Estrogen receptor Beta – an introduction

From Seminars in Reproductive Medicine

The Role of Estrogen Receptor-β in Breast Cancer

Leigh C. Murphy, Ph.D.; Etienne Leygue, Ph.D.

 

Posted: 02/09/2012; Semin Reprod Med. 2012;30(1):5-13. © 2012 Thieme Medical Publishers

 

Abstract and Introduction

Abstract

The discovery of a second estrogen receptor (ER), ERβ, has led to a reevaluation of estrogen action. The widespread expression of ERβ-like proteins in normal and neoplastic mammary tissues suggests a role of ERβ in the breast. Little progress has been made in elucidating this role or roles, but the presence of two ERs and variant isoforms in breast cancers presents challenges and opportunities to tease out complexities in understanding the estrogen signaling pathway in breast tissues. Identification of two groups of ERβ-expressing tumors in vivo, and the possibility of differential function, has already raised expectations that targeting ERβ may offer new treatment options for breast cancer patients where previously only aggressive chemotherapies were available. This supports continued efforts to understand the nature and function of ERβ in breast cancer, but it also suggests that ER status may need to be redefined to include an assessment of ERβ isoforms in addition to ERα.

Introduction

The estrogen receptor (ER) is central to the biology of most human breast cancer. As such, ER has been and continues to be the target for the treatment and prevention of breast cancer. However, the ER is no longer a stand-alone molecule associated with a linear signaling pathway. ER signaling is now known to be remarkably complex and multifaceted. In particular, the discovery of a second ER, called ERβ, has been instrumental in a reevaluation of estrogen action in all tissues, either normal or neoplastic. Gene knockout studies clearly identify ERα expression as essential for normal mammary gland development. ERα today remains the primary target for endocrine therapies in breast cancer. As such, its levels are measured to determine the ER status of primary tumors and to predict the likelihood of patients responding positively to tamoxifen treatment, for example. Such data, in addition to >30 years of discovery and clinical experimentation, have established a central role of ERα in both normal and neoplastic mammary growth. In contrast, although 15 years have passed since the discovery of ERβ, the significance of its expression and its potential roles in normal mammary development, breast tumorigenesis, or tumor progression remain controversial and unclear. This article reviews the current research focused on elucidating the role of ERβ in breast cancer.

 

Estrogen Receptor-β and Normal Mammary Gland

The expression of ERβ in the normal mammary gland of both mouse and human has been investigated.[1] In both cases ERβ is widely expressed in multiple normal mammary gland cellular compartments, including luminal and myoepithelium. This is in stark contrast to ERα, where only infrequent luminal epithelial staining is reported.[2] Similarly, if the ERα knockout mouse models show no mammary gland development, only small effects on mammary gland differentiation and overall none on its function are reported for the ERβ knockout.[3,4] It should be noted, however, that differences between ERβ knockout models and different laboratories have been observed.[3,5,6]

Therefore, somewhat paradoxically, despite ERβ being the most frequently expressed ER in the normal mammary gland, it does not appear to be the most important with respect to normal development and function, at least as determined from mouse gene ablation studies.

 

Estrogen Receptor-β and Breast Cancer

The expression of ERβ in human breast tumors was first reported in 1997 when ERβ RNAs were detected in multiple human breast cancer biopsy samples.[7,8] Demonstration of ERβ-like protein expression in breast tumors by Western blotting and by immunohistochemistry followed quickly thereafter.[9,10] These reports established the platform for many subsequent studies in which clinical samples were investigated for ERβ expression. Relationships between this expression and clinicopathological parameters and patient outcome were investigated to obtain insight into the possible function of ERβ in breast cancer. Although results that have emerged from the latter type approach together with results generated from laboratory studies have provided some significant insights, several important complexities associated with measuring ERβ expression and establishing its exact functions have also emerged. These complexities are likely to have an impact on our understanding of the overall roles that ERβ plays in breast cancer. These complexities include the presence of multiple variant isoforms of ERβ, consideration of differential functions when ERβ is expressed alone versus those when it is coexpressed with ERα, and methods of determining ERβ expression.[11-15]

Estrogen Receptor-β Variants

Fig. 1 illustrates schematically the similarities and differences in the structure of ERβ1 and ERα. Also illustrated are two C-terminally truncated ERβ variants, ERβ 2/cx and ERβ 5, and an N-terminally truncated short form of ERβ 1. Most of the variant isoforms have been detected at the RNA level, even though in some cases, the predicted protein has also been detected in breast tumors.[16-19] Most of these isoforms are generated by alternative splicing mechanisms.[20] In contrast, an N-terminally truncated short form of ERβ 1 may be generated posttranslationally by proteolysis.[21] Other posttranslational modifications have been reported, including detection of a phosphorylated form of ERβ in multiple human breast tumors.[22,23]

 

 

(Enlarge Image)

Figure 1.

Schematic representation of human estrogen receptor (ER)α (hERα) and some ER-β (hERβ) isoform proteins. Human ERβ 1 (hERβ 1) is the full-length ligand-binding form, and ERβ 2 (hERβ 2/cx) and ERβ 5 (hERβ 5) are two common C-terminally truncated variants unable to bind ligands.

