15 results on '"Oesterreich S"'
Search Results
2. Liver tropism of ER mutant breast cancer is characterized by unique molecular changes and immune infiltration.
- Author
-
Wu Y, Li Z, Lee AV, Oesterreich S, and Luo B
- Subjects
- Humans, Female, Gene Expression Regulation, Neoplastic, DNA Copy Number Variations, Gene Expression Profiling, Biomarkers, Tumor genetics, Liver pathology, Liver immunology, Liver metabolism, Transcriptome, Breast Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms immunology, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Mutation, Liver Neoplasms secondary, Liver Neoplasms genetics, Liver Neoplasms immunology, Liver Neoplasms pathology
- Abstract
Purpose: Hotspot estrogen receptor alpha (ER/ESR1) mutations are recognized as the driver for both endocrine resistance and metastasis in advanced ER-positive (ER+) breast cancer, but their contributions to metastatic organ tropism remain insufficiently understood. In this study, we aim to comprehensively profile the organotropic metastatic pattern for ESR1 mutant breast cancer., Methods: The organ-specific metastatic pattern of ESR1 mutant breast cancer was delineated using multi-omics data from multiple publicly available cohorts of ER+ metastatic breast cancer patients. Gene mutation/copy number variation (CNV) and differential gene expression analyses were performed to identify the genomic and transcriptomic alterations uniquely associated with ESR1 mutant liver metastasis. Upstream regulator, downstream pathway, and immune infiltration analysis were conducted for subsequent mechanistic investigations., Results: ESR1 mutation-driven liver tropism was revealed by significant differences, encompassing a higher prevalence of liver metastasis in patients with ESR1 mutant breast cancer and an enrichment of mutations in liver metastatic samples. The significant enrichment of AGO2 copy number amplifications (CNAs) and multiple gene expression changes were revealed uniquely in ESR1 mutant liver metastasis. We also unveiled alterations in downstream signaling pathways and immune infiltration, particularly an enrichment of neutrophils, suggesting potential therapeutic vulnerabilities., Conclusion: Our data provide a comprehensive characterization of the behaviors and mechanisms of ESR1 mutant liver metastasis, paving the way for the development of personalized therapy to target liver metastasis for patients with ESR1 mutant breast cancer., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
3. SNAIL is induced by tamoxifen and leads to growth inhibition in invasive lobular breast carcinoma.
- Author
-
Bossart EA, Tasdemir N, Sikora MJ, Bahreini A, Levine KM, Chen J, Basudan A, Jacobsen BM, Burns TF, and Oesterreich S
- Subjects
- Apoptosis drug effects, Breast Neoplasms genetics, Breast Neoplasms pathology, Carcinoma, Lobular genetics, Carcinoma, Lobular pathology, Cell Cycle Checkpoints drug effects, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, Epithelial-Mesenchymal Transition genetics, Estradiol pharmacology, Estrogen Receptor alpha genetics, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Neoplasm Invasiveness pathology, Signal Transduction drug effects, Tamoxifen analogs & derivatives, Tamoxifen pharmacology, Breast Neoplasms drug therapy, Carcinoma, Lobular drug therapy, Neoplasm Invasiveness genetics, Snail Family Transcription Factors genetics
- Abstract
