Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
Nat Commun ; 13(1): 514, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082299

ABSTRACT

The molecular events and transcriptional plasticity driving brain metastasis in clinically relevant breast tumor subtypes has not been determined. Here we comprehensively dissect genomic, transcriptomic and clinical data in patient-matched longitudinal tumor samples, and unravel distinct transcriptional programs enriched in brain metastasis. We report on subtype specific hub genes and functional processes, central to disease-affected networks in brain metastasis. Importantly, in luminal brain metastases we identify homologous recombination deficiency operative in transcriptomic and genomic data with recurrent breast mutational signatures A, F and K, associated with mismatch repair defects, TP53 mutations and homologous recombination deficiency (HRD) respectively. Utilizing PARP inhibition in patient-derived brain metastatic tumor explants we functionally validate HRD as a key vulnerability. Here, we demonstrate a functionally relevant HRD evident at genomic and transcriptomic levels pointing to genomic instability in breast cancer brain metastasis which is of potential translational significance.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Neoplasm Metastasis , Adult , Breast , Female , Gene Regulatory Networks , Genes, p53/genetics , Humans , Middle Aged , Mutation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Transcriptome
3.
Oncogene ; 40(7): 1318-1331, 2021 02.
Article in English | MEDLINE | ID: mdl-33420368

ABSTRACT

Steroid regulated cancer cells use nuclear receptors and associated regulatory proteins to orchestrate transcriptional networks to drive disease progression. In primary breast cancer, the coactivator AIB1 promotes estrogen receptor (ER) transcriptional activity to enhance cell proliferation. The function of the coactivator in ER+ metastasis however is not established. Here we describe AIB1 as a survival factor, regulator of pro-metastatic transcriptional pathways and a promising actionable target. Genomic alterations and functional expression of AIB1 associated with reduced disease-free survival in patients and enhanced metastatic capacity in novel CDX and PDX ex-vivo models of ER+ metastatic disease. Comparative analysis of the AIB1 interactome with complementary RNAseq characterized AIB1 as a transcriptional repressor. Specifically, we report that AIB1 interacts with MTA2 to form a repressive complex, inhibiting CDH1 (encoding E-cadherin) to promote EMT and drive progression. We further report that pharmacological and genetic inhibition of AIB1 demonstrates significant anti-proliferative activity in patient-derived models establishing AIB1 as a viable strategy to target endocrine resistant metastasis. This work defines a novel role for AIB1 in the regulation of EMT through transcriptional repression in advanced cancer cells with a considerable implication for prognosis and therapeutic interventions.


Subject(s)
Breast Neoplasms/drug therapy , Cadherins/genetics , Histone Deacetylases/genetics , Nuclear Receptor Coactivator 3/genetics , Repressor Proteins/genetics , Antigens, CD/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Metastasis , Nuclear Receptor Coactivator 3/antagonists & inhibitors , Phenotype , Prognosis , Tamoxifen/pharmacology
4.
BMC Med ; 18(1): 349, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33208158

ABSTRACT

BACKGROUND: Metastatic breast cancer is a major cause of cancer-related deaths in woman. Brain metastasis is a common and devastating site of relapse for several breast cancer molecular subtypes, including oestrogen receptor-positive disease, with life expectancy of less than a year. While efforts have been devoted to developing therapeutics for extra-cranial metastasis, drug penetration of blood-brain barrier (BBB) remains a major clinical challenge. Defining molecular alterations in breast cancer brain metastasis enables the identification of novel actionable targets. METHODS: Global transcriptomic analysis of matched primary and metastatic patient tumours (n = 35 patients, 70 tumour samples) identified a putative new actionable target for advanced breast cancer which was further validated in vivo and in breast cancer patient tumour tissue (n = 843 patients). A peptide mimetic of the target's natural ligand was designed in silico and its efficacy assessed in in vitro, ex vivo and in vivo models of breast cancer metastasis. RESULTS: Bioinformatic analysis of over-represented pathways in metastatic breast cancer identified ADAM22 as a top ranked member of the ECM-related druggable genome specific to brain metastases. ADAM22 was validated as an actionable target in in vitro, ex vivo and in patient tumour tissue (n = 843 patients). A peptide mimetic of the ADAM22 ligand LGI1, LGI1MIM, was designed in silico. The efficacy of LGI1MIM and its ability to penetrate the BBB were assessed in vitro, ex vivo and in brain metastasis BBB 3D biometric biohybrid models, respectively. Treatment with LGI1MIM in vivo inhibited disease progression, in particular the development of brain metastasis. CONCLUSION: ADAM22 expression in advanced breast cancer supports development of breast cancer brain metastasis. Targeting ADAM22 with a peptide mimetic LGI1MIM represents a new therapeutic option to treat metastatic brain disease.


