Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nucleic Acids Res ; 45(19): 11056-11069, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28977491

ABSTRACT

Cancer genome sequencing has implicated the cytosine deaminase activity of apolipoprotein B mRNA editing enzyme catalytic polypeptide-like (APOBEC) genes as an important source of mutations in diverse cancers, with APOBEC3B (A3B) expression especially correlated with such cancer mutations. To better understand the processes directing A3B over-expression in cancer, and possible therapeutic avenues for targeting A3B, we have investigated the regulation of A3B gene expression. Here, we show that A3B expression is inversely related to p53 status in different cancer types and demonstrate that this is due to a direct and pivotal role for p53 in repressing A3B expression. This occurs through the induction of p21 (CDKN1A) and the recruitment of the repressive DREAM complex to the A3B gene promoter, such that loss of p53 through mutation, or human papilloma virus-mediated inhibition, prevents recruitment of the complex, thereby causing elevated A3B expression and cytosine deaminase activity in cancer cells. As p53 is frequently mutated in cancer, our findings provide a mechanism by which p53 loss can promote cancer mutagenesis.


Subject(s)
Cytidine Deaminase/genetics , Gene Expression Regulation, Neoplastic , Minor Histocompatibility Antigens/genetics , Tumor Suppressor Protein p53/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytidine Deaminase/metabolism , HCT116 Cells , Humans , Immunoblotting , Minor Histocompatibility Antigens/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
2.
Nucleic Acids Res ; 44(2): 582-94, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26400164

ABSTRACT

Liver receptor homologue 1 (LRH-1) is an orphan nuclear receptor that has been implicated in the progression of breast, pancreatic and colorectal cancer (CRC). To determine mechanisms underlying growth promotion by LRH-1 in CRC, we undertook global expression profiling following siRNA-mediated LRH-1 knockdown in HCT116 cells, which require LRH-1 for growth and in HT29 cells, in which LRH-1 does not regulate growth. Interestingly, expression of the cell cycle inhibitor p21 (CDKN1A) was regulated by LRH-1 in HCT116 cells. p21 regulation was not observed in HT29 cells, where p53 is mutated. p53 dependence for the regulation of p21 by LRH-1 was confirmed by p53 knockdown with siRNA, while LRH-1-regulation of p21 was not evident in HCT116 cells where p53 had been deleted. We demonstrate that LRH-1-mediated p21 regulation in HCT116 cells does not involve altered p53 protein or phosphorylation, and we show that LRH-1 inhibits p53 recruitment to the p21 promoter, likely through a mechanism involving chromatin remodelling. Our study suggests an important role for LRH-1 in the growth of CRC cells that retain wild-type p53.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/genetics , Tumor Suppressor Protein p53/genetics , Binding Sites , Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Deletion , HCT116 Cells , HT29 Cells , Humans , Mutation , Organ Specificity , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
3.
Breast Cancer Res Treat ; 159(2): 215-27, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27514395

ABSTRACT

Differential prognostic roles of Androgen Receptor (AR) have been proposed in breast cancer (BC) depending on tumour oestrogen receptor (ER) status. This study aimed to evaluate the prognostic and/or predictive significance of AR expression in invasive BC. In this study AR expression was studied on a large (n = 1141) consecutive series of early-stage (I-III) BC using tissue microarray and immunohistochemistry (IHC). AR mRNA expression was assessed in a subset of cases. The prognostic impact of AR mRNA expression was externally validated using the online BC gene expression data sets (n = 25 data sets, 4078 patients). Nuclear AR IHC expression was significantly associated with features of good prognosis including older age, smaller tumour size, lower grade and lobular histology particularly in the ER-positive tumours. AR was associated with ER-related markers GATA3, FOXa1, RERG and BEX1. Negative association was observed with HER2, p53, Ki67, TK1, CD71 and AGTR1. AR Overexpression was associated with longer survival (p < 0.001), independent of tumour size, grade, stage [p = 0.033, hazard ratio (HR) = 0.80 95 % CI = 0.64-0.98]. Similar associations were maintained in ER+ tumours in univariate and multivariate analysis (p < 0.01) both in patients with and without adjuvant endocrine or chemotherapy. AR mRNA expression showed significant association with tumour grade, molecular subtypes, and longer 10 and 15 years survival in luminal BC. In the external validation cohorts, AR gene expression data were associated with improved patients' outcome (p < 0.001, HR = 0.84, 95 % CI 0.79-0.90). AR is not only an independent prognostic factor in ER-positive luminal BC but is also expressed in ER-negative tumours. AR could act as a molecular target in patients with ER-positive disease predicting response to adjuvant therapy.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Nucleus , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Grading , Prognosis , Receptors, Estrogen/metabolism , Retrospective Studies , Survival Analysis , Tissue Array Analysis , Tumor Burden
4.
Breast Cancer Res Treat ; 150(3): 511-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25794775