Purpose: Invasive lobular carcinoma (ILC) is a histological subtype of breast cancer that is predominantly estrogen receptor alpha (ER)-positive (+) and is thus treated with endocrine therapies. Herein, we sought to understand the molecular underpinnings of the 4-hydroxytamoxifen (4OHT) resistance in ILC by assessing the potential role of the epithelial-to-mesenchymal transition transcription factor (EMT-TF) SNAIL (SNAI1)., Methods: Using a series of breast cancer cell lines, we measured the basal, estrogen and 4OHT-induced expression of SNAIL and other EMT-TF family members by quantitative reverse transcription-polymerase chain reaction and immunoblotting. Chromatin immunoprecipitation experiments were performed to assess ER binding to the SNAIL promoter. Cell proliferation, cell cycle and apoptosis were assessed in 2D cultures. 3D growth was assessed in Matrigel and Collagen I cultures., Results: Estrogen and 4OHT induced SNAIL expression, but not that of the other EMT-TF family members SLUG (SNAI2) and SMUC (SNAI3), with the 4OHT effect being specific to the lobular but not the ductal subtype. We observed estrogen and 4OHT-induced ER recruitment to the SNAI1 promoter and high endogenous basal levels of SNAIL and several EMT-TFs in ILC cell lines. While SNAIL knockdown had a minor impact on the 4OHT partial agonism in estrogen-depleted conditions, it led to a surprising increase in cell proliferation in full serum. In complementary experiments, inducible SNAI1 overexpression caused decreased proliferation, associated with a cell cycle arrest in G
0 /G1 . Additionally, apoptosis was observed in BCK4 cells., Conclusion: These data suggest a previously unrecognized role for SNAIL in ILC, substantiating a context-dependent behavior for this EMT-TF.- Published
- 2019
- Full Text
- View/download PDF
4. Associations between genetic variants and the effect of letrozole and exemestane on bone mass and bone turnover.
- Author
-
Oesterreich S, Henry NL, Kidwell KM, Van Poznak CH, Skaar TC, Dantzer J, Li L, Hangartner TN, Peacock M, Nguyen AT, Rae JM, Desta Z, Philips S, Storniolo AM, Stearns V, Hayes DF, and Flockhart DA
- Subjects
- Adult, Aged, Aged, 80 and over, Alleles, Androstadienes therapeutic use, Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Hormonal therapeutic use, Aromatase Inhibitors pharmacology, Aromatase Inhibitors therapeutic use, Biomarkers, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Female, Genetic Association Studies, Genotype, Humans, Letrozole, Middle Aged, Nitriles therapeutic use, Polymorphism, Single Nucleotide, Postmenopause, Triazoles therapeutic use, Androstadienes pharmacology, Bone Density drug effects, Bone Density genetics, Bone Remodeling drug effects, Bone Remodeling genetics, Bone and Bones drug effects, Bone and Bones metabolism, Genetic Variation, Nitriles pharmacology, Triazoles pharmacology
- Abstract
Adjuvant therapy for hormone receptor (HR) positive postmenopausal breast cancer patients includes aromatase inhibitors (AI). While both the non-steroidal AI letrozole and the steroidal AI exemestane decrease serum estrogen concentrations, there is evidence that exemestane may be less detrimental to bone. We hypothesized that single nucleotide polymorphisms (SNP) predict effects of AIs on bone turnover. Early stage HR-positive breast cancer patients were enrolled in a randomized trial of exemestane versus letrozole. Effects of AI on bone mineral density (BMD) and bone turnover markers (BTM), and associations between SNPs in 24 candidate genes and changes in BMD or BTM were determined. Of the 503 enrolled patients, paired BMD data were available for 123 and 101 patients treated with letrozole and exemestane, respectively, and paired BTM data were available for 175 and 173 patients, respectively. The mean change in lumbar spine BMD was significantly greater for letrozole-treated (-3.2 %) compared to exemestane-treated patients (-1.0 %) (p = 0.0016). Urine N-telopeptide was significantly increased in patients treated with exemestane (p = 0.001) but not letrozole. Two SNPs (rs4870061 and rs9322335) in ESR1 and one SNP (rs10140457) in ESR2 were associated with decreased BMD in letrozole-treated patients. In the exemestane-treated patients, SNPs in ESR1 (Rs2813543) and CYP19A1 (Rs6493497) were associated with decreased bone density. Exemestane had a less negative impact on bone density compared to letrozole, and the effects of AI therapy on bone may be impacted by genetic variants in the ER pathway.
