing substituents on the oxazoline ring were able to produce H2O2 at the micromolar range. However, we demonstrated using a powerful HyPer protein redox sensor-based flow cytometry assay that most complexes rapidly raised intracellular levels of H2O2. Hence, this study shows that oxidative stress can partly explain the cytotoxicity of these complexes on the HeLa cell line and gives a first entry to their mechanism of action.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Iridium/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Iridium/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
4.
J Cell Mol Med ; 24(14): 8091-8102, 2020 07.
Article in English | MEDLINE | ID: mdl-32515546

ABSTRACT

Membrane receptor intracellular trafficking and signalling are frequently altered in cancers. Our aim was to investigate whether clathrin-dependent trafficking modulates signalling of the ErbB receptor family in response to amphiregulin (AR), EGF, heparin-binding EGF-like growth factor (HB-EGF) and heregulin-1ß (HRG). Experiments were performed using three hepatocellular carcinoma (HCC) cell lines, Hep3B, HepG2 and PLC/PRF/5, expressing various levels of EGFR, ErbB2 and ErbB3. Inhibition of clathrin-mediated endocytosis (CME), by down-regulating clathrin heavy chain expression, resulted in a cell- and ligand-specific pattern of phosphorylation of the ErbB receptors and their downstream effectors. Clathrin down-regulation significantly decreased the ratio between phosphorylated EGFR (pEGFR) and total EGFR in all cell lines when stimulated with AR, EGF, HB-EGF or HRG, except in HRG-stimulated Hep3B cells in which pEGFR was not detectable. The ratio between phosphorylated ErbB2 and total ErbB2 was significantly decreased in clathrin down-regulated Hep3B cells stimulated with any of the ligands, and in HRG-stimulated PLC/PRF/5 cells. The ratio between phosphorylated ErbB3 and total ErbB3 significantly decreased in clathrin down-regulated cell lines upon stimulation with EGF or HB-EGF. STAT3 phosphorylation levels significantly increased in all cell lines irrespective of stimulation, while that of AKT remained unchanged, except in AR-stimulated Hep3B and HepG2 cells in which pAKT was significantly decreased. Finally, ERK phosphorylation was insensitive to clathrin inhibition. Altogether, our observations indicate that clathrin regulation of ErbB signalling in HCC is a complex process that likely depends on the expression of ErbB family members and on the autocrine/paracrine secretion of their ligands in the tumour environment.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Clathrin/metabolism , Liver Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Ligands , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Receptor, ErbB-3/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
5.
Science ; 358(6364): 803-806, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28982797

ABSTRACT

Cell division and differentiation depend on massive and rapid organelle remodeling. The mitotic oscillator, centered on the cyclin-dependent kinase 1-anaphase-promoting complex/cyclosome (CDK1-APC/C) axis, spatiotemporally coordinates this reorganization in dividing cells. Here we discovered that nondividing cells could also implement this mitotic clocklike regulatory circuit to orchestrate subcellular reorganization associated with differentiation. We probed centriole amplification in differentiating mouse-brain multiciliated cells. These postmitotic progenitors fine-tuned mitotic oscillator activity to drive the orderly progression of centriole production, maturation, and motile ciliation while avoiding the mitosis commitment threshold. Insufficient CDK1 activity hindered differentiation, whereas excessive activity accelerated differentiation yet drove postmitotic progenitors into mitosis. Thus, postmitotic cells can redeploy and calibrate the mitotic oscillator to uncouple cytoplasmic from nuclear dynamics for organelle remodeling associated with differentiation.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CDC2 Protein Kinase/metabolism , Cilia/physiology , Mitosis , Animals , Brain/cytology , Cell Differentiation , Centrioles/metabolism , Mice , Organelles/metabolism
6.
Nat Commun ; 6: 6894, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25902731

ABSTRACT

Caprine-like Generalized Hypoplasia Syndrome (SHGC) is an autosomal-recessive disorder in Montbéliarde cattle. Affected animals present a wide range of clinical features that include the following: delayed development with low birth weight, hind limb muscular hypoplasia, caprine-like thin head and partial coat depigmentation. Here we show that SHGC is caused by a truncating mutation in the CEP250 gene that encodes the centrosomal protein C-Nap1. This mutation results in centrosome splitting, which neither affects centriole ultrastructure and duplication in dividing cells nor centriole function in cilium assembly and mitotic spindle organization. Loss of C-Nap1-mediated centriole cohesion leads to an altered cell migration phenotype. This discovery extends the range of loci that constitute the spectrum of autosomal primary recessive microcephaly (MCPH) and Seckel-like syndromes.