Subject(s)
ADAM Proteins/metabolism , Biomimetic Materials/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Peptides/pharmacology , ADAM Proteins/biosynthesis , ADAM Proteins/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Molecular Targeted Therapy , Neoplasm Recurrence, Local/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics
5.
Mol Cancer Ther ; 18(10): 1731-1743, 2019 10.
Article in English | MEDLINE | ID: mdl-31289138

ABSTRACT

Divergent roles for androgen receptor (AR) in breast cancer have been reported. Following aromatase inhibitor (AI) treatment, the conversion of circulating androgens into estrogens can be diminished by >99%. We wished to establish whether the steroid environment can dictate the role of AR and the implications of this for subsequent therapy. This study utilizes models of AI resistance to explore responsiveness to PI3K/mTOR and anti-AR therapy when cells are exposed to unconverted weak androgens. Transcriptomic alterations driven by androstenedione (4AD) were assessed by RNA-sequencing. AR and estrogen receptor (ER) recruitment to target gene promoters was evaluated using ChIP, and relevance to patient profiles was performed using publicly available data sets. Although BEZ235 showed decreased viability across AI-sensitive and -resistant cell lines, anti-AR treatment elicited a decrease in cell viability only in the AI-resistant model. Serum and glucocorticoid-regulated kinase 3 (SGK3) and cAMP-dependent protein kinase inhibitor ß (PKIB) were confirmed to be regulated by 4AD and shown to be mediated by AR; crucially, reexposure to estradiol suppressed expression of these genes. Meta-analysis of transcript levels showed high expression of SGK3 and PKIB to be associated with poor response to endocrine therapy (HR = 2.551, P = 0.003). Furthermore, this study found levels of SGK3 to be sustained in patients who do not respond to AI therapy. This study highlights the importance of the tumor steroid environment. SGK3 and PKIB are associated with poor response to endocrine therapy and could have utility in tailoring therapeutic approaches.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Receptors, Androgen/metabolism , Steroids/metabolism , Adaptation, Physiological/drug effects , Androstenedione/pharmacology , Aromatase Inhibitors/pharmacology , Cell Survival/drug effects , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Postmenopause/drug effects , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects
6.
Oncogene ; 37(15): 2008-2021, 2018 04.
Article in English | MEDLINE | ID: mdl-29367763

ABSTRACT

Steroid receptor coactivator 1 (SRC-1) interacts with nuclear receptors and other transcription factors (TFs) to initiate transcriptional networks and regulate downstream genes which enable the cancer cell to evade therapy and metastasise. Here we took a top-down discovery approach to map out the SRC-1 transcriptional network in endocrine resistant breast cancer. First, rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) was employed to uncover new SRC-1 TF partners. Next, RNA sequencing (RNAseq) was undertaken to investigate SRC-1 TF target genes. Molecular and patient-derived xenograft studies confirmed STAT1 as a new SRC-1 TF partner, important in the regulation of a cadre of four SRC-1 transcription targets, NFIA, SMAD2, E2F7 and ASCL1. Extended network analysis identified a downstream 79 gene network, the clinical relevance of which was investigated in RNAseq studies from matched primary and local-recurrence tumours from endocrine resistant patients. We propose that SRC-1 can partner with STAT1 independently of the estrogen receptor to initiate a transcriptional cascade and control regulation of key endocrine resistant genes.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Regulatory Networks , Nuclear Receptor Coactivator 1/physiology , Animals , Breast Neoplasms/pathology , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/drug effects , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Microarray Analysis , Transcriptional Activation/genetics , Transcriptome/drug effects , Tumor Cells, Cultured
7.
BMC Med ; 15(1): 79, 2017 Apr 12.
Article in English | MEDLINE | ID: mdl-28399921

