Somatic Mutation of P53 Leads to Estrogen Receptor Alpha-Positive

Total Page:16

File Type:pdf, Size:1020Kb

Somatic Mutation of P53 Leads to Estrogen Receptor Alpha-Positive [CANCER RESEARCH 64, 3525–3532, May 15, 2004] Somatic Mutation of p53 Leads to Estrogen Receptor ␣-Positive and -Negative Mouse Mammary Tumors with High Frequency of Metastasis Suh-Chin J. Lin,1,2 Kuo-Fen Lee,4 Alexander Yu. Nikitin,5 Susan G. Hilsenbeck,6 Robert D. Cardiff,7 Aihua Li,1,2 Keon-Wook Kang,1,2 Steven A. Frank,3 Wen-Hwa Lee,2 and Eva Y-H. P. Lee1,2 1Departments of Developmental and Cell Biology, 2Biological Chemistry, and 3Ecology and Evolutionary Biology, University of California, Irvine, California; 4The Salk Institute for Biological Studies, La Jolla, California; 5Department of Biomedical Sciences, Cornell University, Ithaca, New York; 6Department of Medicine and Department of Molecular and Cellular Biology, Breast Center, Baylor College of Medicine, Houston, Texas; and 7Center for Comparative Medicine, University of California, Davis, California ABSTRACT tion of p53 in mouse mammary epithelial cells is necessary to generate a mouse model mimicking human carcinogenesis. ␣ Approximately 70% of human breast cancers are estrogen receptor In addition to genetic mutations, steroid hormones play a critical role (ER␣)-positive, but the origins of ER␣-positive and -negative tumors remain in breast carcinogenesis (13). About 70% of human breast cancers are unclear. Hormonal regulation of mammary gland development in mice is ␣ ␣ similar to that in humans; however, most mouse models produce only ER␣- estrogen receptor (ER )-positive and estrogen-dependent (14), and ␣ negative tumors. In addition, these mouse tumors metastasize at a low rate ER and progesterone receptor (PR) expression is an important indicator relative to human breast tumors. We report here that somatic mutations of of potential responses to hormonal therapy (15). However, the factors that p53 in mouse mammary epithelial cells using the Cre/loxP system leads to control ER␣ expression in tumor cells are unknown. Thus far, most ER␣-positive and -negative tumors. p53 inactivation under a constitutive established mouse models seldom produce ER␣-positive mammary tu- active WAPCrec in prepubertal/pubertal mice, but not under MMTVCre in mors (16). In a C3(1)/SV40 T-antigen-transgenic model, ER␣ expression adult mice, leads to the development of ER␣-positive tumors, suggesting that decreases during early mammary tumor progression (from low- to high- target cells or developmental stages can determine ER␣ status in mammary grade mammary intraepithelial neoplasia and becomes undetectable in tumors. Importantly, these tumors have a high rate of metastasis. An inverse invasive tumors (17). In Brca1- and Brca2-linked mammary tumors, the relationship between the number of targeted cells and median tumor latency majority of tumors show no detectable ER␣ expression (18–20). was also observed. Median tumor latency reaches a plateau when targeted cell numbers exceed 20%, implying the existence of saturation kinetics for In humans, breast cancers frequently metastasize to other organs such breast carcinogenesis. Genetic alterations commonly observed in human as liver, lung, and specifically bone (21). Metastasis rather than primary breast cancer including c-myc amplification and Her2/Neu/erbB2 activation tumors are responsible for most cancer mortality (21, 22). Less than 5% were seen in these mouse tumors. Thus, this tumor system reproduces many of patients with metastatic breast cancer have a long-term remission after important features of human breast cancer and provides tools for the study treatment (22). In established mouse mammary tumor models, the tumor of the origins of ER␣-positive and -negative breast tumors in mice. cells infrequently colonize other organs. Only 10% Brca1 tumors (18), 10% ptenϩ/Ϫ tumors (23), and 0% Brca2 tumors (12, 20) metastasize. Many mouse mammary tumor virus (MMTV)/oncogene-bearing trans- INTRODUCTION genic mice have a rare occurrence of metastasis (24), but metastatic Breast