Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
BMC Cancer ; 15: 588, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26272591

ABSTRACT

BACKGROUND: Estrogen Receptor alpha (ERα)-positive breast cancer patients receive endocrine therapy, often in the form of tamoxifen. However, resistance to tamoxifen is frequently observed. A signalling cascade that leads to tamoxifen resistance is dictated by activation of the Protein Kinase A (PKA) pathway, which leads to phosphorylation of ERα on Serine 305 and receptor activation, following tamoxifen binding. Thus far, it remains elusive what protein complexes enable the PKA-ERα interaction resulting in ERα Serine 305 phosphorylation. METHODS: We performed immunohistochemistry to detect ERαSerine 305 phosphorylation in a cohort of breast cancer patients who received tamoxifen treatment in the metastatic setting. From the same tumor specimens, Agilent 44 K gene expression analyses were performed and integrated with clinicopathological data and survival information. In vitro analyses were performed using MCF7 breast cancer cells, which included immunoprecipitations and Fluorescence Resonance Energy Transfer (FRET) analyses to illustrate ERα complex formation. siRNA mediated knockdown experiments were performed to assess effects on ERαSerine 305 phosphorylation status, ERα/PKA interactions and downstream responsive gene activity. RESULTS: Stratifying breast tumors on ERα Serine 305 phosphorylation status resulted in the identification of a gene network centered upon AKAP13. AKAP13 mRNA expression levels correlate with poor outcome in patients who received tamoxifen treatment in the metastatic setting. In addition, AKAP13 mRNA levels correlate with ERαSerine 305 phosphorylation in breast tumor samples, suggesting a functional connection between these two events. In a luminal breast cancer cell line, AKAP13 was found to interact with ERα as well as with a regulatory subunit of PKA. Knocking down of AKAP13 prevented PKA-mediated Serine 305 phosphorylation of ERα and abrogated PKA-driven tamoxifen resistance, illustrating that AKAP13 is an essential protein in this process. CONCLUSIONS: We show that the PKA-anchoring protein AKAP13 is essential for the phosphorylation of ERαS305, which leads to tamoxifen resistance both in cell lines and tamoxifen-treated breast cancer patients.


Subject(s)
A Kinase Anchor Proteins/metabolism , Breast Neoplasms/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Resistance, Neoplasm/drug effects , Proto-Oncogene Proteins/metabolism , Tamoxifen/pharmacology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Immunohistochemistry , Middle Aged , Minor Histocompatibility Antigens , Phosphorylation , Signal Transduction/drug effects
2.
Proc Natl Acad Sci U S A ; 110(22): 8894-9, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23676274

ABSTRACT

Estrogen receptor alpha (ERα) is involved in numerous physiological and pathological processes, including breast cancer. Breast cancer therapy is therefore currently directed at inhibiting the transcriptional potency of ERα, either by blocking estrogen production through aromatase inhibitors or antiestrogens that compete for hormone binding. Due to resistance, new treatment modalities are needed and as ERα dimerization is essential for its activity, interference with receptor dimerization offers a new opportunity to exploit in drug design. Here we describe a unique mechanism of how ERα dimerization is negatively controlled by interaction with 14-3-3 proteins at the extreme C terminus of the receptor. Moreover, the small-molecule fusicoccin (FC) stabilizes this ERα/14-3-3 interaction. Cocrystallization of the trimeric ERα/14-3-3/FC complex provides the structural basis for this stabilization and shows the importance of phosphorylation of the penultimate Threonine (ERα-T(594)) for high-affinity interaction. We confirm that T(594) is a distinct ERα phosphorylation site in the breast cancer cell line MCF-7 using a phospho-T(594)-specific antibody and by mass spectrometry. In line with its ERα/14-3-3 interaction stabilizing effect, fusicoccin reduces the estradiol-stimulated ERα dimerization, inhibits ERα/chromatin interactions and downstream gene expression, resulting in decreased cell proliferation. Herewith, a unique functional phosphosite and an alternative regulation mechanism of ERα are provided, together with a small molecule that selectively targets this ERα/14-3-3 interface.