- Published
- 2015
- Full Text
- View/download PDF
5. Developing in vitro models of human ductal carcinoma in situ from primary tissue explants.
- Author
-
Brown DD, Dabbs DJ, Lee AV, McGuire KP, Ahrendt GM, Bhargava R, Davidson NE, Brufsky AM, Johnson RR, Oesterreich S, and McAuliffe PF
- Subjects
- Aged, Animals, Biomarkers, Tumor, Biopsy, Needle, Breast Neoplasms metabolism, Breast Neoplasms surgery, Carcinoma, Intraductal, Noninfiltrating metabolism, Carcinoma, Intraductal, Noninfiltrating surgery, Female, Humans, Mastectomy, Middle Aged, Neoplasm Grading, Neoplasm Staging, Primary Cell Culture, Sentinel Lymph Node Biopsy, Breast Neoplasms pathology, Carcinoma, Intraductal, Noninfiltrating pathology, In Vitro Techniques, Tissue Culture Techniques
- Abstract
Because there are currently no reliable predictors for progression of ductal carcinoma in situ (DCIS) to invasive disease, nearly all patients receive comprehensive therapy, leading to over-treatment in many cases. Few in vitro models for studying DCIS progression have been developed. We report here the successful culture and expansion of primary DCIS from surgical specimens using a conditional reprogramming protocol. Patients with percutaneous core-needle biopsy demonstrating DCIS were enrolled in a tissue banking protocol after informed consent was received. Fresh tissue was taken from lumpectomy or mastectomy specimens, mechanically and enzymatically dissociated, cultured in medium conditioned by irradiated mouse fibroblasts and supplemented with rho-associated protein kinase (ROCK) inhibitor, and characterized by immunocytochemistry. Out of 33 DCIS cases, 58% (19) were expanded for up to 2 months in culture, and 42% (14) were frozen immediately after mechanical dissociation for future growth. The cultures are almost exclusively composed of cytokeratin 8- and EpCAM-positive luminal and cytokeratin 14-, cytokeratin 5-, and p63-positive basal mammary epithelial cells, suggesting maintenance of heterogeneity in vitro. Furthermore, as assessed by luminal and basal marker expression, these cells retain their cellular identities both in the "conditionally reprogrammed" proliferative state and after conditioned media and ROCK inhibitor withdrawal. When grown to 100 % confluency, the cultures organize into luminal and basal layers as well as luminal compartments surrounded by basal cells. Primary cultures of DCIS derived directly from patient tissues can be generated and may serve as in vitro models for the study of DCIS.
- Published
- 2015
- Full Text
- View/download PDF
6. Inhibition of histone demethylase, LSD2 (KDM1B), attenuates DNA methylation and increases sensitivity to DNMT inhibitor-induced apoptosis in breast cancer cells.