carcinogenesis requires multiple genetic changes including in- tumors are observed in polyomavirus middle T antigen and neu proto- activation of tumor suppressor genes and activation of oncogenes (1). oncogene transgenic mice (25, 26). Mutations in p53 are observed in close to half of human cancers including We have generated a mouse breast tumor model by using Cre/loxp breast carcinomas (2). p53 is also mutated in families with Li-Fraumeni method to specifically inactivate p53 in mammary epithelial cells. This ␣ syndrome in which early-onset female breast cancer is the most prevalent conditional inactivation of p53 leads to ER -positive and -negative type of tumor (3). p53 is a transcription factor that regulates genes critical mammary tumors with a high rate of metastasis. We found that p53 for cell cycle arrest and for apoptosis after genotoxic stress, thus prevent- inactivation during specific developmental stages critically determines ␣ ing genome instability (4–6). In addition, amplification and/or overex- ER expression in mammary tumors. This breast tumor system provides pression of c-myc, Her2/Neu/erbB2, and cyclin D1 oncogenes are seen in a close model of the human disease and will be useful for both mecha- ␣ a significant portion of breast cancers (2, 7). nistic and therapeutic studies of ER -positive breast cancer. p53 knockout mice are cancer prone and develop early-onset lym- phoma and sarcoma (8, 9) but rarely mammary tumors (10) because of MATERIALS AND METHODS early mortality. To circumvent this problem, p53null mammary epithe- lium is transplanted into the fat pad of wild-type recipients and leads to Targeting Vector Construction. A 10.75-kb clone covering exons 1–10 of the formation of breast tumors (11). Although this offers a potential p53 was isolated from a 129sv mouse genomic library. The 3.4-kb XhoI- model to study breast tumors, the influence of the transplantation process HindIII fragment containing exons 2–9 was subcloned into pBR322. A re- on carcinogenesis is not clear. Also, the transplanted cells are p53null, placement-type targeting vector was made by inserting the first loxP site into whereas in human tumors somatic mutations are acquired in a subset of a BamHI site located in intron 6. A neo-cassette flanked by two loxP sites was cells during tumor progression. Previously, Jonkers et al. (12) reported inserted into the PvuII site in intron 4. The resulting construct was cleaved with that no mammary tumor formation was observed in p53 conditional- XhoI and HindIII, blunt-ended and subcloned into p2TK (27). The finished targeting construct is designated p53neoloxp32TK. mutant mice carrying K14Cre transgene. Therefore, conditional inactiva- MMTVCre Transgene Construction. The backbone of the MMTVCre transgene is pBSpKCR3 (28) containing part of the rabbit ␤-globin gene (the Received 11/10/03; revised 1/28/04; accepted 3/2/04. end of exon 2, intron 2, and exon 3), and the polyadenylation site. A 1.6-kb Grant support: National Cancer Institute mouse consortium Grant CA04964, DOD BglII-HindIII fragment composed of the Cre transgene with a nuclear local- BC013020, NIH CA84241, Breast Cancer Research Foundation to E. Lee and NCIP30 CA93373 to R. Cardiff. ization signal was inserted within exon 3 of the globin gene of pBSpKCR3.A The costs of publication of this article were defrayed in part by the payment of page 1.5-kb HindIII-NheI fragment containing the MMTV-LTR was cut from pMAM charges. This article must therefore be hereby marked advertisement in accordance with (Clontech, Palo Alto, California) and subcloned 5Ј to exon 2 of the globin 18 U.S.C. Section 1734 solely to indicate this fact. gene. The finished transgene construct is designated pMMTV Cre. Requests for reprints: Eva Y-H. P. Lee, University of California-Irvine, Sprague ␤ Hall, Room 140, 839 Health Science Court, Irvine, CA 92697-4037. Phone: (949) 824- MMTVCre-Transgenic Mice Production. The 4.3-kb XhoI fragment 9766; Fax: (949) 824-9767; E-mail: [email protected]. containing the MMTVCre transgene was excised and purified. The transgenic 3525 ER␣ AND