Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Div ; 7(1): 2, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22305267

ABSTRACT

BACKGROUND: Aneuploidy has been acknowledged as a major source of genomic instability in cancer, and it is often considered the result of chromosome segregation errors including those caused by defects in genes controlling the mitotic spindle assembly, centrosome duplication and cell-cycle checkpoints. Aneuploidy and chromosomal instability has been also correlated with epigenetic alteration, however the molecular basis of this correlation is poorly understood. RESULTS: To address the functional connection existing between epigenetic changes and aneuploidy, we used RNA-interference to silence the DNMT1 gene, encoding for a highly conserved member of the DNA methyl-transferases. DNMT1 depletion slowed down proliferation of near-diploid human tumor cells (HCT116) and triggered G1 arrest in primary human fibroblasts (IMR90), by inducing p53 stabilization and, in turn, p21waf1 transactivation. Remarkably, p53 increase was not caused by DNA damage and was not observed after p14-ARF post-transcriptional silencing. Interestingly, DNMT1 silenced cells with p53 or p14-ARF depleted did not arrest in G1 but, instead, underwent DNA hypomethylation and became aneuploid. CONCLUSION: Our results suggest that DNMT1 depletion triggers a p14ARF/p53 dependent cell cycle arrest to counteract the aneuploidy induced by changes in DNA methylation.

2.
J Cell Physiol ; 227(9): 3324-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22170163

ABSTRACT

The spindle assembly checkpoint (SAC) is a cellular surveillance mechanism that ensures faithful chromosome segregation during mitosis and its failure can result in aneuploidy. Previously, it was suggested that reduction of the MAD2 gene, encoding a major component of the SAC, induced aneuploidy in human tumor cells. However, tumor cell lines contain multiple mutations that might affect or exacerbate the cellular response to Mad2 depletion. Thus, the scenario resulting by Mad2 depletion in primary human cells could be different and more complex that the one depicted so far. We used primary human fibroblasts (IMR90) and epithelial breast cells (MCF10A) to gain further insight on the effects of genomic instability caused by transient Mad2 depletion. To this aim we depleted Mad2 by RNAi to a level shown by Mad2 haplo-insufficient cells and found that induced aneuploidy caused premature cellular senescence in IMR90 cells. IMR90 cells showed typical features of senescent cells, like senescence-associated (SA) beta galactosidase expression, including up-regulation of p53 and p14ARF proteins and of p21(waf1) as well, but not of p16(INK4A) cyclin-dependent kinase (Cdk) inhibitor. In contrast, after MAD2 post-transcriptional silencing MCF10A cells in which the INK4A/ARF locus is deleted, showed both aneuploidy and a small increase of p53 and p21(waf1) proteins, but not premature cellular senescence. Finally, our results provides an explanation of how a p53 controlled pathway, involving initially p21(waf1) and then p14ARF, could minimize the occurrence of genomic alterations derived from chromosome instability induced by low amounts of MAD2 protein.


Subject(s)
Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Cellular Senescence/genetics , M Phase Cell Cycle Checkpoints/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/metabolism , Aneuploidy , Calcium-Binding Proteins/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromosomal Instability/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Mad2 Proteins , RNA, Small Interfering/genetics , Repressor Proteins/metabolism , Signal Transduction , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics , beta-Galactosidase/genetics
3.
Cell Cycle ; 9(20): 4174-82, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20948316

ABSTRACT

Highly Expressed in Cancer protein 1 (Hec1) is a subunit of the Ndc80 complex, a constituent of the mitotic kinetochore. HEC1 has been shown to be overexpressed in many cancers, suggesting that HEC1 upregulation is involved in the generation and/or maintenance of the tumour phenotype. However, the regulation of Hec1 expression in normal and tumour cells and the molecular alterations promoting accumulation of this protein in cancer cells are still unknown. Here we show that elevated Hec1 protein levels are characteristic of transformed cell lines of different origins and that kinetochore recruitment of this protein is also increased in cancer cell lines in comparison with normal human cells. Using different cell synchronization strategies, Hec1 expression was found to be tightly regulated during the cell cycle in both normal and cancer cells. A limited proteasome-dependent degradation of Hec1 cellular content was observed at mitotic exit, with no evident differences between normal and cancer cells. Interestingly, increased expression of HEC1 mRNA and Hec1 protein was observed after transient silencing of the retinoblastoma gene by siRNA or following microRNA-mediated permanent depletion of the retinoblastoma protein in HCT116 cells. Our data provide evidence for a functional link between Hec1 expression and the pRb pathway. These observations suggest that disruption of pRb function may lead to chromosome segregation errors and mitotic defects through Hec1 overexpression. This may importantly contribute to aneuploidy and chromosomal instability in RB-defective cancer cells.