ABSTRACT

BACKGROUND: Endocrine therapy is standard treatment for estrogen receptor (ER)-positive breast cancer. However, its efficacy is limited by intrinsic and acquired resistance. Here the potential of S100ß as a biomarker and inhibition of its signaling network as a therapeutic strategy in endocrine treated patients was investigated. METHODS: The expression of S100ß in tissue and serum was assessed by immunohistochemistry and an enzyme-linked immunosorbent assay, respectively. The S100ß signaling network was investigated in cell line models of endocrine resistance by western blot, PCR, immunoprecipitation, and chromatin-immunoprecipitation. Endocrine resistant xenografts and tumor explants from patients with resistant tumors were treated with endocrine therapy in the presence and absence of the p-Src kinase inhibitor, dasatinib. RESULTS: Tissue and serum levels of S100ß were found to predict poor disease-free survival in endocrine-treated patients (n = 509, HR 2.32, 95% CI is 1.58-3.40, p < 0.0001 and n = 187, HR 4.009, 95% CI is 1.66-9.68, p = 0.002, respectively). Moreover, elevated levels of serum S100ß detected during routine surveillance over the patient treatment period significantly associated with subsequent clinically confirmed disease recurrence (p = 0.019). In vivo studies demonstrated that endocrine treatment induced transcriptional regulation of S100ß which was successfully disrupted with tyrosine kinase inhibition. In endocrine resistant xenografts and tumor explants from patients with endocrine resistant breast cancer, combined endocrine and dasatinib treatment reduced tumor proliferation and down-regulated S100ß protein expression in comparison to endocrine treatment alone. CONCLUSIONS: S100ß has potential as a new surveillance tool for patients with ER-positive breast cancer to monitor ongoing response to endocrine therapy. Moreover, endocrine resistant breast cancer patients with elevated S100ß may benefit from combined endocrine and tyrosine-kinase inhibitor treatment. TRIAL REGISTRATION: ClinicalTrials.gov,  NCT01840293 ). Registered on 23 April 2013. Retrospectively registered.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers/blood , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , S100 Calcium Binding Protein beta Subunit/blood , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Drug Resistance, Neoplasm/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , MCF-7 Cells , Mice , Middle Aged , Neoplasm Recurrence, Local , S100 Calcium Binding Protein beta Subunit/genetics , Signal Transduction/drug effects , Tamoxifen/therapeutic use , Tissue Array Analysis , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 21(23): 5371-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26240272

ABSTRACT

PURPOSE: Disease recurrence is a common problem in breast cancer and yet the mechanisms enabling tumor cells to evade therapy and colonize distant organs remain unclear. We sought to characterize global expression changes occurring with metastatic disease progression in the endocrine-resistant setting. EXPERIMENTAL DESIGN: Here, for the first time, RNAsequencing has been performed on matched primary, nodal, and liver metastatic tumors from tamoxifen-treated patients following disease progression. Expression of genes commonly elevated in the metastases of sequenced patients was subsequently examined in an extended matched patient cohort with metastatic disease from multiple sites. The impact of tamoxifen treatment on endocrine-resistant tumors in vivo was investigated in a xenograft model. RESULTS: The extent of patient heterogeneity at the gene level was striking. Less than 3% of the genes differentially expressed between sequential tumors were common to all patients. Larger divergence was observed between primary and liver tumors than between primary and nodal tumors, reflecting both the latency to disease progression and the genetic impact of intervening therapy. Furthermore, an endocrine-resistant in vivo mouse model demonstrated that tamoxifen treatment has the potential to drive disease progression and establish distant metastatic disease. Common functional pathways altered during metastatic, endocrine-resistant progression included extracellular matrix receptor interactions and focal adhesions. CONCLUSIONS: This novel global analysis highlights the influence of primary tumor biology in determining the transcriptomic profile of metastatic tumors, as well as the need for adaptations in cell-cell communications to facilitate successful tumor cell colonization of distant host organs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Transcriptome , Adult , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers , Breast Neoplasms/drug therapy , Cell Communication , Cell Line, Tumor , Cluster Analysis , Combined Modality Therapy , Computational Biology/methods , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Immunohistochemistry , Liver Neoplasms/secondary , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Cancer Res ; 74(13): 3477-88, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24769444