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARγ) is an adopted orphan receptor that belongs to the nuclear receptor superfamily of transcription factors. PPARγ is regarded as a differentiation factor and it plays an important role in regulating adipogenesis, cell growth, proliferation and tumour progression. In breast cancer (BC), PPARγ agonists were reported to inhibit proliferation and growth invasion and promote phenotypic changes associated with a less malignant and more differentiated status. This study aims to assess the prognostic and biological roles of PPARγ protein expression in a large cohort of BC patients (n = 1100) with emphasis on the luminal oestrogen receptor (ER) positive class. Immunohistochemistry was used to assess the levels of PPARγ expression in BC series prepared as tissue microarrays (TMAs). PPARγ antibody specificity was confirmed using Western blotting. PPARγ nuclear expression was detected in 79 % of the cases and its expression was positively correlated with the hormonal receptors (ER, progesterone receptor and androgen receptor). PPARγ levels were significantly higher in tumours with lobular subtype, smaller size and lower grade, while HER2-positive, ductal or medullary tumours were associated with lower PPARγ levels. Survival analysis showed that PPARγ is associated with better outcome in the whole series as well as in luminal ER-positive class. Cox regression model showed that PPARγ is an independent predictor of outcome. Higher PPARγ was associated with longer survival in patients with ER-positive tumours who did not receive hormone therapy. PPARγ is a good prognostic marker associated with hormone receptors. In patients with luminal BCs, PPARγ is a marker of better prognosis and is associated with longer survival.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , PPAR gamma/metabolism , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Female , Humans , Middle Aged , Neoplasm Grading , Prognosis , Receptors, Estrogen/metabolism , Survival Analysis , Tissue Array Analysis
5.
Nucleic Acids Res ; 41(22): 10228-40, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24049078

ABSTRACT

Oestrogen receptor α (ERα) is a nuclear receptor that is the driving transcription factor expressed in the majority of breast cancers. Recent studies have demonstrated that the liver receptor homolog-1 (LRH-1), another nuclear receptor, regulates breast cancer cell proliferation and promotes motility and invasion. To determine the mechanisms of LRH-1 action in breast cancer, we performed gene expression microarray analysis following RNA interference for LRH-1. Interestingly, gene ontology (GO) category enrichment analysis of LRH-1-regulated genes identified oestrogen-responsive genes as the most highly enriched GO categories. Remarkably, chromatin immunoprecipitation coupled to massively parallel sequencing (ChIP-seq) to identify genomic targets of LRH-1 showed LRH-1 binding at many ERα binding sites. Analysis of select binding sites confirmed regulation of ERα-regulated genes by LRH-1 through binding to oestrogen response elements, as exemplified by the TFF1/pS2 gene. Finally, LRH-1 overexpression stimulated ERα recruitment, while LRH-1 knockdown reduced ERα recruitment to ERα binding sites. Taken together, our findings establish a key role for LRH-1 in the regulation of ERα target genes in breast cancer cells and identify a mechanism in which co-operative binding of LRH-1 and ERα at oestrogen response elements controls the expression of oestrogen-responsive genes.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Breast Neoplasms/metabolism , COS Cells , Chlorocebus aethiops , Female , MCF-7 Cells , Response Elements
6.
Pflugers Arch ; 466(7): 1421-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24114173