Subject(s)
14-3-3 Proteins/metabolism , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Estrogen Receptor alpha/metabolism , Glycosides/pharmacology , Models, Molecular , Protein Conformation , Amino Acid Sequence , Crystallization , Dimerization , Estrogen Receptor alpha/genetics , Female , Fluorescence Polarization , Gene Components , Gene Expression Regulation/drug effects , Humans , MCF-7 Cells , Mass Spectrometry , Molecular Sequence Data , Phosphorylation , Protein Isoforms/metabolism , Sequence Alignment
3.
Mol Cancer Ther ; 11(4): 805-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22319200

ABSTRACT

With current techniques, it remains a challenge to assess coregulator binding of nuclear receptors, for example, the estrogen receptor alpha (ERα). ERα is critical in many breast tumors and is inhibited by antiestrogens such as tamoxifen in cancer therapy. ERα is also modified by acetylation and phosphorylation that affect responses to the antiestrogens as well as interactions with coregulators. Phosphorylation of ERα at Ser305 is one of the mechanisms causing tamoxifen resistance. Detection of resistance in patient samples would greatly facilitate clinical decisions on treatment, in which such patients would receive other treatments such as aromatase inhibitors or fulvestrant. Here we describe a coregulator peptide array that can be used for high-throughput analysis of full-length estrogen receptor binding. The peptide chip can detect ERα binding in cell and tumor lysates. We show that ERα phosphorylated at Ser305 associates stronger to various coregulator peptides on the chip. This implies that ERαSer305 phosphorylation increases estrogen receptor function. As this is also detected in a breast tumor sample of a tamoxifen-insensitive patient, the peptide array, as described here, may be applicable to detect tamoxifen resistance in breast tumor samples at an early stage of disease and contribute to personalized medicine.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estrogen Receptor alpha/metabolism , Serine/metabolism , Tamoxifen/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Humans , Microarray Analysis , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Phosphorylation , Transfection
4.
Breast Cancer Res Treat ; 125(1): 1-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20213082

ABSTRACT

Phosphorylation of estrogen receptor α at serine 305 (ERαS305-P) by protein kinase A (PKA) or p21-activated kinase 1 (PAK1) has experimentally been associated with tamoxifen sensitivity. Here, we investigated the clinical application of this knowledge to predict tamoxifen resistance in ER-positive breast cancer patients. Using immunohistochemistry, a score including PAK1 and co-expression of PKA and ERαS305-P (PKA/ERαS305-P) was developed on a training set consisting of 103 patients treated with tamoxifen for metastatic disease, and validated on 231 patients randomized between adjuvant tamoxifen or no treatment. In the training set, PAK1 levels were associated with tumor progression after tamoxifen (HR 1.57, 95% CI 0.99-2.48), as was co-expression of PKA and ERαS305-P (HR 2.00, 95% CI 1.14-3.52). In the validation set, a significant tamoxifen benefit was found among the 73% patients negative for PAK1 and PKA/ERαS305-P (HR 0.54, 95% CI 0.34-0.87), while others (27%) were likely to have no benefit from tamoxifen (HR 0.88, 95% 0.42-1.82). The test for interaction showed a significant difference in recurrence-free survival between groups defined by PAK1 and PKA/ERαS305-P (P = 0.037). Elevated PAK1 and PKA/ERαS305-P appeared to influence tamoxifen sensitivity. Both PAK1 and PKA/ERαS305-P levels were associated with sensitivity to tamoxifen in breast tumors and the combination of these variables should be considered in predicting tamoxifen benefit.


Subject(s)
Breast Neoplasms/drug therapy , Cyclic AMP-Dependent Protein Kinases/metabolism , Estrogen Receptor Modulators/therapeutic use , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/metabolism , Tamoxifen/therapeutic use , p21-Activated Kinases/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Chemotherapy, Adjuvant , Cyclic AMP-Dependent Protein Kinases/genetics , Disease-Free Survival , Drug Resistance, Neoplasm , Europe , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Patient Selection , Phosphorylation , Predictive Value of Tests , Proportional Hazards Models , Reproducibility of Results , Risk Assessment , Risk Factors , Serine , Time Factors , Tissue Array Analysis , Transfection , Treatment Outcome , Up-Regulation , p21-Activated Kinases/genetics
5.
Int J Breast Cancer ; 2011: 232435, 2011.
Article in English | MEDLINE | ID: mdl-22295213

ABSTRACT

About two thirds of all human breast cancer cases are estrogen receptor positive. The drug of first choice for these patients is tamoxifen. However, about half of the recurrences after removal of the primary tumor are or become resistant to this drug. While many mechanisms have been identified for tamoxifen resistance in the lab, at present only a few have been translated to the clinic. This paper highlights the role in tamoxifen resistance of phosphorylation by different kinases on different sites of the estrogen receptor. We will discuss the molecular pathways and kinases that are involved in phosphorylation of ERα and how these affect tamoxifen resistance. Finally, we will elaborate on the clinical translation of these observations and the possibility to predict tamoxifen responses in patient tumor samples before treatment onset. The findings made originally on the bench may translate into a better and personalized treatment of breast cancer patients using an old and safe anticancer drug: tamoxifen.