ABSTRACT

In breast cancer, overexpression of the nuclear coactivator NCOA1 (SRC-1) is associated with disease recurrence and resistance to endocrine therapy. To examine the impact of NCOA1 overexpression on morphogenesis and carcinogenesis in the mammary gland (MG), we generated MMTV-hNCOA1 transgenic [Tg(NCOA1)] mice. In the context of two distinct transgenic models of breast cancer, NCOA1 overexpression did not affect the morphology or tumor-forming capability of MG epithelial cells. However, NCOA1 overexpression increased the number of circulating breast cancer cells and the efficiency of lung metastasis. Mechanistic investigations showed that NCOA1 and c-Fos were recruited to a functional AP-1 site in the macrophage attractant CSF1 promoter, directly upregulating colony-simulating factor 1 (CSF1) expression to enhance macrophage recruitment and metastasis. Conversely, silencing NCOA1 reduced CSF1 expression and decreased macrophage recruitment and breast cancer cell metastasis. In a cohort of 453 human breast tumors, NCOA1 and CSF1 levels correlated positively with disease recurrence, higher tumor grade, and poor prognosis. Together, our results define an NCOA1/AP-1/CSF1 regulatory axis that promotes breast cancer metastasis, offering a novel therapeutic target for impeding this process.


Subject(s)
Breast Neoplasms/pathology , Macrophage Colony-Stimulating Factor/genetics , Nuclear Receptor Coactivator 1/genetics , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Lymphatic Metastasis , MCF-7 Cells , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/immunology , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Neoplasm Recurrence, Local/genetics , Neoplastic Cells, Circulating/pathology , Nuclear Receptor Coactivator 1/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , RNA Interference , RNA, Small Interfering , Transcription Factor AP-1/genetics
10.
Clin Cancer Res ; 18(12): 3305-15, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22550166

ABSTRACT

PURPOSE: The use of aromatase inhibitors (AI) in the treatment of estrogen receptor (ER)-positive, postmenopausal breast cancer has proven efficacy. However, inappropriate activation of ER target genes has been implicated in the development of resistant tumors. The ER coactivator protein AIB1 has previously been associated with initiation of breast cancer and resistance to endocrine therapy. EXPERIMENTAL DESIGN: Here, we investigated the role of AIB1 in the deregulation of ER target genes occurring as a consequence of AI resistance using tissue microarrays of patients with breast cancer and cell line models of resistance to the AI letrozole. RESULTS: Expression of AIB1 associated with disease recurrence (P = 0.025) and reduced disease-free survival time (P = 0.0471) in patients treated with an AI as first-line therapy. In a cell line model of resistance to letrozole (LetR), we found ERα/AIB1 promoter recruitment and subsequent expression of the classic ER target genes pS2 and Myc to be constitutively upregulated in the presence of both androstenedione and letrozole. In contrast, the recruitment of the ERα/AIB1 transcriptional complex to the nonclassic ER target cyclin D1 and its subsequent expression remained sensitive to steroid treatment and could be inhibited by treatment with letrozole. Molecular studies revealed that this may be due in part to direct steroid regulation of c-jun-NH(2)-kinase (JNK), signaling to Jun and Fos at the cyclin D1 promoter. CONCLUSION: This study establishes a role for AIB1 in AI-resistant breast cancer and describes a new mechanism of ERα/AIB1 gene regulation which could contribute to the development of an aggressive tumor phenotype.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Androstenedione/pharmacology , Aromatase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Disease-Free Survival , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Letrozole , MAP Kinase Signaling System , Neoplasm Recurrence, Local , Nitriles/pharmacology , Nuclear Receptor Coactivator 3/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Trefoil Factor-1 , Triazoles/pharmacology , Tumor Suppressor Proteins/biosynthesis , Up-Regulation
11.
Endocr Relat Cancer ; 17(1): 255-64, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20032008