 

A common observation is that ERβ expression is decreased in cancer compared with the equivalent normal tissue.[24-27] Mechanisms potentially associated with this downregulation of ERβ expression include increased methylation of the ERβ promoter, microRNAs, and other posttranscriptional regulatory mechanisms.[28-30] As a direct result of these overall low ERβ levels in breast cancer cells, there are few, if any, cell line models where reliable ERβ expression has been demonstrated. Most of the models developed to study ERβ protein function have therefore been generated using either upregulated transient, constitutive stable, or inducible stable expression of ERβ-like proteins in cultured cell lines and xenografts from these cell lines.[31-37] A common conclusion has been that ERβ 1, the full-length ligand binding isoform (ERβ 1), is antiproliferative and proapoptotic.[32,34,38] However, in stark contrast to the latter conclusions are models in which ERβ 1 overexpression is growth stimulatory or has no effect on proliferation and/or cell death.[36,37,39] Furthermore, there are published data reporting correlations of ERβ expression with both pro- and antiproliferative activity in normal mammary cells.[40,41] The reasons for such differences remain unknown, even though current data indicate that differences in species and/or cell background, in particular p53 status, type of ERβ, unknown clonal selection variables, and level of expression, may influence ERβ’s effect on proliferation.[42] It is unclear whether the increased expression of ERβ 1 in experimental systems is relevant to the levels of ERβ seen in tumors and/or normal tissues in vivo, especially because ERβ is often downregulated in tumors compared with normal tissues.[24-26]

It is interesting that the only models showing an association of overexpression of ERβ 1 with increased proliferation, invasiveness, and metastasis is when it has been introduced into a breast cancer cell line that is ERα negative.[36,37] This may be highly relevant when interpreting the differences seen in vivo when the expression of ERβ 1 alone in human breast tumors is compared with its coexpression with ERα.[11,12,43]

Most studies of ERβ function and mechanisms of action have focused on ERβ 1, and considerably less is known about putative roles played by ERβ variants. The most studied variant is the ERβ 2/cx. Here again, different activities in apparently similar model systems have been observed using overexpression approaches. Generally, the C-terminally truncated ERβ variants such as ERβ 2/cx and ERβ 5 are thought to have little activity of their own. They can, however, influence the activity of either ERβ 1 and ERα when coexpressed, either inhibiting or enhancing these two ligand binding forms of ERs.[17,44-46] Using stable overexpression of ERβ 2/cx in MCF7 cells, inhibition of cell cycling and colony growth in soft agar were reported.[47] Part of the mechanism involved was thought to be inhibition of ERα activity due to ERα/ERβ 2/cx heterodimer formation, resulting in a proteasome-dependent degradation of ERα.[47] In contrast, this was not seen in another model of overexpression of ERβ 2/cx in MCF7 cells despite decreased expression of ERα downstream targets such as the progesterone receptor.[17]

More recently, the existence of an N-terminally truncated shorter form of ERβ 1 was reported that may be functionally distinct from the longer form of ERβ 1.[21,48] Other limited experimental studies can be interpreted to suggest that the long and short forms of ERβ 1 may have differential effects on inflammatory/immune activities.[49] Given the anti-inflammatory activities of ERβ 1, the relative expression of the long and short forms of ERβ 1 might have functional implications in breast cancer.[50-52]

Expression of Estrogen Receptor-β in Breast Tumors in vivo

Consistently decreased expression of ERβ 1 protein levels from normal to invasive breast cancer has been described in several studies.[24,53] In contrast, ERα expression is increased during mammary tumorigenesis, and ERβ 2/cx expression has also be shown to be increased in breast tumors compared with normal breast tissue.[54,55] Overall, there appears to be a marked alteration of the relative expression of not only ERβ 1 and ERα but also the relative expression of ERβ isoforms during breast tumorigenesis.[56,57] This is consistent with the known alteration of estrogen signaling that occurs during breast cancer development and suggests that the altered levels of ERβ isoforms could also, in part, underlie this altered estrogen signaling. On a side note, although originally thought to possibly result from so-called hiccups of the splicing machinery, alternative splicing events have now been shown to be highly controlled and to introduce a new layer of complexity in the regulation of gene expression.[58-61] Alternative splicing as such is suspected to participate in various pathologies including skin diseases, neurodegeneration, and cancer.[62-66] One might speculate that modifications of the splicing events controlling the respective proportion of ERβ isoforms, through the resulting alterations of ER signaling and hence response to estrogen, contribute not only to breast tumorigenesis but also to resistance to specific endocrine therapies.

Despite ERβ 1 downregulation in invasive breast cancer, a significant number of invasive breast cancers still express ERβ 1.[11,15] Several studies have now been published in which expression of ERβ-like proteins in tumors was correlated with known histopathological and clinical parameters to gain further insight into the potential role played by these proteins in breast cancer.

Multiple studies along these lines have now been published. However, they led to varied and often contradictory results. This may, in part, be due to how ERβ expression was determined (RNA or protein), which antibody was used to determine protein expression, definitions of ERβ positivity and negativity, and a lack of appreciation that when considering ERβ expression, there are clearly two groups to take into account: one in which ERβ is coexpressed with ERα and the second where ERβ is expressed alone.

Because ERβ can, in addition to actual tumor cells, also be expressed in other cell types, such as infiltrating lymphocytes, other immune cells, and fat and vascular cells that make up a heterogeneous breast tumor in vivo, studies investigating globally extracted RNAs may be difficult to interpret.[67] Studies using microdissected invasive cells can overcome this issue. Approaches where ERβ expression is assessed taking into account the cellular location within a tumor sample are likely to be the most informative, as are those that accurately determine the exact nature of the ERβ isoforms expressed. Therefore, immunohistochemical studies using well-characterized antibodies to specific ERβ epitopes located in the N-terminus and therefore detecting “total” ERβ-like (i.e., the sum of ERβ 1 long and short, ERβ 2/cx, ER β 5, etc.) proteins or those using antibodies specific for a given ERβ isoform (e.g., ERβ 1 only) are the focus of the following discussion.

