Regulatory Mechanisms in Breast Cancer

Regulatory Mechanisms in Breast Cancer

Author: Marc E. Lippman

Publisher: Springer Science & Business Media

Published: 2012-12-06

Total Pages: 455

ISBN-13: 1461539404

DOWNLOAD EBOOK

In Breast Cancer: Cellular and Molecular Biology [Kluwer Academic Pub lishers, 1988], we tried to present an introduction to the emerging basic studies on steroid receptors, oncogenes, and growth factors in the regulation of normal and malignant mammary epithelium. The response to this volume was superb, indicating a tremendous interest in basic growth regulatory mechanisms governing breast cancer and controlling its malignant progres sion. In the two years since its publication, much new and exciting in formation has been published and the full interplay of regulatory mechanisms is now beginning to emerge. We have divided this book into four sections that we hope will unify important concepts and help to crystallize areas of consensus and/or disagreement among a diverse group of basic and clinical scientists working on the disease. The first section is devoted to studies on oncogenes, antioncogenes, proliferation, and tumor prognosis. The first chapter, by Sunderland and McGuire, introduces the characteristics of breast cancer as studied by patho logists to establish prognostic outcome. Of particular interest is a new proto oncogene called HER-2 (or neu), which is rapidly becoming accepted as a valuable new tumor marker of poor prognosis. The second chapter, by Lee Bookstein and Lee, introduces the best known antioncogene, the retinoblas toma antioncogene, whose expression is sometimes lost in breast cancer. Malignant progression appears to be influenced by the balance of proto oncogene and antioncogene expression.


Molecular Aspects of Antiestrogen Resistance and Autophagy in Breast Cancer Cells

Molecular Aspects of Antiestrogen Resistance and Autophagy in Breast Cancer Cells

Author: Irina Nayvelt

Publisher:

Published: 2009

Total Pages: 141

ISBN-13:

DOWNLOAD EBOOK

The major objective of this thesis was to examine the molecular aspects of estrogenic growth and autophagy in estrogen receptor [alpha] (ER[alpha])-positive breast cancer cells. We first examined the role of autophagy mediator, Beclin 1, in estrogenic signaling and antiestrogen resistance in Beclin 1-overexpressing MCF-7 cells. We found that a potential interaction between ER[alpha] and Beclin 1 rendered Beclin 1-transfected cells less sensitive to estradiol (E2)-induced growth stimulation, and to antiestrogen-mediated growth inhibition. Thus, a novel function for Beclin 1 might involve down-regulation of the action of ER[alpha], contributing to resistance of breast cancer cells to antiestrogens. In an attempt to develop novel therapeutic agents for breast cancer, we explored the effect of the polyamine analogue, 1,15-bis(ethylamino)-4,8,12-triazapentadecane (BE-3-3-3-3), on MCF-7 cell growth in the presence and absence of E2. BE-3-3-3-3 caused growth inhibition in the presence of E2. However, it mimicked estradiol and stimulated cell growth in the absence of E2, and induced growth response genes, such as c-fos, c-jun, and c-myc. This also induced autophagy, and increased levels of autophagy-related proteins, Beclin 1 and MAP LC3-II. In another approach to introduce gene therapy for breast cancer treatment, we explored the physico-chemical aspects of DNA nanoparticle formation. In an effort to optimize gene delivery systems, we investigated DNA condensation to nanoparticles in the presence of [alpha], [alpha]'-methylated spermine analogues, and characterized the size, shape and stability of the resultant nanoparticles. Although some analogues proved more efficacious DNA condensing agents than spermine, hydrodynamic radii of nanoparticles produced by analogues were comparable to those produced by spermine. We also compared the DNA condensing abilities of poly-L-lysine (PLL) and oligolysines, and characterized the physico-chemical properties of their condensates. PLL was a more effective condensing agent than oligolysines, and produced more stable nanoparticles. We conclude that PLL and oligolysines bind and condense DNA through different mechanisms. In summary, our research provides new insights into the mechanism of antiestrogen resistance and autophagy in breast cancer. We also provide mechanistic insight into DNA nanoparticle formation in the presence of polyamine analogues and lysines.