- Author
-
Katz TA, Vasilatos SN, Harrington E, Oesterreich S, Davidson NE, and Huang Y
- Subjects
- Antineoplastic Agents pharmacology, Cell Line, Tumor, Female, Gene Silencing, Histones metabolism, Humans, Inhibitory Concentration 50, Methylation, RNA, Small Interfering genetics, Transcriptional Activation drug effects, Tumor Stem Cell Assay, Apoptosis drug effects, Apoptosis genetics, Breast Neoplasms genetics, Breast Neoplasms metabolism, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, DNA Methylation, Drug Resistance, Neoplasm, Histone Demethylases genetics
- Abstract
Increasing evidence suggests that dysfunction of histone lysine demethylase is associated with abnormal chromatin remodeling and gene silencing, contributing to breast tumorigenesis. In silico analysis shows that the newly identified histone demethylase lysine-specific demethylase 2 is highly expressed in breast cancer, especially in invasive tumors. However, it is currently unknown how LSD2 regulates chromatin remodeling and gene expression regulation in breast cancer. Using short hairpin RNA, we stably knocked down LSD2 (LSD2-KD) in MDA-MB-231 breast cancer cells. LSD2-KD led to accumulation of H3K4me1/2 without changing methylation levels of other key histone lysine residues, suggesting that LSD2 acts as a bona fide H3K4 demethylase in breast cancer cells. LSD2-KD resulted in decreased colony formation and attenuated global DNA methylation in MDA-MB-231 cells. Additionally, treatment with the DNMT inhibitor, 5-aza-deoxycytidine (DAC), synergistically increased mRNA expression of aberrantly silenced genes important in breast cancer development, including PR, RARβ, ERα, SFRP1, SFRP2, and E-cadherin in LSD2-KD cells. Furthermore, LSD2-KD cells are more susceptible to cell death than scramble controls, and combined treatment with tranylcypromine, an LSD2 inhibitor, and DAC resulted in synergistic growth inhibition of breast cancer cells. DNMT inhibition by DAC in LSD2-KD cells led to internucleosomal DNA fragmentation, enhanced PARP cleavage and increased sub-G1 apoptotic cell population. These results demonstrate an important role for LSD2 in regulation of DNA methylation and gene silencing in breast cancer, and suggest that inhibition of LSD2 in combination with DNA methyltransferase inhibition represents a novel approach for epigenetic therapy of breast cancer.
- Published
- 2014
- Full Text
- View/download PDF
7. Genetic associations with toxicity-related discontinuation of aromatase inhibitor therapy for breast cancer.
- Author
-
Henry NL, Skaar TC, Dantzer J, Li L, Kidwell K, Gersch C, Nguyen AT, Rae JM, Desta Z, Oesterreich S, Philips S, Carpenter JS, Storniolo AM, Stearns V, Hayes DF, and Flockhart DA
- Subjects
- Adult, Aged, Aged, 80 and over, Androstadienes adverse effects, Breast Neoplasms metabolism, Female, Humans, Introns, Letrozole, Middle Aged, Models, Genetic, Multivariate Analysis, Nitriles adverse effects, Polymorphism, Single Nucleotide, Prospective Studies, Treatment Failure, Triazoles adverse effects, Aromatase Inhibitors adverse effects, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Estrogen Receptor alpha genetics
- Abstract
Up to 25 % of patients discontinue adjuvant aromatase inhibitor (AI) therapy due to intolerable symptoms. Predictors of which patients will be unable to tolerate these medications have not been defined. We hypothesized that inherited variants in candidate genes are associated with treatment discontinuation because of AI-associated toxicity. We prospectively evaluated reasons for treatment discontinuation in women with hormone receptor-positive breast cancer initiating adjuvant AI through a multicenter, prospective, randomized clinical trial of exemestane versus letrozole. Using multiple genetic models, we evaluated potential associations between discontinuation of AI therapy because of toxicity and 138 variants in 24 candidate genes, selected a priori, primarily with roles in estrogen metabolism and signaling. To account for multiple comparisons, statistical significance was defined as p < 0.00036. Of the 467 enrolled patients with available germline DNA, 152 (33 %) discontinued AI therapy because of toxicity. Using a recessive statistical model, an intronic variant in ESR1 (rs9322336) was associated with increased risk of musculoskeletal toxicity-related exemestane discontinuation [HR 5.0 (95 % CI 2.1-11.8), p < 0.0002]. An inherited variant potentially affecting estrogen signaling may be associated with exemestane-associated toxicity, which could partially account for intra-patient differences in AI tolerability. Validation of this finding is required.
