Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Br J Cancer ; 109(12): 3034-41, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24178758

ABSTRACT

BACKGROUND: Persin is a plant toxin that displays synergistic cytotoxicity with tamoxifen in human breast cancer cell lines. Here, we examined the ability of persin to circumvent tamoxifen resistance and delineated the intracellular signalling pathways involved. METHODS: The induction of apoptosis in tamoxifen-resistant and -sensitive breast cancer cells was measured by flow cytometry following treatment with persin±tamoxifen. Markers of endoplasmic reticulum stress (ERS) were analysed following treatment, and their causal role in mediating persin-induced apoptosis was determined using chemical inhibitors and RNA interference. RESULTS: Cells that were resistant to an apoptotic concentration of tamoxifen maintained an apoptotic response to persin. Persin-induced apoptosis was associated with an increase in markers of ERS, that is, CHOP expression and XBP-1 splicing and was decreased by CHOP siRNA. The CASP-4 inhibitor Z-YVAD-FMK markedly inhibited persin-induced apoptosis in both tamoxifen-sensitive and -resistant cells. CONCLUSION: The cytotoxic effects of persin are CASP-4 dependent and mediated by CHOP-dependent and -independent ERS signalling cascades. Increased ERS signalling contributes to persin-induced reversal of tamoxifen resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Endoplasmic Reticulum Stress/drug effects , Fatty Alcohols/pharmacology , Plant Extracts/pharmacology , Tamoxifen/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Fatty Alcohols/administration & dosage , Female , Humans , MCF-7 Cells , Signal Transduction , Tamoxifen/administration & dosage
2.
Ann Oncol ; 23(7): 1713-22, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22241899

ABSTRACT

BACKGROUND: Current staging methods for pancreatic cancer (PC) are inadequate, and biomarkers to aid clinical decision making are lacking. Despite the availability of the serum marker carbohydrate antigen 19.9 (CA19.9) for over two decades, its precise role in the management of PC is yet to be defined, and as a consequence, it is not widely used. METHODS: We assessed the relationship between perioperative serum CA19.9 levels, survival and adjuvant chemotherapeutic responsiveness in a cohort of 260 patients who underwent operative resection for PC. RESULTS: By specifically assessing the subgroup of patients with detectable CA19.9, we identified potential utility at key clinical decision points. Low postoperative CA19.9 at 3 months (median survival 25.6 vs 14.8 months, P=0.0052) and before adjuvant chemotherapy were independent prognostic factors. Patients with postoperative CA 19.9 levels>90 U/ml did not benefit from adjuvant chemotherapy (P=0.7194) compared with those with a CA19.9 of ≤90 U/ml (median 26.0 vs 16.7 months, P=0.0108). Normalization of CA19.9 within 6 months of resection was also an independent favorable prognostic factor (median 29.9 vs 14.8 months, P=0.0004) and normal perioperative CA19.9 levels identified a good prognostic group, which was associated with a 5-year survival of 42%. CONCLUSIONS: Perioperative serum CA19.9 measurements are informative in patients with detectable CA19.9 (defined by serum levels of >5 U/ml) and have potential clinical utility in predicting outcome and response to adjuvant chemotherapy. Future clinical trials should prioritize incorporation of CA19.9 measurement at key decision points to prospectively validate these findings and facilitate implementation.


Subject(s)
Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Carcinoma, Pancreatic Ductal/blood , Neoplasm Recurrence, Local , Pancreatic Neoplasms/blood , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/therapy , Chemotherapy, Adjuvant , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Perioperative Period , Prognosis , Proportional Hazards Models , Retrospective Studies
3.
Oncogene ; 30(28): 3186-97, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21383694

