Expression of Human Estrogen Receptor

Total Page:16

File Type:pdf, Size:1020Kb

Expression of Human Estrogen Receptor Proc. Natl. Acad. Sci. USA Vol. 96, pp. 5722–5727, May 1999 Medical Sciences Expression of human estrogen receptor-a and -b, progesterone receptor, and androgen receptor mRNA in normal and malignant ovarian epithelial cells (mutationysequence deletionyestrogen actionyandrogen actionyprogesterone action) KIN-MANG LAU*, SAMUEL C. MOK†, AND SHUK-MEI HO*‡ *Department of Biology, Tufts University, Medford, MA 02155; and †Laboratory of Gynecological Oncology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 Communicated by Elwood V. Jensen, Karolinska Institute, Huddinge, Sweden, March 15, 1999 (received for review September 15, 1998) b a ABSTRACT Our understanding of the roles played by sex estradiol-17 (E2), progesterone, 20 -hydroxyprogesterone, hormones in ovarian carcinogenesis has been limited by a lack testosterone, and androstenedione have all been shown to of data concerning the mode of sex hormone action in human correlate with OC tumor masses (9–11). Taken together, these ovarian surface epithelial (HOSE) cells, the tissue of origin of findings suggest that steroid hormones are likely involved in >90% of ovarian cancers. We have compared the relative the genesis and progression of the disease, yet their mecha- abundance of estrogen receptor (ER)a,ERb, progesterone nisms of action remain unclear. receptor (PR), and androgen receptor (AR) mRNA in four The classical estrogen receptor (ER), recently renamed ERa primary cultures of HOSE cells obtained from postmeno- (12), and the progesterone receptor (PR) were found in ,50% pausal women to those found in late serous adenocarcinoma of OC specimens, whereas androgen receptor (AR) was de- primary cell cultures and established ovarian cancer cell lines. a b tected in most cases ( 80%) (1–4). In recent studies, tran- We observed coexpression of ER and ER mRNA along with scripts of the newly discovered ER subtype, ERb (12), were AR and PR transcripts in normal HOSE cells and disruption a found in normal human ovaries and benign and malignant of ER mRNA expression as well as dramatic down-regulation ovarian tumors (13, 14), as well as in primary cultures of of PR and AR transcript expression in most ovarian cancer normal human ovarian surface epithelial (HOSE) cells (15). cells. In contrast, levels of ERb mRNA were unaffected by the Unfortunately, none of the aforementioned studies demon- malignant state. Additionally, a novel mutation involving a strated a strong association between ERa, PR, or AR status 32-bp deletion in exon 1 of ERa transcripts was detected in the and OC histological types or grades. Furthermore, treatments SKOV3 cell line. This mutation would explain why SKOV3 was reported to be ER-positive but estrogen-insensitive. Taken of OCs with tamoxifen, antiandrogens, or progestins produced together, these findings suggest that estrogens, signaling via very dismal responses (for review, see refs. 1–4). Conse- either or both ER subtypes, may play an indispensable role in quently, it is widely believed that levels of sex hormone regulating normal HOSE cell functions. Therefore, loss of receptors have little prognostic value and are poor predictors ERa, PR, and AR mRNA expression in HOSE cells may be of hormone-manipulation outcomes for OCs. responsible for neoplastic transformation in this cell type. In A major challenge in assessing the significance of sex contrast, the roles played by ERb in normal and malignant hormones and their receptors in ovarian carcinogenesis is the HOSE cells remain elusive. Finally, the coexistence of mutated paucity of information about their expression levels in the ERa mRNA and normal ERb transcripts in SKOV3 argues in normal HOSE. Over 90% of OCs arise from the HOSE, which favor of a dependency of ERb action on functional ERas. shares a common embryonic origin with epithelia of Mullerian