METASTASIS IN p53-MEDIATED BREAST CANCER MODELS founders were generated by microinjecting the MMTVCre transgene fragment of exons 5 and 6 led to an in-frame deletion of 99 codons (codons into the male pronucleus of fertilized eggs derived from CB6F1 ϫ C57BL/6 123–221) that encode part of the DNA-binding domain. The presence intercrosses. Transgenic founders were identified by Southern analysis or PCR of loxP sites in introns 4 and 6 did not interfere with the transcription of tail DNA. The primers for the Cre transgene (363-bp amplified) were CreF of p53, and the full-length p53 protein was detected in p53fp/fp mouse Ј Ј Ј (5 -GGTGTCCAATTTACTGACCGTACA-3 ) and CreR (5 -CGGATCCGC- embryonic fibroblasts in the absence of Cre recombinase (Fig. 1C). A CGCATAACCAGTG-3Ј). All mice were maintained in accordance with the smaller protein product of expected mass of 39 kDa, designated guidelines of Laboratory Animal Research of The University of Texas Health ⌬5,6 fp/fp Science Center at San Antonio and Institutional Animal Care and Use Com- p53 , was detected in p53 mouse embryonic fibroblasts infected mittee of University of California, Irvine. with Cre adenoviruses (Fig. 1C). IR-induced responses demonstrate ⌬5,6 Generation of Conditional p53 Mutant Mice. J1 embryonic stem cells that p53 is transcriptionally inactive and fails to increase p21 ⌬5,6 were electroporated with SalI-linearized p53neoloxp32TK and selected with G418 target gene transcription. In contrast to wild-type p53, mutant p53 and 1-(2-deoxy-2-fluoro-␤-D-arabinofuranosyl)-5-iodouracil. Embryonic stem protein was not stabilized after IR (Fig. 1D). No p53 protein was cells harboring homologous recombination were identified by Southern blotting detected from mouse embryonic fibroblasts derived from p53-null using a 3Ј probe external to the targeting region. The neo-cassette was removed (p53Ϫ/Ϫ) mice (Fig. 1, C and D). from targeted embryonic stem cells by transient expression of pSPCre.Of231 Characterization of Cre-Transgenic Mice. To introduce somatic clones analyzed by Southern blotting, two contained a recombination that removed p53 mutation in the mammary gland, different lines of mice express- only neo.
Recommended publications
  • CDC25B Mediates Rapamycin-Induced Oncogenic Responses in Cancer Cells
    Published OnlineFirst March 10, 2009; DOI: 10.1158/0008-5472.CAN-08-3222 Research Article CDC25B Mediates Rapamycin-Induced Oncogenic Responses in Cancer Cells Run-qiang Chen,1 Qing-kai Yang,1 Bing-wen Lu,2 Wei Yi,1 Greg Cantin,2 Yan-ling Chen,1 Colleen Fearns,3 John R. Yates III,2 and Jiing-Dwan Lee1 Departments of 1Immunology and Microbial Science, 2Chemical Physiology, and 3Chemistry, The Scripps Research Institute, La Jolla, California Abstract expression of PTEN, increased PI3K activity, and increased expression or activation of AKT in advanced prostate cancer Because the mammalian target of rapamycin (mTOR) pathway (8–10). These aberrations also are indicators of a poor prognosis is commonly deregulated in human cancer, mTOR inhibitors, for prostate cancer patients (11, 12). More importantly, long-term rapamycin and its derivatives, are being actively tested in androgen deprivation treatment for prostate cancer patients that cancer clinical trials. Clinical updates indicate that the reinforces the PI3K/AKT pathway also up-regulates mTOR anticancer effect of these drugs is limited, perhaps due to activation in prostate tumor (9, 10). These abovementioned rapamycin-dependent induction of oncogenic cascades by an experimental and clinical data lead to the supposition that mTOR as yet unclear mechanism. As such, we investigated rapamy- inhibitors (rapamycin and its derivatives) should be effective in cin-dependent phosphoproteomics and discovered that 250 treating human cancer. Unfortunately, recent clinical data indicates phosphosites in 161 cellular proteins were sensitive to that rapamycin shows therapeutic potential in only few types of rapamycin. Among these, rapamycin regulated four kinases human cancer: endometrial carcinoma, renal cell carcinoma, and and four phosphatases.