- Published
- 2013
- Full Text
- View/download PDF
8. Elevated nuclear expression of the SMRT corepressor in breast cancer is associated with earlier tumor recurrence.
- Author
-
Smith CL, Migliaccio I, Chaubal V, Wu MF, Pace MC, Hartmaier R, Jiang S, Edwards DP, Gutiérrez MC, Hilsenbeck SG, and Oesterreich S
- Subjects
- Cell Line, Tumor, Cell Nucleolus genetics, Cell Nucleolus metabolism, Estrogen Receptor alpha metabolism, Female, Humans, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local metabolism, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Nuclear Receptor Co-Repressor 2 genetics, Survival Analysis, Tamoxifen administration & dosage, Tissue Array Analysis, Breast Neoplasms drug therapy, Breast Neoplasms epidemiology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Gene Expression Regulation, Neoplastic, Nuclear Receptor Co-Repressor 2 metabolism
- Abstract
Silencing mediator of retinoic acid and thyroid hormone receptor (SMRT), also known as nuclear corepressor 2 (NCOR2) is a transcriptional corepressor for multiple members of the nuclear receptor superfamily of transcription factors, including estrogen receptor-α (ERα). In the classical model of corepressor action, SMRT binds to antiestrogen-bound ERα at target promoters and represses ERα transcriptional activity and gene expression. Herein SMRT mRNA and protein expression was examined in a panel of 30 breast cancer cell lines. Expression of both parameters was found to vary considerably amongst lines and the correlation between protein and mRNA expression was very poor (R (2) = 0.0775). Therefore, SMRT protein levels were examined by immunohistochemical staining of a tissue microarray of 866 patients with stage I-II breast cancer. Nuclear and cytoplasmic SMRT were scored separately according to the Allred score. The majority of tumors (67 %) were negative for cytoplasmic SMRT, which when detected was found at very low levels. In contrast, nuclear SMRT was broadly detected. There was no significant difference in time to recurrence (TTR) according to SMRT expression levels in the ERα-positive tamoxifen-treated patients (P = 0.297) but the difference was significant in the untreated patients (P = 0.01). In multivariate analysis, ERα-positive tamoxifen-untreated patients with high nuclear SMRT expression (SMRT 5-8, i.e., 2nd to 4th quartile) had a shorter TTR (HR = 1.94, 95 % CI, 1.24-3.04; P = 0.004) while there was no association with SMRT expression for ERα-positive tamoxifen-treated patients. There was no association between SMRT expression and overall survival for patients, regardless of whether they received tamoxifen. Thus while SMRT protein expression was not predictive of outcome after antiestrogen therapy, it may have value in predicting tumor recurrence in patients not receiving adjuvant tamoxifen therapy.
- Published
- 2012
- Full Text
- View/download PDF
9. BRCA1 promoter methylation status does not predict response to tamoxifen in sporadic breast cancer patients.
- Author
-
Cerne JZ, Zong L, Jelinek J, Hilsenbeck SG, Wang T, Oesterreich S, and McGuire SE
- Subjects
- BRCA1 Protein genetics, Breast Neoplasms genetics, Breast Neoplasms mortality, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Female, Humans, Middle Aged, Retrospective Studies, Treatment Outcome, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms drug therapy, DNA Methylation, Genes, BRCA1, Promoter Regions, Genetic, Tamoxifen therapeutic use
- Abstract
The purpose of this study is to investigate whether BRCA1 promoter methylation is associated with poorer outcome in sporadic breast cancer cases treated with tamoxifen. BRCA1 promoter methylation was determined by bisulfite pyrosequencing in two groups of sporadic breast cancer patients, systemically untreated (N = 497) and treated with adjuvant tamoxifen (N = 497). Associations of BRCA1 promoter methylation with clinopathological characteristics and the effect of BRCA1 promoter methylation on time to first recurrence (TTR) and overall survival (OS) were examined. No significant differences were observed between BRCA1 promoter methylation and clinopathological characteristics in untreated and tamoxifen-treated groups. Cut point analysis did not find any promising cut point for BRCA1 promoter methylation that would differentially influence TTR and OS in untreated and tamoxifen-treated group. Using the median (2.53 %) and an arbitrary value of 10 % as a cut point for methylation, we still found no significant effect of BRCA1 promoter methylation on TTR and OS in untreated and tamoxifen-treated group. Despite data suggesting that BRCA1 levels impact estrogen receptor response to tamoxifen, our results indicate that BRCA1 promoter methylation is not associated with poorer outcome in sporadic breast cancer cases treated with tamoxifen.
- Published
- 2012
- Full Text
- View/download PDF
10. Estrogen and insulin-like growth factor-I (IGF-I) independently down-regulate critical repressors of breast cancer growth.
- Author
-
Casa AJ, Potter AS, Malik S, Lazard Z, Kuiatse I, Kim HT, Tsimelzon A, Creighton CJ, Hilsenbeck SG, Brown PH, Oesterreich S, and Lee AV
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Benzimidazoles pharmacology, Biomarkers, Tumor genetics, Breast Neoplasms pathology, Cell Line, Tumor, Disease-Free Survival, Down-Regulation, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogen Receptor alpha antagonists & inhibitors, Estrogen Receptor alpha metabolism, Female, Fulvestrant, Gene Expression Profiling, Genes, Tumor Suppressor, Humans, Insulin-Like Growth Factor I pharmacology, Kaplan-Meier Estimate, Oligonucleotide Array Sequence Analysis, Prognosis, Pyridones pharmacology, Receptor, IGF Type 1 antagonists & inhibitors, Receptor, IGF Type 1 metabolism, Breast Neoplasms metabolism, Cell Proliferation, Estradiol physiology, Gene Expression Regulation, Neoplastic, Insulin-Like Growth Factor I physiology
- Abstract
Although estrogen receptor alpha (ERα) and insulin-like growth factor (IGF) signaling are important for normal mammary development and breast cancer, cross-talk between these pathways, particularly at the level of transcription, remains poorly understood. We performed microarray analysis on MCF-7 breast cancer cells treated with estradiol (E2) or IGF-I for 3 or 24 h. IGF-I regulated mRNA of five to tenfold more genes than E2, and many genes were co-regulated by both ligands. Importantly, expression of these co-regulated genes correlated with poor prognosis of human breast cancer. Closer examination revealed enrichment of repressed transcripts. Interestingly, a number of potential tumor suppressors, for example, B-cell linker (BLNK), were down-regulated by IGF-I and E2. Analysis of three down-regulated genes showed that E2-mediated repression occurred independently of IGF-IR, and IGF-I-mediated repression occurred independently of ERα. However, repression by IGF-I or E2 required common kinases, such as PI3K and MEK, suggesting downstream convergence of the two pathways. In conclusion, E2 and IGF-I co-regulate a set of genes that affect breast cancer outcome. There is enrichment of repressed transcripts, and, for some genes, the down-regulation is independent at the receptor level. This may be important clinically, as tumors with active ERα and IGF-IR signaling may require co-targeting of both pathways.
- Published
- 2012
- Full Text
- View/download PDF
11. The p160 ER co-regulators predict outcome in ER negative breast cancer.
- Author
-
Spears M, Oesterreich S, Migliaccio I, Guiterrez C, Hilsenbeck S, Quintayo MA, Pedraza J, Munro AF, Thomas JS, Kerr GR, Jack WJ, Kunkler IH, Cameron DA, Chetty U, and Bartlett JM
- Subjects
- Adult, Aged, Breast Neoplasms metabolism, Breast Neoplasms pathology, Female, Humans, Middle Aged, Nuclear Receptor Coactivator 3 metabolism, Prognosis, Receptor, ErbB-2 metabolism, Survival Analysis, Tissue Array Analysis, Breast Neoplasms mortality, Nuclear Receptor Coactivators metabolism, Receptors, Estrogen analysis
- Abstract
The SRC family of ER co-regulators are frequently overexpressed in breast cancer. Overexpression of AIB1 appears to be linked to hormone resistance in HER2 positive breast cancer. However, the role of these co-regulators in ER negative disease is poorly understood. SRC1, SRC2 and AIB1 expression was determined by immunohistochemical analysis of tissue microarrays constructed from tumours within the Edinburgh Breast Conservation Series (BCS). The BCS represents a fully documented consecutive cohort of 1,812 patients treated by breast conservation surgery in a single institution. Our results demonstrate tumours that overexpress both HER2 and AIB1 were associated with markedly reduced relapse free, distant relapse free and overall survival compared to HER2 and AIB1 only overexpressing tumours irrespective of ER status. In ER negative disease both SRC1 and AIB1 were linked to early relapse and death. The SRC family of ER co-regulators is involved in early relapse and resistance in both ER negative and ER positive breast cancer challenging the conventional concept that this effect is mediated solely via the ER.