duct-derived tissues (Fallopian tube, endometrium, and endo- Ovarian carcinoma (OC) is the second most common and the cervix) but is distinctly different from the granulosa–thecal most deadly malignancy of the female reproductive tract (for cells of the ovary (1–4). In terms of tissue mass, this layer review, see refs. 1–4). Etiological factors involved in ovarian represents only a small fraction of the whole ovary. Thus, data carcinogenesis remain poorly defined, and effective treatment generated from studies that compare levels of a molecular protocols are limited (1–3). Epidemiological data suggest that marker found in OCs with those observed in whole ovaries are endogenous and exogenous sex hormones may play important difficult to interpret because expression pattern in HOSE roles in the pathogenesis of the disease. In this regard, could easily be masked by those in other ovarian cell types. In estrogens taken as oral contraceptives during premenopausal this regard, although previous studies have demonstrated years offer protection, but when used postmenopausally as localization of ERb in ovarian granulosal cells and ERa hormone replacement therapies elevate risk (1–6). The risk of throughout the ovary (12, 16, 17), only recently have both ER developing invasive OC increases with ever-use of hormone subtypes been found in normal HOSE cells (15). However, it replacement therapy and has been shown to depend on the remains unclear as to whether their expression levels are duration of usage (5, 6). In addition to estrogens, other ovarian altered after neoplastic transformation. Importantly, little is or adrenal steroids such as androstenedione, testosterone, and known about the relationships between the expression patterns progestins have all been implicated as risk factors for OC of the two ER subtypes and those of other steroid receptors in (1–4). Androstenedione and progesterone are present at normal and malignant HOSE cells. higher concentrations in the ovarian vein draining the affected ovary when compared with levels found in the vein draining Abbreviations: HOSE, human ovarian surface epithilial; ER, estrogen the contralateral, disease-free gland (7, 8). Plasma levels of receptor; PR, progesterone receptor; AR androgen receptor; OC, ovarian carcinoma; RT-PCR, reverse transcription–PCR; GAPDH, The publication costs of this article were defrayed in part by page charge glyceraldehyde-3-phosphate dehydrogenase. ‡To whom reprint requests should be addressed at: Department of payment. This article must therefore be hereby marked ‘‘advertisement’’ in Surgery, Division of Urology, University Campus, University of accordance with 18 U.S.C. §1734 solely to indicate this fact. Massachusetts Memorial Medical Center, 55 Lake Avenue North, PNAS is available online at www.pnas.org. Worcester, MA 01655. 5722 Downloaded by guest on September 23, 2021 Medical Sciences: Lau et al. Proc. Natl. Acad. Sci. USA 96 (1999) 5723 To fill this data gap, in the present study, we have compared hyde-6-phosphate dehydrogenase (GAPDH), a housekeeping the expression of ERa,ERb, PR, and AR transcripts in gene. Genomic DNA was eliminated by digestion with DNase primary cultures of normal HOSE cells, obtained from post- I. One microgram of total RNA was reverse-transcribed by menopausal women, to those found in primary ovarian cancer using the GeneAmp RNA PCR kit (Perkin–Elmer). Subse- cell cultures and in established OC cell lines. This approach has quently, 2 ml of the resulting cDNA samples was used in each allowed us to observe that (i)ERa and ERb mRNA, as well PCR. as AR and PR transcripts, are coexpressed in normal HOSE The primer sequences for GAPDH, ERa,ERb, PR, and AR cells, (ii) whole exon-deletion variants of ERa and ERb are are given in Table 1 (20–22). Three pairs of primers were used commonly found in HOSE and ovarian cancer cells, and (iii) to detect ERa wild-type and variant mRNAs, and two pairs of PR