    [Show full text]
  • Analysis of Estrogen Receptor Isoforms and Variants in Breast Cancer Cell Lines
    EXPERIMENTAL AND THERAPEUTIC MeDICINE 2: 537-544, 2011 Analysis of estrogen receptor isoforms and variants in breast cancer cell lines MAIE AL-BADER1, CHRISTOPHER FORD2, BUSHRA AL-AYADHY3 and ISSAM FRANCIS3 Departments of 1Physiology, 2Surgery, and 3Pathology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait Received November 22, 2010; Accepted February 14, 2011 DOI: 10.3892/etm.2011.226 Abstract. In the present study, the expression of estrogen domain C, the DNA binding domain; domains D/E, bearing receptor (ER)α and ERβ isoforms in ER-positive (MCF7, both the activation function-2 (AF-2) and the ligand binding T-47D and ZR-75-1) and ER-negative (MDA-MB-231, SK-BR-3, domains; and finally, domain F, the C-terminal domain (6,7). MDA-MB-453 and HCC1954) breast cancer cell lines was The actions of estrogens are mediated by binding to ERs investigated. ERα mRNA was expressed in ER-positive and (ERα and/or ERβ). These receptors, which are co-expressed some ER-negative cell lines. ERα ∆3, ∆5 and ∆7 spliced in a number of tissues, form functional homodimers or variants were present in MCF7 and T-47D cells; ERα ∆5 heterodimers. When bound to estrogens as homodimers, the and ∆7 spliced variants were detected in ZR-75-1 cells. transcription of target genes is activated (8,9), while as heterodi- MDA-MB-231 and HCC1954 cells expressed ERα ∆5 and ∆7 mers, ERβ exhibits an inhibitory action on ERα-mediated gene spliced variants. The ERβ1 variant was expressed in all of the expression and, in many instances, opposes the actions of ERα cell lines and the ERβ2 variant in all of the ER-positive and (7,9).
    [Show full text]
  • Expression of Aromatase, Estrogen Receptor and , Androgen
    0031-3998/06/6006-0740 PEDIATRIC RESEARCH Vol. 60, No. 6, 2006 Copyright © 2006 International Pediatric Research Foundation, Inc. Printed in U.S.A. Expression of Aromatase, Estrogen Receptor ␣ and ␤, Androgen Receptor, and Cytochrome P-450scc in the Human Early Prepubertal Testis ESPERANZA B. BERENSZTEIN, MARI´A SONIA BAQUEDANO, CANDELA R. GONZALEZ, NORA I. SARACO, JORGE RODRIGUEZ, ROBERTO PONZIO, MARCO A. RIVAROLA, AND ALICIA BELGOROSKY Research Laboratory [E.B.B., M.S.B., C.R.G., N.I.S., J.R., M.A.R., A.B.], Hospital de Pediatria Garrahan, Buenos Aires C124 5AAM, Argentina; Centro de Investigaciones en Reproduccion [R.P.], Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C112 1ABG, Argentina ABSTRACT: The expression of aromatase, estrogen receptor ␣ might affect adult testicular cell mass, as well as testicular (ER␣) and ␤ (ER␤), androgen receptor (AR), and cytochrome P-450 function (8). side chain cleavage enzyme (cP450scc) was studied in prepubertal In humans (9), there are three growth phases of LCs testis. Samples were divided in three age groups (GRs): GR1, during testicular development. Fetal LCs produce testoster- ϭ newborns (1- to 21-d-old neonates, n 5); GR2, postnatal activation one required for fetal masculinization and Insl-3, necessary ϭ stage (1- to 7-mo-old infants, n 6); GR3, childhood (12- to for testicular descent (10). They regress during the third ϭ ␣ 60-mo-old boys, n 4). Absent or very poor detection of ER by trimester of pregnancy. A second wave of infantile LCs has immunohistochemistry in all cells and by mRNA expression was been described during the postnatal surge of luteinizing observed.