ABSTRACT

Recognition of the pivotal role of estrogen in the aetiology of breast cancer has led to the development of antiestrogens (AE), such as tamoxifen (TAM) as effective therapies for the treatment and prevention of this disease. However, despite their widespread clinical efficacy, response to AEs is often short-lived, and acquired or innate therapeutic resistance remains a major obstacle in the successful treatment of breast cancer. Thus, delineating the intracellular pathways that mediate the cellular response to estrogen could potentially lead to new, more effective approaches to the treatment of breast cancer, particularly endocrine-resistant disease. Here, we have identified the BCL-2 homology 3 (BH3)-only, pro-apoptotic regulator, PUMA (p53 upregulated modulator of apoptosis) as an estrogen target gene that is acutely downregulated in response to estrogen in breast cancer cell lines, independently of their p53 status. PUMA is transcriptionally upregulated following treatment with TAM, and knock down of PUMA expression in these cells attenuates the apoptotic response to TAM. Furthermore, low PUMA expression in breast carcinomas is significantly associated with breast cancer-specific death (P=0.0014 and P=0.0115, for mRNA and protein, respectively), and worse outcome in TAM-treated patients (mRNA, P=1.49e-05). These findings suggest that the dysregulation of apoptotic signaling pathways such as those executed through PUMA, can significantly impact on both the progression and therapeutic responsiveness of breast cancer. Moreover, they provide a convincing rationale for exploring new therapeutic approaches involving endocrine and non-endocrine therapies that target apoptotic pathways as an effective strategy for tackling endocrine refractory disease.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tamoxifen/pharmacology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cohort Studies , Disease Progression , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Antagonists/therapeutic use , Estrogens/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Transcription, Genetic/drug effects , Treatment Outcome , Young Adult
4.
Mol Endocrinol ; 24(7): 1380-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20519331

ABSTRACT

Prolactin and progesterone act together to regulate mammary alveolar development, and both hormones have been implicated in breast cancer initiation and progression. Here we show that Elf5, a prolactin-induced ETS transcription factor that specifies the mammary secretory cell lineage, is also induced by progestins in breast cancer cells via a direct mechanism. To define the transcriptional response to progestin elicited via Elf5, we made an inducible Elf5 short hairpin-RNA knock-down model in T47D breast cancer cells and used it to prevent the progestin-induction of Elf5. Functional analysis of Affymetrix gene expression data using Gene Ontologies and Gene Set Enrichment Analysis showed enhancement of the progestin effects on cell cycle gene expression. Cell proliferation assays showed a more efficacious progestin-induced growth arrest when Elf5 was kept at baseline levels. These results showed that progestin induction of Elf5 expression tempered the antiproliferative effects of progestins in T47D cells, providing a further mechanistic link between prolactin and progestin in the regulation of mammary cell phenotype.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Progestins/pharmacology , Progestins/therapeutic use , Proto-Oncogene Proteins c-ets/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Female , Humans , Mifepristone/pharmacology , Oligonucleotide Array Sequence Analysis , RNA Interference , Transcription Factors
5.
Oncogene ; 28(15): 1812-20, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19287456

ABSTRACT

Aberrant expression of cyclin D1 protein is a common feature of breast cancer. However, the CCND1 gene encodes two gene products, cyclin D1a and cyclin D1b, which have discrete mechanisms of regulation and impact on cell behavior. A polymorphism at nucleotide 870 in the CCND1 gene, rs603965, influences the relative production of the encoded proteins and can impart increased risk for tumor development. Here, the impact of both the G/A870 polymorphism and cyclin D1b protein production on breast cancer risk, disease phenotype and patient outcome was analysed. In a large multiethnic case-control study, the G/A870 polymorphism conferred no significant risk for breast cancer overall or by stage or estrogen receptor (ER) status. However, the cyclin D1b protein was found to be upregulated in breast cancer, independent of cyclin D1a levels, and exhibited heterogeneous levels in breast cancer specimens. High cyclin D1a expression inversely correlated with the Ki67 proliferation marker and was not associated with clinical outcome. In contrast, elevated cyclin D1b expression was independently associated with adverse outcomes, including recurrence, distant metastasis and decreased survival. Interestingly, cyclin D1b was particularly associated with poor outcome in the context of ER-negative breast cancer. Thus, specific cyclin D1 isoforms are associated with discrete forms of breast cancer and high cyclin D1b protein levels hold prognostic potential.


Subject(s)
Breast Neoplasms/chemistry , Cyclin D1/analysis , Breast Neoplasms/etiology , Breast Neoplasms/mortality , Cyclin D1/genetics , Genes, erbB-2 , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Polymorphism, Genetic , Prognosis , Protein Isoforms , Receptors, Estrogen/analysis
6.
Br J Cancer ; 100(1): 123-33, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19066611