and AR mRNA expression are significantly down- primers were used in the identification of wild-type and exon regulated in ovarian cancer cells that exhibit altered ERa, but 5 deletion variant of ERb (Table 1). Hot start PCR using normal ERb, message expression. A previously unknown ERa AmpliTaq Gold DNA polymerase (Perkin–Elmer) was used in mutation, leading to a 32-bp deletion in exon 1, has been all amplification reactions. The enzyme was activated by identified in an ovarian cancer cell line, SKOV3. This cell line preheating the reaction mixtures at 95°C for 6 minutes before a has been shown to express ER but is estrogen- and anties- thermal cycling. This protocol was chosen to minimize non- trogen-resistant (18). Taken together, these findings implicate specific product amplification. The routine PCR program was regulation of normal HOSE cell functions by estrogens, and 30 cycles of 1 min at 94°C, 1 min at 60°C (annealing temper- possibly by progestins and androgens. Additionally, the emer- ature), and 1 min at 72°C. mRNA-specific modifications gence of sex hormone resistance, via down-regulation or included (i) an annealing temperature of 58°C used to amplify mutational inactivation of receptors, may be a key feature of ERb cDNA, (ii) an annealing temperature of 55°C in the ovarian epithelial transformation. amplification of ERa and AR cDNAs, (iii) a cycle number of 35 for ERa cDNA amplification, (iv) GAPDH cDNA ampli- MATERIALS AND METHODS fied for 22–26 cycles, and its levels served as loading control. After PCR, the products were resolved on 2% agarose gel with Cell Cultures and Cell Lines. Four primary cultures of ethidium bromide. The images was captured under UV tran- normal HOSE cells (HOSE27, HOSE20, HOSE17, and sillumination. In initial experiments, after amplification, PCR HOSE13), one primary culture of normal mesothelial cells products were excised, purified, and subjected to direct se- (MesO13), four primary cultures of ovarian carcinoma cells quencing to verify amplification of the correct sequences. (OVCA420, OVCA429, OVCA432, and OVCA433), and Serial dilutions of total RNA and cDNA were used in early three ovarian cancer cell lines (DOV13, SKOV3, and CAOV3) experiments to establish the range of linearity between signal were used in this study.
Recommended publications
  • RUNX1 Control of Mammary Epithelial and Breast Cancer Cell Phenotypes
    University of Massachusetts Medical School eScholarship@UMMS GSBS Dissertations and Theses Graduate School of Biomedical Sciences 2017-12-12 RUNX1 Control of Mammary Epithelial and Breast Cancer Cell Phenotypes Deli Hong University of Massachusetts Medical School Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/gsbs_diss Part of the Cancer Biology Commons, and the Cell Biology Commons Repository Citation Hong D. (2017). RUNX1 Control of Mammary Epithelial and Breast Cancer Cell Phenotypes. GSBS Dissertations and Theses. https://doi.org/10.13028/M21Q2F. Retrieved from https://escholarship.umassmed.edu/gsbs_diss/949 This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in GSBS Dissertations and Theses by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. RUNX1 CONTROL OF MAMMARY EPITHELIAL AND BREAST CANCER CELL PHENOTYPES A Dissertation Presented By Deli Hong Submitted to the Faculty of the University of Massachusetts Graduate School of Biomedical Sciences, Worcester in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY December 12, 2017 Program of Cell Biology RUNX1 CONTROL OF MAMMARY EPITHELIAL AND BREAST CANCER CELL PHENOTYPES A Dissertation Presented By Deli Hong This work was undertaken in the Graduate School of Biomedical Sciences Program of Cell Biology The signature of the Thesis Advisor signifies validation of Dissertation content Gary S. Stein, Ph. D., Thesis Advisor The signatures of the Dissertation Defense Committee signify completion and approval as to style and content of the Dissertation Leslie Shaw, Ph.