ABSTRACT

The endogenous lipid agent N-arachidonoylethanolamine (anandamide), among other effects, has been shown to be involved in nociceptive processing both in the central and peripheral nervous systems. Anandamide is thought to be synthesised by several enzymatic pathways both in a Ca(2+)-sensitive and Ca(2+)-insensitive manner, and rat primary sensory neurons produce anandamide. Here, we show for the first time, that cultured rat primary sensory neurons express at least four of the five known Ca(2+)-insensitive enzymes implicated in the synthesis of anandamide, and that application of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-arachidonoyl, the common substrate of the anandamide-synthesising pathways, results in anandamide production which is not changed by the removal of extracellular Ca(2+). We also show that anandamide, which has been synthesised in primary sensory neurons following the application of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-arachidonoyl induces a transient receptor potential vanilloid type 1 ion channel-mediated excitatory effect that is not inhibited by concomitant activation of the cannabinoid type 1 receptor. Finally, we show that sub-populations of transient receptor potential vanilloid type 1 ion channel-expressing primary sensory neurons also express some of the putative Ca(2+)-insensitive anandamide-synthesising enzymes. Together, these findings indicate that anandamide synthesised by primary sensory neuron via a Ca(2+)-insensitive manner has an excitatory rather than an inhibitory role in primary sensory neurons and that excitation is mediated predominantly through autocrine signalling. Regulation of the activity of the Ca(2+)-insensitive anandamide-synthesising enzymes in these neurons may be capable of regulating the activity of these cells, with potential relevance to controlling nociceptive processing.


Subject(s)
Action Potentials , Arachidonic Acids/metabolism , Calcium/metabolism , Endocannabinoids/metabolism , Phosphatidylethanolamines/pharmacology , Polyunsaturated Alkamides/metabolism , Sensory Receptor Cells/metabolism , Animals , Arachidonic Acids/biosynthesis , Cells, Cultured , Endocannabinoids/biosynthesis , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Ganglia, Spinal/metabolism , Group IB Phospholipases A2/genetics , Group IB Phospholipases A2/metabolism , Lysophospholipase/genetics , Lysophospholipase/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphatidylethanolamines/chemistry , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/enzymology , Sensory Receptor Cells/physiology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
7.
Annu Rev Med ; 62: 217-32, 2011.
Article in English | MEDLINE | ID: mdl-21054173

ABSTRACT

The identification of the link between breast cancer and estrogens has led to the development of antiestrogens, in particular tamoxifen, to inhibit the activities of estrogen receptors (ERs) in breast cancer cells. The clinical use of tamoxifen has played a major part in decreasing breast cancer mortality over the past 30 years. Though antiestrogenic in the breast, some antiestrogens have estrogen-like actions in other tissues, acting to promote bone density and protect against cardiovascular disease, thus raising the possibility of their use in counteracting the effects of estrogen loss following menopause. Moreover, antiestrogens show efficacy as chemopreventive agents in women at high risk of developing breast cancer. Thus, antiestrogens define an important and well-understood class of cancer drug, which continue to be a mainstay in breast cancer treatment.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor Modulators/therapeutic use , Bone Density/drug effects , Breast/drug effects , Cardiovascular Diseases/drug therapy , Estrogens/physiology , Estrogens/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors
8.
Nat Commun ; 13(1): 2011, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440136

ABSTRACT

Estrogen receptor alpha (ER/ESR1) is frequently mutated in endocrine resistant ER-positive (ER+) breast cancer and linked to ligand-independent growth and metastasis. Despite the distinct clinical features of ESR1 mutations, their role in intrinsic subtype switching remains largely unknown. Here we find that ESR1 mutant cells and clinical samples show a significant enrichment of basal subtype markers, and six basal cytokeratins (BCKs) are the most enriched genes. Induction of BCKs is independent of ER binding and instead associated with chromatin reprogramming centered around a progesterone receptor-orchestrated insulated neighborhood. BCK-high ER+ primary breast tumors exhibit a number of enriched immune pathways, shared with ESR1 mutant tumors. S100A8 and S100A9 are among the most induced immune mediators and involve in tumor-stroma paracrine crosstalk inferred by single-cell RNA-seq from metastatic tumors. Collectively, these observations demonstrate that ESR1 mutant tumors gain basal features associated with increased immune activation, encouraging additional studies of immune therapeutic vulnerabilities.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Female , Humans , Mutation
9.
Cancer Res ; 82(7): 1321-1339, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35078818