ABSTRACT

BAG-1 (bcl-2-associated athanogene) enhances oestrogen receptor (ER) function and may influence outcome and response to endocrine therapy in breast cancer. We determined relationships between BAG-1 expression, molecular phenotype, response to tamoxifen therapy and outcome in a cohort of breast cancer patients and its influence on tamoxifen sensitivity in MCF-7 breast cancer cells in vitro. Publically available gene expression data sets were analysed to identify relationships between BAG-1 mRNA expression and patient outcome. BAG-1 protein expression was assessed using immunohistochemistry in 292 patients with invasive ductal carcinoma and correlated with clinicopathological variables, therapeutic response and disease outcome. BAG-1-overexpressing MCF-7 cells were treated with antioestrogens to assess its effects on cell proliferation. Gene expression data demonstrated a consistent association between high BAG-1 mRNA and improved survival. In ER+ cancer (n=189), a high nuclear BAG-1 expression independently predicted improved outcome for local recurrence (P=0.0464), distant metastases (P=0.0435), death from breast cancer (P=0.009, hazards ratio 0.29, 95% CI: 0.114-0.735) and improved outcome in tamoxifen-treated patients (n=107; P=0.0191). BAG-1 overexpression in MCF-7 cells augmented antioestrogen-induced growth arrest. A high BAG-1 expression predicts improved patient outcome in ER+ breast carcinoma. This may reflect both a better definition of the hormone-responsive phenotype and a concurrent increased sensitivity to tamoxifen.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , DNA-Binding Proteins/physiology , Estrogen Antagonists/therapeutic use , Receptors, Estrogen/analysis , Tamoxifen/therapeutic use , Transcription Factors/physiology , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Humans , Immunohistochemistry , Neoplasm Invasiveness , RNA, Messenger/analysis , Transcription Factors/analysis , Transcription Factors/genetics
7.
Br J Cancer ; 98(3): 537-41, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18231110

ABSTRACT

Identification of a biomarker of prognosis and response to therapy that can be assessed preoperatively would significantly improve overall outcomes for patients with pancreatic cancer. In this study, patients whose tumours exhibited high LMO4 expression had a significant survival advantage following operative resection, whereas the survival of those patients whose tumours had low or no LMO4 expression was not significantly different when resection was compared with operative biopsy alone.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Homeodomain Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/surgery , Cohort Studies , Female , Humans , LIM Domain Proteins , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/surgery , Prognosis , Survival Analysis , Treatment Outcome
8.
Oncogene ; 27(1): 126-38, 2008 Jan 03.
Article in English | MEDLINE | ID: mdl-17599043

ABSTRACT

Progesterone regulates the proliferation and differentiation of normal mammary epithelium. In breast cancer cells, progesterone and its synthetic analogs, progestins, induce long-term growth inhibition and differentiation. However, the mechanisms responsible are not fully understood. When T-47D breast cancer cells were treated with the synthetic progestin ORG 2058 (16alpha-ethoxy-21-hydroxy-19-norpregn-4-en-3,20-dione), all isoforms of Wilms' tumor protein 1 (Wt1) mRNA and protein were rapidly downregulated. We reasoned that the decrease in Wt1 levels may contribute to the long-term antiproliferative and differentiative effects of progestins as proliferation and differentiation are known end points of Wt1 action. Consistent with this idea, Wt1 small interfering RNA led to a decrease in S phase and cyclin D1 levels, and increased Oil-Red-O staining, indicating increased lipogenesis. Conversely, overexpression of Wt1 attenuated the decrease in S phase induced by ORG 2058 at 48-96 h. This was accompanied by the sustained expression of cyclin D1 despite progestin treatment, and increased levels of retinoblastoma (Rb) phosphorylation at sites targeted by cyclin D1-Cdk4 (Ser249/Thr252). Wt1 overexpression also attenuated the ORG 2058-mediated increase in fatty acid synthase levels and reduced lipogenesis. Thus, Wt1 downregulation was sufficient to mimic the effects of progestin and was necessary for complete progestin-mediated proliferative arrest and subsequent differentiation. Furthermore, Wt1 overexpression modulated the effects of progestins but not anti-estrogens or androgens. These results indicate that Wt1 is an important early target of progestins that regulates both proliferation and differentiation in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Proliferation , Progestins/physiology , WT1 Proteins/biosynthesis , Cell Differentiation/genetics , Cell Line, Transformed , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Lipogenesis/genetics , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , WT1 Proteins/physiology
9.
Endocr Relat Cancer ; 10(2): 179-86, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12790780