ABSTRACT

Epidemiological, clinical, and molecular studies suggest a role for oestrogen in thyroid cancer. How oestrogen mediates its effects and the consequence of it on clinical outcome has not been fully elucidated. The participation of coregulatory proteins in modulating oestrogen receptor (ER) function and input of crosstalk with the tyrosine kinase receptor HER2 was investigated. Oestrogen induced cell proliferation in the follicular thyroid cancer (FTC)-133 cells, but not in the anaplastic 8305C cell line. Knockdown of the coactivator steroid receptor coactivator (SRC)-1 inhibited FTC-133 basal, but not oestrogen induced, cell proliferation. Oestrogen also increased protein expression of SRC-1 and the ER target gene cyclin D1 in the FTC-133 cell line. ERalpha, ERbeta, the coregulatory proteins SRC-1 and nuclear corepressor (NCoR), and the tyrosine kinase receptor HER2 were localised by immunohistochemistry and immnofluorescence in paraffin-embedded tissue from thyroid tumour patients (n=111). ERalpha was colocalised with both SRC-1 and NCoR to the nuclei of the tumour epithelial cells. Expression of ERalpha and NCoR was found predominantly in non-anaplastic tumours and was significantly associated with well-differentiated tumours and reduced incidence of disease recurrence. In non-anaplastic tumours, HER2 was significantly associated with SRC-1, and these proteins were associated with poorly differentiated tumours, capsular invasion and disease recurrence. Totally, 87% of anaplastic tumours were positive for SRC-1. Kaplan-Meier estimates of disease-free survival indicated that in thyroid cancer, SRC-1 strongly correlates with reduced disease-free survival (P<0.001), whereas NCoR predicted increased survival (P<0.001). These data suggest opposing roles for the coregulators SRC-1 and NCoR in thyroid tumour progression.


Subject(s)
Adenocarcinoma, Follicular/etiology , Estrogen Receptor alpha/physiology , Receptor, ErbB-2/physiology , Thyroid Neoplasms/etiology , Transcription Factors/physiology , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma/etiology , Carcinoma/metabolism , Carcinoma/mortality , Case-Control Studies , Child , Child, Preschool , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/physiology , Estrogen Receptor alpha/metabolism , Female , Humans , Male , Middle Aged , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptor, ErbB-2/metabolism , Survival Analysis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/mortality , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factors/metabolism , Tumor Cells, Cultured , Young Adult
12.
Clin Cancer Res ; 15(6): 2098-106, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276281