ABSTRACT

Constitutively active estrogen receptor α (ER/ESR1) mutations have been identified in approximately one-third of ER+ metastatic breast cancers. Although these mutations are known as mediators of endocrine resistance, their potential role in promoting metastatic disease has not yet been mechanistically addressed. In this study, we show the presence of ESR1 mutations exclusively in distant but not local recurrences in five independent breast cancer cohorts. In concordance with transcriptomic profiling of ESR1-mutant tumors, genome-edited ESR1 Y537S and D538G-mutant cell models exhibited a reprogrammed cell adhesive gene network via alterations in desmosome/gap junction genes and the TIMP3/MMP axis, which functionally conferred enhanced cell-cell contacts while decreasing cell-extracellular matrix adhesion. In vivo studies showed ESR1-mutant cells were associated with larger multicellular circulating tumor cell (CTC) clusters with increased compactness compared with ESR1 wild-type CTCs. These preclinical findings translated to clinical observations, where CTC clusters were enriched in patients with ESR1-mutated metastatic breast cancer. Conversely, context-dependent migratory phenotypes revealed cotargeting of Wnt and ER as a vulnerability in a D538G cell model. Mechanistically, mutant ESR1 exhibited noncanonical regulation of several metastatic pathways, including secondary transcriptional regulation and de novo FOXA1-driven chromatin remodeling. Collectively, these data provide evidence for ESR1 mutation-modulated metastasis and suggest future therapeutic strategies for targeting ESR1-mutant breast cancer. SIGNIFICANCE: Context- and allele-dependent transcriptome and cistrome reprogramming in mutant ESR1 cell models elicit diverse metastatic phenotypes related to cell adhesion and migration, which can be pharmacologically targeted in metastatic breast cancer.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Neoplasms, Second Primary , Neoplastic Cells, Circulating , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Humans , Mutation
10.
Breast Cancer Res Treat ; 127(2): 385-96, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20607599

ABSTRACT

Estrogen receptor-α (ER) is expressed in the great majority of breast cancers, and the inhibition of ER action is a key part of breast cancer treatment. The inhibition of ER action is achieved using anti-estrogens, primarily tamoxifen, and with aromatase inhibitors that inhibit estrogen biosynthesis, thereby preventing ER activation. However, resistance to these therapies is common. With the aim of identifying new molecular targets for breast cancer therapy, we have identified the liver receptor homolog-1 (LRH-1) as an estrogen-regulated gene. RNA interference and over-expression studies were used to investigate the role of the LRH-1 in regulating breast cancer growth and to identify the targets of an LRH-1 action. Promoter recruitment was determined using reporter gene and chromatin immunoprecipitation (ChIP) assays. We show that LRH-1 regulates breast cancer cell growth by regulating the ER expression. Reporter gene and in vitro DNA-binding assays identified an LRH-1-binding site in the ER gene promoter, and ChIP assays have demonstrated in vivo binding at this site. We also provide evidence for new LRH-1 variants in breast cancer cells arising from the use of alternative promoters. Previous studies have shown that LRH-1 functions in estrogen biosynthesis by regulating aromatase expression. Our findings extend this by highlighting LRH-1 as a key regulator of the estrogen response in breast cancer cells through the regulation of ER expression. Hence, inhibition of LRH-1 could provide a powerful new approach for the treatment of endocrine-resistant breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Amino Acid Sequence , Animals , Aromatase/metabolism , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , COS Cells , Cell Line, Tumor , Cell Proliferation , Chlorocebus aethiops , Female , Gene Order , Hep G2 Cells , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Estrogen/genetics , Sequence Alignment
11.
Breast Cancer Res Treat ; 128(2): 357-68, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20730598

ABSTRACT

Estrogen receptor-α (ERα) positive breast cancer frequently responds to inhibitors of ERα activity, such as tamoxifen, and/or to aromatase inhibitors that block estrogen biosynthesis. However, many patients become resistant to these agents through mechanisms that remain unclear. Previous studies have shown that expression of ERα in ERα-negative breast cancer cell lines frequently inhibits their growth. In order to determine the consequence of ERα over-expression in ERα-positive breast cancer cells, we over-expressed ERα in the MCF-7 breast cancer cell line using adenovirus gene transduction. ERα over-expression led to ligand-independent expression of the estrogen-regulated genes pS2 and PR and growth in the absence of estrogen. Interestingly, prolonged culturing of these cells in estrogen-free conditions led to the outgrowth of cells capable of growth in cultures from ERα transduced, but not in control cultures. From these cultures a line, MLET5, was established which remained ERα-positive, but grew in an estrogen-independent manner. Moreover, MLET5 cells were inhibited by anti-estrogens showing that ERα remains important for their growth. Gene expression microarray analysis comparing MCF-7 cells with MLET5 highlighted apoptosis as a major functional grouping that is altered in MLET5 cells, such that cell survival would be favoured. This conclusion was further substantiated by the demonstration that MLET5 show resistance to etoposide-induced apoptosis. As the gene expression microarray analysis also shows that the apoptosis gene set differentially expressed in MLET5 is enriched for estrogen-regulated genes, our findings suggest that transient over-expression of ERα could lead to increased cell survival and the development of estrogen-independent growth, thereby contributing to resistance to endocrine therapies in breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Adenoviridae/genetics , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/therapeutic use , Tumor Cells, Cultured
12.
Sci Rep ; 8(1): 5237, 2018 Mar 22.
Article in English | MEDLINE | ID: mdl-29568076

