Differential Regulation of Cell Cycle Progression in Human Breast Cancer Cell Lines by the Estrogen Receptor

Differential Regulation of Cell Cycle Progression in Human Breast Cancer Cell Lines by the Estrogen Receptor

Author:

Publisher:

Published: 1999

Total Pages: 0

ISBN-13:

DOWNLOAD EBOOK

Critical predictions as to the biological behavior, and thus the appropriate therapeutic strategy, of breast cancers can be made based upon the status of the estrogen receptor (ER). In support of DOD grant # DAMD-17-97-1-7069, our goal is to better understand the mechanisms by which ER controls the expression of target genes and therefore mediates the biological effects upon gene regulation and cell cycle progression. Our detailed studies of the regions of ER that control cell cycle progression in breast cancer cell lines have indicated an absolute requirement for the Activating Function-2 (AF-2) region of ER for hormone-dependent cell cycle progression. In many different classes of nuclear receptors, this area has been vigorously studied and has been shown to be important for the physical interaction between hormone bound receptors and coactivators. Our studies have demonstrated that mechanisms involving the chemical and structural modification of chromatin are critical for transcriptional responses to estrogen and may also be important for estrogen-dependent cell cycle progression.


Regulation of E2F-1 Gene Expression in Human Breast Cancer Cells

Regulation of E2F-1 Gene Expression in Human Breast Cancer Cells

Author: Sharon Khethiwe Ngwenya

Publisher:

Published: 2005

Total Pages:

ISBN-13:

DOWNLOAD EBOOK

17[beta]-Estradiol induces E2F-1 gene expression in ZR-75 and MCF-7 human breast cancer cells. Analysis of the E2F-1 gene promoter in MCF-7 cells previously showed that hormone-induced transactivation required interactions between estrogen receptor [alpha] (ER[alpha])/Sp1 bound to upstream GC-rich sites and NFYA bound to downstream CCAAT sites within the -169 to -54 promoter region. This promoter region was also E2-responsive in ER[alpha]-positive ZR-75 cells; however, further analysis of the promoter showed that cooperative ER[alpha]/Sp1/NFY interactions were not necessary for hormone-inducedtransactivation in ZR-75 cells. The upstream GC-rich motifs are activated independently by ER[alpha]/Sp1 in ZR-75 but not MCF-7 cells, and the downstream CCAAT sites were also E2-responsive. E2 also induced reporter gene activity in ZR-75 cells transfected with an expression plasmid containing the yeast GAL4 DNA binding domain fused to pM-NFYA and a construct containing five tandem GAL4 response elements. Subsequent studies showed that hormonal activation of pE2F-1j[subscript]m1 and pM-NFYA are dependent on non-genomic pathways in which E2 activates cAMP/protein kinase A. Hormone-dependent regulation of E2F-1gene expression in ZR-75 and MCF-7 involves different mechanisms, demonstrating the importance of cell context on transactivation pathways, even among ER-positive breast cancer cell lines. TCDD inhibited ER[alpha]-mediated responses in MCF-7 and ZR-75 cells. E2-induced E2F-1protein and mRNA levels in MCF-7 and ZR-75 cells and this response was inhibited by TCDD. Constructs containing GC-rich sites alone orin combination with the downstream NFY sites were used in transactivation studies to investigate the mechanism of inhibitory AhR-ER[alpha] crosstalk. AlthoughTCDD inhibited E2-induced mRNA, protein and reporter gene actitivity, it was not possible to determine if the inhibitory response was due to limiting ER[alpha]protein levels due to proteasome degradation since proteaome inhibitors aloneblocke hormone-dependent responses. TCDD also inhibited the cAMP/PKApathway by inhibiting adenyl cyclase activity. In Drosophila SL-2 cells cotransfected with the GC-rich -169 to -54 region, ER[alpha] and Sp1 plasmids E2 induced transactivation in cells cotransfected with AhR/Arnt expression plasmids suggesting that the AhR complex suppressed ER[alpha]/Sp1 action. These results demonstrate that TCDD inhibits E2-dependent activation of both non-genomic and genomic pathways of ER-mediated E2F-1 gene expression.