- Published
- 2012
- Full Text
- View/download PDF
12. Low SAFB levels are associated with worse outcome in breast cancer patients.
- Author
-
Hammerich-Hille S, Bardout VJ, Hilsenbeck SG, Osborne CK, and Oesterreich S
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms mortality, Disease-Free Survival, Female, Humans, Immunoblotting, Kaplan-Meier Estimate, Middle Aged, Prognosis, Selective Estrogen Receptor Modulators therapeutic use, Tamoxifen therapeutic use, Treatment Outcome, Biomarkers, Tumor analysis, Breast Neoplasms metabolism, Matrix Attachment Region Binding Proteins biosynthesis, Nuclear Matrix-Associated Proteins biosynthesis, Receptors, Estrogen biosynthesis
- Abstract
The scaffold attachment factors SAFB1 and SAFB2 have been shown to function as estrogen receptor (ERalpha) co-repressors in breast cancer cells, and to affect many cellular processes such as stress response, RNA processing, and apoptosis. SAFB1 and SAFB2 have also been implicated in breast tumorigenesis: Their shared chromosomal locus at 19p13 is frequently lost in breast cancer, mutations have been identified, and overexpression results in growth inhibition. The purpose of this study was to determine SAFB1/SAFB2 protein expression in human breast tumors, to correlate their expression with either natural progression ("prognostic factor") or with response to Tamoxifen ("predictive factor"), and to analyze potential correlations with tumor characteristics. SAFB1/SAFB2 protein were measured by immunoblotting using a pan-SAFB antibody in tumor extracts from patients with long-term clinical follow-up (n = 289), a subset of whom had received no adjuvant systemic therapy after breast cancer surgery (n = 117) and another subset of whom were treated with adjuvant Tamoxifen (n = 172). SAFB levels were correlated with clinico-pathological variables and patient outcome. SAFB levels varied widely, with 25 tumors not expressing detectable levels of SAFB. SAFB expression was significantly correlated with ERalpha, HER-2, bcl-2 and with expression of other ERalpha coregulators such as SRC-3. There was no association between SAFB expression and disease free survival, however, low SAFB expression was significantly associated with worse overall survival in patients who did not receive adjuvant therapy. This study shows that low SAFB protein levels predict poor prognosis of breast cancer patients, suggesting critical functions of SAFB1 and SAFB2 in breast cancer cells.