    [Show full text]
  • Estrogen-Related Receptor Alpha: an Under-Appreciated Potential Target for the Treatment of Metabolic Diseases
    International Journal of Molecular Sciences Review Estrogen-Related Receptor Alpha: An Under-Appreciated Potential Target for the Treatment of Metabolic Diseases Madhulika Tripathi, Paul Michael Yen and Brijesh Kumar Singh * Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore; [email protected] (M.T.); [email protected] (P.M.Y.) * Correspondence: [email protected] Received: 7 February 2020; Accepted: 24 February 2020; Published: 28 February 2020 Abstract: The estrogen-related receptor alpha (ESRRA) is an orphan nuclear receptor (NR) that significantly influences cellular metabolism. ESRRA is predominantly expressed in metabolically-active tissues and regulates the transcription of metabolic genes, including those involved in mitochondrial turnover and autophagy. Although ESRRA activity is well-characterized in several types of cancer, recent reports suggest that it also has an important role in metabolic diseases. This minireview focuses on the regulation of cellular metabolism and function by ESRRA and its potential as a target for the treatment of metabolic disorders. Keywords: estrogen-related receptor alpha; mitophagy; mitochondrial turnover; metabolic diseases; non-alcoholic fatty liver disease (NAFLD); adipogenesis; adaptive thermogenesis 1. Introduction When the estrogen-related receptor alpha (ESRRA) was first cloned, it was found to be a nuclear receptor (NR) that had DNA sequence homology to the estrogen receptor alpha (ESR1) [1]. There are several examples of estrogen-related receptor (ESRR) and estrogen-signaling cross-talk via mutual transcriptional regulation or reciprocal binding to each other’s response elements of common target genes in a context-specific manner [2,3].
    [Show full text]
  • Bioinformatics-Based Screening of Key Genes for Transformation of Liver
    Jiang et al. J Transl Med (2020) 18:40 https://doi.org/10.1186/s12967-020-02229-8 Journal of Translational Medicine RESEARCH Open Access Bioinformatics-based screening of key genes for transformation of liver cirrhosis to hepatocellular carcinoma Chen Hao Jiang1,2, Xin Yuan1,2, Jiang Fen Li1,2, Yu Fang Xie1,2, An Zhi Zhang1,2, Xue Li Wang1,2, Lan Yang1,2, Chun Xia Liu1,2, Wei Hua Liang1,2, Li Juan Pang1,2, Hong Zou1,2, Xiao Bin Cui1,2, Xi Hua Shen1,2, Yan Qi1,2, Jin Fang Jiang1,2, Wen Yi Gu4, Feng Li1,2,3 and Jian Ming Hu1,2* Abstract Background: Hepatocellular carcinoma (HCC) is the most common type of liver tumour, and is closely related to liver cirrhosis. Previous studies have focussed on the pathogenesis of liver cirrhosis developing into HCC, but the molecular mechanism remains unclear. The aims of the present study were to identify key genes related to the transformation of cirrhosis into HCC, and explore the associated molecular mechanisms. Methods: GSE89377, GSE17548, GSE63898 and GSE54236 mRNA microarray datasets from Gene Expression Omni- bus (GEO) were analysed to obtain diferentially expressed genes (DEGs) between HCC and liver cirrhosis tissues, and network analysis of protein–protein interactions (PPIs) was carried out. String and Cytoscape were used to analyse modules and identify hub genes, Kaplan–Meier Plotter and Oncomine databases were used to explore relationships between hub genes and disease occurrence, development and prognosis of HCC, and the molecular mechanism of the main hub gene was probed using Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis.