Grainyhead-like Protein 2 Regulates the Transcriptional Activity of Estrogen Receptor Alpha Phosphorylated at Serine 118 in Breast Cancer

Grainyhead-like Protein 2 Regulates the Transcriptional Activity of Estrogen Receptor Alpha Phosphorylated at Serine 118 in Breast Cancer

Author: Rebecca M. Reese

Publisher:

Published: 2022

Total Pages: 0

ISBN-13:

DOWNLOAD EBOOK

The Grainyhead-like protein family, composed of GRHL1, GRHL2, and GRHL3, are nuclear transcription factors that regulate epithelial differentiation. GRHL2 is characterized as having oncogenic and tumor suppressive roles across many cancers. GRHL2 is also associated with several nuclear hormone receptors, including progesterone receptor (PR), androgen receptor (AR), and more recently, estrogen receptor (ER). ER is expressed in over 70% of breast cancers and is a major therapeutic target. GRHL2 is more highly expressed in ER-positive over ER-negative breast cancers, and studies suggest that GRHL2 modulates ER recruitment to chromatin. GRHL2 is hypothesized to enhance ER-transcriptional activity by recruiting histone modifiers like MLL3 to enhancers, but it can also repress ER-transcriptional activity by suppressing the catalytic activity of the histone acetyltransferase p300. Our group found a specific association of the GRHL2 motif with ER phosphorylated at serine 118 (pS118-ER), a post-translational modification activated by estrogen (E2) that is required for maximal ER-transcriptional activity. As a whole, the mechanisms by which GRHL2 regulates ER-transcriptional activity and, more specifically, pS118-ER transcriptional activity are not well understood. In the work presented here, I take cistromic and transcriptomic approaches to explore the role of GRHL2 in facilitating pS118-ER recruitment to chromatin and downstream transcriptional activity. I find that GRHL2 is critical for maximal pS118-ER chromatin-recruitment, GRHL2 can both enhance and antagonize E2-mediated pS118-ER transcriptional activity, and ER/GRHL2 co-regulated genes are involved in cellular migration. The dual roles of GRHL2 in pS118-ER transcriptional regulation may be due to the pioneering activities of GRHL2 which allow the factor to promote an open chromatin structure and subsequently recruit or modulate coregulators present at a given locus. I also explore the function of a portion of the poorly defined GRHL2 transactivation domain and find a 52 amino acid portion of the domain is important for GRHL2 transactivation activity. These studies further our understanding of the function of GRHL2 in regulating pS118-ER transcriptional activity in breast cancer and provide a basis for future studies to expand our knowledge of the interaction between these two transcription factors.


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.


Regulation of Mammary Stem Cells and Breast Cancer by the Co-factor of LIM Domains (Clim) Transcriptional Regulator

Regulation of Mammary Stem Cells and Breast Cancer by the Co-factor of LIM Domains (Clim) Transcriptional Regulator

Author: Michael Salmans

Publisher:

Published: 2014

Total Pages: 113

ISBN-13: 9781321022223

DOWNLOAD EBOOK

Recent advancements in mammary gland biology demonstrate conflicting models in maintenance of basal and luminal cell compartments by either unipotent or bipotent mammary stem cells. However, the molecular mechanisms underlying control of the basal cell compartment, including stem cells, remain poorly understood. Here we explore the currently unknown transcriptional mechanisms of basal stem cell (BSC) maintenance, in addition to addressing the role of the basal cell compartment in preserving luminal cell fate and promoting development of human breast tumors of luminal origin. We discover a novel function for the Co-factor of LIM domains (Clim) transcriptional regulator in promoting mammary gland branching morphogenesis and breast tumorigenesis through maintenance of the basal stem cell population. The transcriptional networks coordinated by Clims in basal mammary epithelial cells also preserve the identity of luminal epithelial cells, demonstrating a crosstalk between these two cellular compartments. Furthermore, we correlate developmental gene expression data with human breast cancer to investigate the role of developmental pathways during the initiation and progression of breast cancer. The gene regulatory networks identified during development, including those specifically coordinated by Clims, correlate with breast cancer patient outcome, suggesting these genes play an important role in the progression of breast cancer. We also explore the role of Agr2 a putative protein disulfide isomerase (PDI) initially identified as an estrogen-responsive gene in breast cancer cell lines, in normal mammary gland development and breast cancer. While Agr2 expression in breast cancer is positively correlated with estrogen receptor (ER) expression, it is upregulated in both hormone dependent and independent carcinomas. Agr2 expression is developmentally regulated in the mammary gland, with maximum expression during late pregnancy and lactation. We show that [Subscript Agr2] facilitates normal lobuloalveolar development by regulating mammary epithelial cell proliferation. In conclusion, the estrogen-responsive Agr2, a candidate breast cancer oncogene, regulates epithelial cell proliferation and lobuloalveolar development in the mammary gland. The pro-proliferative effects of Agr2 may explain its actions in early tumorigenesis.


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.