ABSTRACT

PURPOSE: This study investigates the role of the p160 coactivators AIB1 and SRC-1 independently, and their interactions with the estrogen receptor, in the development of resistance to endocrine treatments. EXPERIMENTAL DESIGN: The expression of the p160s and the estrogen receptor, and their interactions, was analyzed by immunohistochemistry and quantitative coassociation immunofluorescent microscopy, using cell lines, primary breast tumor cell cultures, and a tissue microarray with breast cancer samples from 560 patients. RESULTS: Coassociation of the p160s and estrogen receptor alpha was increased in the LY2 endocrine-resistant cell line following treatment with tamoxifen in comparison with endocrine-sensitive MCF-7 cells. In primary cultures, there was an increase in association of the coactivators with estrogen receptor alpha following estrogen treatment but dissociation was evident with tamoxifen. Immunohistochemical staining of the tissue microarray revealed that SRC-1 was a strong predictor of reduced disease-free survival (DFS), both in patients receiving adjuvant tamoxifen treatment and untreated patients (P < 0.0001 and P = 0.0111, respectively). SRC-1 was assigned a hazard ratio of 2.12 using a Cox proportional hazards model. Endocrine-treated patients who coexpressed AIB1 with human epidermal growth factor receptor 2 had a significantly shorter DFS compared with all other patients (P = 0.03). Quantitative coassociation analysis in the patient tissue microarray revealed significantly stronger colocalization of AIB1 and SRC-1 with estrogen receptor alpha in patients who have relapsed in comparison with those patients who did not recur (P = 0.026 and P = 0.00001, respectively). CONCLUSIONS: SRC-1 is a strong independent predictor of reduced DFS, whereas the interactions of the p160 proteins with estrogen receptor alpha can predict the response of patients to endocrine treatment.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor alpha/physiology , Histone Acetyltransferases/physiology , Neoplasm Recurrence, Local/etiology , Nuclear Proteins/physiology , Nucleocytoplasmic Transport Proteins/physiology , Tamoxifen/therapeutic use , Transcription Factors/physiology , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Cell Line, Tumor , DNA-Binding Proteins , Disease-Free Survival , Drug Resistance, Neoplasm , Estrogen Receptor alpha/analysis , Female , Histone Acetyltransferases/analysis , Humans , Nuclear Proteins/analysis , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 3 , Nucleocytoplasmic Transport Proteins/analysis , Prognosis , RNA-Binding Proteins , Tissue Array Analysis , Trans-Activators/analysis , Trans-Activators/physiology , Transcription Factors/analysis
13.
J Pharmacol Exp Ther ; 329(1): 38-47, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19131583

ABSTRACT

Microtubule assembly and disassembly is required for the maintenance of cell structure, mobility, and division. However, the cellular and biochemical implications of microtubule disruption are not fully understood. Using a proteomic approach, we found that the peptidyl-prolyl isomerase, cyclophilin A, was increased in plasma membrane extracts from chronic myeloid leukemia cells after microtubule disruption. In addition, we found that two peptidyl-prolyl isomerases, cyclophilin A and pin1, are overexpressed up to 10-fold in hematological malignancies compared with normal peripheral blood mononuclear cells. Although previous reports suggest that cyclophilin A is localized to the cytosol of mammalian cells, we found that cyclophilin A and pin1 are both localized to the nucleus and nuclear domains in hematopoietic cells. Microtubule disruption of hematopoietic cells caused a dramatic subcellular redistribution of cyclophilin A and pin1 from the nucleus to the cytosol and plasma membrane. We suggest that this accounts for the increased cyclophilin A at the plasma membrane of chronic myeloid leukemia cells after microtubule disruption. The subcellular redistribution of cyclophilin A and pin1 occurred in a c-Jun NH(2)-terminal kinase- and serine protease-dependent manner. Moreover, the altered subcellular localization of the peptidyl-prolyl isomerases occurred in a dose- and time-dependent manner after microtubule disruption and was found to correlate with G(2)/M arrest and precede induced cell death. These results suggest that the function of peptidyl-prolyl isomerases may be influenced by microtubule dynamics throughout the cell cycle, and their altered localization may be an important part of the mechanism by which microtubule-disrupting agents exert their cytostatic effects.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Nucleus/metabolism , Cyclophilin A/metabolism , Cytoplasm/metabolism , Hematologic Neoplasms/metabolism , Microtubules/drug effects , Oxazepines/pharmacology , Paclitaxel/pharmacology , Peptidylprolyl Isomerase/metabolism , Pyrroles/pharmacology , Blotting, Western , Cell Division/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cytoplasm/drug effects , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , G2 Phase/drug effects , Hematologic Neoplasms/pathology , Humans , Hydrolysis , Monocytes/drug effects , Monocytes/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Peptide Mapping , Silver Staining , Trypsin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...