    [Show full text]
  • The Regulatory Roles of Phosphatases in Cancer
    Oncogene (2014) 33, 939–953 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW The regulatory roles of phosphatases in cancer J Stebbing1, LC Lit1, H Zhang, RS Darrington, O Melaiu, B Rudraraju and G Giamas The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3--kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies. Oncogene (2014) 33, 939–953; doi:10.1038/onc.2013.80; published online 18 March 2013 Keywords: cancer; phosphatases; solid tumours GASTROINTESTINAL MALIGNANCIES abs in sera were significantly associated with poor survival in Oesophageal cancer advanced ESCC, suggesting that they may have a clinical utility in Loss of PTEN (phosphatase and tensin homologue deleted on ESCC screening and diagnosis.5 chromosome 10) expression in oesophageal cancer is frequent, Cao et al.6 investigated the role of protein tyrosine phosphatase, among other gene alterations characterizing this disease. Zhou non-receptor type 12 (PTPN12) in ESCC and showed that PTPN12 et al.1 found that overexpression of PTEN suppresses growth and protein expression is higher in normal para-cancerous tissues than induces apoptosis in oesophageal cancer cell lines, through in 20 ESCC tissues.
    [Show full text]
  • The Role of Constitutive Androstane Receptor and Estrogen Sulfotransferase in Energy Homeostasis
    THE ROLE OF CONSTITUTIVE ANDROSTANE RECEPTOR AND ESTROGEN SULFOTRANSFERASE IN ENERGY HOMEOSTASIS by Jie Gao Bachelor of Engineering, China Pharmaceutical University, 1999 Master of Science, China Pharmaceutical University, 2002 Submitted to the Graduate Faculty of School of Pharmacy in partial fulfillment of the requirements for the degree of Doctor of Philosophy University of Pittsburgh 2012 UNIVERSITY OF PITTSBURGH School of Pharmacy This dissertation was presented by Jie Gao It was defended on January 18, 2012 and approved by Billy W. Day, Professor, Pharmaceutical Sciences Donald B. DeFranco, Professor, Pharmacology & Chemical Biology Samuel M. Poloyac, Associate Professor, Pharmaceutical Sciences Song Li, Associate Professor, Pharmaceutical Sciences Dissertation Advisor: Wen Xie, Professor, Pharmaceutical Sciences ii Copyright © by Jie Gao 2012 iii THE ROLE OF CONSTITUTIVE ANDROSTANE RECEPTOR AND ESTROGEN SULFOTRANSFERASE IN ENERGY HOMEOSTASIS Jie Gao, PhD University of Pittsburgh, 2012 Obesity and type 2 diabetes are related metabolic disorders of high prevalence. The constitutive androstane receptor (CAR) was initially characterized as a xenobiotic receptor regulating the responses of mammals to xenotoxicants. In this study, I have uncovered an unexpected role of CAR in preventing obesity and alleviating type 2 diabetes. Activation of CAR prevented obesity and improved insulin sensitivity in both the HFD-induced type 2 diabetic model and the ob/ob mice. In contrast, CAR null mice maintained on a chow diet showed spontaneous insulin insensitivity. The metabolic benefits of CAR activation may have resulted from inhibition of hepatic lipogenesis and gluconeogenesis. The molecular mechanism through which CAR activation suppressed hepatic gluconeogenesis might be mediated via peroxisome proliferator- activated receptor gamma coactivator-1 alpha (PGC-1α).
    [Show full text]
  • Genetic Alterations of Protein Tyrosine Phosphatases in Human Cancers
    Oncogene (2015) 34, 3885–3894 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW Genetic alterations of protein tyrosine phosphatases in human cancers S Zhao1,2,3, D Sedwick3,4 and Z Wang2,3 Protein tyrosine phosphatases (PTPs) are enzymes that remove phosphate from tyrosine residues in proteins. Recent whole-exome sequencing of human cancer genomes reveals that many PTPs are frequently mutated in a variety of cancers. Among these mutated PTPs, PTP receptor T (PTPRT) appears to be the most frequently mutated PTP in human cancers. Beside PTPN11, which functions as an oncogene in leukemia, genetic and functional studies indicate that most of mutant PTPs are tumor suppressor genes. Identification of the substrates and corresponding kinases of the mutant PTPs may provide novel therapeutic targets for cancers harboring these mutant PTPs. Oncogene (2015) 34, 3885–3894; doi:10.1038/onc.2014.326; published online 29 September 2014 INTRODUCTION tyrosine/threonine-specific phosphatases. (4) Class IV PTPs include Protein tyrosine phosphorylation has a critical role in virtually all four Drosophila Eya homologs (Eya1, Eya2, Eya3 and Eya4), which human cellular processes that are involved in oncogenesis.1 can dephosphorylate both tyrosine and serine residues. Protein tyrosine phosphorylation is coordinately regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases 1 THE THREE-DIMENSIONAL STRUCTURE AND CATALYTIC (PTPs). Although PTKs add phosphate to tyrosine residues in MECHANISM OF PTPS proteins, PTPs remove it. Many PTKs are well-documented oncogenes.1 Recent cancer genomic studies provided compelling The three-dimensional structures of the catalytic domains of evidence that many PTPs function as tumor suppressor genes, classical PTPs (RPTPs and non-RPTPs) are extremely well because a majority of PTP mutations that have been identified in conserved.5 Even the catalytic domain structures of the dual- human cancers are loss-of-function mutations.