Molecular Mechanisms Underlying the Regulation of Gene Expression and Growth in Breast Cancer Cells

Molecular Mechanisms Underlying the Regulation of Gene Expression and Growth in Breast Cancer Cells

Author: Miao Sun

Publisher:

Published: 2013

Total Pages: 175

ISBN-13:

DOWNLOAD EBOOK

Breast cancer is a serious public health issue, and a full understanding of its etiology and pathophysiology is a primary focus in the field. Molecularly, the combined action of a plethora of factors in multiple pathways is involved in the regulation of the breast tumoriogenic process. Characterization of a more complete spectrum of the molecular factors will provide insights into the development of new and improved diagnostic, prognostic and therapeutic tools for treating breast cancer. To this end, my studies utilize a combination of molecular biology and bioinformatic methods, to uncover the mechanisms underlying the regulation of gene expression and growth in human breast cancer cells. To investigate the molecular crosstalk of the estrogen and c-Jun N-terminal kinase 1 (JNK1) signaling pathways, I monitored the genomic localization of estrogen receptor [alpha] (ER[alpha]) and JNK1 in basal and estrogen-stimulated MCF-7 breast cancer cells. I found that JNK1 binds to the promoter of many genes. ER[alpha] is required for the binding of JNK1 to the estrogen-induced sites, and JNK1 in turn functions as a coregulator of ER[alpha]. The convergence of ER[alpha] and JNK1 at target promoters regulates estrogen-dependent gene expression, as well as downstream estrogendependent cell growth responses. Furthermore, the implication of long noncoding RNAs (lncRNAs) in breast cancer is also coming to light. I developed a computational approach that integrates information from multiple genomic datasets, and generated a comprehensive catalog of 1888 expressed lncRNA genes in MCF-7 cells. Almost half of them are first annotated in this study, and more than a quarter are estrogen-regulated. Close examination revealed many interesting features. Interestingly, cell type-specific expression of lncRNAs predicts the intrinsic molecular subtypes of breast cancer, suggesting its potential utility as prognostic marker. Lastly, by selecting lncRNAs with elevated expression in breast tumors, and whose differential expression across a wide spectrum of tissues and cell types correlates with important cell viability genes, we identified a number of lncRNAs that are required for the normal growth of human breast cancer cells. Collectively, my studies expanded our understanding of the molecular mechanisms underlying breast cancer biology, and suggested new targets for therapeutic interventions.


Epigenetic Regulation by Estrogen Receptor in Breast Cancer Cells

Epigenetic Regulation by Estrogen Receptor in Breast Cancer Cells

Author: Athéna Sklias

Publisher:

Published: 2019

Total Pages: 0

ISBN-13:

DOWNLOAD EBOOK

Previous epidemiological and experimental studies have strongly implicated estrogens in breast cancer risk and Estrogen Receptor (ER), the transcription factor to which estrogen binds, is considered as the major molecular driver of around 70% breast cancers. The importance of the deregulated estrogen signalling is further highlighted by increasing evidence that current chemopreventive and therapeutic strategies that target hormonally responsive breast cancers frequently result in the development of resistance to anti-estrogens and metastatic progression, highlighting the need for understanding the molecular underlying mechanisms. While until recently, ER was believed to act as a stand-alone transcription factor, which can directly bind its motifs in DNA, it is now accepted that ER activity is a complex and dynamic process that requires highly concerted actions of a dozen transcriptional cofactors and various chromatin regulators at DNA. Recent studies focused on characterising ER-associated cofactors and their role in opening the chromatin provided a remarkable insight into transcriptional regulation mediated by ER. However DNA methylation and histone acetylation are poorly understood in the context of ER's dynamic binding. In this thesis, I combined a cell culture protocol adapted for studying estradiol (E2) deprivation and re-stimulation in stricto sensu in ER-positive breast cancer cells with the latest methylation array, that allowed a genome-wide interrogation of DNA methylation (including a comprehensive panel of enhancers). I further investigated histone acetylation (ChIP-seq) and transcriptome (RNA-seq) after E2 deprivation and re-stimulation to better characterise the ability of ER to coordinate gene regulation. I found that E2 deprivation and re-stimulation result in time-dependent DNA methylation changes and in histone acetylation across diverse genomic regions, many of which overlap with enhancers. Further enrichment analysis of transcription factor (TF) binding and motif occurrence highlights the importance of ER tethering mainly through two partner TF families, AP-1 and FOX, in the proximity of enhancers that are differentially methylated and acetylated. This is the first study that comprehensively characterized DNA methylation at enhancers in response to inhibition and activation of ER signalling. The transcriptome and genome occupancy data further reinforced the notion that ER activity may orchestrate a broad transcriptional programme through regulating a limited panel of critical enhancers. Finally, the E2 re-stimulation experiments revealed that although the majority of the observed epigenetic changes induced by E2 deprivation could be largely reversed when the cells were re-stimulated we show that DNA hypermethylation and H3K27 acetylation at enhancers as well as several gene expression changes are selectively retained. The partial reversibility can be interpreted as a sign of treatment efficiency but also as a mechanism by which ER activity may contribute to endocrine resistance. This study provides entirely new information that constitutes a major advance in our understanding of the events by which ER and its cofactors mediate changes in DNA methylation and chromatin states at enhancers. These findings should open new avenues for studying role of the deregulated estrogen signalling in the mechanism underlying the “roots” of endocrine resistance that commonly develops in response to anti-estrogen therapy.