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

13.
Mol Cancer Ther ; 17(6): 1156-1166, 2018 06.
Article in English | MEDLINE | ID: mdl-29545334

ABSTRACT

Recent reports indicate that some cancer types are especially sensitive to transcription inhibition, suggesting that targeting the transcriptional machinery provides new approaches to cancer treatment. Cyclin-dependent kinase (CDK)7 is necessary for transcription, and acts by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (PolII) to enable transcription initiation. CDK7 additionally regulates the activities of a number of transcription factors, including estrogen receptor (ER)-α. Here we describe a new, orally bioavailable CDK7 inhibitor, ICEC0942. It selectively inhibits CDK7, with an IC50 of 40 nmol/L; IC50 values for CDK1, CDK2, CDK5, and CDK9 were 45-, 15-, 230-, and 30-fold higher. In vitro studies show that a wide range of cancer types are sensitive to CDK7 inhibition with GI50 values ranging between 0.2 and 0.3 µmol/L. In xenografts of both breast and colorectal cancers, the drug has substantial antitumor effects. In addition, combination therapy with tamoxifen showed complete growth arrest of ER-positive tumor xenografts. Our findings reveal that CDK7 inhibition provides a new approach, especially for ER-positive breast cancer and identify ICEC0942 as a prototype drug with potential utility as a single agent or in combination with hormone therapies for breast cancer. ICEC0942 may also be effective in other cancers that display characteristics of transcription factor addiction, such as acute leukaemia and small-cell lung cancer. Mol Cancer Ther; 17(6); 1156-66. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Xenograft Model Antitumor Assays , Cyclin-Dependent Kinase-Activating Kinase
14.
Sci Rep ; 7(1): 16115, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170437

ABSTRACT

Polo-like kinase-1 (PLK1) plays a major role in driving mitotic events, including centrosome disjunction and separation, and is frequently over-expressed in human cancers. PLK1 inhibition is a promising therapeutic strategy and works by arresting cells in mitosis due to monopolar spindles. The p53 tumour suppressor protein is a short-lived transcription factor that can inhibit the growth, or stimulate the death, of developing cancer cells. Curiously, although p53 normally acts in an anti-cancer capacity, it can offer significant protection against inhibitors of PLK1, but the events underpinning this effect are not known. Here, we show that functional p53 reduces the sensitivity to PLK1 inhibitors by permitting centrosome separation to occur, allowing cells to traverse mitosis and re-enter cycle with a normal complement of 2N chromosomes. Protection entails the activation of p53 through the DNA damage-response enzymes, ATM and ATR, and requires the phosphorylation of p53 at the key regulatory site, Ser15. These data highlight a previously unrecognised link between p53, PLK1 and centrosome separation that has therapeutic implications for the use of PLK1 inhibitors in the clinic.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Benzimidazoles/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Survival/drug effects , Centrosome/drug effects , Fluorescent Antibody Technique , Gene Silencing , HCT116 Cells , Humans , Mitosis/drug effects , Mitosis/genetics , Mitosis/physiology , Morpholines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazines/pharmacology , Pyrones/pharmacology , Sulfones/pharmacology , Thiophenes/pharmacology , Tumor Suppressor Protein p53/genetics , Polo-Like Kinase 1
15.
Nat Commun ; 8(1): 1865, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29192207