Since last reviewed in 2008, several other studies have been published.[11] As such, there are at least 23 studies now reporting detection of ERβ-like proteins in tumors from >3000 human breast cancer cases with correlations to clinical outcome ( Table 1 ).[68,69] Most but not all of these studies find associations of higher levels of ERβ-like proteins with better clinical outcome, often in patients who have been treated with tamoxifen therapy. In many of these cases ERβ 1 was determined using specific antibodies to the C-terminal region of this receptor. However, similar conclusions were made using antibodies to an N-terminal epitope that would recognize total ERβ-like proteins, and some studies have used antibodies to the ERβ 2/cx C-terminal region with similar conclusions.[70] Of the seven studies in which both ERβ 1 and ERβ 2/cx were compared, two[12,71] found no association of either isoform with clinical outcome, one[72] found that high levels of ERβ 2/cx and ERβ 1 were associated with better clinical outcome, one[18] found that high levels of ERβ 2/cx but not ERβ 1 were associated with better clinical outcome, and three[19,68,73] found that high levels of ERβ 1 but not ERβ 2/cx were associated with better clinical outcome. In most cases only nuclear staining of the ERβ proteins was determined, although two studies determined both nuclear and cytoplasmic staining of the ERβ isoforms and provided evidence that subcellular localization of ERβ 2/cx was differentially associated with clinical outcome.[18,19] Both these latter studies identified the cytoplasmic localization of ERβ 2/cx as associated with poor clinical outcome. Several of the studies also suggest that the level of ERβ was predictive of outcome due to tamoxifen treatment.[68,69] More recently, higher levels of ERβ 1 were also associated with better clinical outcome in patients treated with aromatase inhibitors.[74,75]

Further insight into the possible reasons for the contradictory results associated with ERβ expression and its association with clinical outcome, in particular in patients treated with tamoxifen, comes with a recent prospective study in which ERβ 1 expression was compared across four intrinsic molecular subtypes, previously classified by expression profiling.[76,77] Interestingly, ERβ 1 expression was evenly distributed across the luminal A, luminal B, HER2, and triple negative subtypes. High levels were differentially associated with clinical outcome, however, depending on whether the patients were node positive or negative. This was most marked between the luminal A and luminal B subtypes, where high ERβ 1 expression was associated with good clinical outcome in luminal A node-negative but not node-positive groups. In contrast, high ERβ 1 expression was associated with poor clinical outcome in luminal B node-positive but not node-negative groups.[76] Luminal A and B subtypes are generally ERα positive and in previous studies would have been considered as one group (ER+) and likely be eligible for endocrine therapy. Differential representation of these two molecular subtypes plus or minus nodal involvement in previous retrospective studies could therefore have significantly affected the results deduced.

Most of the studies already cited focused on ER+(i.e., ERα-positive breast cancer), so they represent ERβ coexpressed with ERα and are often treated with endocrine therapies. So a general conclusion is that higher levels of ERβ-like proteins in the presence of ERα in human breast cancer are associated with a better prognosis and the increased likelihood of responsiveness to endocrine therapies. These data also support the idea that ER profiling (determination of ERα, ERβ 1, and ERβ 2/cx and the development of an ER isoform code) may provide a more accurate biomarker for prognosis and treatment response prediction than what is currently available.

The mechanism underlying this conclusion may be associated with the ability of several ERβ isoforms to heterodimerize with ERα and negatively modulate ERα activity.[20,45,47] Furthermore, one model of inducible ERβ 1 expression in ERα-positive MCF7 cells reported that increased ERβ 1 is associated with increased sensitivity to the growth inhibitory effects of 4-hydroxy-tamoxifen, and more recently for increased sensitivity of breast cancer to endoxifen, an active metabolite of tamoxifen in vivo.[31,78] These results further support a direct mechanistic role of ERβ 1 in endocrine therapy sensitivity in vivo.

Despite the relatively few studies undertaken to date with a focus on ERα-negative but ERβ-positive breast cancer, there is a generally consistent conclusion that ERβ 1 likely has a different role in these tumors and could be a targeted treatment option for patients who otherwise have few options except aggressive chemotherapies.[12,43,69,79] The first data supporting a differential role for ERβ in ERα-negative breast cancer showed a positive correlation of ERβ 1 with markers of proliferation (Ki 67) and cell cycling (cyclin A).[12,43,79] At least in one study, the association was found for ERβ 1 but not ERβ 2/cx, supporting the idea that a selective ligand targeting was feasible.[12] Given the often found antiproliferative and proapoptotic activities of overexpressed ERβ 1 in cells in culture, irrespective of the ERα status, the mechanism of ERβ1’s positive association with proliferation is unclear at this point. It must be pointed out, however, that the nature of ERβ 1 in ERα-negative tumors (i.e., short versus long form, any posttranslational modifications or mutated ERβ 1) has not been investigated.