6.
J Cell Sci ; 123(Pt 8): 1253-61, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20332105

ABSTRACT

Human estrogen receptors alpha and beta (ERalpha and ERbeta) greatly differ in their target genes, transcriptional potency and cofactor-binding capacity, and are differentially expressed in various tissues. In classical estrogen response element (ERE)-mediated transactivation, ERbeta has a markedly reduced activation potential compared with ERalpha; the mechanism underlying this difference is unclear. Here, we report that the binding of steroid receptor coactivator-1 (SRC-1) to the AF-1 domain of ERalpha is essential but not sufficient to facilitate synergy between the AF-1 and AF-2 domains, which is required for a full agonistic response to estradiol (E2). Complete synergy is achieved through the distinct hinge domain of ERalpha, which enables combined action of the AF-1 and AF-2 domains. AF-1 of ERbeta lacks the capacity to interact with SRC-1, which prevents hinge-mediated synergy between AF-1 and AF-2, thereby explaining the reduced E2-mediated transactivation of ERbeta. Transactivation of ERbeta by E2 requires only the AF-2 domain. A weak agonistic response to tamoxifen occurs for ERalpha, but not for ERbeta, and depends on AF-1 and the hinge-region domain of ERalpha.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Tamoxifen/pharmacology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/metabolism , Humans , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Receptor Coactivator 1/metabolism , Protein Binding/drug effects , Protein Conformation , Protein Structure, Tertiary , Protein Transport/drug effects , RNA Polymerase II/metabolism , Structure-Activity Relationship , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
7.
J Natl Cancer Inst ; 101(24): 1725-9, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-19940281

ABSTRACT

Although estrogen receptor-alpha (ER) [corrected] is a marker used to identify breast cancer patients most likely to benefit from endocrine therapy, approximately 50% of ER-positive [corrected] breast carcinomas are resistant to tamoxifen. Preclinical studies have shown that phosphorylation of ER [corrected] at serine-118 (ER alpha S118-P) is required for tamoxifen-mediated inhibition of ER-induced [corrected] gene expression. We evaluated the association between recurrence-free survival after tamoxifen treatment and ER alpha S118-P expression by use of Cox proportional hazards regression. Data were from 239 premenopausal patients with breast cancer who participated in a randomized trial of 2 years of adjuvant tamoxifen treatment vs no systemic treatment. ER alpha S118-P expression was assessed by immunohistochemistry and categorized by use of the Allred score (low expression = score of 0-6; high expression = score of 7-8). All statistical tests were two-sided. Compared with systemically untreated patients, we found evidence of a benefit from adjuvant tamoxifen among patients whose tumors had high ER alpha S118-P expression (23.7 recurrences per 1000 person-years versus 72.2 recurrences per 1000 person-years, hazard ratio [HR] of recurrence = 0.36, 95% confidence interval [CI] = 0.20 to 0.65) but not among patients whose tumors had low expression (51.0 recurrences per 1000 person-years versus 57.0 recurrences per 1000 person-years, HR of recurrence = 0.87, 95% CI = 0.51 to 1.48), a statistically significant difference (P for interaction = .037). ER alpha 118-P was not associated with recurrence-free survival among untreated patients. Thus, ER alpha S118-P expression appears to be associated with response to tamoxifen. [corrected]


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Receptor alpha/drug effects , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Disease-Free Survival , Estrogen Receptor alpha/metabolism , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Multivariate Analysis , Odds Ratio , Phosphorylation/drug effects , Premenopause , Proportional Hazards Models , Serine/metabolism , Tamoxifen/therapeutic use
8.
Chem Biol ; 16(7): 702-11, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19635407

ABSTRACT

The interaction of estrogen receptor alpha (ERalpha) with the consensus LXXLL motifs of transcriptional coactivators provides an entry for functional ERalpha inhibition. Here, synthetic cell-permeable LXXLL peptide probes are brought forward that allow evaluation of the interaction of specific recognition motifs with ERalpha in the context of the cell. The probes feature a nona-arginine tag that facilitates cellular entry and induces probe localization in nucleoli. The nucleoli localization provides an explicit tool for evaluating the LXXLL motif interaction with ERalpha. The probes compete with coactivators, bind ERalpha, and recruit it into the nucleoli. The physical inhibition of the ERalpha-coactivator interaction by the probes is shown to be correlated with the inhibition of ERalpha-mediated gene transcription. This chemical biology approach allows evaluating the ERalpha-coactivator interaction and inhibitor binding directly in cells.


Subject(s)
Estrogen Receptor alpha/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Motifs , Cell Nucleolus/metabolism , Humans , Molecular Probe Techniques , Oligopeptides/pharmacology , Peptides/chemical synthesis , Protein Binding , Transcription, Genetic/drug effects
9.
Mol Endocrinol ; 23(9): 1335-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19477949

ABSTRACT

Resistance to tamoxifen treatment occurs in approximately 50% of the estrogen receptor (ER)alpha-positive breast cancer patients. Resistant patients would benefit from treatment with other available antiestrogens. Arzoxifene is an effective growth inhibitor of ERalpha-positive breast cancer cells, including tamoxifen-resistant tumors. In this study, we show that overexpression of a regular component of the ERalpha transcription factor complex, cyclin D1, which occurs in approximately 40% of breast cancer patients, renders cells resistant to the new promising antiestrogen, arzoxifene. Overexpression of cyclin D1 alters the conformation of ERalpha in the presence of arzoxifene. In this altered conformation, ERalpha still recruits RNA polymerase II to an estrogen response element-containing promoter, inducing transcription of an ERalpha-dependent reporter gene and of endogenous pS2, and promoting arzoxifene-stimulated growth of MCF-7 cells. Arzoxifene is then converted from an ERalpha antagonist into an agonist. This can be explained by a stabilization of the ERalpha/steroid receptor coactivator-1 complex in the presence of arzoxifene, only when cyclin D1 is overexpressed. These results indicate that subtle changes in the conformation of ERalpha upon binding to antiestrogen are at the basis of resistance to antiestrogens.


Subject(s)
Cyclin D1/biosynthesis , Drug Resistance , Estrogen Receptor Modulators/pharmacology , Gene Expression Regulation , Piperidines/pharmacology , Thiophenes/pharmacology , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor alpha/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Microscopy, Confocal/methods , Models, Chemical
10.
EMBO J ; 26(15): 3534-44, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17627277

ABSTRACT

Resistance to tamoxifen is observed in half of the recurrences in breast cancer, where the anti-estrogen tamoxifen acquires agonistic properties for transactivating estrogen receptor alpha (ERalpha). In a previous study, we showed that protein kinase A (PKA)-mediated phosphorylation of serine 305 (S305) of ERalpha results in resistance to tamoxifen. Now, we demonstrate that phosphorylation of S305 in ERalpha by PKA leads to an altered orientation between ERalpha and its coactivator SRC-1, which renders the transcription complex active in the presence of tamoxifen. This altered orientation involves the C-termini of ERalpha and SRC-1, which required a prolonged AF-1-mediated interaction. This intermolecular reorientation as a result of PKA-mediated phosphorylation of ERalpha-S305 and tamoxifen binding provides a unique model for resistance to the anticancer drug tamoxifen.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Histone Acetyltransferases/metabolism , Tamoxifen/pharmacology , Transcription Factors/metabolism , Base Sequence , Cell Line, Tumor , DNA Primers , DNA, Neoplasm/metabolism , Fluorescence Resonance Energy Transfer , Humans , In Situ Hybridization, Fluorescence , Nuclear Receptor Coactivator 1 , Phosphorylation , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
11.
Mol Cancer Ther ; 6(5): 1526-33, 2007 May.
Article in English | MEDLINE | ID: mdl-17513601