ABSTRACT

Resistance to endocrine therapy remains a major clinical problem in breast cancer. Genetic studies highlight the potential role of estrogen receptor-α (ESR1) mutations, which show increased prevalence in the metastatic, endocrine-resistant setting. No naturally occurring ESR1 mutations have been reported in in vitro models of BC either before or after the acquisition of endocrine resistance making functional consequences difficult to study. We report the first discovery of naturally occurring ESR1 Y537C and ESR1 Y537S mutations in MCF7 and SUM44 ESR1-positive cell lines after acquisition of resistance to long-term-estrogen-deprivation (LTED) and subsequent resistance to fulvestrant (ICIR). Mutations were enriched with time, impacted on ESR1 binding to the genome and altered the ESR1 interactome. The results highlight the importance and functional consequence of these mutations and provide an important resource for studying endocrine resistance.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estradiol/analogs & derivatives , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/genetics , Cell Line, Tumor , Estradiol/therapeutic use , Female , Fulvestrant , Humans , MCF-7 Cells , Mutation , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use
16.
Clin Cancer Res ; 22(23): 5929-5938, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27301701

ABSTRACT

PURPOSE: CDK-activating kinase (CAK) is required for the regulation of the cell cycle and is a trimeric complex consisting of cyclin-dependent kinase 7 (CDK7), Cyclin H, and the accessory protein, MAT1. CDK7 also plays a critical role in regulating transcription, primarily by phosphorylating RNA polymerase II, as well as transcription factors such as estrogen receptor-α (ER). Deregulation of cell cycle and transcriptional control are general features of tumor cells, highlighting the potential for the use of CDK7 inhibitors as novel cancer therapeutics. EXPERIMENTAL DESIGN: mRNA and protein expression of CDK7 and its essential cofactors cyclin H and MAT1 were evaluated in breast cancer samples to determine if their levels are altered in cancer. Immunohistochemical staining of >900 breast cancers was used to determine the association with clinicopathologic features and patient outcome. RESULTS: We show that expressions of CDK7, cyclin H, and MAT1 are all closely linked at the mRNA and protein level, and their expression is elevated in breast cancer compared with the normal breast tissue. Intriguingly, CDK7 expression was inversely proportional to tumor grade and size, and outcome analysis showed an association between CAK levels and better outcome. Moreover, CDK7 expression was positively associated with ER expression and in particular with phosphorylation of ER at serine 118, a site important for ER transcriptional activity. CONCLUSIONS: Expressions of components of the CAK complex, CDK7, MAT1, and Cyclin H are elevated in breast cancer and correlate with ER. Like ER, CDK7 expression is inversely proportional to poor prognostic factors and survival. Clin Cancer Res; 22(23); 5929-38. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cyclin H/genetics , Cyclin-Dependent Kinases/genetics , Gene Expression/genetics , Receptors, Estrogen/genetics , Adult , Cell Cycle Proteins , Female , Humans , Middle Aged , Phosphorylation/genetics , Prognosis , Signal Transduction/genetics , Transcription Factors , Transcription, Genetic/genetics , Cyclin-Dependent Kinase-Activating Kinase
17.
Oncogene ; 23(45): 7561-70, 2004 Sep 30.
Article in English | MEDLINE | ID: mdl-15334066

ABSTRACT

The androgen receptor (AR) is a member of the nuclear receptor superfamily of ligand-activated transcription factors and plays a key role in the development and progression of prostate cancer. Current therapies include the use of antiandrogens aimed at inhibiting the transcriptional activation of AR-regulated genes by AR. Here, we explore a strategy aimed at obtaining silencing of AR-regulated genes, based on the properties of the transcriptional repressor promyelocytic leukamia zinc-finger protein (PLZF). In order to do this, we have made a fusion protein between PLZF and AR, named PLZF-AR, and show that PLZF-AR is able to bring about silencing of genomically encoded AR-regulated genes and inhibit the androgen-regulated growth of LNCaP prostate cancer cells. Together, our results show that this strategy is able to bring about potent repression of AR-regulated responses and, therefore, could be of value in the development of new therapies for prostate cancer.