Mechanisms of Estrogen Receptor Alpha Mediated Transcriptional Repression

Mechanisms of Estrogen Receptor Alpha Mediated Transcriptional Repression

Author: Joseph Sin

Publisher:

Published: 2009

Total Pages: 42

ISBN-13:

DOWNLOAD EBOOK

Prolonged exposure to increased levels of estrogen has been shown to increase the risk of breast cancer. In addition, estrogen has been shown to cause breast cancer cell proliferation. A common form of breast cancer treatment involved selective estrogen receptor modulation. A molecular explanation of how this works is that estrogen regulates and binds to estrogen receptor (ER), a ligand-dependent transcription factor. ER associated with estrogen induces gene transcription by translocating into the nucleus and binding to estrogen response element. ER also recruits cofactor proteins, which results in chromatin remodeling and gene expression regulation through interacting with histone acetylases or transcriptional machinery. Most studies have focused on the study of how ER can activate gene transcription. Recently, ER has been shown to also repress gene transcription. my research has two parts. The first part was to find genes that were down regulated by estrogen in order to increase the data pool of genes down-regulated by estrogen. Four target genes, ARGN, MGC16169, CALML5, and NFIB are suspected to be involved in down-regulation by ER. However, after conducting validation tests, these genes were determined to not be repressed. The second part includes characterizing the specific effects of co-repressors NCoR, NRIP1, and SMRT. Removal of these co-repressors and subsequent effect of their removal on following four ER target sites, HES1, PSCA, SLC35A1, and MME were studied. A knock down of a single co-repressor did not affect the majority of transcriptional activity in ER repressed target genes. A triple knock down was also conducted in hope that removal of multiple co-repressors might affect repression. However, the triple knock down was a failure and future experiments need to be done. Understanding the mechanisms of ER transcriptional repression would significantly aid the creation of effective treatments for breast cancer.


Regulation of Estrogen Receptor-alpha Mediated Gene Expression and Endocrine Resistance Through Estrogen Receptor-alpha Phosphorylation and Micro-RNA in Breast Cancer

Regulation of Estrogen Receptor-alpha Mediated Gene Expression and Endocrine Resistance Through Estrogen Receptor-alpha Phosphorylation and Micro-RNA in Breast Cancer

Author: Kyuri Kim

Publisher:

Published: 2011

Total Pages:

ISBN-13:

DOWNLOAD EBOOK

Estrogens are associated with the development and progression of breast cancer in addition to their role in normal reproductive physiology, and estrogen receptors (ER) mediate the actions of estrogen in target tissues by regulating the expression of numerous biologically important target genes. The progression of human breast cancer and the development of resistance to endocrine therapies are thought to be associated with ER phosphorylation. We generated multiple combinations of ER phospho-mutants, at residues serine 104, 106, 118, 167, 236, and 305, and examined their impact on receptor half-life, the agonist and antagonist balance of selective estrogen receptor modulators (SERMs) and selective estrogen receptor downregulators (SERDs), the regulation of ER transcriptional activity, and stimulation of cell proliferation in response to estradiol and SERMs/SERD. We showed that changes in ER affecting the phosphorylation status of the receptor greatly impact receptor function and differential SERM and SERD modulated cellular responses that could contribute to resistance to endocrine therapies in breast cancer. We also studied the regulation of microRNAs (miRNAs) by estradiol and growth factors through ER and extracellular signal-regulated kinase 2 (ERK2) in order to understand their physiological impact on breast cancer. We identified nine miRNA- encoding genes harboring overlapping ER and ERK2 binding sites close to their transcription start sites, which require ER and ERK2 for transcriptional induction as well as estradiol- mediated miRNA regulation. We then identified TP63, a target of miR-101, miR-190 and miR- 196a2, and showed that TP63 plays an important role in estradiol- or growth factor-mediated cellular response in breast cancer cells (MCF-7 and MDA-MB-231) by increasing tumor cell growth and in vitro invasion mainly controlled by miR-196a2 action. These results suggest a tumor-suppressive role of miR-196a2 in regulating TP63 expression and the aggressive behavior of breast cancers.