The importance of understanding the role of ERβ 1 expression alone in breast cancer has been heightened with the publication of two studies where ERβ expression was determined by immunohistochemistry using archival pathology blocks representing ERα-negative breast cancer patients who had been treated with adjuvant tamoxifen for 2 years with long-term follow-up.[68,69] Both studies reported that ERβ positivity was associated with significantly better survival. Because one of them measured ERβ 1, tamoxifen binding to ERβ 1 being directly involved in the mechanism mediating the treatment effect is a real and exciting possibility.[68] As well, ERβ 1 expression was found to be an important prognostic marker in triple-negative breast cancer patients treated with tamoxifen in a subgroup analysis.[68] As mentioned previously, the implications are that targeting ERβ 1 pathway(s) may be a treatment option for such patients who generally have few options other than aggressive chemotherapies, and some clinical trials are underway to address such a possibility, including the ANZ 1001 SORBET study coordinated by the Australian New Zealand Breast Cancer Trials Group (ANZ CTRN: 12610000506099).

Markers of Estrogen Receptor-β Functionality in Breast Cancer

The emerging idea that the function of ERβ 1 may be different in breast cancer when it is expressed alone to that when its coexpressed with ERα, underscores the need to identify downstream targets of ERβ 1 functionality in human breast cancer. Historically, downstream markers of ERα functionality such as the progesterone receptor and pS2, have proven useful clinically in more accurately predicting prognosis and responsiveness to endocrine therapies.[80,81] Therefore, reliable downstream specific markers of ERβ activity would be extremely valuable in elucidating roles of ERβ and in providing more accurate biomarkers of a functionally intact ERβ signaling pathway in both ERα-positive and -negative tumors. In 2008, a unique ERβ-associated gene expression profile was described, only in ERα-negative but not ERα-positive breast tumors.[69] This not only supports a differential function of ERβ 1 when expressed alone, but it also supports the likelihood that distinct downstream targets of ERβ 1 exist and are relevant to human breast cancer in vivo. A few gene expression profiling, chromatin-immunoprecipitation (ChiP)-on-chip and ChiP-seq studies have been published using ERβ 1 overexpressing ERα-positive MCF7 breast cancer cell models to identify ERβ 1 regulated genes and gene networks.[32,33,82,83] This of course models ERβ 1 coexpression with ERα, and only one study has been reported on expression profiling of the ERα-negative breast cancer cell line, Hs578T, stably expressing inducible ERβ 1 expression.[84] This models ERβ 1 expression alone in breast cancer.

These global expression and genome-wide binding site analyses are still in the early stages and as yet have not provided unique molecular signatures that have been applied to large numbers of breast cancer cases and correlated with ERβ expression and clinical outcome. Some general themes are emerging, however. In some cases ERβ has been identified to regulate genes that are part of transforming growth factor (TGF)-β-like signaling pathways.[33] If this is supported by further analyses, it is possible that altered ERβ action during cancer progression may be mediated in part through alteration of TGF-β-like signaling.[27,85,86] Regulation of individual genes and gene networks associated with proliferation, the cell cycle, and apoptosis is a common feature in estrogen-treated breast cancer models of ERβ overexpression with ERα.[32,33,87] Substantial overlap of ERα and ERβ 1 binding sites using genome-wide binding analyses, which are enriched for estrogen response element (ERE) motifs, has also been reported. However, ERα can displace ERβ 1 to different sites that are less enriched for ERE motifs when ERα and ERβ 1 are coexpressed and estrogen treated.[32,82,88] Interestingly, in the only study published so far where an originally ER-negative breast cancer cell line was engineered to stably overexpress either ERα or ERβ 1, it was concluded that each receptor mainly regulated unique gene sets.[84] As well in this same analysis, a model of ERβ 2/cx expression alone was found to have no effect on either proliferation or gene expression in breast cancer cells, suggesting this variant requires coexpression with either ERα or ERβ 1 to mediate activity.[84] There is an obvious need to develop and analyze more models of ERβ 1 expression alone using both global expression profiling and genome-wide binding analyses, and to compare results with those obtained in ERβ 1 and ERα coexpressing models. Equally and perhaps more important is to determine the relevance of the biomarkers of ERβ 1 functionality, either unique gene signatures and/or individual genes, in breast cancer in vivo. Furthermore, when established as relevant, correlative studies of these biomarkers of ERβ 1 functionality with ERβ 1 expression in large (well powered statistically) well-annotated breast cancer cohorts with long clinical follow-up using standard protocols and cut points should be undertaken, perhaps using a international collaborative network approach.[89]

Summary and Conclusions

Although ERα remains today the primary target for endocrine therapies in breast cancer and seems to be the most critical in terms of development of the normal mammary gland, the widespread expression of ERβ-like proteins in both normal and neoplastic mammary tissues suggest that ERβ has a role in the breast. Little progress has been made in definitively elucidating this role. The presence of two ERs and their variant isoforms presents obvious challenges but also opportunities to tease out complexities in terms of understanding the estrogen signaling pathway in both normal and neoplastic breast tissues. The availability of more accurate tools, such as specific antibodies and the identification of downstream markers of functionality, will be significant in advancing knowledge in this field as well as determining relevance in human breast cancer in vivo.

The recognition of two distinct ERβ-expressing types of breast tumors in vivo and the possibility of differential function has already raised expectations that targeting ERβ in some cases may offer new treatment options for groups of breast cancer patients where previously only aggressive chemotherapies were available. If nothing else, this supports the continued efforts to understand the nature and function of ERβ in breast cancer, but it also suggests that ER status may need to be redefined to include an assessment of ERβ isoforms in addition to ERα.