Subject(s)
Cattle Diseases/genetics , Cell Cycle Proteins/genetics , Cell Movement/genetics , Centrioles/metabolism , Hypopigmentation/veterinary , Microcephaly/veterinary , Morphogenesis/genetics , Muscular Diseases/veterinary , Animals , Cattle , Hypopigmentation/genetics , Microcephaly/genetics , Muscular Diseases/genetics , Mutation , Syndrome
7.
Am J Pathol ; 180(1): 131-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22056911

ABSTRACT

Mitotic kinesin-like protein 2 (MKlp2), a microtubule-associated motor, is required during mitosis exit for the final step of cytokinesis. It also contributes to retrograde vesicular trafficking from the Golgi apparatus to the endoplasmic reticulum in interphase. The KIF20A gene encoding MKlp2 is controlled by the E2F-retinoblastoma protein-p16 pathway, and its widely expressed mRNA is found in fetal and proliferating adult tissues. The expression pattern and function of MKlp2 in the adult liver, however, have not been investigated. We report herein that MKlp2 transiently accumulates in vivo during mouse liver regeneration after partial hepatectomy and is strongly overexpressed in preneoplastic and neoplastic mouse liver. In vitro in mitogen-stimulated primary hepatocytes, MKlp2 accumulated in the nucleus during the G2 phase of the cell cycle coincident with the mitotic kinase Aurora B. Human hepatoma cell lines exhibited high levels of MKlp2; however, it was undetectable in normal human hepatocytes. RNAi-mediated MKlp2 knockdown in hepatoma cells induced polyploidization consistent with its essential function in promoting cytokinesis and inhibited cell proliferation without inducing apoptosis. KIF20A mRNA was strongly accumulated in a large series of human hepatocellular carcinomas, with the highest expression observed in tumors with genomic instability. Accumulation of MKlp2 in normal proliferating, preneoplastic, and transformed hepatocytes suggests that MKlp2 contributes to both normal and pathologic hepatocyte proliferation and is linked to tumor aggressiveness in human hepatocellular carcinomas.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Kinesins/metabolism , Liver Neoplasms/metabolism , Liver Regeneration/physiology , Precancerous Conditions/metabolism , Aged , Animals , Carcinoma, Hepatocellular/pathology , Cell Cycle/physiology , Cell Line, Tumor , Female , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitogens/pharmacology , Precancerous Conditions/pathology , Tumor Cells, Cultured
8.
Int J Oncol ; 39(2): 393-400, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21567080

ABSTRACT

We have previously shown that the genotoxin-induced apoptosis in mouse embryo fibroblasts was enhanced by the extracellular matrix protein fibronectin (FN). In the present study, we tested the hypothesis that FN regulates the DNA damage response (DDR) signaling pathways in HCT116 (p53-wt) and HT29 (p53-mut) human colon cancer cells and tumor-derived myofibroblasts. DNA damage recognition mechanisms were analyzed by immunofluorescence staining, cell cycle analysis and immunoblotting addressed at specific molecular sensors and executors involved in the DDR pathways. The results show that FN, but not collagen type IV or Matrigel, initiates and potentiates the DDR to the anticancer drug cisplatin in an α5 integrin and cell cycle-dependent manner (S and G2/M phases) in human colon cancer cells. Accordingly, we demonstrate that adhesion of HCT116 cells to FN upregulated the expression of α5 integrin FN receptors at the cell surface. These FN-induced DDR pathways include the concerted phosphorylation of histone H2AX on Ser139 detected as nuclear foci (γ-H2AX, 15 and 25 kDa forms), of ataxia telangiectasia mutated (ATM-Ser1981), checkpoint kinase 2 (CHK2-Thr68, 62 and 67 kDa) and p53-Ser15. These FN-induced γ-H2AX signals were interrupted or attenuated by selective inhibitors acting on the DDR pathway kinases, including wortmannin (targeting the phosphatidylinositol-3-kinase-related protein kinases, PIKK), KU55933 (ATM), NU7026 (DNA-dependent protein kinase catalytic subunit, DNA-PK-cs) and SP600125 (JNK2, stress activated protein kinase/c-Jun N-terminal kinase-2). Adhesion to FN also potentiated the γ-H2AX signals and the cytotoxic effects of cisplatin in human colon tumor-derived myofibroblasts. These data provide evidence that FN promotes DNA damage recognition and chemosensitization to cisplatin via the potentiation of the DNA damage signaling responses in human colon cancer cells and tumor-derived myofibroblasts.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA Damage/drug effects , DNA Damage/genetics , Fibronectins/metabolism , Fibronectins/pharmacology , Myofibroblasts/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 2 , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , HCT116 Cells , HT29 Cells , Histones/metabolism , Humans , Myofibroblasts/pathology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Fibronectin/metabolism , Signal Transduction/drug effects
9.
J Biol Chem ; 283(22): 15201-8, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18378678