    [Show full text]
  • GATA3 As an Adjunct Prognostic Factor in Breast Cancer Patients with Less Aggressive Disease: a Study with a Review of the Literature
    diagnostics Article GATA3 as an Adjunct Prognostic Factor in Breast Cancer Patients with Less Aggressive Disease: A Study with a Review of the Literature Patrizia Querzoli 1, Massimo Pedriali 1 , Rosa Rinaldi 2 , Paola Secchiero 3, Paolo Giorgi Rossi 4 and Elisabetta Kuhn 5,6,* 1 Section of Anatomic Pathology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44124 Ferrara, Italy; [email protected] (P.Q.); [email protected] (M.P.) 2 Section of Anatomic Pathology, ASST Mantova, Ospedale Carlo Poma, 46100 Mantova, Italy; [email protected] 3 Surgery and Experimental Medicine and Interdepartmental Center of Technology of Advanced Therapies (LTTA), Department of Morphology, University of Ferrara, 44121 Ferrara, Italy; [email protected] 4 Epidemiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; [email protected] 5 Division of Pathology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy 6 Department of Biomedical, Surgical, and Dental Sciences, University of Milan, 20122 Milano, Italy * Correspondence: [email protected]; Tel.: +39-02-5032-0564; Fax: +39-02-5503-2860 Abstract: Background: GATA binding protein 3 (GATA3) expression is positively correlated with Citation: Querzoli, P.; Pedriali, M.; estrogen receptor (ER) expression, but its prognostic value as an independent factor remains unclear. Rinaldi, R.; Secchiero, P.; Rossi, P.G.; Thus, we undertook the current study to evaluate the expression of GATA3 and its prognostic value Kuhn, E. GATA3 as an Adjunct in a large series of breast carcinomas (BCs) with long-term follow-up. Methods: A total of 702 Prognostic Factor in Breast Cancer consecutive primary invasive BCs resected between 1989 and 1993 in our institution were arranged Patients with Less Aggressive in tissue microarrays, immunostained for ER, progesterone receptor (PR), ki-67, HER2, p53, and Disease: A Study with a Review of GATA3, and scored.
    [Show full text]
  • Progesterone Receptor Isoform B Regulates the Oxtr-Plcl2-Trpc3 Pathway to Suppress Uterine Contractility
    Progesterone receptor isoform B regulates the Oxtr-Plcl2-Trpc3 pathway to suppress uterine contractility Mary C. Peaveya,1, San-Pin Wub,1, Rong Lib, Jian Liub, Olivia M. Emeryb, Tianyuan Wangc, Lecong Zhouc, Margeaux Wetendorfd, Chandra Yallampallie, William E. Gibbonse, John P. Lydond, and Francesco J. DeMayob,2 aDepartment of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC 27599; bReproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709; cIntegrative Bioinformatic Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709; dDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030; and eDepartment of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030 Edited by R. Michael Roberts, University of Missouri, Columbia, MO, and approved January 19, 2021 (received for review June 6, 2020) Uterine contractile dysfunction leads to pregnancy complications The “progesterone receptor isoform switch” concept was posited such as preterm birth and labor dystocia. In humans, it is hypoth- to explain the transition from a quiescent to a contractile myometrial esized that progesterone receptor isoform PGR-B promotes a re- phenotype in the presence of high levels of progesterone (10, 11, laxed state of the myometrium, and PGR-A facilitates uterine 26–30). Progesterone acts via the nuclear receptor isoforms, PGR-A contraction. This hypothesis was tested in vivo using transgenic and PGR-B, which are coexpressed at different levels throughout mouse models that overexpress PGR-A or PGR-B in smooth muscle pregnancy (31–35). Progesterone can transactivate different tran- cells. Elevated PGR-B abundance results in a marked increase in scriptional programs determined by the relative levels of PGR-A and gestational length compared to control mice (21.1 versus 19.1 d PGR-B isoforms in the myometrium (36–41).
    [Show full text]
  • The Novel Progesterone Receptor
    0013-7227/99/$03.00/0 Vol. 140, No. 3 Endocrinology Printed in U.S.A. Copyright © 1999 by The Endocrine Society The Novel Progesterone Receptor Antagonists RTI 3021– 012 and RTI 3021–022 Exhibit Complex Glucocorticoid Receptor Antagonist Activities: Implications for the Development of Dissociated Antiprogestins* B. L. WAGNER†, G. POLLIO, P. GIANGRANDE‡, J. C. WEBSTER, M. BRESLIN, D. E. MAIS, C. E. COOK, W. V. VEDECKIS, J. A. CIDLOWSKI, AND D. P. MCDONNELL Department of Pharmacology and Cancer Biology (B.L.W., G.P., P.G., D.P.M.), Duke University Medical Center, Durham, North Carolina 27710; Molecular Endocrinology Group (J.C.W., J.A.C.), NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Biochemistry and Molecular Biology (M.B., W.V.V.), Louisiana State University Medical School, New Orleans, Louisiana 70112; Ligand Pharmaceuticals, Inc. (D.E.M.), San Diego, California 92121; Research Triangle Institute (C.E.C.), Chemistry and Life Sciences, Research Triangle Park, North Carolina 27709 ABSTRACT by agonists for DNA response elements within target gene promoters. We have identified two novel compounds (RTI 3021–012 and RTI Accordingly, we observed that RU486, RTI 3021–012, and RTI 3021– 3021–022) that demonstrate similar affinities for human progeste- 022, when assayed for PR antagonist activity, accomplished both of rone receptor (PR) and display equivalent antiprogestenic activity. As these steps. Thus, all three compounds are “active antagonists” of PR with most antiprogestins, such as RU486, RTI 3021–012, and RTI function. When assayed on GR, however, RU486 alone functioned as 3021–022 also bind to the glucocorticoid receptor (GR) with high an active antagonist.
    [Show full text]
  • Expression of the Apoptosis-Related Genes Bcl-2 and P53 in Clinical Samples from Endometrial Carcinoma Patients
    ANTICANCER RESEARCH 31: 4191-4194 (2011) Expression of the Apoptosis-related Genes Bcl-2 and p53 in Clinical Samples from Endometrial Carcinoma Patients IRENE GONZÁLEZ-RODILLA1, VIRGINIA VERNA2, ANA-BELÉN MUÑOZ2, JOSÉ ESTÉVEZ2, MERCEDES BOIX2 and JOSÉ SCHNEIDER2 Departments of 1Pathology and 2Obstetrics and Gynecology, Marqués de Valdecilla University Hospital, Cantabria University, Santander, Spain Abstract. Background: Although alterations in the mechanisms biological features indicative of a less aggressive tumor of apoptosis are an integral part of the tumor phenotype, their phenotype (1). Furthermore, in that same paper, any level of precise role in endometrial carcinoma is still obscure. The aim expression of Bcl-2, from the lowest upwards, was found to was to determine whether Bcl-2 plays a similar biological role be biologically significant. Finally, an explanation for the so- in endometrial cancer as in breast cancer, endometrial cancer called ‘Bcl-2 paradox in breast cancer’, where Bcl-2 being an being also a hormone-dependent tumor. Materials and anti-apoptotic gene, thus promoting the immortalisation of Methods: The expression of the apoptosis-related Bcl-2 and p53 tumor cells, should in principle represent a negative biological genes, together with Ki67, E-cadherin, c-erb-B2 and estrogen feature of tumors expressing it, virtually all published results and progesterone receptors were studied in 136 formalin-fixed, point to the opposite, i.e. that Bcl-2 expression is associated paraffin-embedded endometrial carcinoma samples by means with a better outcome in breast cancer, was put forward. Bcl- of immunohistochemistry. Results: Bcl-2 expression correlated 2 was found to be predominantly expressed in well- directly and significantly with E-cadherin (r=0.22, p=0.011) differentiated, low-proliferating breast carcinomas expressing estrogen receptor (r=0.18, p=0.04) and progesterone receptor hormone receptors, all of them highly favorable prognostic expression (r=0.30, p=0.0006), and inversely with surgical factors.