 

Acknowledgments ӬThis work was supported by the Canadian Institutes of Health Research (CIHR), the CancerCare Manitoba Foundation (CCMF), the Canadian Breast Cancer Foundation (CBCF), and the Canadian Breast Cancer Research Alliance (CBCRA).

Semin Reprod Med. 2012;30(1):5-13. © 2012 Thieme Medical Publishers

 

References

Speirs V, Skliris G, Burdall S, Carder P. Distinct expression patterns of ERa and ERb in normal human mammary gland. J Clin Pathol 2002;255;371-374

Clarke RB, Howell A, Potten CS, Anderson E. Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Res 1997;57(22):4987-4991

Couse JF, Korach KS. Estrogen receptor null mice: what have we learned and where will they lead us? Endocr Rev 1999;20(3):358-417

Förster C, Mäkela S, Wärri A, et al. Involvement of estrogen receptor beta in terminal differentiation of mammary gland epithelium. Proc Natl Acad Sci U S A 2002;99(24):15578-15583

Antal MC, Krust A, Chambon P, Mark M. Sterility and absence of histopathological defects in nonreproductive organs of amouse ERbeta-null mutant. Proc Natl Acad Sci U S A 2008;105(7):2433-2438

Nilsson S, Mäkelä S, Treuter E, et al. Mechanisms of estrogen action. Physiol Rev 2001;81(4):1535-1565

Dotzlaw H, Leygue E, Watson PH, Murphy LC. Expression of estrogen receptor-beta in human breast tumors. J Clin Endocrinol Metab 1997;82(7):2371-2374

Enmark E, Pelto-Huikko M, Grandien K, et al. Human estrogen receptor β-gene structure, chromosomal localization, and expression pattern. J Clin Endocrinol Metab 1997;82(12):4258-4265

Fuqua SA, Schiff R, Parra I, et al. Expression of wild-type estrogen receptor beta and variant isoforms in human breast cancer. Cancer Res 1999;59(21):5425-5428

Järvinen TA, Pelto-Huikko M, Holli K, Isola J. Estrogen receptor beta is coexpressed with ERalpha and PR and associated with nodal status, grade, and proliferation rate in breast cancer. Am J Pathol 2000;156(1):29-35

Fox EM, Davis RJ, ShupnikMA. ERbeta in breast cancer””onlooker, passive player, or active protector? Steroids 2008;73(11):1039-1051

Skliris GP, Leygue E, Curtis-Snell L, Watson PH, Murphy LC. Expression of oestrogen receptor-beta in oestrogen receptoralpha negative human breast tumours. Br J Cancer 2006;95(5):616-626

Monroe DG, Secreto FJ, Subramaniam M, Getz BJ, Khosla S, Spelsberg TC. Estrogen receptor alpha and beta heterodimers exert unique effects on estrogen- and tamoxifen-dependent gene expression in human U2OS osteosarcoma cells. Mol Endocrinol 2005;19(6):1555-1568

Weitsman GE, Skliris G, Ung K, et al. Assessment of multiple different estrogen receptor-beta antibodies for their ability to immunoprecipitate under chromatin immunoprecipitation conditions. Breast Cancer Res Treat 2006;100(1):23-31

Murphy LC,Watson PH. Is oestrogen receptor-beta a predictor of endocrine therapy responsiveness in human breast cancer? Endocr Relat Cancer 2006;13(2):327-334

Chi A, Chen X, Chirala M, Younes M. Differential expression of estrogen receptor beta isoforms in human breast cancer tissue. Anticancer Res 2003;23(1A):211-216

Saji S, Omoto Y, Shimizu C, et al. Expression of estrogen receptor (ER) (beta)cx protein in ER(alpha)-positive breast cancer: specific correlation with progesterone receptor. Cancer Res 2002;62(17):4849-4853

Shaaban AM, Green AR, Karthik S, et al. Nuclear and cytoplasmic expression of ERbeta1, ERbeta2, and ERbeta5 identifies distinct prognostic outcome for breast cancer patients. Clin Cancer Res 2008;14(16):5228-5235

Yan M, Rayoo M, Takano EA, Fox SB; kConFab Investigators. Nuclear and cytoplasmic expressions of ERβ1 and ERβ2 are predictive of response to therapy and alters prognosis in familial breast cancers. Breast Cancer Res Treat 2011;126(2):395-405

Moore JT, McKee DD, Slentz-Kesler K, et al. Cloning and characterization of human estrogen receptor beta isoforms. Biochem Biophys Res Commun 1998;247(1):75-78

Savinov AY, Remacle AG, Golubkov VS, et al. Matrix metalloproteinase 26 proteolysis of the NH2-terminal domain of the estrogen receptor beta correlates with the survival of breast cancer patients. Cancer Res 2006;66(5):2716-2724

Sanchez M, Picard N, Sauvé K, Tremblay A. Challenging estrogen receptor beta with phosphorylation. Trends Endocrinol Metab 2010;21(2):104-110

Hamilton-Burke W, Coleman L, Cummings M, et al. Phosphorylation of estrogen receptor beta at serine 105 is associated with good prognosis in breast cancer. Am J Pathol 2010;177(3):1079-1086

Roger P, Sahla ME, Mäkelä S, Gustafsson JA, Baldet P, Rochefort H. Decreased expression of estrogen receptor beta protein in proliferative preinvasive mammary tumors. Cancer Res 2001;61(6):2537-2541

Rutherford T, Brown WD, Sapi E, Aschkenazi S, Muñoz A, Mor G. Absence of estrogen receptor-beta expression in metastatic ovarian cancer. Obstet Gynecol 2000;96(3):417-421

Foley EF, Jazaeri AA, ShupnikMA, Jazaeri O, Rice LW. Selective loss of estrogen receptor beta in malignant human colon. Cancer Res 2000;60(2):245-248

Leav I, Lau KM, Adams JY, et al. Comparative studies of the estrogen receptors beta and alpha and the androgen receptor in normal human prostate glands, dysplasia, and in primary and metastatic carcinoma. Am J Pathol 2001;159(1):79-92

Zhu X, Leav I, Leung YK, et al. Dynamic regulation of estrogen receptor-beta expression by DNA methylation during prostate cancer development and metastasis. Am J Pathol 2004;164(6):2003-2012

Al-Nakhle H, Burns PA, Cummings M, et al. Estrogen receptor beta1 expression is regulated by miR-92 in breast cancer. Cancer Res 2010;70(11):4778-4784

Smith L, Brannan RA, Hanby AM, et al. Differential regulation of estrogen receptor beta isoforms by 5′ untranslated regions in cancer. J Cell Mol Med 2009; July 28 (Epub ahead of print)

Murphy LC, Peng B, Lewis A, et al. Inducible upregulation of oestrogen receptor-beta1 affects oestrogen and tamoxifen responsiveness in MCF7 human breast cancer cells. J Mol Endocrinol 2005;34(2):553-566

Zhao C, Dahlman-Wright K, Gustafsson JA. Estrogen signaling via estrogen receptor beta. J Biol Chem 2010;285(51):39575-39579

Chang EC, Frasor J, Komm B, Katzenellenbogen BS. Impact of estrogen receptor beta on gene networks regulated by estrogen receptor alpha in breast cancer cells. Endocrinology 2006;147(10):4831-4842

Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL, Leitman DC. Estrogen receptor beta inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest. Cancer Res 2004;64(1):423-428

Lazennec G, Bresson D, Lucas A, Chauveau C, Vignon F. ER beta inhibits proliferation and invasion of breast cancer cells. Endocrinology 2001;142(9):4120-4130

Tonetti DA, Rubenstein R, DeLeon M, et al. Stable transfection of an estrogen receptor beta cDNA isoform into MDA-MB-231 breast cancer cells. J Steroid Biochem Mol Biol 2003;87(1):47-55

Hou YF, Yuan ST, Li HC, et al. ERbeta exerts multiple stimulative effects on human breast carcinoma cells. Oncogene 2004;23 (34):5799-5806

Warner M, Gustafsson JA. The role of estrogen receptor beta (ERbeta) in malignant diseases””a new potential target for anti-proliferative drugs in prevention and treatment of cancer. Biochem Biophys Res Commun 2010;396(1):63-66

Rousseau C, Nichol JN, Pettersson F, Couture MC, Miller WH Jr. ERbeta sensitizes breast cancer cells to retinoic acid: evidence of transcriptional crosstalk. Mol Cancer Res 2004;2(9):523-531

Helguero LA, Faulds MH, Gustafsson JA, Haldosén LA. Estrogen receptors alfa (ERalpha) and beta (ERbeta) differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11. Oncogene 2005;24(44):6605-6616

Cheng G,Weihua Z,WarnerM, Gustafsson JA. Estrogen receptors ER alpha and ER beta in proliferation in the rodent mammary gland. Proc Natl Acad Sci U S A 2004;101(11):3739-3746

Skliris GP, Lewis A, Emberley E, et al. Estrogen receptor-beta regulates psoriasin (S100A7) in human breast cancer. Breast Cancer Res Treat 2007;104(1):75-85

O’Neill PA, Davies MP, Shaaban AM, et al. Wild-type oestrogen receptor beta (ERbeta1) mRNA and protein expression in Tamoxifen-treated post-menopausal breast cancers. Br J Cancer 2004;91(9):1694-1702

Leung YK, Mak P, Hassan S, Ho SM. Estrogen receptor (ER)-beta isoforms: a key to understanding ER-beta signaling. Proc Natl Acad Sci U S A 2006;103(35):13162-13167

Peng B, Lu B, Leygue E, Murphy LC. Putative functional characteristics of human estrogen receptor-beta isoforms. JMol Endocrinol 2003;30(1):13-29

Inoue S, Ogawa S, Horie K, et al. An estrogen receptor beta isoform that lacks exon 5 has dominant negative activity on both ERalpha and ERbeta. Biochem Biophys Res Commun 2000;279(3):814-819

Zhao C, Matthews J, Tujague M, et al. Estrogen receptor beta2 negatively regulates the transactivation of estrogen receptor alpha in human breast cancer cells. Cancer Res 2007;67(8):3955-3962

Tateishi Y, Sonoo R, Sekiya Y, et al. Turning off estrogen receptor beta-mediated transcription requires estrogen-dependent receptor proteolysis. Mol Cell Biol 2006;26(21):7966-7976

Bhat RA, Harnish DC, Stevis PE, Lyttle CR, Komm BS. A novel human estrogen receptor beta: identification and functional analysis of additional N-terminal amino acids. J Steroid Biochem Mol Biol 1998;67(3):233-240

Gilmore TD, Herscovitch M. Inhibitors of NF-kappaB signaling: 785 and counting. Oncogene 2006;25(51):6887-6899

Cvoro A, Tatomer D, Tee MK, Zogovic T, Harris HA, Leitman DC. Selective estrogen receptor-beta agonists repress transcription of proinflammatory genes. J Immunol 2008;180(1):630-636

Harris HA, Albert LM, Leathurby Y, et al. Evaluation of an estrogen receptor-beta agonist in animal models of human disease. Endocrinology 2003;144(10):4241-4249

Skliris GP, Munot K, Bell SM, et al. Reduced expression of oestrogen receptor beta in invasive breast cancer and its re-expression using DNA methyl transferase inhibitors in a cell line model. J Pathol 2003;201(2):213-220

Esslimani-Sahla M, Kramar A, Simony-Lafontaine J, Warner M, Gustafsson JA, Rochefort H. Increased estrogen receptor betacx expression during mammary carcinogenesis. Clin Cancer Res 2005;11(9):3170-3174

Omoto Y, Kobayashi S, Inoue S, et al. Evaluation of oestrogen receptor beta wild-type and variant protein expression, and relationship with clinicopathological factors in breast cancers. Eur J Cancer 2002;38(3):380-386

Leygue E, Dotzlaw H, Watson PH, Murphy LC. Expression of estrogen receptor beta1, beta2, and beta5 messenger RNAs in human breast tissue. Cancer Res 1999;59(6):1175-1179

Leygue E, Dotzlaw H, Watson PH, Murphy LC. Altered estrogen receptor alpha and beta messenger RNA expression during human breast tumorigenesis. Cancer Res 1998;58(15):3197-3201

Brett D, Pospisil H, Valcárcel J, Reich J, Bork P. Alternative splicing and genome complexity. Nat Genet 2002;30(1):29-30

Lemischka IR, Pritsker M. Alternative splicing increases complexity of stem cell transcriptome. Cell Cycle 2006;5(4):347-351

Modrek B, Lee C. A genomic view of alternative splicing. Nat Genet 2002;30(1):13-19

Roberts GC, Smith CW. Alternative splicing: combinatorial output from the genome. Curr Opin Chem Biol 2002;6(3):375-383

Wessagowit V, Nalla VK, Rogan PK, McGrath JA. Normal and abnormalmechanisms of gene splicing and relevance to inherited skin diseases. J Dermatol Sci 2005;40(2):73-84

Gallo JM, Jin P, Thornton CA, et al. The role of RNA and RNA processing in neurodegeneration. J Neurosci 2005;25(45):10372-10375

Lee CJ, Irizarry K. Alternative splicing in the nervous system: an emerging source of diversity and regulation. Biol Psychiatry 2003;54(8):771-776

Hall PA, Russell SH. New perspectives on neoplasia and the RNA world. Hematol Oncol 2005;23(2):49-53

Kalnina Z, Zayakin P, Silina K, Linē A. Alterations of pre-mRNA splicing in cancer. Genes Chromosomes Cancer 2005;42(4):342-357

CummingsM, Iremonger J, Green CA, Shaaban AM, Speirs V. Gene expression of ERbeta isoforms in laser microdissected human breast cancers: implications for gene expression analyses. Cell Oncol 2009;31(6):467-473

Honma N, Horii R, Iwase T, et al. Clinical importance of estrogen receptor-beta evaluation in breast cancer patients treated with adjuvant tamoxifen therapy. J Clin Oncol 2008;26(22):3727-3734

Gruvberger-Saal SK, Bendahl PO, Saal LH, et al. Estrogen receptor beta expression is associated with tamoxifen response in ERalpha-negative breast carcinoma. Clin Cancer Res 2007;13(7):1987-1994

Palmieri C, Lam EW, Mansi J, et al. The expression of ER beta cx in human breast cancer and the relationship to endocrine therapy and survival. Clin Cancer Res 2004;10(7):2421-2428

Miller WR, Anderson TJ, Dixon JM, Saunders PT. Oestrogen receptor beta and neoadjuvant therapy with tamoxifen: prediction of response and effects of treatment. Br J Cancer 2006;94(9):1333-1338

Sugiura H, Toyama T, Hara Y, et al. Expression of estrogen receptor beta wild-type and its variant ERbetacx/beta2 is correlated with better prognosis in breast cancer. Jpn J Clin Oncol 2007;37(11):820-828

Esslimani-SahlaM, Simony-Lafontaine J, Kramar A, et al. Estrogen receptor beta (ER beta) level but not its ER beta cx variant helps to predict tamoxifen resistance in breast cancer. Clin Cancer Res 2004;10(17):5769-5776

Yamashita H, Takahashi S, Ito Y, et al. Predictors of response to exemestane as primary endocrine therapy in estrogen receptor-positive breast cancer. Cancer Sci 2009;100(11):2028-2033

Motomura K, Ishitobi M, Komoike Y, et al. Expression of estrogen receptor beta and phosphorylation of estrogen receptor alpha serine 167 correlate with progression-free survival in patients with metastatic breast cancer treated with aromatase inhibitors. Oncology 2010;79(1-2):55-61

Novelli F, Milella M, Melucci E, et al. A divergent role for estrogen receptor-beta in node-positive and node-negative breast cancer classified according to molecular subtypes: an observational prospective study. Breast Cancer Res 2008;10(5):R74

Perou CM, Sørlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000;406(6797):747-752

