Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Oncogene ; 35(41): 5446-5455, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27065322

ABSTRACT

Cell cycle genes are often aberrantly expressed in cancer, but how their misexpression drives tumorigenesis mostly remains unclear. From S phase to early mitosis, EMI1 (also known as FBXO5) inhibits the anaphase-promoting complex/cyclosome, which controls cell cycle progression through the sequential degradation of various substrates. By analyzing 7403 human tumor samples, we find that EMI1 overexpression is widespread in solid tumors but not in blood cancers. In solid cancers, EMI1 overexpression is a strong prognostic marker for poor patient outcome. To investigate causality, we generated a transgenic mouse model in which we overexpressed Emi1. Emi1-overexpressing animals develop a wide variety of solid tumors, in particular adenomas and carcinomas with inflammation and lymphocyte infiltration, but not blood cancers. These tumors are significantly larger and more penetrant, abundant, proliferative and metastatic than control tumors. In addition, they are highly aneuploid with tumor cells frequently being in early mitosis and showing mitotic abnormalities, including lagging and incorrectly segregating chromosomes. We further demonstrate in vitro that even though EMI1 overexpression may cause mitotic arrest and cell death, it also promotes chromosome instability (CIN) following delayed chromosome alignment and anaphase onset. In human solid tumors, EMI1 is co-expressed with many markers for CIN and EMI1 overexpression is a stronger marker for CIN than most well-established ones. The fact that Emi1 overexpression promotes CIN and the formation of solid cancers in vivo indicates that Emi1 overexpression actively drives solid tumorigenesis. These novel mechanistic insights have important clinical implications.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinogenesis/genetics , Cell Cycle Proteins/biosynthesis , Chromosomal Instability/genetics , F-Box Proteins/biosynthesis , Neoplasms/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Mitosis/genetics , Neoplasms/pathology , Phosphorylation
2.
Curr Cancer Drug Targets ; 2(4): 337-53, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12470210

ABSTRACT

The anticancer properties of histone deacetylase inhibitors have been known for some time. However, it is only recently that the functional identities of the intracellular targets mediating the anticancer properties have started to be revealed. These targets appear to play significant roles in cell cycle control, apoptosis and differentiation. Importantly, the modulation of these activities is likely to be mediated by alterations in the acetylation status of both histone and non-histone targets. Identification of these targets, and the specific histone deacetylase enzymes that modulate them, is an important step in designing rational-based therapies for the treatment of cancer. In this review we discuss the state of progress in identifying the molecular pathways/events mediating the anticancer activity of histone deacetylase inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Animals , Clinical Trials as Topic/statistics & numerical data , Histone Deacetylases/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/enzymology
3.
Melanoma Res ; 12(6): 539-47, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12459643

ABSTRACT

While mutations of CDKN2A are associated with melanoma predisposition, the precise role of its gene product p16 in the development of sporadic melanoma is less clearly understood. We sought to determine the prevalence of p16 expression using immunohistochemical analysis in a population-based sample of melanoma tumours, and also to identify histological, phenotypic and environmental factors associated with the presence or absence of p16 expression. We conducted face-to-face interviews with 108 patients newly diagnosed with melanoma to ascertain their history of sun exposure, and recorded various phenotypic parameters. Paraffin sections of tumours from these patients were stained with an anti-p16 monoclonal antibody following antigen retrieval. Overall, 52 (48%) tumours expressed p16; nodular melanomas had significantly lower levels of p16 immunoreactivity than superficial spreading melanomas (P = 0.015). While no association was found between p16 expression and host phenotype, loss of p16 staining was associated with thicker lesions (p = 0.084) and a high mitotic index (P = 0.013). Taken together, these findings are consistent with loss of p16 being a late event in the progression of sporadic primary melanomas, being associated with tumours of a more aggressive nature.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Melanoma/metabolism , Melanoma/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Melanoma/genetics , Middle Aged , Mitotic Index , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Risk Factors , Skin Neoplasms/genetics
4.
Mol Pharmacol ; 60(4): 828-37, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562446

ABSTRACT

Histone deacetylase inhibitors show promise as chemotherapeutic agents and have been demonstrated to block proliferation in a wide range of tumor cell lines. Much of this antiproliferative effect has been ascribed to the up-regulated expression of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1). In this article, we report that p21 expression was up-regulated by relatively low doses of the histone deacetylase inhibitor azelaic bishydroxamic acid (ABHA) and correlated with a proliferative arrest. Higher doses of ABHA were cytotoxic. Cells that did not up-regulate p21 expression were hypersensitive to killing by ABHA and died via apoptosis, whereas up-regulation of p21 correlated with reduced sensitivity and a block in the apoptotic mechanism, and these cells seemed to die by necrosis. Using isogenic p21(+/+) and p21(-/-) cell lines and direct inhibition of caspase activity, we demonstrate that the reduced sensitivity to killing by ABHA is a consequence of inhibition of apoptosis by up-regulated p21 expression. These data indicate the enormous potential of therapeutic strategies that bypass the cytoprotective effect of p21 and act on the same molecular targets as the histone deacetylase inhibitors.


Subject(s)
Apoptosis , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Boron Compounds , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , HeLa Cells , Humans , Methacrylates , Methylmethacrylates , Tumor Cells, Cultured , Up-Regulation/drug effects
5.
J Biol Chem ; 276(25): 22491-9, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11304533

ABSTRACT

Use of specific histone deacetylase inhibitors has revealed critical roles for the histone deacetylases (HDAC) in controlling proliferation. Although many studies have correlated the function of HDAC inhibitors with the hyperacetylation of histones, few studies have specifically addressed whether the accumulation of acetylated histones, caused by HDAC inhibitor treatment, is responsible for growth inhibition. In the present study we show that HDAC inhibitors cause growth inhibition in normal and transformed keratinocytes but not in normal dermal fibroblasts. This was despite the observation that the HDAC inhibitor, suberic bishydroxamate (SBHA), caused a kinetically similar accumulation of hyperacetylated histones. This cell type-specific response to SBHA was not due to the inactivation of SBHA by fibroblasts, nor was it due to differences in the expression of specific HDAC family members. Remarkably, overexpression of HDACs 1, 4, and 6 in normal human fibroblasts resulted in cells that could be growth-inhibited by SBHA. These data suggest that, although histone acetylation is a major target for HDAC inhibitors, the accumulation of hyperacetylated histones is not sufficient to cause growth inhibition in all cell types. This suggests that growth inhibition, caused by HDAC inhibitors, may be the culmination of histone hyperacetylation acting in concert with other growth regulatory pathways.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/metabolism , Skin/drug effects , Acetylation , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/enzymology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , RNA, Messenger/metabolism , Skin/cytology , Skin/enzymology
6.
Exp Cell Res ; 257(1): 11-21, 2000 May 25.
Article in English | MEDLINE | ID: mdl-10854050

ABSTRACT

The activation of cdc2/cyclin B is the trigger for entry into mitosis. The mechanism of cdc2/cyclin B activation is complex, but the final step is the dephosphorylation of the Thr14 and Tyr15 residues on the cdc2 subunit, catalyzed by a member of the Cdc25 family of phosphatases. Cdc2/cyclin B1 accumulates at the centrosome in late G2 phase and has been implicated in the conversion of the centrosome from an interphase to a mitotic microtubule organizing center. Here we demonstrate biochemically that cdc2/cyclin B1 accumulates at the centrosome in late G2 as the inactive, phosphotyrosine 15 form and that the centrosomal cdc2/cyclin B1 can be activated in vitro by recombinant cdc25B. We provide evidence that a portion of the cdc2/cyclin B1 translocated into the nucleus in prophase is the inactive tyrosine-15-phosphorylated form. At this time the centrosomal and cytoplasmic cdc2/cyclin B1 is already active. This provides evidence that the activation of cdc2/cyclin B1 is initiated in the cytoplasm and that full activation of the translocated pool occurs in the nucleus.


Subject(s)
CDC2 Protein Kinase/metabolism , Centrosome/metabolism , Cyclin B/metabolism , Mitosis , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Centrosome/ultrastructure , Cyclin B1 , Enzyme Activation , Humans , Signal Transduction , Tumor Cells, Cultured
7.
Mol Biol Cell ; 11(6): 2069-83, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848630

ABSTRACT

Important aspects of cell cycle regulation are the checkpoints, which respond to a variety of cellular stresses to inhibit cell cycle progression and act as protective mechanisms to ensure genomic integrity. An increasing number of tumor suppressors are being demonstrated to have roles in checkpoint mechanisms, implying that checkpoint dysfunction is likely to be a common feature of cancers. Here we report that histone deacetylase inhibitors, in particular azelaic bishydroxamic acid, triggers a G2 phase cell cycle checkpoint response in normal human cells, and this checkpoint is defective in a range of tumor cell lines. Loss of this G2 checkpoint results in the tumor cells undergoing an aberrant mitosis resulting in fractured multinuclei and micronuclei and eventually cell death. This histone deacetylase inhibitor-sensitive checkpoint appears to be distinct from G2/M checkpoints activated by genotoxins and microtubule poisons and may be the human homologue of a yeast G2 checkpoint, which responds to aberrant histone acetylation states. Azelaic bishydroxamic acid may represent a new class of anticancer drugs with selective toxicity based on its ability to target a dysfunctional checkpoint mechanism in tumor cells.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Signal Transduction/drug effects , Cell Cycle/drug effects , G1 Phase/drug effects , G2 Phase/drug effects , HeLa Cells , Humans , Mitosis/drug effects , Tumor Cells, Cultured
8.
Int J Cancer ; 82(2): 305-12, 1999 Jul 19.
Article in English | MEDLINE | ID: mdl-10389768

ABSTRACT

CDKN2A appears to be the major melanoma susceptibility gene, and is also mutated/deleted in sporadic tumours of various types including melanoma. Thus far most approaches to assessing the functionality of mutations in this gene have used in vitro methods such as CDK4 binding and kinase inhibition assays, with sometimes disparate conclusions about functional significance of some variants between studies. We have used a melanoma cell line (MM96L) with no functional p16, as the basis for a "semi-in vivo" transfection-based assay for exogenous p16 functionality based on the growth parameters of the cells and the behaviour of variant proteins after transfection of different CDKN2A cDNAs. Colony counts performed on these transfectants revealed that all but the wild type, + 24 bp ad A148T variants have a diminished ability to inhibit cell growth. All other variants detected either constitutionally in familial melanoma patients (I49T, R87P, G101W and V126D) or somatically in melanomas (N71S, and P81L), appeared functionally impaired in this assay. This diminution of function was independent of CDK4 and CDK6 binding ability. Furthermore, the predominant localization of these variants within the cell was different from that of wt p16. This mislocalization may provide an explanation for their lack of function, or alternatively, it may also be an indicator that the cells are processing unstable, misfolded p16 proteins. This novel assay for assessment of functionality of p16 variants may better reflect the role of some of these mutations in vivo, and as such is a useful adjunct to other in vitro assays.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Genes, p16 , Melanoma/genetics , Neoplasm Proteins/deficiency , Cell Nucleus/chemistry , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cytoplasm/chemistry , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Fluorescent Antibody Technique, Indirect , Gene Deletion , Genetic Predisposition to Disease , Genetic Variation , Humans , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Polymorphism, Genetic , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Tumor Stem Cell Assay
9.
Biochem Biophys Res Commun ; 260(2): 510-5, 1999 Jul 05.
Article in English | MEDLINE | ID: mdl-10403798

ABSTRACT

Progression through G2 phase into mitosis is regulated by the activation of the mitotic cyclin/cdk complexes, which are in turn activated cdc25B and cdc25C phosphatases. Here we report that alternate splicing produces at least five variants of cdc25B, although only cdc25B2 and cdc25B3 are detectable as proteins. Analysis of these two variants shows that cdc25B2 is expressed at lower levels relative to cdc25B3 in all cell lines tested, and the expression of both increased markedly during G2 and mitosis. Overexpression of the catalytically inactive version of either cdc25B variant produced a G2 arrest implicating both in regulating G2/M progression.


Subject(s)
Cell Cycle Proteins/genetics , G2 Phase/genetics , Mitosis/genetics , Phosphoprotein Phosphatases/genetics , Protein Isoforms/genetics , RNA Splicing , cdc25 Phosphatases , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Cell Line , Humans , Molecular Weight , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/physiology , Protein Isoforms/chemistry , Protein Isoforms/physiology
10.
J Biol Chem ; 274(20): 13961-9, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318807

ABSTRACT

Cyclin D-Cdk4 complexes have a demonstrated role in G1 phase, regulating the function of the retinoblastoma susceptibility gene product (Rb). Previously, we have shown that following treatment with low doses of UV radiation, cell lines that express wild-type p16 and Cdk4 responded with a G2 phase cell cycle delay. The UV-responsive lines contained elevated levels of p16 post-treatment, and the accumulation of p16 correlated with the G2 delay. Here we report that in UV-irradiated HeLa and A2058 cells, p16 bound Cdk4 and Cdk6 complexes with increased avidity and inhibited a cyclin D3-Cdk4 complex normally activated in late S/early G2 phase. Activation of this complex was correlated with the caffeine-induced release from the UV-induced G2 delay and a decrease in the level of p16 bound to Cdk4. Finally, overexpression of a dominant-negative mutant of Cdk4 blocked cells in G2 phase. These data indicate that the cyclin D3-Cdk4 activity is necessary for cell cycle progression through G2 phase into mitosis and that the increased binding of p16 blocks this activity and G2 phase progression after UV exposure.


Subject(s)
Cyclin-Dependent Kinases/metabolism , G2 Phase/radiation effects , Proto-Oncogene Proteins , Ultraviolet Rays , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclins/metabolism , Enzyme Activation , Flow Cytometry , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , S Phase/radiation effects , Tumor Cells, Cultured
11.
Mutat Res ; 422(1): 43-53, 1998 Nov 09.
Article in English | MEDLINE | ID: mdl-9920427

ABSTRACT

Ultraviolet (UV) radiation contributes to the aetiology of melanoma, but the precise mechanistic details are still unclear. The CDKN2A gene which is associated with familial and sporadic melanoma, encodes a tumour suppressor, p16. We have previously shown that in response to low doses of UV radiation the level of p16 increases, and that this correlates with a G2 delay. Here we report that in melanoma cell lines which do not express p16, or express a mutant p16, no G2 delay is observed in response to UV. The loss of functional p16 also correlates with an increase in DNA damage as judged by increased numbers of bi- and multinuclear cells and cells containing 1-2 micronuclei following UV irradiation. This work provides a further link between UV radiation, CDKN2A and melanoma, suggesting that the functional inactivation of CDKN2A disrupts a p16-dependent G2 cell cycle checkpoint, thus contributing to the development of this neoplasm.


Subject(s)
Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Genes, p16 , Ultraviolet Rays , Cell Cycle/physiology , Cell Division/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/radiation effects , Gene Deletion , HeLa Cells , Humans , Keratinocytes/cytology , Keratinocytes/radiation effects , Kinetics , Melanoma/genetics , Time Factors , Tumor Cells, Cultured
12.
Cancer Res ; 57(21): 4868-75, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9354451

ABSTRACT

The CDKN2A gene maps to chromosome 9p21-22 and is responsible for melanoma susceptibility in some families. Its product, p16, binds specifically to CDK4 and CDK6 in vitro and in vivo, inhibiting their kinase activity. CDKN2A is homozygously deleted or mutated in a large proportion of tumor cell lines and some primary tumors, including melanomas. The aim of this study was to investigate the involvement of CDKN2A and elucidate the mechanisms of p16 inactivation in a panel of 60 cell lines derived from sporadic melanomas. Twenty-six (43%) of the melanoma lines were homozygously deleted for CDKN2A, and an additional 15 (25%) lines carried missense, nonsense, or frameshift mutations. All but one of the latter group were shown by microsatellite analysis to be hemizygous for the region of 9p surrounding CDKN2A. p16 was detected by Western blotting in only five of the cell lines carrying mutations. Immunoprecipitation of p16 in these lines, followed by Western blotting to detect the coprecipitation of CDK4 and CDK6, revealed that p16 was functionally compromised in all cell lines but the one that carried a heterozygous CDKN2A mutation. In the remaining 19 lines that carried wild-type CDKN2A alleles, Western blot analysis and immunoprecipitation indicated that 11 cell lines expressed a wild-type protein. Northern blotting was performed on the remaining eight cell lines and revealed that one cell line carried an aberrantly sized RNA transcript, and two other cell lines failed to express RNA. The promoter was found to be methylated in five cell lines that expressed CDKN2A transcript but not p16. Presumably, the message seen by Northern blotting in these cell lines is the result of cross-hybridization of the total cDNA probe with the exon 1beta transcript. Microsatellite analysis revealed that the majority of these cell lines were hemi/homozygous for the region surrounding CDKN2A, indicating that the wild-type allele had been lost. In the 11 cell lines that expressed functional p16, microsatellite analysis revealed loss of heterozygosity at the markers immediately surrounding CDKN2A in five cases, and the previously characterized R24C mutation of CDK4 was identified in one of the remaining 6 lines. These data indicate that 55 of 60 (92%) melanoma cell lines demonstrated some aberration of CDKN2A or CDK4, thus suggesting that this pathway is a primary genetic target in melanoma development.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Gene Deletion , Genes, p16/genetics , Melanoma/genetics , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Neoplasm Proteins/metabolism , Sequence Deletion , Tumor Cells, Cultured
13.
J Biol Chem ; 272(45): 28607-14, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9353326

ABSTRACT

Cdc25 regulates entry into mitosis by regulating the activation of cyclin B/cdc2. In humans, at least two cdc25 isoforms have roles in controlling the G2/M transition. Here we show, using bacterially expressed recombinant proteins, that two cdc25B splice variants, cdc25B2 and cdc25B3, are capable of activating cyclin A/cdk2 and cyclin B/cdc2, but that mitotic hyperphosphorylation of these proteins increases their activity toward only cyclin B1/cdc2. Cdc25C has only very low activity in its unphosphorylated form, and following hyperphosphorylation it will efficiently catalyze the activation of only cyclin B/cdc2. This was reflected by the in vivo activity of the immunoprecipitated cdc25B and cdc25C from interphase and mitotic HeLa cells. The increased activity of the hyperphosphorylated cdc25s toward cyclin B1/cdc2 was in large part due to increased binding of this substrate. The substrate specificity, activities, and timing of the hyperphosphorylation of cdc25B and cdc25C during G2 and M suggest that these two mitotic cdc25 isoforms are activated by different kinases and perform different functions during progression through G2 into mitosis.


Subject(s)
CDC2 Protein Kinase/metabolism , CDC2-CDC28 Kinases , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , Cyclin-Dependent Kinases/metabolism , GTP-Binding Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/genetics , Cyclin B1 , Cyclin-Dependent Kinase 2 , HeLa Cells , Humans , Interphase , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Recombinant Fusion Proteins/metabolism , cdc25 Phosphatases
14.
Oncogene ; 15(7): 749-58, 1997 Aug 14.
Article in English | MEDLINE | ID: mdl-9266961

ABSTRACT

In response to low doses of ultraviolet (U.V.) radiation, cells undergo a G2 delay. In this study we have shown that the G2 delay results in the accumulation of inactive forms of cyclin B1/cdc2 and both the G2 and mitotic complexes of cyclin A/cdk. This appears to be through a block in the cdc25-dependent activation of these complexes. The expression and localisation of cyclin A and cyclin B1/cdk complexes are similar in U.V.-induced G2 delay and normal early G2 phase cells. Cdc25B and cdc25C also accumulate to normal G2 levels in U.V. irradiated cells, but the mitotic phosphorylation associated with increased activity of both cdc25B and cdc25C is absent. The cdc25B accumulates in the nucleus of U.V. irradiated cells and in normal G2 phase cells. Thus the block in cyclin B/cdc2 activation is in part due to the physical separation of cyclin B/cdc2, localised in the cytoplasm, from the cdc25B and cdc25C phosphatases localised in the nucleus. The data positions the U.V.-induced G2 checkpoint at either the S/G2 transition or early G2 phase, prior to the activation of cyclin A/cdk2.


Subject(s)
CDC2 Protein Kinase/radiation effects , Cell Cycle Proteins/radiation effects , Cyclin B , Cyclins/radiation effects , G2 Phase/radiation effects , Phosphoprotein Phosphatases/radiation effects , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , HeLa Cells/metabolism , HeLa Cells/radiation effects , Humans , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , S Phase/radiation effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Ultraviolet Rays , cdc25 Phosphatases
15.
Int J Cancer ; 74(4): 407-15, 1997 Aug 22.
Article in English | MEDLINE | ID: mdl-9291430

ABSTRACT

Paraffin sections (n = 168, 27 benign, 16 low malignant potential [LMP] and 125 malignant tumours) from epithelial ovarian tumours were evaluated immunohistochemically for expression of retinoblastoma gene product (pRB) and p53 protein, and the relationship among pRB, p53 and cyclin-dependent kinase inhibitor 2 (CDKN2) gene product p16INK4A (p16) was analysed, following our previous study of p16. Forty-one percent of the benign, 50% of the LMP and most (71%) of the malignant tumours showed high pRB expression. High expression of pRB (>50% pRB-positive cells) significantly correlated with non-mucinous histological subtypes. Reduced pRB expression, substage and residual disease were significant predictors for poor prognosis in stage I patients. All the benign and most of the LMP (81%) tumours were in either the p53-negative or low p53-positive category, but nearly half of the malignant tumours had high p53 expression. High p53 accumulation was found in non-mucinous, high grade and late stage tumours. For well-differentiated carcinomas, high p53 expression was a predictor of poor prognosis. However, even though high p53 expression was not associated with histological subtype, stage or the presence of residual disease, high p53 expression was not an independent predictor when all clinical parameters were combined. For all ovarian cancers, a close correlation was found between high p53 and high p16 expression. The relationship between the expression of pRB and p16 depended on tumour stage. In stage I tumours, high pRB was associated with low p16 reactivity. On the other hand, most advanced tumours showed both high pRB and high p16 reactivity.


Subject(s)
Ovarian Neoplasms/pathology , Retinoblastoma Protein/analysis , Tumor Suppressor Protein p53/analysis , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16 , Female , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Life Tables , Neoplasm Invasiveness , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/surgery , Predictive Value of Tests , Prognosis , Regression Analysis , Retinoblastoma Protein/biosynthesis , Survival Rate , Time Factors , Tumor Suppressor Protein p53/biosynthesis
16.
J Invest Dermatol ; 109(1): 61-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9204956

ABSTRACT

CDKN2A is a melanoma susceptibility gene that is mutated and/or deleted in familial and sporadic melanoma as well as in a range of other tumors. It encodes a cell cycle regulator, p16, whose function is to inhibit activity of cyclin-dependent kinases 4 and 6. We set out to investigate the effect of reintroducing CDKN2A into MM96L, a melanoma cell line that does not express p16, by electroporation of wt CDKN2A cDNA. Our results show that transfection of the CDKN2A cDNA has a dramatic effect on cell morphology, inducing a more dendritic phenotype resembling that of adult melanocytes. This effect on cell morphology was not cell line specific because it was reproduced in another melanoma line (SK-MEL-13), which has a homozygous deletion of CDKN2A. It was abolished by mutations that abrogate p16 function, as shown by transfection of a Pro81Leu p16 variant. Reintroduction of levels of p16 protein similar to those of cultured neonatal foreskin melanocytes decreased the growth rate of the transfected clones. Surprisingly, we did not see any effect on anchorage-independent growth or on the following melanoma markers tested by western blotting: p21/WAF1, tyrosinase-related antigen 1, HMB45, and intermediate filament antigen. These data indicate that reintroduction into melanoma cells of wild type p16 at levels similar to cultured melanocytes can induce morphologic changes and suppress growth but is not sufficient to affect anchorage-independent growth.


Subject(s)
Carrier Proteins/genetics , Aged , Biomarkers, Tumor/analysis , Clone Cells/metabolism , Cyclin-Dependent Kinase Inhibitor p16 , Enzyme Inhibitors/pharmacology , Female , Gene Expression , Genes, Tumor Suppressor , Genetic Variation , Humans , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors , Transfection , Tumor Cells, Cultured/drug effects
17.
Int J Cancer ; 74(1): 57-63, 1997 Feb 20.
Article in English | MEDLINE | ID: mdl-9036870

ABSTRACT

Paraffin sections from 190 epithelial ovarian tumours, including 159 malignant and 31 benign epithelial tumours, were analysed immunohistochemically for expression of cyclin-dependent kinase inhibitor 2 (CDKN2A) gene product p16INK4A (p16). Most benign tumours showed no p16 expression in the tumour cells, whereas only 11% of malignant cancers were p16 negative. A high proportion of p16-positive tumour cells was associated with advanced stage and grade, and with poor prognosis in cancer patients. For FIGO stage I tumours, a high proportion of p16-positive tumour cells was associated with poorer survival, suggesting that accumulation of p16 is an early event of ovarian tumorigenesis. In contrast to tumour cells, high expression of p16 in the surrounding stromal cells was not associated with the stage and grade, but was associated with longer survival. When all parameters were combined in a multivariate analysis, high p16 expression in stromal cells was not an independent predictor for survival, indicating that low p16 expression in stromal cells is associated with other markers of tumour progression. High expression of p16 in the stromal cells of tumours from long-term survivors suggests that tumour growth is limited to some extent by factors associated with p16 expression in the matrix.


Subject(s)
Carrier Proteins/analysis , Ovarian Neoplasms/pathology , Biomarkers, Tumor/analysis , Blotting, Western , Carrier Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16 , Disease Progression , Female , Genes, Tumor Suppressor , HeLa Cells , Humans , Immunohistochemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Ovarian Neoplasms/surgery , Prognosis , Survival Rate , Time Factors
18.
Anal Biochem ; 254(2): 231-5, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9417782

ABSTRACT

Study of the function and regulation of the important cell cycle regulator cdc25 phosphatase has been hampered by the lack of a sensitive and specific substrate and assay. Here we report the production of a specific and sensitive substrate for the cdc25 phosphatase. The substrate is human cyclin B1/cdc2 phosphorylated on the inhibitory Thr14 and Tyr15 residues and activating Thr161 on cdc2, and is relatively simple to produce from readily available materials. The assay is based on the cdc25-specific dephosphorylation and activation of the phosphorylated cyclin B1/cdc2 substrate (PY15), using the increased histone H1 kinase activity of the activated PY15 as a read-out of cdc25 activity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , CDC2 Protein Kinase/biosynthesis , Cell Cycle Proteins/biosynthesis , Cell Fractionation , Cyclin B/biosynthesis , Cyclin B1 , Glutathione Transferase/metabolism , HeLa Cells , Humans , Phosphoprotein Phosphatases/biosynthesis , Phosphothreonine/metabolism , Phosphotyrosine/metabolism , Recombinant Fusion Proteins/metabolism , Solubility , Substrate Specificity , Xenopus , cdc25 Phosphatases
19.
Cancer Res ; 56(11): 2510-4, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8653687

ABSTRACT

p16 is the product of the CDKN2A locus, which is mutated or deleted in many human tumors. In response to nonlethal UVC irradiation, HeLa cells accumulate elevated levels of p16. The accumulation of p16 is delayed 8-12 h following irradiation and correlates with S-phase and G2 delays, decreasing as the cells recover and recommence normal cell growth. The maximum levels of p16 correlated with G2 delay. The UVC-induced cell cycle delay was absent in cell lines derived from HeLa that did not express p16 and in a melanoma line deleted for p16.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle/radiation effects , HeLa Cells/radiation effects , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor p16 , DNA Repair , Dose-Response Relationship, Radiation , Fluorescent Antibody Technique, Indirect , Genes, Tumor Suppressor , HeLa Cells/cytology , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Ultraviolet Rays
20.
J Cell Sci ; 109 ( Pt 5): 1081-93, 1996 May.
Article in English | MEDLINE | ID: mdl-8743955

ABSTRACT

The formation of the mitotic spindle is an essential prerequisite for successful mitosis. The dramatic changes in the level of microtubule (Mt) nucleation at the centrosomes and Mt dynamics that occur in prophase are presumed to be initiated through the activity of cdc2/cyclin B. Here we present data that the cdc25B isoform functions to activate the cytoplasmic pool of cdc2/cyclin B responsible for these events. In contrast to cdc25C, cdc25B is present at low levels in HeLa cells during interphase, but sharply increases in prophase, when cdc25B accumulation in the cytoplasm correlates with prophase spindle formation. Overexpression of wild type and dominant negative mutants of cdc25B and cdc25C shows that prophase Mt nucleation is a consequence of cytoplasmic cdc25B activity, and that cdc25C regulates nuclear G2/M events. Our data also suggest that the functional status of the centrosome can regulate nuclear mitotic events.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/ultrastructure , Cytoplasm/enzymology , G2 Phase/physiology , Metaphase/physiology , Microtubules/ultrastructure , Phosphoprotein Phosphatases/metabolism , Spindle Apparatus , cdc25 Phosphatases , Antibodies, Monoclonal , Base Sequence , Genes, Dominant , HeLa Cells , Humans , Molecular Sequence Data , Prophase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...