Subject(s)
Cell Cycle/physiology , Neoplasms/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Cytoskeletal Proteins , Gene Silencing , Humans , Kinetochores/metabolism , Neoplasms/genetics , Nuclear Proteins/genetics , RNA Interference , Retinoblastoma Protein/genetics
4.
Mol Cancer ; 8: 119, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-20003272

ABSTRACT

BACKGROUND: Aneuploidy is a hallmark of most human cancers that arises as a consequence of chromosomal instability and it is frequently associated with centrosome amplification. Functional inactivation of the Retinoblastoma protein (pRb) has been indicated as a cause promoting chromosomal instability as well centrosome amplification. However, the underlying molecular mechanism still remains to be clarified. RESULTS: Here we show that pRb depletion both in wild type and p53 knockout HCT116 cells was associated with the presence of multipolar spindles, anaphase bridges, lagging chromosomes and micronuclei harbouring whole chromosomes. In addition aneuploidy caused by pRb acute loss was not affected by p53 loss.Quantitative real-time RT-PCR showed that pRB depletion altered expression of genes involved in centrosome duplication, kinetochore assembly and in the Spindle Assembly Checkpoint (SAC). However, despite MAD2 up-regulation pRb-depleted cells seemed to have a functional SAC since they arrested in mitosis after treatments with mitotic poisons. Moreover pRb-depleted HCT116 cells showed BRCA1 overexpression that seemed responsible for MAD2 up-regulation.Post-transcriptional silencing of CENPA by RNA interference, resulting in CENP-A protein levels similar to those present in control cells greatly reduced aneuploid cell numbers in pRb-depleted cells. CONCLUSION: Altogether our findings indicate a novel aspect of pRb acute loss that promotes aneuploidy mainly by inducing CENPA overexpression that in turn might induce micronuclei by affecting the correct attachment of spindle microtubules to kinetochores.


Subject(s)
Autoantigens/genetics , Chromosomal Proteins, Non-Histone/genetics , Genomic Instability , Retinoblastoma Protein/genetics , Autoantigens/physiology , Base Sequence , Blotting, Western , Cell Line, Tumor , Centromere Protein A , Chromosomal Proteins, Non-Histone/physiology , DNA Primers , Humans , Microscopy, Fluorescence , RNA Interference , RNA Processing, Post-Transcriptional , Reverse Transcriptase Polymerase Chain Reaction
5.
BMC Cell Biol ; 10: 79, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19883508

ABSTRACT

BACKGROUND: Changes in chromosome number or structure as well as supernumerary centrosomes and multipolar mitoses are commonly observed in human tumors. Thus, centrosome amplification and mitotic checkpoint dysfunctions are believed possible causes of chromosomal instability. The Retinoblastoma tumor suppressor (RB) participates in the regulation of synchrony between DNA synthesis and centrosome duplication and it is involved in transcription regulation of some mitotic genes. Primary human fibroblasts were transfected transiently with short interfering RNA (siRNA) specific for human pRb to investigate the effects of pRb acute loss on chromosomal stability. RESULTS: Acutely pRb-depleted fibroblasts showed altered expression of genes necessary for cell cycle progression, centrosome homeostasis, kinetochore and mitotic checkpoint proteins. Despite altered expression of genes involved in the Spindle Assembly Checkpoint (SAC) the checkpoint seemed to function properly in pRb-depleted fibroblasts. In particular AURORA-A and PLK1 overexpression suggested that these two genes might have a role in the observed genomic instability. However, when they were post-transcriptionally silenced in pRb-depleted fibroblasts we did not observe reduction in the number of aneuploid cells. This finding suggests that overexpression of these two genes did not contribute to genomic instability triggered by RB acute loss although it affected cell proliferation. Acutely pRb-depleted human fibroblasts showed the presence of micronuclei containing whole chromosomes besides the presence of supernumerary centrosomes and aneuploidy. CONCLUSION: Here we show for the first time that RB acute loss triggers centrosome amplification and aneuploidy in human primary fibroblasts. Altogether, our results suggest that pRb-depleted primary human fibroblasts possess an intact spindle checkpoint and that micronuclei, likely caused by mis-attached kinetochores that in turn trigger chromosome segregation errors, are responsible for aneuploidy in primary human fibroblasts where pRb is acutely depleted.