ABSTRACT

The LIM-only protein FHL2 acts as a transcriptional modulator that positively or negatively regulates multiple signaling pathways. We recently reported that FHL2 cooperates with CREB-binding protein/p300 in the activation of beta-catenin/T cell factor target gene cyclin D1. In this paper, we demonstrate that FHL2 is associated with the cyclin D1 promoter at the T cell factor/CRE site, providing evidence that cyclin D1 is a direct target of FHL2. We show that deficiency of FHL2 greatly reduces the proliferative capacity of spontaneously immortalized mouse fibroblasts, which is associated with decreased expression of cyclin D1 and p16(INK4a), and hypophosphorylation of Rb. Reexpression of FHL2 in FHL2-null fibroblasts efficiently restores cyclin D1 levels and cell proliferative capacity, indicating that FHL2 is critical for cyclin D1 activation and cell growth. Moreover, ectopic cyclin D1 expression is sufficient to override growth inhibition of immortalized FHL2-null fibroblasts. Gene expression profiling revealed that FHL2 deficiency triggers a broad change of the cell cycle program that is associated with down-regulation of several G(1)/S and G(2)/M cyclins, E2F transcription factors, and DNA replication machinery, thus correlating with reduced cell proliferation. This change also involves down-regulation of the negative cell cycle regulators, particularly INK4 inhibitors, which could counteract the decreased expression of cyclins, allowing cells to grow. Our study illustrates that FHL2 can act on different aspects of the cell cycle program to finely regulate cell proliferation.


Subject(s)
Cell Cycle/physiology , Cyclins/biosynthesis , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Response Elements/physiology , Transcription Factors/metabolism , Animals , Cell Line , Cyclin D , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclins/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fibroblasts/cytology , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mice , Mice, Knockout , Muscle Proteins/genetics , Phosphorylation , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription Factors/genetics , beta Catenin/genetics , beta Catenin/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
10.
J Med Chem ; 50(12): 2842-50, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17511440

ABSTRACT

We have designed and evaluated 45 linear analogues of the natural constrained cyclopeptide TMC-95A. These synthetically less demanding molecules are based on the tripeptide sequence Y-N-W of TMC-95A. Structural variations in the amino acid side chains and termini greatly influenced both the efficiency and selectivity of action on a given type of active site. Inhibition constants were submicromolar (Ki approximately 300 nM) despite the absence of the entropically favorable constrained conformation that is characteristic of TMC-95A and its cyclic analogues. These linear compounds were readily prepared and reasonably stable in culture medium and could be optimized to inhibit one, two, or all three proteasome catalytic sites. Cytotoxicity assays performed on a series of human tumor cell lines identified the most potent inhibitors in cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Peptides, Cyclic/chemical synthesis , Proteasome Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Structure-Activity Relationship
11.
Cytometry A ; 71(1): 1-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17211879

ABSTRACT

BACKGROUND: Cell division or cytokinesis, which results from a series of events starting in metaphase, is the mechanism by which the mother cell cytoplasm is divided between the two daughter cells. Hence it is the final step of the cell division cycle. The aim of the present study was to demonstrate that mammalian cells undergoing cytokinesis can be sorted selectively by flow cytometry. MATERIALS AND METHODS: Cultures of HeLa cells were arrested in prometaphase by nocodazole, collected by mitotic shake-off and released for 90 min into fresh medium to enrich for cells undergoing cytokinesis. After ethanol fixation and DNA staining, cells were sorted based on DNA content and DNA fluorescence signal height. RESULTS: We define a cell population that transiently accumulates when synchronized cells exit mitosis before their entry into G1. We show that this population is highly enriched in cells undergoing cytokinesis. In addition, this population of cells can be sorted and analyzed by immunofluorescence and western blotting. CONCLUSIONS: This method of cell synchronization and sorting provides a simple means to isolate and biochemically analyze cells in cytokinesis, a period of the cell cycle that has been difficult to study by cell fractionation.