- Published
- 2010
- Full Text
- View/download PDF
13. Hsp27 overexpression inhibits doxorubicin-induced apoptosis in human breast cancer cells.
- Author
-
Hansen RK, Parra I, Lemieux P, Oesterreich S, Hilsenbeck SG, and Fuqua SA
- Subjects
- Antigens, Neoplasm, Breast Neoplasms enzymology, Breast Neoplasms genetics, Clone Cells, DNA Fragmentation, DNA-Binding Proteins, HSP27 Heat-Shock Proteins, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes biosynthesis, Molecular Chaperones, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Topoisomerase II Inhibitors, Transfection, Tumor Cells, Cultured drug effects, Antineoplastic Agents antagonists & inhibitors, Antineoplastic Agents pharmacology, Apoptosis drug effects, Breast Neoplasms metabolism, Breast Neoplasms pathology, DNA Topoisomerases, Type II biosynthesis, Doxorubicin antagonists & inhibitors, Doxorubicin pharmacology, Heat-Shock Proteins, Neoplasm Proteins biosynthesis
- Abstract
Previously we demonstrated that heat shock protein 27 (hsp27) overexpression confers resistance to the chemotherapeutic agent doxorubicin in MDA-MB-231 breast cancer cells. Since induction of apoptosis is one underlying mechanism of chemotherapeutic drug action, we investigated the effect of hsp27 overexpression on doxorubicin-induced apoptosis, finding that hsp27 protects MDA-MB-231 cells from apoptosis. We also examined expression of the doxorubicin target, topoisomerase II (topo II), in control and hsp27-overexpressing stable transfectants, as topo II expression is important for both drug sensitivity and the initiation of apoptosis by doxorubicin. The relative levels of both topo IIalpha and beta were higher in the controls than the hsp27-overexpressing clones, suggesting that the apoptotic protective effect of hsp27 overexpression in MDA-MB-231 cells is associated with altered topo II expression.
- Published
- 1999
- Full Text
- View/download PDF
14. Heat shock proteins and drug resistance.
- Author
-
Fuqua SA, Oesterreich S, Hilsenbeck SG, Von Hoff DD, Eckardt J, and Osborne CK
- Subjects
- Antineoplastic Agents pharmacology, Breast Neoplasms genetics, Drug Resistance, Gene Expression Regulation, HSP70 Heat-Shock Proteins drug effects, HSP70 Heat-Shock Proteins physiology, Humans, Prognosis, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Heat-Shock Proteins drug effects, Heat-Shock Proteins physiology
- Abstract
Heat shock proteins (hsp's) are induced in cells when exposed to different environmental stressful conditions. We have found that breast cancer cells sometimes express high levels of several hsp's, which may both augment the aggressiveness of these tumors and make them more resistant to treatment. We have shown that hsp70 is an ominous prognostic sign as detected by Western blot assays in node-negative breast tumors, and that hsp27 increases specific resistance to doxorubicin in breast cancer cell lines. These findings have direct clinical application, and suggest that modulating hsp expression may be a therapeutic target for reversal of hsp-associated detrimental cellular effects.
- Published
- 1994
- Full Text
- View/download PDF
15. Constitutive overexpression of the 27,000 dalton heat shock protein in late passage human breast cancer cells.
- Author
-
Fuqua SA, Benedix MG, Krieg S, Weng CN, Chamness GC, and Oesterreich S
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Base Sequence, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Division, Chromosomes, Human, Pair 3, DNA, Neoplasm genetics, DNA, Neoplasm isolation & purification, Estradiol pharmacology, Heat-Shock Proteins genetics, Hot Temperature, Humans, Molecular Sequence Data, Neoplasm Proteins genetics, Adenocarcinoma pathology, Breast Neoplasms pathology, Gene Expression Regulation, Neoplastic drug effects, Heat-Shock Proteins biosynthesis, Neoplasm Proteins biosynthesis
- Abstract
We present evidence that the mechanisms controlling induction of heat shock transcription factors (HSFs) and mRNA expression of the 27,000 molecular weight heat shock protein, hsp27, are diverse in human breast cancer cells. Heat shock accumulation of hsp27 RNA is associated with the activation of HSF in MDA-MB-231 cells. We have later passage MCF-7 breast cancer cell lines with elevated, constitutive expression of hsp27 mRNA, perhaps due to hsp27 gene amplification. Estradiol and heat shock treatment no longer affect the level of hsp27 mRNA in these cells. Heat induction of HSF is inhibited in cells overexpressing hsp27, although metal ions and amino acid analogs are still capable of activating HSF. These cells will provide a useful system for characterizing alternative pathways in HSF inhibition and activation.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.