Subject(s)
Androgens/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Gene Silencing , Receptors, Androgen/genetics , Recombinant Fusion Proteins/physiology , Transcription Factors/genetics , Animals , Base Sequence , COS Cells , DNA Primers , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Humans , Kruppel-Like Transcription Factors , Promyelocytic Leukemia Zinc Finger Protein , Recombinant Fusion Proteins/genetics
18.
Clin Cancer Res ; 10(23): 8094-104, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585645

ABSTRACT

PURPOSE: Estrogen receptor alpha (ERalpha)-positive breast cancer cell lines are up to 10 times more sensitive than ERalpha-negative cell lines to the antiproliferative activity of the histone deacetylase inhibitor trichostatin A (TSA). The purpose of the study was to investigate the mechanisms underlying this differential response. EXPERIMENTAL DESIGN AND RESULTS: In the ERalpha-positive MCF-7 cell line, TSA repressed ERalpha and cyclin D1 transcription and induced ubiquitin dependent proteasomal degradation of cyclin D1, leading primarily to G(1)-S-phase cell cycle arrest. By contrast, cyclin D1 degradation was enhanced but its transcription unaffected by TSA in the ERalpha-negative MDA-MB-231 cell line, which arrested in G(2)-M phase. Cyclin D1 degradation involved Skp2/p45, a regulatory component of the Skp1/Cullin/F-box complex; silencing SKP2 gene expression by RNA interference stabilized cyclin D1 and abrogated the cyclin D1 down-regulation response to TSA. CONCLUSIONS: Tamoxifen has been shown to inhibit ERalpha-mediated cyclin D1 transcription, and acquired resistance to tamoxifen is associated with a shift to ERalpha-independent cyclin D1 up-regulation. Taken together, our data show that TSA effectively induces cyclin D1 down-regulation through both ERalpha-dependent and ERalpha-independent mechanisms, providing an important new strategy for combating resistance to antiestrogens.


Subject(s)
Breast Neoplasms/metabolism , Cyclin D1/metabolism , Estrogen Receptor alpha/metabolism , Hydroxamic Acids/pharmacology , Transcription, Genetic/drug effects , Uterine Neoplasms/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cysteine Proteinase Inhibitors/pharmacology , Drug Resistance, Neoplasm , Endopeptidases/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors , Humans , Leupeptins/pharmacology , RNA Interference , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , S-Phase Kinase-Associated Proteins/genetics , Tamoxifen/pharmacology , Tumor Cells, Cultured , Uterine Neoplasms/pathology
19.
Cell Rep ; 12(5): 837-49, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26212333

ABSTRACT

LMTK3 is an oncogenic receptor tyrosine kinase (RTK) implicated in various types of cancer, including breast, lung, gastric, and colorectal cancer. It is localized in different cellular compartments, but its nuclear function has not been investigated so far. We mapped LMTK3 binding across the genome using ChIP-seq and found that LMTK3 binding events are correlated with repressive chromatin markers. We further identified KRAB-associated protein 1 (KAP1) as a binding partner of LMTK3. The LMTK3/KAP1 interaction is stabilized by PP1α, which suppresses KAP1 phosphorylation specifically at LMTK3-associated chromatin regions, inducing chromatin condensation and resulting in transcriptional repression of LMTK3-bound tumor suppressor-like genes. Furthermore, LMTK3 functions at distal regions in tethering the chromatin to the nuclear periphery, resulting in H3K9me3 modification and gene silencing. In summary, we propose a model where a scaffolding function of nuclear LMTK3 promotes cancer progression through chromatin remodeling.


Subject(s)
Breast Neoplasms/metabolism , Chromatin Assembly and Disassembly , Membrane Proteins/metabolism , Models, Biological , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Heterografts , Humans , MCF-7 Cells , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
20.
Cell Rep ; 13(1): 108-121, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26411678

ABSTRACT

Estrogen receptor α (ERα) is the key transcriptional driver in a large proportion of breast cancers. We report that APOBEC3B (A3B) is required for regulation of gene expression by ER and acts by causing C-to-U deamination at ER binding regions. We show that these C-to-U changes lead to the generation of DNA strand breaks through activation of base excision repair (BER) and to repair by non-homologous end-joining (NHEJ) pathways. We provide evidence that transient cytidine deamination by A3B aids chromatin modification and remodelling at the regulatory regions of ER target genes that promotes their expression. A3B expression is associated with poor patient survival in ER+ breast cancer, reinforcing the physiological significance of A3B for ER action.


Subject(s)
Breast Neoplasms/genetics , Cytidine Deaminase/genetics , Cytidine/metabolism , DNA End-Joining Repair , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/metabolism , DNA/genetics , DNA/metabolism , DNA Damage , Deamination , Estrogen Receptor alpha/metabolism , Female , Humans , Minor Histocompatibility Antigens , Prognosis , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Transcription, Genetic , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL