Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Oncogene ; 32(30): 3543-51, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-22907427

ABSTRACT

Protein kinase A (PKA)-induced estrogen receptor alpha (ERα) phosphorylation at serine residue 305 (ERαS305-P) can induce tamoxifen (TAM) resistance in breast cancer. How this phospho-modification affects ERα specificity and translates into TAM resistance is unclear. Here, we show that S305-P modification of ERα reprograms the receptor, redirecting it to new transcriptional start sites, thus modulating the transcriptome. By altering the chromatin-binding pattern, Ser305 phosphorylation of ERα translates into a 26-gene expression classifier that identifies breast cancer patients with a poor disease outcome after TAM treatment. MYC-target genes and networks were significantly enriched in this gene classifier that includes a number of selective targets for ERαS305-P. The enhanced expression of MYC increased cell proliferation in the presence of TAM. We demonstrate that activation of the PKA signaling pathway alters the transcriptome by redirecting ERα to new transcriptional start sites, resulting in altered transcription and TAM resistance.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Genes, Neoplasm , Promoter Regions, Genetic , Tamoxifen/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/drug effects , Humans , Phosphorylation , Protein Binding
2.
J Pathol ; 217(3): 372-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18991335

ABSTRACT

Phosphorylation of oestrogen receptor alpha at serine 305 (ERalphaS305-P) induces tamoxifen resistance in experimental studies, but does not influence response to other endocrine agents, such as fulvestrant. We evaluated ERalphaS305-P using immunohistochemistry in 377 breast carcinomas from premenopausal participants of a randomized trial (n=248) and patients with advanced disease (n=129). Among the premenopausal patients, adjuvant tamoxifen improved recurrence-free survival (RFS) for ERalphaS305-P-negative tumours (multivariate HR=0.53, 95% CI 0.32-0.86, p=0.010), but not for ERalphaS305-P-positive tumours (multivariate HR=1.01, 95% CI 0.33-3.05, p=0.99) (interaction p=0.131). Notably, ERalphaS305-P was not significantly associated with RFS in patients not treated with tamoxifen (multivariate HR=0.64, 95% CI 0.30-1.37, p=0.248), indicating that ERalphaS305-P is a marker for treatment outcome rather than tumour progression. Given the direct experimental link between ERalphaS305-P and tamoxifen resistance and these first clinical data suggesting that premenopausal patients with ERalphaS305-P-positive breast cancer are resistant to adjuvant tamoxifen, further research is encouraged to study whether alternative endocrine treatment should be considered for this subgroup.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Serine/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/analysis , Blotting, Western/methods , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Cell Line, Tumor , Estrogen Receptor alpha/analysis , Female , Humans , Immunohistochemistry , Phosphorylation , Retrospective Studies , Tamoxifen/therapeutic use , Tissue Array Analysis , Treatment Outcome
4.
Eur J Cancer ; 38(13): 1775-82, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12175695

ABSTRACT

The Rho-like guanine triphosphate (GTP)ases become activated by extracellular ligands and regulate a wide variety of biological processes, including cell motility, spreading of cells, cytoskeletal organisation and transcriptional activity. We studied the effect of expression of WtRac and Cdc42 and of their constitutive active V12 variants on cell cycle transition using the isopropylthiogalactoside (IPTG) inducible Rac and Cdc42 transfectants of porcine aortic endothelial (PAE) cells. Expression of V12Rac or V12Cdc42 resulted initially in an enrichment of cells in G2/M, followed by the appearance of multinucleated cells with some of the nuclei still being able to incorporate bromodeoxyuridine (BrdU). By fluorescent activated cell sorter (FACS) analysis, these cells appeared as polyploid cells. Prolonged activation of V12Rac or V12Cdc42 resulted in genomic instability and these cells finally detached from the culture plate. These findings indicate that induction of the constitutive active V12 forms of Rac and Cdc42 results in 'mitotic slippage', where endoreplication takes place irrespective of the exit from cytokinesis.


Subject(s)
GTP Phosphohydrolases/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Division , Cell Line , DNA/biosynthesis , Fluorescent Antibody Technique , Swine
5.
Br J Cancer ; 86(3): 402-8, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11875707

ABSTRACT

Overexpression of G1-S regulators cyclin D1 or cyclin A is frequently observed in breast cancer and is also to result in ligand-independent activation of oestrogen receptor in vitro. This might therefore, provide a mechanism for failure of tamoxifen treatment. We examined by immunohistochemical staining the effect of deregulation of these, and other cell cycle regulators on tamoxifen treatment in a group of 394 patients with early stage breast cancer. In univariate analysis, expression of cyclin A, Neu, Ki-67 index, and lack of OR expression were significantly associated with worse prognosis. When adjusted by the clinical model (for lymph node status, age, performance status, T-classification, grade, prior surgery, oestrogen receptor status and tamoxifen use), only overexpression of cyclin A and Neu were significantly associated with worse prognosis with hazard ratios of, respectively, 1.709 (P=0.0195) and 1.884 (P=0.0151). Overexpression of cyclin A was found in 86 out of the 201 OR-positive cases treated with tamoxifen, and was the only independent marker associated with worse prognosis (hazard ratio 2.024, P=0.0462). In conclusion, cyclin A is an independent predictor of recurrence of early stage breast cancer and is as such a marker for response in patients treated with tamoxifen.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin A/analysis , Tamoxifen/therapeutic use , Aged , Analysis of Variance , Biomarkers/analysis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle , Female , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Staging , Prognosis , Receptors, Estrogen/analysis , Survival Rate , Time Factors
6.
Breast Cancer Res Treat ; 67(1): 23-33, 2001 May.
Article in English | MEDLINE | ID: mdl-11518463

ABSTRACT

Overexpression of the cyclin D1 (CCND1) gene, encoding a downstream effector of mitogenic signals that plays a central role in G1 phase progression, is often found in cancerous cells. In sporadic breast cancer (BC), this is one of the most frequent and early genetic lesions identified so far, found in more than 50% of the tumors. Inhibitors of the mevalonate/protein prenylation pathway belong to a new family of cancer therapeutic agents that act by blocking intracellular mitogenic signal transduction pathways, thereby preventing expansion of pre-cancerous foci and inhibiting growth of transformed cells. It is not known at present whether constitutively high intracellular levels of cyclin D1 might interfere with the cytostatic actions of mevalonate/protein prenylation inhibitors. This possibility was investigated here by assessing the cell cycle effects of Simvastatin, a non-toxic upstream inhibitor of the mevalonate pathway, on human BC MCF-7 cells expressing either normal or enhanced levels of cyclin D1 from of a stably transfected, tet-inducible expression vector. Results show that constitutive overexpression of this protein, such as that found in sporadic BCs, does not influence the growth inhibitory effects of Simvastatin in vitro. In addition, D1-overexpressing embryo fibroblasts were also found to be responsive to the cell cycle effects of mevalonate/protein prenylation pathway blockade, further suggesting that high intracellular levels of cyclin D1 do not prevent the cytostatic actions of compounds targeting this metabolic pathway.


Subject(s)
Breast Neoplasms/pathology , Cell Division/drug effects , Cyclin D1/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Neoplasm Proteins/metabolism , Protein Prenylation/drug effects , Simvastatin/therapeutic use , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Humans , Mevalonic Acid , Rats
7.
J Pathol ; 194(3): 327-33, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11439365

ABSTRACT

There is increasing evidence that there are different progression routes leading to invasive breast cancer, depending on histology and differentiation grade. The aim of this study was to determine alterations in the expression of proteins involved in proliferation and apoptosis in non-invasive and invasive ductal breast lesions. Immunohistochemistry was performed on 106 usual ductal hyperplasias (UDH), 61 DCIS lesions and 53 invasive ductal breast carcinomas. Increased proliferation (Ki67), overexpression of cyclin D1, HER-2/neu, p21 and p53, and decreased expression of bcl-2 and p27 could already be found in UDH. Significant differences between UDH and DCIS lesions were found for only one protein when UDH was compared with well-differentiated DCIS (p27), for three proteins when compared with intermediately differentiated DCIS (p21, cyclin D1, Ki-67), and for all proteins when compared with poorly-differentiated DCIS. Comparing DCIS with invasive lesions of same differentiation grade, proliferation was elevated in the invasive lesions. Altered expression of the other proteins was in general only slightly increased in the invasive lesion compared with DCIS. The number of proteins with altered expression per lesion was highest in poorly-differentiated lesions and was comparable between DCIS and invasive cancer of the same differentiation grade. In conclusion, the biggest changes in expression of these proliferation and apoptosis related proteins appear to occur during the transition from hyperplasia to DCIS; they probably play a minor role in the transition from DCIS to invasive breast lesion of same differentiation grade. Well-differentiated in situ and invasive breast lesions share many of the aberrations in expression of these proteins, as do poorly-differentiated in situ and invasive lesions. However, there are many differences between the well and poorly-differentiated lesions. This further supports the existence of different progression routes leading to breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Cycle Proteins/metabolism , Apoptosis , Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Cyclin D1/analysis , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Receptor, ErbB-2/analysis , Tumor Suppressor Protein p53/analysis
8.
Int J Cancer ; 89(3): 236-41, 2000 May 20.
Article in English | MEDLINE | ID: mdl-10861499

ABSTRACT

p27(Kip1) (p27), a cyclin-dependent kinase inhibitor, has an important role in the progression of cells from G(1) into S phase of the cell cycle. p27 may act as a tumor suppressor, and several reports suggest that loss of its expression in breast carcinoma is related to tumor progression and poor prognosis. We evaluated p27 immunohistochemical expression in 512 consecutive cases of breast carcinoma with 9 years of median-term follow-up. p27 expression was heterogeneous and frequently less intense than in normal cells. Low p27 expression (<50% of reacting cells) was associated with grade III tumors, N0 status, estrogen receptor-negative status, and low cyclin D1 expression. In the whole series of cases, p27 expression did not predict outcome. In node-negative cases (249 patients), high p27 expression indicated poor prognosis. p27 was not prognostically relevant in the group of 223 patients with pT1 disease or in the group of 154 patients <50 years of age. We also investigated the prognostic value of the combined expression of p27 and cyclin D1, but no differences in survival were seen in this bivariate analysis.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins , Microtubule-Associated Proteins/biosynthesis , Tumor Suppressor Proteins , Adult , Aged , Carcinoma, Ductal, Breast/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunohistochemistry , Lymphatic Metastasis , Menopause , Middle Aged , Recurrence , Time Factors
9.
Mol Pathol ; 52(2): 78-83, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10474686

ABSTRACT

AIMS: To investigate the expression of the genes encoding cyclin D1 and p21 in proliferative and non-proliferative cells, as demonstrated by the Ki67 antibody, and to correlate these findings with differentiation. METHODS: Immunohistochemistry and immunofluorescence double staining were performed on three breast cancers, two squamous cell cancers of the head and neck, and one ovarium cystadenocarcinoma. In addition, the in vitro effect of cyclin D1 on p21 gene expression in MCF7 breast cancer cells was evaluated. RESULTS: Immunofluorescence double staining showed a differentiation related gradient in the detection of the Ki67 antigen, cyclin D1, and p21 in squamous cell cancers of the head and neck: Ki67 was detected in the basal layers of the tumour and the cyclin D1 and p21 genes were coexpressed in the higher, more differentiated layers of the tumour. The breast and ovarian cancers often had cells that coexpressed the p21 and cyclin D1 genes, whereas coexpression of cyclin D1 and Ki67 did not occur. Western blot analysis of the MCF7 breast cancer cells showed an upregulation of p21 production when cyclin D1 gene expression was induced. CONCLUSION: Overexpression of the cyclin D1 gene seems to lead to growth arrest in a variety of human cancers, possibly through the induction of p21 by cyclin D1. In squamous cell cancer, concerted overexpression of the genes encoding cyclin D1 and p21 might also induce differentiation.


Subject(s)
Cyclin D1/metabolism , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Cells, Cultured , Up-Regulation
10.
J Pathol ; 188(4): 341-3, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10440742

ABSTRACT

Cancer cells arise from an increasing genetic disarray affecting control over proliferation, self-defence, and senescence. Multiple mechanisms with a multitude of players are invoked in the genetic distortion which leads to tumour progression in different tissues. Tumour pathology is in need of a clear, distinctive classification of tumour samples for assessment of risk and treatment. What for, and how should these two fields be combined? Markers which indicate the overall genetic disorder in cancer cells may be sufficient, in addition to a morphological description, to assess risk of cancer patients, in particular in cases with lymph node-negative disease. However, a refinement of risk is at hand when a better evaluation of genetic alterations is achieved. The status of genetic malfunctioning may also provide target(s) for therapy as well. In this editorial, the genetic alterations implicated in disturbed regulation of the G1 phase of the cell cycle by the Rb/cyclin D1/p16/cdk4 pathway are considered to provide relevant information to assess risk as well as to target therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , G1 Phase , Neoplasms/metabolism , Humans , Neoplasm Proteins/metabolism , Prognosis
11.
Cancer Res ; 59(5): 1134-40, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10070974

ABSTRACT

Overexpression of cyclin D1, a G1 cell cycle regulator, is often found in many different tumor types, such as breast carcinoma and squamous cell carcinoma of the head and neck. The overexpression of this protein is, in several cases, associated with a poor prognosis. In this study, the effect of cyclin D1 on radiosensitivity was investigated in a breast tumor cell line, MCF7, containing a cyclin D1 gene construct under the control of a tetracycline-sensitive regulator. MCF7 cells cultured without tetracycline resulted in a 6-fold increase in the cyclin D1 protein. Cyclin D1-overexpressing MCF7 cells were more sensitive to ionizing radiation than the nonoverexpressing counterparts. The cyclin D1-overexpressing cells also exhibited a higher induction of apoptosis. Treatment with a dose of 5 Gy resulted in a rapid increase of p53 and p21 in the cyclin D1-overexpressing cells. Nonoverexpressing cells showed a more transient expression of these proteins after ionizing radiation. A pronounced G2-M block was observed in both cell lines. The cyclin D1-overexpressing cells were, however, released earlier from the block than the control cells. These data suggest that overexpression of cyclin D1 alters sensitivity toward ionizing radiation by modulating gamma-radiation-induced G2-M transition.


Subject(s)
Apoptosis/radiation effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Cell Survival/radiation effects , Cyclin D1/biosynthesis , Female , Flow Cytometry , Gamma Rays , Humans , Kinetics , Radiation Tolerance , Tumor Cells, Cultured , Tumor Stem Cell Assay
13.
Cell Adhes Commun ; 6(2-3): 185-209, 1998.
Article in English | MEDLINE | ID: mdl-9823470

ABSTRACT

We have identified a novel gene, EMS1, that is consistently amplified and overexpressed in human carcinomas with an amplification of the chromosome 11q13 region. Comparisons of the EMS1 sequences with those present in the GenBank databases revealed a high identity with chicken cortactin. Southern and western blot analyses confirm the high sequence conservation during evolution. An antiserum specific for human cortactin, showed in gene transfer experiments that both human p80 and p85 isoforms are encoded by the EMS1 cDNA. Further comparisons demonstrated an high sequence and structural homology with HS1 that is implicated in signal transduction in lymphoid cells only. Expression of EMS1/cortactin mRNA was restricted to tumor cell lines derived from non-lymphoid origin. Cortactin contains (i) a filamentous actin binding tandem repeat domain, (ii) a proline-rich SH3-binding and (iii) a SH3 domain that is common in proteins involved in signal transduction. Our data suggest that human EMS1/cortactin has a function in signal transmission between cell-matrix contact sites and the cytoskeleton and, as such, its overexpression due to 11q13 amplification might effect adhesive properties of human carcinomas.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 11 , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Animals , Antibodies , Base Sequence , Blotting, Western , Cell Adhesion/physiology , Cortactin , Cyclin D1/physiology , Evolution, Molecular , Female , Gene Amplification , Humans , Microfilament Proteins/analysis , Microfilament Proteins/immunology , Molecular Sequence Data , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , RNA, Messenger/analysis , Rabbits , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction/physiology , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology
14.
Am J Clin Pathol ; 110(5): 647-52, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9802351

ABSTRACT

Mutations in the tumor suppressor gene p53, analyzed in bladder washings, have positive predictive value for the progression of superficial bladder cancer to invasive disease. Bladder washings reflect the general status of the urothelium, and because sampling of bladder washings can be performed as an outpatient procedure, patients can be monitored more carefully. To determine the actual value of bladder washing specimens in assessing the p53 status of histologic specimens, we used the technique of polymerase chain reaction-single-strand conformation polymorphism to analyze bladder washings and the synchronous tumors of 15 patients for the presence of p53 mutations. A significant correlation (2-tailed Fisher's exact test) between the p53 status of bladder washings and histologic specimens was observed if the 2 were compared among the specimens of a single patient. Overall, in 2 patients the mutation present in the tumors was not detected in the bladder washings, and in 1 patient the mutation in the bladder washing was not detected in the histologic specimens. These conflicting results obtained with bladder washings and histologic specimens could be explained mainly by the architecture of the tumors. The observed specificity of 86% and sensitivity of 75% emphasizes that although the correlation between the 2 methods is good, in a number of cases they are complementary to each other. The analysis of p53 mutations in at least 2 bladder washings gives insight into the p53 status of the synchronous tumors.


Subject(s)
Genes, p53/genetics , Mutation , Therapeutic Irrigation , Urinary Bladder Neoplasms/genetics , Humans , Immunohistochemistry , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Sensitivity and Specificity , Sequence Analysis , Tissue Embedding , Tumor Suppressor Protein p53/analysis , Urinary Bladder Neoplasms/pathology
15.
Am J Surg Pathol ; 22(10): 1267-76, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9777989

ABSTRACT

Neural cell adhesion molecules (NCAM) represent specific markers of neuroendocrine (NE) differentiation in lung cancer. Because the polysialic acid form (NCAM-PSA) has reduced adhesion properties, we hypothesized that NCAM-PSA expression could favor metastatic spread. Immunostaining of NCAM and NCAM-PSA were therefore compared in 120 NE lung tumors, including 17 typical carcinoids, 3 atypical carcinoids, 30 large cell NE carcinomas and 70 small cell lung carcinomas, as compared with 25 adenocarcinomas and 25 squamous cell carcinomas. Neural cell adhesion molecules were negative in adenocarcinomas and squamous cell carcinomas but were constantly expressed in all NE tumors from typical carcinoids to small cell lung carcinomas. NCAM-PSA expression was significantly more frequent in high-grade tumors, with 24 of 30 positive cases in large cell NE carcinomas and 65 of 70 positive cases in small cell lung carcinoma, than in carcinoids with 10 of 17 and 2 of 3 positive cases in typical carcinoids and atypical carcinoids, respectively. The neural cell adhesion molecule-polysialic acid form scores of staining were significantly higher in high-grade as compared with low-grade tumors (p = 0.002), and were correlated with nodal spread (p = 0.04) and metastasis (p = 0.016) across histologic classes but not in individual tumor type. We conclude that NCAM-PSA connotes poor differentiation and aggressive clinical behavior in the spectrum of NE lung tumors, but cannot be regarded as a prognostic factor in individual tumor classes.


Subject(s)
Lung Neoplasms/metabolism , Neural Cell Adhesion Molecules/metabolism , Neuroendocrine Tumors/metabolism , Sialic Acids/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antibodies, Monoclonal/analysis , Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Count , Chromogranin A , Chromogranins/metabolism , Diagnosis, Differential , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphatic Metastasis , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/secondary , Retrospective Studies , Survival Rate
16.
Mol Pathol ; 51(1): 30-4, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9624417

ABSTRACT

BACKGROUND: In vitro studies have shown that amplification and overexpression of the cyclin D1 gene can accelerate the progress of cells through the G1 phase. Therefore, cyclin D1 may have an apoptosis inhibiting effect. The retinoblastoma (Rb) gene was shown recently to be an important regulator of apoptosis. AIMS: To evaluate whether expression of the cyclin D1 and Rb genes correlated with apoptotic counts in a group of 97 invasive breast cancers. METHODS: Expression of the cyclin D1 and Rb genes was detected by standard immunnohistochemistry using paraffin wax embedded sections. Apoptotic cells were counted according to a strict protocol, in 10 fields of vision systematically spread over the most poorly differentiated area of the tumour, at a magnification of x630. Apoptotic cells counts were expressed as apoptotic cells/mm2. RESULTS: Cyclin D1 overexpression was found in 49% of cases. Loss of Rb expression was found in 44% of cases, and occurred particularly in poorly differentiated tumours. Cyclin D1 and Rb expression showed a positive correlation (p = 0.003). Apoptotic counts varied from 1 to 62/mm2. There were no significant correlations between cyclin D1 overexpression and apoptotic counts in the total group or in the retinoblastoma protein (pRb) positive tumours. Loss of Rb expression also showed no correlation with apoptotic counts. CONCLUSIONS: Cyclin D1 is frequently overexpressed in pRb positive tumours, but no evidence has been found for an anti-apoptotic effect of cyclin D1 overexpression or Rb expression in invasive breast cancer.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/metabolism , Cyclin D1/metabolism , Neoplasm Proteins/metabolism , Retinoblastoma Protein/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Neoplasm Invasiveness
17.
Cancer Res ; 58(5): 871-6, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9500441

ABSTRACT

Cyclin D1 is a target for positive regulation by estrogens in growth-responsive cells, in which it mediates their mitogenic effects. Amplification and overexpression of the cyclin D1 gene (CCND1) might thus represent a genetic lesion inducing hormone-independent growth of transformed cells. Indeed, cyclin D1 overexpression has been found in up to 50% of primary breast cancers, and in about one-third of these cases, this is linked to amplification of the 11q13 chromosomal region, which also includes the CCND1 gene. These tumors are predominantly estrogen receptor-positive, and for this reason, these patients are often selected for adjuvant antiestrogen therapy. No information is available, however, as to whether cyclin D1 overexpression due to gene amplification might interfere with and reduce antiestrogen efficacy. This was investigated here by taking advantage of an experimental model that reproduces cyclin D1 overexpression resulting from increased CCND1 gene dosage in hormone-responsive human breast cancer cells. For this, MCF-7 cells stably transfected with a tet-inducible cyclin D1 expression vector were tested for their in vitro response to steroidal (ICI 182,780) and nonsteroidal (trans-4-hydroxytamoxifen) antiestrogens under condition of low (endogenous only) or high (exogenous) cyclin D1 levels. Results show that although cyclin D1 overexpression seems to interfere with the early cell cycle effects of antiestrogens, it does not prevent their cytostatic actions, so that growth of cyclin-overexpressing MCF-7 cells is still efficiently inhibited in vitro by these drugs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chromosomes, Human, Pair 11 , Cyclin D1/genetics , Estrogen Antagonists/pharmacology , Gene Expression Regulation, Neoplastic , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Division/genetics , Cyclin D1/biosynthesis , Female , Gene Dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Tumor Cells, Cultured
18.
Arch Otolaryngol Head Neck Surg ; 123(5): 497-502, 1997 May.
Article in English | MEDLINE | ID: mdl-9158396

ABSTRACT

OBJECTIVES: To evaluate the overexpression of cyclin D1 and p53 as a prognostic marker of squamous cell carcinoma of the head and neck and to investigate whether deregulation of these genes is associated with an unfavorable course of disease. DESIGN: Retrospective study. MATERIALS AND METHODS: Formalin-fixed, paraffin-embedded tumor materials that were obtained from a well-characterized series of 115 patients with resectable head and neck cancer at The Netherlands Cancer Institute, Amsterdam, were analyzed by immunohistochemical methods using antiserum samples that were directed against 2 proteins (ie, cyclin D1 and p53), which are crucial in the regulation of the G1 phase of the cell cycle. RESULTS: Overexpression of cyclin D1 protein was found in 49% of the patients with squamous cell carcinoma of the head and neck. This overexpression was not associated with known prognostic factors (eg, the T and N stages). Tumors recurred more frequently and in a shorter period in patients whose primary tumors showed an overexpression of cyclin D1 protein. This difference (P = .05) was statistically significant in a stepwise proportional hazard regression analysis. However, since a discrepancy in staining results was observed between the biopsy and resection materials that were taken from the same patient, this result may not have been applicable in the evaluation of biopsy specimens only. This discrepancy is most likely owing to tissue heterogeneity. The overexpression of p53 that was found in 42% of the patients was of no prognostic significance. CONCLUSIONS: These data provide evidence that overexpression of cyclin D1 protein in resection material of squamous cell carcinomas of the head and neck is indicative of a poor prognosis, independently of other known prognostic factors. Whether overexpression of cyclin D1 may therefore be used to select patients for more intensive treatment should be examined in the context of a clinical trial.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic/physiology , Head and Neck Neoplasms/genetics , Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cyclin D1 , Cyclins/metabolism , Disease-Free Survival , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Oncogene Proteins/metabolism , Prognosis , Retrospective Studies
19.
Am J Pathol ; 150(2): 705-11, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9033283

ABSTRACT

Cyclin D1 overexpression, detected by standard immunohistochemistry, was correlated with other prognostic variables and its prognostic value was evaluated in a group of 148 invasive breast cancers with long-term follow-up. Overexpression of cyclin D1 (59% of cases) was negatively correlated (chi 2 test) with histological grade (P = 0.0001), mean nuclear area (P = 0.004), mean nuclear volume (P = 0.02), and mitotic activity (P = 0.03) and positively correlated with estrogen receptor (P = 0.0001). There was a strong correlation between cyclin D1 overexpression and histological type (P = 0.0001). Positive cyclin D1 staining was seen in 11 of 13 tubular carcinomas, 3 of 3 mucinous carcinomas, 4 of 4 invasive cribriform carcinomas, and 17 of 20 lobular carcinomas. Of 102 ductal cancers, 52 were positive, and all 6 medullary carcinomas were negative. There were no significant correlations with lymph node status, tumor size, or DNA ploidy. In survival analysis, cyclin D1 overexpression did not provide significant univariate or multivariate prognostic value. In conclusion, cyclin D1 is mainly overexpressed in the well differentiated and lobular types of invasive breast cancer and is strongly associated with estrogen receptor positivity. It is negatively correlated with the proliferation marker mitoses count and with the differentiation markers nuclear area and nuclear volume. However, cyclin D1 overexpression does not seem to have prognostic value in invasive breast cancer when no adjuvant treatment is given.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclins/metabolism , Oncogene Proteins/metabolism , Cyclin D1 , Female , Flow Cytometry , Humans , Immunohistochemistry , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Survival Analysis
20.
Cell ; 88(3): 405-15, 1997 Feb 07.
Article in English | MEDLINE | ID: mdl-9039267

ABSTRACT

Both cyclin D1 and estrogens have an essential role in regulating proliferation of breast epithelial cells. We show here a novel role for cyclin D1 in growth regulation of estrogen-responsive tissues by potentiating transcription of estrogen receptor-regulated genes. Cyclin D1 mediates this activation independent of complex formation to a CDK partner. Cyclin D1 activates estrogen receptor-mediated transcription in the absence of estrogen and enhances transcription in its presence. The activation of estrogen receptor by cyclin D1 is not inhibited by anti-estrogens. A direct physical binding of cyclin D1 to the hormone binding domain of the estrogen receptor results in an increased binding of the receptor to estrogen response element sequences, and upregulates estrogen receptor-mediated transcription. These results highlight a novel role for cyclin D1 as a CDK-independent activator of the estrogen receptor.


Subject(s)
Cyclin-Dependent Kinases/physiology , Cyclins/pharmacology , Oncogene Proteins/pharmacology , Receptors, Estrogen/metabolism , Base Sequence , Cyclin D1 , Cyclins/metabolism , Enzyme Activation/drug effects , Estradiol/pharmacology , HeLa Cells , Humans , Ligands , Oncogene Proteins/metabolism , Protein Binding/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Transcription, Genetic/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...