    [Show full text]
  • Correlation of Wilms' Tumor 1 Isoforms with HER2 and ER-Α and Its Oncogenic Role in Breast Cancer
    ANTICANCER RESEARCH 34: 1333-1342 (2014) Correlation of Wilms’ Tumor 1 Isoforms with HER2 and ER-α and its Oncogenic Role in Breast Cancer TAPANAWAN NASOMYON1, SRILA SAMPHAO2, SURASAK SANGKHATHAT2, SOMRIT MAHATTANOBON2 and POTCHANAPOND GRAIDIST1 Departments of 1Biomedical Sciences and 2Surgery, Faculty of Medicine, Prince of Songkla University, Hat-Yai, Songkhla, Thailand Abstract. Background: Wilms’ tumor 1 (WT1) gene has solid tumors, such as those of the lung (2) and breast (3-5), different functional properties depending on the isoform type. and other non-solid tumors such as leukemia (6-8). This has This gene correlates with cell proliferation in various types raised the possibility that WT1 could have tumorigenic of cancer. Here, we investigated the expression of WT1 activity rather than tumor-suppressor activity (9-13). isoforms in breast cancer tissues, and focused on the Moreover, WT1 mRNA and protein are expressed in nearly oncogenic role through estrogen receptor-alpha (ER-α) and 90% of breast carcinoma tissues but with low detection in human epidermal growth factor receptor 2 (HER2). adjacent normal breast samples (3). High expression levels Materials and Methods: Expression of WT1(17AA+) and of WT1 mRNA are related to poor prognosis of breast cancer (17AA−) was investigated in adjacent normal breast and (14) and leukemia (7). These phenomena could be due to a breast cancer using Reverse transcription-polymerase chain growth and survival effect from WT1. In addition, down- reaction and western blotting. The correlation of WT1 regulation of WT1 inhibits breast cancer cell proliferation isoforms with HER2 and ER-α was examined using MCF-7 (11).
    [Show full text]
  • Low Levels of Estrogen Receptor Protein Predict Resistance To
    7490 Vol. 10, 7490–7499, November 15, 2004 Clinical Cancer Research Featured Article Low Levels of Estrogen Receptor ␤ Protein Predict Resistance to Tamoxifen Therapy in Breast Cancer Torsten A. Hopp,1,3 Heidi L. Weiss,1,3 improved disease-free and overall survival in patients Irma S. Parra,3 Yukun Cui,1,3 treated with adjuvant tamoxifen therapy. 1,2,3 Conclusions: These findings provide evidence that C. Kent Osborne, and ␤ 1,2,3 ER- may be an independent predictor of response to ta- Suzanne A. W. Fuqua moxifen in breast cancer. Furthermore, these results suggest Departments of 1Medicine, 2Molecular and Cellular Biology, and ␤ 3 that ER- may influence tumor progression in ways differ- Breast Center, Baylor College of Medicine and the Methodist ent from those mediated by the ER-␣ isoform. Hospital, Houston, Texas INTRODUCTION ABSTRACT For more than 30 years, the classical estrogen receptor, Purpose: Breast cancer is a hormone-dependent cancer, called ER-␣, has been extensively studied as a prognostic and and the presence of estrogen receptor ␣ (ER-␣) in tumors is predictive marker in clinical breast cancer, making this nuclear used clinically to predict the likelihood of response to hor- receptor the most valuable target for the treatment of human monal therapies. The clinical value of the second recently breast cancer with selective estrogen receptor modulators or the identified ER isoform, called ER-␤, is less clear, and there is newer generation aromatase inhibitors. Patients with ER-␣– currently conflicting data concerning its potential role as a positive tumors have a significantly prolonged overall and re- prognostic or predictive factor.