ABSTRACT

The central involvement of estrogen in the development of the mammary gland and in the genesis of breast cancer has lent impetus to studies of the links between estrogen action and the cell cycle machinery. Recent studies of the estrogenic regulation of molecules with known roles in the control of G1/S phase progression have resulted in significant advances in understanding these links. Estrogens independently regulate the expression and function of c-Myc and cyclin D1 and the induction of either c-Myc or cyclin D1 is sufficient to recapitulate the effects of estrogen on cell cycle progression. These pathways converge at the activation of cyclin E-Cdk2 complexes. The active cyclin E-Cdk2 complexes are depleted of the cyclin dependent kinase (CDK) inhibitor p21(WAF1/CIP1) because of estrogen-mediated inhibition of nascent p21(WAF1/CIP1). Insulin and estrogen synergistically stimulate cell cycle progression, and the ability of estrogen to antagonize an insulin-induced increase in p21(WAF1/CIP1) gene expression appears to underlie this effect. Antiestrogen treatment of MCF-7 cells leads to an acute decrease of c-Myc expression, a subsequent decline in cyclin D1, and ultimately arrest of cells in a state with features characteristic of quiescence. An antisense-mediated decrease in c-Myc expression results in decreased cyclin D1 expression and inhibition of DNA synthesis, mimicking the effects of antiestrogen treatment and emphasizing the importance of c-Myc as an estrogen/antiestrogen target. These data identify c-Myc, cyclin D1, p21(WAF1/CIP1) and cyclin E-Cdk2 as central components of estrogen regulation of cell cycle progression and hence as potential downstream targets that contribute to the role of estrogen in oncogenesis.


Subject(s)
Breast Neoplasms/metabolism , CDC2-CDC28 Kinases , Cell Cycle Proteins/metabolism , Cell Cycle/drug effects , Estrogen Receptor Modulators/pharmacology , Estrogens/pharmacology , Animals , Breast Neoplasms/pathology , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Female , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism
10.
J Biol Chem ; 276(50): 47675-83, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11590147

ABSTRACT

Long-term growth inhibition, arrest in G(1) phase and reduced activity of both cyclin D1-Cdk4 and cyclin E-Cdk2 are elicited by progestin treatment of breast cancer cells in culture. Decreased cyclin expression, induction of p18(INK4c) and increased association of the CDK inhibitors p21(WAF1/Cip1) and p27(Kip1) with cyclin E-Cdk2 have been implicated in these responses. To determine the role of decreased cyclin expression, T-47D human breast cancer cells constitutively expressing cyclin D1 or cyclin E were treated with the progestin ORG 2058. Overexpression of cyclin E had only a modest effect on growth inhibition. Although cyclin E expression was maintained during progestin treatment, cyclin E-Cdk2 activity decreased by approximately 60%. This was accompanied by p27(Kip1) association with cyclin E-Cdk2, indicating that both cyclin E down-regulation and p27(Kip1) recruitment contribute to the decrease in activity. In contrast, overexpression of cyclin D1 induced progestin resistance and cell proliferation continued despite decreased cyclin E-Cdk2 activity. Progestin treatment of cyclin D1-overexpressing cells was associated with increased p27(Kip1) association with cyclin E-Cdk2. Thus the ability of cyclin D1 to confer progestin resistance does not depend on sequestration of p27(Kip1) away from cyclin E-Cdk2, providing evidence for a critical function of cyclin D1 other than as a high-capacity "sink" for p27(Kip1). These data indicate that regulation of cyclin D1 is a critical element of progestin inhibition in breast cancer cells and suggest that breast cancers overexpressing cyclin D1 may respond poorly to progestin therapy.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , Drug Resistance, Neoplasm , Progestins/antagonists & inhibitors , Progestins/pharmacology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Division , Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , Down-Regulation , Drug Resistance , Flow Cytometry , G1 Phase , Phosphorylation , Precipitin Tests , Pregnenediones/pharmacology , Progesterone Congeners/pharmacology , Prognosis , Protein Binding , Retroviridae/genetics , Retroviridae/metabolism , S Phase , Time Factors
11.
J Biol Chem ; 276(49): 45945-51, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11595732

ABSTRACT

The mechanism by which all-trans retinoic acid (ATRA) leads to a G(1) arrest of the cell cycle remains unclear. We show here that the decrease in D-type cyclin levels observed following ATRA treatment correlates with an increase in the rate of cyclin D1 ubiquitylation in both T-47D and MCF-7 breast cancer cell lines. However, MCF-7 cells are more resistant to ATRA than T-47D cells indicating that cyclin D1 degradation is not sufficient for ATRA-mediated arrest. We found a striking difference between these cells in that while ATRA induces an elevation in the cdk inhibitor p27 in T-47D cells, this is not observed in the ATRA-resistant MCF-7 cells. Furthermore, we demonstrate that ATRA promotes the ubiquitylation of Skp2, an F-box protein that targets p27 for degradation. Moreover, overexpression of Skp2 in T-47D cells prevents accumulation of p27 and promotes resistance to ATRA. In addition, overexpression of cyclin D1 in T-47D cells also promotes ATRA resistance. We found that the mechanism of ATRA-induced ubiquitylation of cyclin D1 and Skp2 is independent of CUL-1 expression and that ATRA can rescue cyclin D1 degradation in the uterine cell line SK-UT-1, where D-type cyclins are stabilized due to a specific defect in proteolysis. These data suggest that ATRA induces a novel pathway of ubiquitylation and that the degradation of the F-box protein Skp2 is the mechanism underlying p27 accumulation and cyclin E-cdk2 inactivation following ATRA treatment.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division/drug effects , Tretinoin/pharmacology , Ubiquitin/metabolism , Cell Cycle/drug effects , Humans , Hydrolysis , Reverse Transcriptase Polymerase Chain Reaction , S-Phase Kinase-Associated Proteins , Tumor Cells, Cultured
12.
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
13.
J Biol Chem ; 275(49): 38221-9, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10991938

ABSTRACT

Estrogen antagonists inhibit cell cycle progression in estrogen-responsive cells, but the molecular mechanisms are not fully defined. Antiestrogen-mediated G(0)/G(1) arrest is associated with decreased cyclin D1 gene expression, inactivation of cyclin D1-cyclin dependent kinase (Cdk) 4 complexes, and decreased phosphorylation of the retinoblastoma protein (pRb). We now show that treatment of MCF-7 breast cancer cells with the pure estrogen antagonist ICI 182780 results in inhibition of cyclin E-Cdk2 activity prior to a decrease in the G(1) to S phase transition. This decrease was dependent on p21(WAF1/Cip1) since treatment with antisense oligonucleotides to p21 attenuated the effect. Recruitment of p21 to cyclin E-Cdk2 complexes was in turn dependent on decreased cyclin D1 expression since it was apparent following treatment with antisense cyclin D1 oligonucleotides. To define where within the G(0) to S phase continuum antiestrogen-treated cells arrested, we assessed the relative abundance and phosphorylation state of pocket protein-E2F complexes. While both pRb and p107 levels were significantly decreased, p130 was increased 4-fold and was accompanied by the formation of p130.E2F4 complexes and the accumulation of hyperphophorylated E2F4, putative markers of cellular quiescence. Thus, ICI 182780 inhibits both cyclin D1-Cdk4 and cyclin E-Cdk2 activity, resulting in the arrest of MCF-7 cells in a state with characteristics of quiescence (G(0)), as opposed to G(1) arrest.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/physiology , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/metabolism , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins , Transcription Factors/metabolism , Breast Neoplasms , Cell Cycle/drug effects , Cell Division , Cyclin D1/genetics , Cyclin D1/physiology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , E2F4 Transcription Factor , Enzyme Inhibitors/metabolism , Estradiol/pharmacology , Female , Fulvestrant , Humans , Kinetics , Models, Biological , Oligodeoxyribonucleotides, Antisense/pharmacology , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/physiology , Retinoblastoma-Like Protein p130 , Tumor Cells, Cultured
14.
Clin Cancer Res ; 6(7): 2777-87, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914724

ABSTRACT

Frequent deletions or mutations of the INK4 gene, which encodes the cyclin-dependent kinase 4 inhibitor p16INK4a, have been documented in various human cancers, but little is known about the role of this tumor suppressor gene in primary breast cancer. We examined p16INK4a mRNA expression and its relationship with cyclin D1 and estrogen receptor (ER) expression in 314 primary breast cancers using Northern blots probed with a p16 exon 1alpha-specific cDNA. Tumor samples overexpressing p16INK4a were predominantly ER negative with low levels of cyclin D1. Cyclin D1 and ER mRNA levels in the high p16INK4a expressers were significantly lower than those in the remainder of the population (P = 0.0001). Furthermore, the mean p16INK4a mRNA level in the ER-negative tumors was significantly higher than that in the ER-positive group (P = 0.0001). Because the INK4 gene is frequently inactivated by de novo methylation, we investigated the frequency of INK4a exon 1alpha methylation in a subset of 120 primary breast cancers using methylation-specific PCR; 24 of these were methylated. These findings indicate that high expression of p16INK4a and reduced expression due to de novo INK4a methylation are frequent events in primary breast cancer. In a subset of 217 patients for whom detailed clinical data were available, high p16INK4a mRNA expression was associated with high tumor grade (P = 0.006), > or = 4 axillary lymph node involvement (P = 0.004), ER negativity (P = 0.0001), and increased risk of relapse (P = 0.006). The significant negative correlation between p16INK4a and ER gene expression raises issues regarding their functional interrelationships and whether high p16INK4a expression may be associated with a lack of hormone responsiveness in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carrier Proteins/genetics , Genes, Tumor Suppressor , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carrier Proteins/analysis , Cyclin D1/analysis , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Disease-Free Survival , Exons , Female , Humans , Middle Aged , Predictive Value of Tests , Prognosis , RNA, Messenger/analysis , Receptors, Estrogen/analysis , Survival Rate , Time Factors , Tumor Cells, Cultured
15.
Mol Cell Biol ; 20(7): 2581-91, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10713180

ABSTRACT

The steroid hormone progesterone regulates proliferation and differentiation in the mammary gland and uterus by cell cycle phase-specific actions. The long-term effect of progestins on T-47D breast cancer cells is inhibition of cellular proliferation. This is accompanied by decreased G(1) cyclin-dependent kinase (CDK) activities, redistribution of the CDK inhibitor p27(Kip1) among these CDK complexes, and alterations in the elution profile of cyclin E-Cdk2 upon gel filtration chromatography, such that high-molecular-weight complexes predominate. This study aimed to determine the relative contribution of CDK inhibitors to these events. Following progestin treatment, the majority of cyclin E- and D-CDK complexes were bound to p27(Kip1) and few were bound to p21(Cip1). In vitro, recombinant His(6)-p27 could quantitatively reproduce the effects on cyclin E-Cdk2 kinase activity and the shift in molecular weight observed following progestin treatment. In contrast, cyclin D-Cdk4 was not inhibited by His(6)-p27 in vitro or p27(Kip1) in vivo. However, an increase in the expression of the Cdk4/6 inhibitor p18(INK4c) and its extensive association with Cdk4 and Cdk6 were apparent following progestin treatment. Recombinant p18(INK4c) led to the reassortment of cyclin-CDK-CDK inhibitor complexes in vitro, with consequent decrease in cyclin E-Cdk2 activity. These results suggest a concerted model of progestin action whereby p27(Kip1) and p18(INK4c) cooperate to inhibit cyclin E-Cdk2 and Cdk4. Since similar models have been developed for growth inhibition by transforming growth factor beta and during adipogenesis, interaction between the Cip/Kip and INK4 families of inhibitors may be a common theme in physiological growth arrest and differentiation.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Cycle/drug effects , Microtubule-Associated Proteins/metabolism , Progestins/pharmacology , Tumor Suppressor Proteins , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Pregnenediones/pharmacology , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Tumor Cells, Cultured
16.
J Physiol ; 529 Pt 3: 541-52, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11195932

ABSTRACT

NCC27 is a nuclear chloride ion channel, identified in the PMA-activated U937 human monocyte cell line. NCC27 mRNA is expressed in virtually all cells and tissues and the gene encoding NCC27 is also highly conserved. Because of these factors, we have examined the hypothesis that NCC27 is involved in cell cycle regulation. Electrophysiological studies in Chinese hamster ovary (CHO-K1) cells indicated that NCC27 chloride conductance varied according to the stage of the cell cycle, being expressed only on the plasma membrane of cells in G2/M phase. We also demonstrate that Cl- ion channel blockers known to block NCC27 led to arrest of CHO-K1 cells in the G2/M stage of the cell cycle, the same stage at which this ion channel is selectively expressed on the plasma membrane. These data strongly support the hypothesis that NCC27 is involved, in some as yet undetermined manner, in regulation of the cell cycle.


Subject(s)
Cell Cycle/physiology , Chloride Channels/physiology , Animals , Anthracenes/pharmacology , CHO Cells , Cell Membrane/metabolism , Cell Size/physiology , Chloride Channels/genetics , Chlorides/physiology , Conserved Sequence/genetics , Cricetinae , Electric Conductivity , Electrophysiology , G2 Phase , Gene Expression , Glycolates/pharmacology , Intracellular Membranes/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Mitosis , Multigene Family , Transfection
17.
Clin Cancer Res ; 5(8): 2069-76, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10473088

ABSTRACT

Cyclin D1 is a key cell cycle regulatory protein with demonstrated oncogenic activity in a variety of malignancies. Cyclin D1 mRNA and protein are overexpressed in approximately 50% of primary breast carcinomas; however, the pathophysiological consequences of increased expression remain unclear. To investigate the functional sequelae of cyclin D1 mRNA overexpression, we analyzed clinical outcome in relation to the cyclin D1 mRNA level in 253 primary breast cancer patients (median follow-up, 75 months) with particular reference to estrogen receptor (ER) status and endocrine response. Overall, with the exception of the relationship between cyclin D1 mRNA expression and the ER, cyclin D1 mRNA was not associated with other clinicopathological features such as age, menopausal status, axillary lymph node status, vascular invasion, tumor size, type, and grade. However, in patients with ER-positive tumors (n = 182), high levels of cyclin D1 mRNA were associated with increased risk of relapse (P = 0.0016), local recurrence (P = 0.025), metastasis (P = 0.019), and death (P = 0.025). In contrast, there were no clinical correlations with cyclin D1 expression in ER-negative disease (n = 71). In 33 patients who received endocrine therapy for their primary or recurrent breast cancers, there was an apparent association between a high cyclin D1 mRNA level and a shorter response duration within the ER-positive subgroup (P = 0.04). Our findings indicate that overexpression of cyclin D1 mRNA correlates with a worse prognosis within the ER-positive breast cancer phenotype and may be a contributing factor to the development of endocrine resistance in ER-positive disease.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Cyclin D1/metabolism , Receptors, Estrogen/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Female , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/biosynthesis , Survival Rate , Tamoxifen/therapeutic use , Treatment Outcome
18.
Oncogene ; 17(8): 1053-9, 1998 Aug 27.
Article in English | MEDLINE | ID: mdl-9747885

ABSTRACT

The EMS1 and CCND1 genes at chromosome 11q13 are amplified in about 15% of primary breast cancers but appear to confer different phenotypes in ER positive and ER negative tumours. Since there are no published data on EMS1 expression in large series of breast cancers we examined the relationship of EMS1 expression with EMS1 gene copy number and expression of mRNAs for cyclin D1 and ER. In a subset of 129 patients, where matched tumour RNA and DNA was available, EMS1 mRNA overexpression was associated predominantly with gene amplification (P = 0.0061), whereas cyclin D1 mRNA overexpression was not (P = 0.3142). In a more extensive series of 351 breast cancers, there was no correlation between cyclin D1 and EMS1 expression in the EMS1 and cyclin D1 overexpressors (P = 0.3503). Although an association between EMS1 mRNA expression and ER positivity was evident (P = 0.0232), when the samples were divided into quartiles of EMS1 or cyclin D1 mRNA expression, the increase in the proportion of ER positive tumours in the ascending EMS1 mRNA quartiles was not statistically significant (P = 0.0951). In marked contrast there was a significant stepwise increase in ER positivity in ascending quartiles of cyclin D1 mRNA (P = 0.030). A potential explanation for this difference was provided by the observation that in ER positive breast cancer cells oestradiol treatment resulted in increased cyclin D1 gene expression but was without effect on EMS1. The relationship between EMS1 expression and clinical outcome was examined in a subset of 234 patients with median follow-up of 74 months. High EMS1 expression was associated with age > 50 years (P = 0.0001), postmenopausal status (P = 0.0008), lymph node negativity (P = 0.019) and an apparent trend for worse prognosis in the ER negative subgroup. These data demonstrate that overexpression of EMS1 mRNA is largely due to EMS1 gene amplification, is independent of cyclin D1 and ER expression and, in contrast to cyclin D1, is not regulated by oestrogen. Independent overexpression of these genes may confer different phenotypes and disease outcomes in breast cancer as has been inferred from recent studies of EMS1 and CCND1 gene amplification.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cyclin D1/biosynthesis , Gene Expression Regulation, Neoplastic , Microfilament Proteins , Neoplasm Proteins/genetics , Receptors, Estrogen/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Chromosomes, Human, Pair 11/genetics , Cortactin , Cyclin D1/genetics , Humans , Middle Aged , RNA, Messenger/biosynthesis
19.
Oncogene ; 16(21): 2803-13, 1998 May 28.
Article in English | MEDLINE | ID: mdl-9652748

ABSTRACT

Members of the erbB family of receptor tyrosine kinases are commonly overexpressed in human breast cancer. However, the relative contribution of particular signalling pathways activated downstream of these receptors to the mitogenic response of transformed breast epithelial cells remains poorly characterized. Administration of heregulin-beta2 (HRG), a ligand for erbB3 and erbB4, to growth arrested T-47D human breast cancer cells leads to activation of both the PI3-kinase and MAP kinase signalling pathways and potent stimulation of cell cycle progression. Specific inhibitors were used to assess the role of these pathways in HRG-induced mitogenesis and to identify underlying mechanisms in terms of regulation of gene expression. Treatment with the MEK inhibitor PD98059 led to a complete block of HRG-induced entry into S-phase, whilst administration of the PI3-kinase inhibitor wortmannin resulted in only modest inhibition. In addition, administration of PD98059 8 h after HRG was equipotent with simultaneous administration in inhibiting entry into S-phase. However, delaying addition for 14-16 h after HRG, when the cells were entering S-phase, was without effect. HRG stimulation led to sequential induction of c-myc, cyclin D1, cyclin E and cyclin A gene expression and hyperphosphorylation of the retinoblastoma protein pRB. p21 (WAF1/CIP1/SDI1) gene expression was rapidly induced by HRG, but significant changes in p27 (KIP1) protein levels were not detected. Preincubation with PD98059 blocked the HRG-dependent induction of cyclin D1 mRNA, p21 and c-Myc protein and pRB phosphorylation. These findings demonstrate that MEK activation is critical to HRG-induced S-phase entry in these cells whilst PI3-kinase plays a minor role. Moreover, these data are compatible with HRG-induced activation of MEK being critical for a mid-G1 transition point and implicate c-myc and cyclin D1 as key targets of the MAP kinase pathway involved in this response.


Subject(s)
Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carrier Proteins/pharmacology , Glycoproteins/pharmacology , Neuregulin-1 , Androstadienes/pharmacology , Breast Neoplasms/enzymology , Cell Cycle/drug effects , Cyclin D1/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Gene Expression , Humans , Mitogen-Activated Protein Kinase Kinases , Mitogens/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors , Protein Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured , Up-Regulation , Wortmannin
20.
Oncogene ; 16(22): 2865-78, 1998 Jun 04.
Article in English | MEDLINE | ID: mdl-9671407

ABSTRACT

The G1 cyclins, cyclin D1 and E, are rate limiting for progression through G1 phase of the cell cycle in breast epithelial cells and are oncogenic when expressed in the mammary epithelium of transgenic mice. These genes are frequently overexpressed in clinical breast cancer where overexpression appears to be associated with specific disease phenotypes, altered responsiveness to therapeutic intervention and patient survival. In order to investigate the functional correlates of cyclin D1 and cyclin E overexpression we employed a panel of normal, immortalized and neoplastic breast epithelial cell lines to examine the relationships between cyclin gene expression, cyclin-CDK complex formation and CDK activity. In agreement with earlier studies cyclin D1 and E expression varied over an approximately tenfold range among the 18 cell lines studied. There was no apparent relationship, however, between cyclin D1 expression and the in vitro activity of its major kinase partner, Cdk4, although MDA-MB-134 cells displayed the highest level of both cyclin D1 expression and Cdk4 activity. Similarly, there was no significant relationship between cyclin E expression and cyclin E-Cdk2 activity. Fractionation of whole cell lysates by gel filtration chromatography revealed that approximately 90% of the cyclin E protein was present in inactive complexes containing the CDK inhibitors p21 and p27. Much of the small fraction of active cyclin E protein was of very high apparent molecular mass, >400 kDa, suggesting that formation of these complexes is a more important determinant of cyclin E-Cdk2 activity than cyclin E abundance. These data suggest that properties of cyclins D1 and E in addition to their ability to activate Cdk4 and Cdk2 may contribute to the effects of overexpression on the breast cancer phenotype.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , Cyclins/biosynthesis , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Breast Neoplasms/enzymology , Cell Division , Chromatography, Gel , Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Cyclin G , Cyclin G1 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/biosynthesis , Enzyme Inhibitors/metabolism , Epithelial Cells , Humans , Microtubule-Associated Proteins/biosynthesis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...