    [Show full text]
  • Characterization and Assay of Progesterone Receptor in Human Mammary Carcinoma1
    [CANCER RESEARCH 37, 464-471 , February 1977] Characterization and Assay of Progesterone Receptor in Human Mammary Carcinoma1 M. F. Pichon and E. Milgrom Groupe de Recherches sur Ia Biochimie Endocrinienne et Ia Reproduction (INSERM U.135), FacultédeMédecine,Paris-Sud, 94270 Bicétre,France ever, only some patients are improved by such treatments. It SUMMARY would thus be of great practical importance to predict in advance which patient will respond and to nationalize the [3H]Pregn-4-ene-3,20-dione ([3Hjpmogestenane)-recepton choice of the surgical or pharmacological technique to be complexes from human mammary carcinoma were found to used. During the last 15 years, the detection and quantifica be stabilized in the presence of glycerol. The dissociation tion of estrogen receptors in mammary tumors have allowed rate constant was lowered and the equilibrium dissociation marked progress in this direction (11, 12, 16, 18, 21, 22, 34). constant was decreased (KD = 3 nM in the absence of However, the growth of mammary carcinoma may be can glycerol and 1.1 nM in the presence of 30% glycerol), traIled not only by estrogens but also by other hormones whereas no clean-cut effect on the association rate was including observed and no change occurred in the concentration andragens, glucacorticaids, pragestagens , and of binding sites. Gortisol was found to compete with prolactin (13). The study of specific receptors for these [3Hjpmagesterone only at concentrations higher than 1 @M. hormones should shed some bight on the problem of tumor This made it possible to distinguish [3H]pmogestemanebind hormonal dependence . The characterization and measu me ing to the receptor from binding to carticosteroid-binding ment of progesterone receptors have been hampered by the globulin.
    [Show full text]
  • Genetic Alterations of Histone Lysine Methyltransferases and Their Significance in Breast Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No.4 Genetic alterations of histone lysine methyltransferases and their significance in breast cancer Lanxin Liu1,*, Sarah Kimball1,*, Hui Liu1, Andreana Holowatyj1 and Zeng-Quan Yang1 1 Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA * These authors contributed equally to this work Correspondence to: Zeng-Quan Yang, email: [email protected] Keywords: breast cancer, histone lysine methyltransferase, gene amplification, deletion, mutation Received: August 27, 2014 Accepted: December 10, 2014 Published: December 11, 2014 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Histone lysine methyltransferases (HMTs), a large class of enzymes that catalyze site-specific methylation of lysine residues on histones and other proteins, play critical roles in controlling transcription, chromatin architecture, and cellular differentiation. However, the genomic landscape and clinical significance of HMTs in breast cancer remain poorly characterized. Here, we conducted a meta-analysis of approximately 50 HMTs in breast cancer and identified associations among recurrent copy number alterations, mutations, gene expression, and clinical outcome. We identified 12 HMTs with the highest frequency of genetic alterations, including 8 with high-level amplification, 2 with putative homozygous deletion, and 2 with somatic mutation. Different subtypes of breast cancer have different patterns of copy number and expression for each HMT gene. In addition, chromosome 1q contains four HMTs that are concurrently or independently amplified or overexpressed in breast cancer. Copy number or mRNA expression of several HMTs was significantly associated with basal- like breast cancer and shorter patient survival.
    [Show full text]
  • Diagnostic Value of Progesterone Receptor and P53 Expression in Uterine Smooth Muscle Tumors Iman H Hewedi*, Nehal a Radwan and Lobna S Shash
    Hewedi et al. Diagnostic Pathology 2012, 7:1 http://www.diagnosticpathology.org/content/7/1/1 RESEARCH Open Access Diagnostic value of progesterone receptor and p53 expression in uterine smooth muscle tumors Iman H Hewedi*, Nehal A Radwan and Lobna S Shash Abstract Background: The diagnosis of uterine smooth muscle tumors depends on a combination of microscopic features. However, a small number of these tumors still pose difficult diagnostic challenges. Aim: To investigate progesterone receptor (PR) and p53 expression in leiomyomas (LMs), atypical leiomyomas (ALMs), smooth muscle tumors of uncertain malignant potential (STUMP), and leiomyosarcomas (LMSs) and to evaluate the potential utility of the selected immunohistochemical markers in differentiating these tumors. Materials and methods: Immunohistochemical expression of PR and p53 was investigated in 41 uterine smooth muscle tumors comprising: 15 LMS, 4 STUMP, 6 ALM and 16 LM. Quantitative evaluation of PR and p53 expression was graded on a scale from 0 to 3+. Results: Leiomyosarcomas showed reduced PR expression. All LMs as well as ALMs and STUMP were stained intensely for PR. Conversely, LMS was strongly stained with p53, while the three non-sarcomatous groups (STUMP, ALM, LM) were either entirely negative or weakly stained for p53. Regarding both PR and p53 expression, the difference between the LMS group and the three non-sarcomatous groups was highly significant (p < 0.001). Combined high PR - low p53 expression was seen in all the 26 examined cases of the non-sarcomatous group including the STUMP cases and none of the LMS cases. Therefore, it represents a “benign” profile with 100% specificity in diagnosis of a non-sarcomatous tumor.
    [Show full text]
  • Progesterone Receptor-A Isoform Interaction with RUNX Transcription Factors Controls
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.17.448908; this version posted June 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Progesterone receptor-A isoform interaction with RUNX transcription factors controls 2 chromatin remodelling at promoters during ovulation 3 Authors: Dinh DT 1 (*), Breen J 2, Nicol B 3, Smith KM 1, Nicholls M 1, Emery A 1, 4 Wong YY 1, Barry SC 1, Yao HHC 3, Robker RL 1, Russell DL 1 (*) 5 Affiliations: 6 1. Robinson Research Institute, Adelaide Medical School, University of Adelaide, Australia 7 2. South Australian Genomics Centre (SAGC), South Australian Health and Medical 8 Research Institute (SAHMRI), Adelaide, South Australia, Australia 9 3. Reproductive Developmental Biology Group, National Institute of Environmental Health 10 Sciences, Research Triangle Park, NC 27709, USA 11 12 Summary: 13 Progesterone receptor (PGR) plays diverse roles in reproductive tissues and thus coordinates 14 mammalian fertility. In the ovary, acutely induced PGR is the key determinant of ovulation 15 through transcriptional control of a unique set of genes that culminates in follicle rupture. 16 However, the molecular mechanisms for PGR’s specialised function in ovulation is poorly 17 understood. To address this, we assembled a detailed genomic profile of PGR action through 18 combined ATAC-seq, RNA-seq and ChIP-seq analysis in wildtype and isoform-specific PGR 19 null mice.
    [Show full text]
  • Maintenance of Mammary Epithelial Phenotype by Transcription Factor Runx1 Through Mitotic Gene Bookmarking Joshua Rose University of Vermont
    University of Vermont ScholarWorks @ UVM Graduate College Dissertations and Theses Dissertations and Theses 2019 Maintenance Of Mammary Epithelial Phenotype By Transcription Factor Runx1 Through Mitotic Gene Bookmarking Joshua Rose University of Vermont Follow this and additional works at: https://scholarworks.uvm.edu/graddis Part of the Biochemistry Commons, and the Genetics and Genomics Commons Recommended Citation Rose, Joshua, "Maintenance Of Mammary Epithelial Phenotype By Transcription Factor Runx1 Through Mitotic Gene Bookmarking" (2019). Graduate College Dissertations and Theses. 998. https://scholarworks.uvm.edu/graddis/998 This Thesis is brought to you for free and open access by the Dissertations and Theses at ScholarWorks @ UVM. It has been accepted for inclusion in Graduate College Dissertations and Theses by an authorized administrator of ScholarWorks @ UVM. For more information, please contact [email protected]. MAINTENANCE OF MAMMARY EPITHELIAL PHENOTYPE BY TRANSCRIPTION FACTOR RUNX1 THROUGH MITOTIC GENE BOOKMARKING A Thesis Presented by Joshua Rose to The Faculty of the Graduate College of The University of Vermont In Partial Fulfillment of the Requirements for the Degree of Master of Science Specializing in Cellular, Molecular, and Biomedical Sciences January, 2019 Defense Date: November 12, 2018 Thesis Examination Committee: Sayyed Kaleem Zaidi, Ph.D., Advisor Gary Stein, Ph.D., Advisor Seth Frietze, Ph.D., Chairperson Janet Stein, Ph.D. Jonathan Gordon, Ph.D. Cynthia J. Forehand, Ph.D. Dean of the Graduate College ABSTRACT Breast cancer arises from a series of acquired mutations that disrupt normal mammary epithelial homeostasis and create multi-potent cancer stem cells that can differentiate into clinically distinct breast cancer subtypes. Despite improved therapies and advances in early detection, breast cancer remains the leading diagnosed cancer in women.