Wu X, Subramaniam M, Grygo SB, et al. Estrogen receptor-beta sensitizes breast cancer cells to the anti-estrogenic actions of endoxifen. Breast Cancer Res 2011;13(2):R27

Jensen EV, Cheng G, Palmieri C, et al. Estrogen receptors and proliferation markers in primary and recurrent breast cancer. Proc Natl Acad Sci U S A 2001;98(26):15197-15202

Bardou VJ, Arpino G, Elledge RM, Osborne CK, Clark GM. Progesterone receptor status significantly improves outcome prediction over estrogen receptor status alone for adjuvant endocrine therapy in two large breast cancer databases. J Clin Oncol 2003;21(10):1973-1979

Foekens JA, van Putten WL, Portengen H, et al. Prognostic value of PS2 and cathepsin D in 710 human primary breast tumors: multivariate analysis. J Clin Oncol 1993;11(5):899-908

Zhao C, Gao H, Liu Y, et al. Genome-wide mapping of estrogen receptor-beta-binding regions reveals extensive cross-talk with transcription factor activator protein-1. Cancer Res 2010;70(12):5174-5183

Grober OM, Mutarelli M, Giurato G, et al. Global analysis of estrogen receptor beta binding to breast cancer cell genome reveals an extensive interplay with estrogen receptor alpha for target gene regulation. BMC Genomics 2011;12;36

Secreto FJ, Monroe DG, Dutta S, Ingle JN, Spelsberg TC. Estrogen receptor alpha/beta isoforms, but not betacx, modulate unique patterns of gene expression and cell proliferation in Hs578T cells. J Cell Biochem 2007;101(5):1125-1147

Bierie B, Moses HL. Gain or loss of TGFbeta signaling in mammary carcinoma cells can promote metastasis. Cell Cycle 2009;8(20):3319-3327

Horvath LG, Henshall SM, Lee C-S, et al. Frequent loss of estrogen receptor-beta expression in prostate cancer. Cancer Res 2001;61(14):5331-5335

Williams C, Edvardsson K, Lewandowski SA, Ström A, Gustafsson JA. A genome-wide study of the repressive effects of estrogen receptor beta on estrogen receptor alpha signaling in breast cancer cells. Oncogene 2008;27(7):1019-1032

Charn TH, Liu ET, Chang EC, Lee YK, Katzenellenbogen JA, Katzenellenbogen BS. Genome-wide dynamics of chromatin binding of estrogen receptors alpha and beta: mutual restriction and competitive site selection. Mol Endocrinol 2010;24(1):47-59

Carder PJ, Murphy CE, Dervan P, et al. A multi-centre investigation towards reaching a consensus on the immunohistochemical detection of ERbeta in archival formalin-fixed paraffin embedded human breast tissue. Breast Cancer Res Treat 2005;92(3):287-293

Mann S, Laucirica R, Carlson N, et al. Estrogen receptor beta expression in invasive breast cancer. Hum Pathol 2001;32(1):113-118

Murphy LC, Leygue E, Niu Y, Snell L, Ho S-M, Watson PH. Relationship of coregulator and oestrogen receptor isoform expression to de novo tamoxifen resistance in human breast cancer. Br J Cancer 2002;87(12):1411-1416

Omoto Y, Inoue S, Ogawa S, et al. Clinical value of the wild-type estrogen receptor beta expression in breast cancer. Cancer Lett 2001;163(2):207-212

Iwase H, Zhang Z, Omoto Y, et al. Clinical significance of the expression of estrogen receptors alpha and beta for endocrine therapy of breast cancer. Cancer Chemother Pharmacol 2003;52(Suppl 1):S34-S38

Hopp TA, Weiss HL, Parra IS, Cui Y, Osborne CK, Fuqua SA. Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer. Clin Cancer Res 2004;10(22):7490-7499

Fleming FJ, Hill AD, McDermott EW, O’Higgins NJ, Young LS. Differential recruitment of coregulator proteins steroid receptor coactivator-1 and silencing mediator for retinoid and thyroid receptors to the estrogen receptor-estrogen response element by beta-estradiol and 4-hydroxytamoxifen in human breast cancer. J Clin Endocrinol Metab 2004;89(1):375-383

Myers E, Fleming FJ, Crotty TB, et al. Inverse relationship between ER-beta and SRC-1 predicts outcome in endocrine-resistant breast cancer. Br J Cancer 2004;91(9):1687-1693

Nakopoulou L, Lazaris AC, Panayotopoulou EG, et al. The favourable prognostic value of oestrogen receptor beta immune-histochemical expression in breast cancer. J Clin Pathol 2004;57(5):523-528

Saji S, Omoto Y, Shimizu C, et al. Clinical impact of assay of estrogen receptor beta cx in breast cancer. Breast Cancer 2002;9(4):303-307

Vinayagam R, Sibson DR, Holcombe C, Aachi V, Davies MP. Association of oestrogen receptor beta 2 (ER beta 2/ER beta cx) with outcome of adjuvant endocrine treatment for primary breast cancer””a retrospective study. BMC Cancer 2007;7;131

Maehle BO, Collett K, Tretli S, Akslen LA, Grotmol T. Estrogen receptor beta””an independent prognostic marker in estrogen receptor alpha and progesterone receptor-positive breast cancer? APMIS 2009;117(9):644-650

1 thought on “Estrogen receptor Beta – an introduction”

  1. Pingback: WeeksMD » Soy is helpful for cancer. (hint: ERbeta)

Leave a Comment

Your email address will not be published. Required fields are marked *