ABSTRACT

Anti-estrogen resistance is a major clinical problem in the treatment of breast cancer. In this study, fluorescence resonance energy transfer (FRET) analysis, a rapid and direct way to monitor conformational changes of estrogen receptor alpha (ERalpha) upon anti-estrogen binding, was used to characterize resistance to anti-estrogens. Nine different anti-estrogens all induced a rapid FRET response within minutes after the compounds have liganded to ERalpha in live cells, corresponding to an inactive conformation of the ERalpha. Phosphorylation of Ser(305) and/or Ser(236) of ERalpha by protein kinase A (PKA) and of Ser(118) by mitogen-activated protein kinase (MAPK) influenced the FRET response differently for the various anti-estrogens. PKA and MAPK are both associated with resistance to anti-estrogens in breast cancer patients. Their respective actions can result in seven different combinations of phospho-modifications in ERalpha where the FRET effects of particular anti-estrogen(s) are nullified. The FRET response provided information on the activity of ERalpha under the various anti-estrogen conditions as measured in a traditional reporter assay. Tamoxifen and EM-652 were the most sensitive to kinase activities, whereas ICI-182,780 (Fulvestrant) and ICI-164,384 were the most stringent. The different responses of anti-estrogens to the various combinations of phospho-modifications in ERalpha elucidate why certain anti-estrogens are more prone than others to develop resistance. These data provide new insights into the mechanism of action of anti-hormones and are critical for selection of the correct individual patient-based endocrine therapy in breast cancer.


Subject(s)
Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogens/chemistry , Fluorescence Resonance Energy Transfer/methods , Mutation , Bacterial Proteins/metabolism , Cell Line, Tumor , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/chemistry , Fulvestrant , Humans , Luminescent Proteins/metabolism , Models, Biological , Models, Chemical , Piperidines/pharmacology , Tamoxifen/pharmacology , Transcriptional Activation
12.
Nucl Recept Signal ; 5: e003, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17464358

ABSTRACT

Transcription controlled by Steroid Hormone Receptors (SHRs) plays a key role in many important physiological processes like organ development, metabolite homeostasis, and response to external stimuli. Understandably, the members of this family have drawn a lot of attention from the scientific community since their discovery, four decades ago. Still, after many years of research we are only beginning to unravel the complex nature of these receptors. The pace at which we do has improved significantly in recent years with the discovery of genetically encoded fluorescent probes, and the accompanying revival of biophysical approaches that allow more detailed study of SHRs. Here, we will look into the different aspects of SHR signalling, and discuss how biophysical techniques have contributed to visualizing their function in their native context, the living cell.


Subject(s)
Cell Nucleus/metabolism , Fluorescence Recovery After Photobleaching/methods , Fluorescence Resonance Energy Transfer/methods , Gene Expression Profiling/methods , Microscopy, Fluorescence/methods , Receptors, Steroid/metabolism , Transcriptional Activation/physiology , Animals , Cell Physiological Phenomena , Humans
13.
Cancer Cell ; 5(6): 597-605, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15193262

ABSTRACT

Using a novel approach that detects changes in the conformation of ERalpha, we studied the efficacy of anti-estrogens to inactivate ERalpha under different experimental conditions. We show that phosphorylation of serine-305 in the hinge region of ERalpha by protein kinase A (PKA) induced resistance to tamoxifen. Tamoxifen bound but then failed to induce the inactive conformation, invoking ERalpha-dependent transactivation instead. PKA activity thus induces a switch from antagonistic to agonistic effects of tamoxifen on ERalpha. In clinical samples, we found that downregulation of a negative regulator of PKA, PKA-RIalpha, was associated with tamoxifen resistance prior to treatment. Activation of PKA by downregulation of PKA-RIalpha converts tamoxifen from an ERalpha inhibitor into a growth stimulator, without any effect on ICI 182780 (Fulvestrant).


Subject(s)
Breast Neoplasms/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Resistance, Neoplasm , Estradiol/analogs & derivatives , Receptors, Estrogen/chemistry , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Binding Sites , Blotting, Western , Breast Neoplasms/enzymology , Cell Division , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha , Female , Fluorescence Resonance Energy Transfer , Fulvestrant , Humans , Luciferases/metabolism , Microscopy, Confocal , Models, Biological , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , RNA Interference , Receptors, Estrogen/metabolism , Serine/chemistry , Time Factors , Transcriptional Activation , Transfection
14.
Oncogene ; 21(53): 8158-65, 2002 Nov 21.
Article in English | MEDLINE | ID: mdl-12444551

ABSTRACT

Estrogen receptor-mediated transcription is enhanced by overexpression of G1/S cyclins D1, E or A in the presence as well in the absence of estradiol. Excess of G1/S cyclins also prevents the inhibition of transactivation of estrogen receptor (ER) by the pure antiestrogen ICI 182780. Cyclin D1 mediates this transactivation independent of complex formation to its CDK4/6 partner. This raises the possibility that overexpression of G1/S cyclins renders growth of ER-positive breast cancer hormone-independent and resistant to treatment with antiestrogens. Transient transfection of ER-positive breast cancer cell lines T47D and MCF7 with G1/S cyclins could overcome the growth arrest induced by ICI 182780 treatment. The ability of various cyclin D1 mutants to overcome the ICI 182780 mediated growth arrest corresponded with their ability to stimulate cyclin A- and E2F- promoter based reporter activities in the presence of ICI 182780. Transfection of a mutant cyclin D1 (cyclin D1-KE) that was unable to bind CDK4 and was reported to transactivate ER in the presence of ICI 182780, could not stimulate proliferation in ICI 182780 treated cells. On the other hand, cyclin D1-LALA, which is unable to stimulate ERE transactivation, could overcome the ICI 182780 cell cycle arrest. Furthermore, transient transfection of T47D cells using cyclin D1 together with a catalytic inactive mutant of CDK4 (CDK4-DN) indicated that the observed effect is due to binding to CDK inhibitors. However, a moderate, sixfold overexpression of cyclin D1 in stably transfected MCF7 cells did not overcome the ICI 182780 mediated growth arrest. These results indicate that CDK-independent transactivation of the estrogen receptor by cyclin D1 is by itself, not sufficient to result in estradiol-independent growth of breast cancer cells, whereas a vast overexpression of G1/S cyclins is able to do so, most likely by capturing of CDK inhibitors.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Cell Cycle Proteins , Cyclin A/physiology , Cyclin D1/physiology , Cyclin E/physiology , DNA-Binding Proteins , Estradiol/analogs & derivatives , Estrogens , G1 Phase/physiology , Neoplasm Proteins/analysis , Neoplasms, Hormone-Dependent/pathology , Proto-Oncogene Proteins , Receptors, Estrogen/analysis , S Phase/physiology , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cyclin D1/genetics , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Fulvestrant , Humans , Neoplasms, Hormone-Dependent/metabolism , Recombinant Fusion Proteins/physiology , Transcription Factors/physiology , Transcriptional Activation , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
15.
Breast Cancer Res Treat ; 74(1): 55-63, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12150453

ABSTRACT

Treatment of MCF7 human breast cancer cells with taxol induces G2/M arrest followed by mitotic death. A moderate overexpression of ectopic cyclin D1 accelerated these G2/M associated events and resulted in a reduced clonogenic survival upon taxol treatment. Taxol treatment resulted in elevated expression of p53 and of p21, which was more pronounced and persistent in cyclin D1 overexpressing cells. Overexpression of cyclin D1 altered sensitivity to taxol by modulating exit from mitosis, which is controlled by p21. These results indicate that overexpression of cyclin D1 sensitizes MCF7 cells to treatment with taxol.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin D1/biosynthesis , Gene Expression Regulation, Neoplastic , Mitosis , Paclitaxel/pharmacology , Female , Humans , Tumor Cells, Cultured
16.
Head Neck ; 24(7): 694-704, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12112544

ABSTRACT

Tumors gradually develop as a result of a multistep acquisition of genetic alterations and ultimately emerge as selfish, intruding and metastatic cells. The genetic defects associated with the process of tumor progression affect control of proliferation, programmed cell death, cell aging, angiogenesis, escape from immune control and metastasis. Fundamental cancer research over the last thirty years has revealed a multitude of genetic alterations which specify more or less separate steps in tumor development and which are collectively responsible for the process of tumor progression. The genes affected play in normal cells a crucial role in control over cell duplication and the interaction between cells, and between cells and their direct surrounding. This is illustrated on control during the G1/S phase of the cell cycle by its ultimate regulators: cyclins and cyclin dependent kinases. These proteins not only control the transition through the G1/S phase of the cell cycle, but also serve as mediators of the interaction between cells, and between cells and their surrounding. Defaults in the regulation of these proteins are associated with tumor progression, and, therefore, serve as targets for therapy. Defaults in those genes are found in various tumor types, although some of those prevail in particular tumor types. In this review emphasis is given to the defaults that occur in head and neck cancer.


Subject(s)
G1 Phase/physiology , Head and Neck Neoplasms/physiopathology , S Phase/physiology , Aneuploidy , Apoptosis/physiology , Cell Differentiation , Cyclins/physiology , DNA Damage/physiology , G1 Phase/genetics , Head and Neck Neoplasms/genetics , Humans , Ki-67 Antigen/physiology , Mitosis/physiology , Phosphorylation , S Phase/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...