    [Show full text]
  • Progesterone Receptor Transcription and Non-Transcription Signaling Mechanisms Susan A
    Steroids 68 (2003) 761–770 Progesterone receptor transcription and non-transcription signaling mechanisms Susan A. Leonhardt a, Viroj Boonyaratanakornkit a, Dean P. Edwards b,∗ a Department of Pathology B216, School of Medicine University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Campus Box B216, Denver, CO 80262, USA b Department of Pathology and Program in Molecular Biology, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Campus Box B216, Denver, CO 80262, USA Abstract The diverse effects of progesterone on female reproductive tissues are mediated by the progesterone receptor (PR), a member of the nuclear receptor family of ligand-dependent transcription factors. Thus, PR is an important therapeutic target in female reproduction and in certain endocrine dependent cancers. This paper reviews our understanding of the mechanism of action of the most widely used PR antagonist RU486. Although RU486 is a competitive steroidal antagonist that can displace the natural hormone for PR, it’s potency derives from additional “active antagonism” that involves inhibiting the activity of PR hormone agonist complexes in trans through heterodimerization and competition for binding to progesterone response elements on target DNA, and by recruitment of corepressors that have the potential to actively repress gene transcription. An additional functional role for PR has recently been defined whereby a subpopulation of PR in the cytoplasm or cell membrane is capable of mediating rapid progesterone induced activation of certain signal transduction pathways in the absence of gene transcription. This paper also reviews recent results on the mechanism of the extra-nuclear action of PR and the potential biological roles and implications of this novel PR signaling pathway.
    [Show full text]
  • (NGS) for Primary Endocrine Resistance in Breast Cancer Patients
    Int J Clin Exp Pathol 2018;11(11):5450-5458 www.ijcep.com /ISSN:1936-2625/IJCEP0084102 Original Article Impact of next-generation sequencing (NGS) for primary endocrine resistance in breast cancer patients Ruoyang Li1*, Tiantian Tang1*, Tianli Hui1, Zhenchuan Song1, Fugen Li2, Jingyu Li2, Jiajia Xu2 1Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China; 2Institute of Precision Medicine, 3D Medicines Inc., Shanghai, China. *Equal contributors. Received August 15, 2018; Accepted September 22, 2018; Epub November 1, 2018; Published November 15, 2018 Abstract: Multiple mechanisms have been detected to account for the acquired resistance to endocrine therapies in breast cancer. In this study we retrospectively studied the mechanism of primary endocrine resistance in estrogen receptor positive (ER+) breast cancer patients by next-generation sequencing (NGS). Tumor specimens and matched blood samples were obtained from 24 ER+ breast cancer patients. Fifteen of them displayed endocrine resistance, including recurrence and/or metastases within 24 months from the beginning of endocrine therapy, and 9 pa- tients remained sensitive to endocrine therapy for more than 5 years. Genomic DNA of tumor tissue was extracted from formalin-fixed paraffin-embedded (FFPE) tumor tissue blocks. Genomic DNA of normal tissue was extracted from peripheral blood mononuclear cells (PBMC). Sequencing libraries for each sample were prepared, followed by target capturing for 372 genes that are frequently rearranged in cancers. Massive parallel
    [Show full text]