Subject(s)
Cell Separation/methods , Cytokinesis/physiology , Flow Cytometry/methods , Aurora Kinases , Cyclin B/biosynthesis , Cyclin B1 , HeLa Cells , Humans , Mitosis/genetics , Mitosis/physiology , Protein Serine-Threonine Kinases/biosynthesis
12.
Mol Cell Biol ; 24(20): 8917-28, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15456866

ABSTRACT

Vertebrates express two A-type cyclins; both associate with and activate the CDK2 protein kinase. Cyclin A1 is required in the male germ line, but its molecular functions are incompletely understood. We observed specific induction of cyclin A1 expression and promoter activity after UV and gamma-irradiation which was mediated by p53. cyclin A1-/- cells showed increased radiosensitivity. To unravel a potential role of cyclin A1 in DNA repair, we performed a yeast triple hybrid screen and identified the Ku70 DNA repair protein as a binding partner and substrate of the cyclin A1-CDK2 complex. DNA double-strand break (DSB) repair was deficient in cyclin A1-/- cells. Further experiments indicated that A-type cyclins activate DNA DSB repair by mechanisms that depend on CDK2 activity and Ku proteins. Both cyclin A1 and cyclin A2 enhanced DSB repair by homologous recombination, but only cyclin A1 significantly activated nonhomologous end joining. DNA DSB repair was specific for A-type cyclins because cyclin E was ineffective. These findings establish a novel function for cyclin A1 and CDK2 in DNA DSB repair following radiation damage.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Cyclin A/metabolism , DNA Repair , DNA/metabolism , DNA/radiation effects , Gene Expression Regulation , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , CDC2-CDC28 Kinases/genetics , Cells, Cultured , Cyclin A/genetics , Cyclin A1 , Cyclin-Dependent Kinase 2 , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Hematopoietic Stem Cells , Humans , Ku Autoantigen , Macromolecular Substances , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Promoter Regions, Genetic , Random Allocation , Recombination, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Ultraviolet Rays
13.
Reproduction ; 127(4): 503-11, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047941

ABSTRACT

In higher eukaryotes, the cyclins constitute a family of proteins involved in progression through the cell cycle. The cyclin A1 gene (Ccna1) is expressed during meiosis and is required for spermatogenesis. Targeted disruption of the Ccna1 gene with a LacZ reporter gene has allowed us to study the expression pattern of this gene in more detail. We have confirmed expression in mouse pre-meiotic spermatocytes and also detected expression in the accessory olfactory bulb, hippocampus and amygdala of the adult brain. We have also found that the amount of cyclin A1 protein influences the fertility of male mice and its action is modulated by genetic background. On an outbred genetic background (129S6/SvEv x MF1), Ccna1 (tm1Col) -/- animals are sterile due to spermatogenic arrest prior to the first meiotic division while Ccna1 (tm1Col) +/- mice show reduced sperm production and fertility. This is even more pronounced on an inbred genetic background (129S6/SvEv) where Ccna1 (tm1Col) +/- male mice are sterile due to a severe reduction in the total number of sperm.


Subject(s)
Cyclin A/physiology , Fertility/physiology , Animals , Brain Chemistry , Cyclin A/analysis , Cyclin A1 , Haploidy , Homozygote , Male , Mice , Mice, Inbred C57BL , Oligospermia/metabolism , Organ Size/genetics , Seminiferous Tubules/chemistry , Sperm Count , Testis/anatomy & histology
14.
Oncogene ; 21(10): 1493-500, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11896577

ABSTRACT

Cyclin A2 is predominantly, but not exclusively, localized in the nucleus from G1/S transition onwards. It is degraded when cells enter mitosis after nuclear envelope breakdown. We previously showed that a fusion protein (S2A) between the hepatitis B virus (HBV) surface antigen protein and a non-degradable fragment of human cyclin A2 (Delta152) resides in the endoplasmic reticulum membranes, escapes degradation and transforms normal rat fibroblasts. The present study investigates whether cytoplasmic cyclin A2 may play a role in oncogenesis. We show that the sequestration of non-degradable cyclin A2-Delta152 by a cellular ER targeting domain (PRL-A2) leads to cell transformation when coexpressed with activated Ha-ras. REF52 cells constitutively expressing PRL-A2 are found to have a high incidence of multinucleate giant cells, polyploidy and abnormal centrosome numbers, giving rise to the nucleation of multipolar spindles. Injection of these cells into athymic nude mice causes tumors, even in the absence of a cooperating Ha-ras oncogene. These results demonstrate that, independently of any viral context, an intracellular redistribution of non-degradable cyclin A2 is capable of deregulating the normal cell cycle to the point where it promotes aneuploidy and cancer.


Subject(s)
Cell Transformation, Neoplastic , Centrosome/ultrastructure , Cyclin A/physiology , Endoplasmic Reticulum/chemistry , Polyploidy , Animals , Cell Cycle , Cells, Cultured , Cyclin A/genetics , Cyclin A/immunology , Cyclin A2 , Genes, ras , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasms, Experimental/etiology , Oncogene Proteins/physiology , Rats , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...