    [Show full text]
  • FOXM1 Is a Transcriptional Target of Erα and Has a Critical Role in Breast
    Oncogene (2010) 29, 2983–2995 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc ORIGINAL ARTICLE FOXM1 is a transcriptional target of ERa and has a critical role in breast cancer endocrine sensitivity and resistance J Millour1, D Constantinidou1, AV Stavropoulou1, MSC Wilson1, SS Myatt1, JM-M Kwok1, K Sivanandan1, RC Coombes1, RH Medema2, J Hartman3, AE Lykkesfeldt4 and EW-F Lam1 1Department of Surgery and Cancer, Imperial College London, London, UK; 2Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands; 3Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden and 4Department of Breast Cancer Research, Institute of Cancer Biology, Danish Cancer Society, Copenhagen, Denmark In this study, we investigated the regulation of FOXM1 Introduction expression by estrogen receptor a (ERa) and its role in hormonal therapy and endocrine resistance. FOXM1 Breast cancer is the second most prevalent cause of protein and mRNA expression was regulated by ER- cancer death in the western hemisphere and displays a ligands, including estrogen, tamoxifen (OHT) and fulves- complex aeitology. The forkhead box (FOX) family trant (ICI182780; ICI) in breast carcinoma cell lines. member FOXM1 was previously reported to be elevated Depletion of ERa by RNA interference (RNAi) in MCF-7 in breast cancer as well as in carcinomas of other origins cells downregulated FOXM1 expression. Reporter gene (Pilarsky et al., 2004). FOXM1 is expressed in prolifer- assays showed that ERa activates FOXM1 transcription ating adult tissues and in response to injury or repair through an estrogen-response element (ERE) located (Korver et al., 1997a, b; Ye et al., 1999; Leung et al., within the proximal promoter region.
    [Show full text]
  • An Activin A/BMP2 Chimera, AB215, Blocks Estrogen Signaling Via Induction of ID Proteins in Breast Cancer Cells
    Jung et al. BMC Cancer 2014, 14:549 http://www.biomedcentral.com/1471-2407/14/549 RESEARCH ARTICLE Open Access An Activin A/BMP2 chimera, AB215, blocks estrogen signaling via induction of ID proteins in breast cancer cells Jae Woo Jung1,3, Sun Young Shim1, Dong Kun Lee1, Witek Kwiatkowski2 and Senyon Choe1,2* Abstract Background: One in eight women will be affected by breast cancer in her lifetime. Approximately 75% of breast cancers express estrogen receptor alpha (ERα) and/or progesterone receptor and these receptors are markers for tumor dependence on estrogen. Anti-estrogenic drugs such as tamoxifen are commonly used to block estrogen-mediated signaling in breast cancer. However, many patients either do not respond to these therapies (de novo resistance) or develop resistance to them following prolonged treatment (acquired resistance). Therefore, it is imperative to continue efforts aimed at developing new efficient and safe methods of targeting ER activity in breast cancer. Methods: AB215 is a chimeric ligand assembled from sections of Activin A and BMP2. BMP2’sandAB215’s inhibition of breast cancer cells growth was investigated. In vitro luciferase and MTT proliferation assays together with western blot, RT_PCR, and mRNA knockdown methods were used to determine the mechanism of inhibition of estrogen positive breast cancer cells growth by BMP2 and AB215. Additionally in vivo xenograft tumor model was used to investigate anticancer properties of AB215. Results: Here we report that AB215, a chimeric ligand assembled from sections of Activin A and BMP2 with BMP2-like signaling, possesses stronger anti-proliferative effects on ERα positive breast cancer cells than BMP2.
    [Show full text]