Subject(s)
Aneuploidy , Cell Nucleus/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cells, Cultured , Centrosome/metabolism , Chromosomal Instability , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mitosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Retinoblastoma Protein/genetics , Polo-Like Kinase 1
6.
Oncol Res ; 17(3): 115-25, 2008.
Article in English | MEDLINE | ID: mdl-18669163

ABSTRACT

Aurora-A is a centrosome-associated serine/threonine kinase that is overexpressed in multiple types of human tumors. Primarily, Aurora-A functions in centrosome maturation and mitotic spindle assembly. Overexpression of Aurora-A induces centrosome amplification and G2/M cell cycle progression. Recently, it was observed that overexpression of Aurora-A renders cells resistant to cisplatin (CDDP)-, etoposide-, and paclitaxel-induced apoptosis. Our results indicate that already in initial stages of cancer progression Aurora-A overexpression could have a major role in inducing supernumerary centrosomes and aneuploidy, as shown by immunohistochemistry on tissue sections from various stages of human colon cancer. Aneuploidy was also observed after Aurora-A ectopic overexpression in colon cancer cells with MIN phenotype. Silencing of Aurora-A by RNA interference in tumor cell lines triggered arrest of the cell cycle associated to apoptosis/ mitotic catastrophe. Finally, Aurora-A transcriptional silencing seems to confer cancer cells a greater sensitivity to chemotherapy by vincristine, indicating Aurora-A as a possible gene target in cancer therapy.


Subject(s)
Apoptosis/drug effects , Carcinoma/genetics , Cell Cycle/drug effects , Colonic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Aneuploidy , Antineoplastic Agents, Phytogenic/administration & dosage , Aurora Kinases , Carcinoma/drug therapy , Carcinoma/pathology , Cell Line, Tumor , Centrosome/chemistry , Centrosome/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Synergism , Gene Expression Regulation, Neoplastic , Gene Silencing , HeLa Cells , Humans , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transcription, Genetic , Transfection , Vincristine/administration & dosage
7.
BMC Cancer ; 7: 212, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17999753

ABSTRACT

BACKGROUND: Genetic instability is a hallmark of tumours and preneoplastic lesions. The predominant form of genome instability in human cancer is chromosome instability (CIN). CIN is characterized by chromosomal aberrations, gains or losses of whole chromosomes (aneuploidy), and it is often associated with centrosome amplification. Centrosomes control cell division by forming a bipolar mitotic spindle and play an essential role in the maintenance of chromosomal stability.However, whether centrosome amplification could directly cause aneuploidy is not fully established. Also, alterations in genes required for mitotic progression could be involved in CIN.A major candidate is represented by Aurora-A/STK15 that associates with centrosomes and is overexpressed in several types of human tumour. METHODS: Centrosome amplification were induced by hydroxyurea treatment and visualized by immunofluorescence microscopy. Aurora-A/STK15 ectopic expression was achieved by retroviral infection and puromycin selection in HCT116 tumour cells. Effects of Aurora-A/STK15 depletion on centrosome status and ploidy were determined by Aurora-A/STK15 transcriptional silencing by RNA interference. Changes in the expression levels of some mitotic genes were determined by Real time RT-PCR. RESULTS: We investigated whether amplification of centrosomes and overexpression of Aurora-A/STK15 induce CIN using as a model system a colon carcinoma cell line (HCT116). We found that in HCT116 cells, chromosomally stable and near diploid cells harbouring a MIN phenotype, centrosome amplification induced by hydroxyurea treatment is neither maintained nor induces aneuploidy. On the contrary, ectopic overexpression of Aurora-A/STK15 induced supernumerary centrosomes and aneuploidy. Aurora-A/STK15 transcriptional silencing by RNA interference in cells ectopically overexpressing this kinase promptly decreased cell numbers with supernumerary centrosomes and aneuploidy. CONCLUSION: Our results show that centrosome amplification alone is not sufficient to induce chromosomal instability in colon cancer cells with a MIN phenotype. Alternatively, centrosome amplification has to be associated with alterations in genes regulating mitosis progression such as Aurora-A/STK15 to trigger CIN.


Subject(s)
Cell Transformation, Neoplastic/genetics , Centrosome , Chromosomal Instability , Colonic Neoplasms/genetics , Microsatellite Instability , Protein Serine-Threonine Kinases/genetics , Aneuploidy , Apoptosis , Aurora Kinase A , Aurora Kinases , Cell Communication , Cell Differentiation , Colonic Neoplasms/metabolism , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Phenotype , Ploidies , Protein Serine-Threonine Kinases/biosynthesis , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Spindle Apparatus , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL