1
|
Rouse WB, Tompkins VS, O’Leary CA, Moss WN. The RNA secondary structure of androgen receptor-FL and V7 transcripts reveals novel regulatory regions. Nucleic Acids Res 2024; 52:6596-6613. [PMID: 38554103 PMCID: PMC11194067 DOI: 10.1093/nar/gkae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
The androgen receptor (AR) is a ligand-dependent nuclear transcription factor belonging to the steroid hormone nuclear receptor family. Due to its roles in regulating cell proliferation and differentiation, AR is tightly regulated to maintain proper levels of itself and the many genes it controls. AR dysregulation is a driver of many human diseases including prostate cancer. Though this dysregulation often occurs at the RNA level, there are many unknowns surrounding post-transcriptional regulation of AR mRNA, particularly the role that RNA secondary structure plays. Thus, a comprehensive analysis of AR transcript secondary structure is needed. We address this through the computational and experimental analyses of two key isoforms, full length (AR-FL) and truncated (AR-V7). Here, a combination of in-cell RNA secondary structure probing experiments (targeted DMS-MaPseq) and computational predictions were used to characterize the static structural landscape and conformational dynamics of both isoforms. Additionally, in-cell assays were used to identify functionally relevant structures in the 5' and 3' UTRs of AR-FL. A notable example is a conserved stem loop structure in the 5'UTR of AR-FL that can bind to Poly(RC) Binding Protein 2 (PCBP2). Taken together, our results reveal novel features that regulate AR expression.
Collapse
Affiliation(s)
- Warren B Rouse
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Van S Tompkins
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Collin A O’Leary
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
- Current Address: Departments of Biology and Chemistry, Cornell College, Mount Vernon, IA 52314, USA
| | - Walter N Moss
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
2
|
Abstract
Androgens are essential sex steroid hormones for both sexes. Testosterone (T) is the predominant androgen in males, while in adult females, T concentrations are about 15-fold lower and androgen precursors are converted to estrogens. T is produced primarily in testicular Leydig cells in men, while in women precursors are biosynthesised in the adrenal cortex and ovaries and converted into T in the periphery. The biosynthesis of T occurs via a series of enzymatic reactions in steroidogenic organs. Notably, the more potent androgen, dihydrotestosterone, may be synthesized from T in the classic pathway, however, alternate metabolic pathways also exist. The classic action of androgens on target organs is mediated through the androgen receptor, which regulates nuclear receptor gene transcription. However, the androgen-androgen receptor complex may also interact directly with membrane proteins or signaling molecules to exert more rapid effects. This review summarizes the current knowledge of androgen biosynthesis, mechanisms of action and endocrine effects in human biology, and relates these effects to respective human congenital and acquired disorders.
Collapse
Affiliation(s)
- Rawda Naamneh Elzenaty
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Switzerland.
| | - Therina du Toit
- Department of Biomedical Research, University of Bern, Switzerland.
| | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Switzerland.
| |
Collapse
|
3
|
Mild androgen insensitivity syndrome: the current landscape. Endocr Pract 2022; 28:911-917. [PMID: 35660466 DOI: 10.1016/j.eprac.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Mild Androgen Insensitivity (MAIS) belongs to the Androgen Insensitivity Syndrome (AIS) spectrum, an X-linked genetic disease that is the most common cause of differences in sex development (DSD). Unfortunately, AIS studies mainly focus on the partial and the complete phenotype, and the mild phenotype (MAIS) has been barely reported. Our purpose is to explore the MAIS facets, clinical features, and molecular aspects. METHODS We collected all reported MAIS cases in the medical literature and presented them based on the phenotype and the molecular diagnosis. RESULTS We identified 49 different AR mutations in 69 individuals in the literature. We compared the AR mutations presented in MAIS individuals with AR mutations previously reported in other AIS phenotypes (CAIS and PAIS) regarding the type, location, genotype-phenotype correlation, and functional studies. CONCLUSION This review provides a landscape of the mild phenotype of AIS. Most MAIS patients present with male infertility. Therefore, AR gene sequencing should be considered during male infertility investigation, even in males with typically male external genitalia. In addition, MAIS can be part of other medical conditions, such as X-linked spinal and bulbar muscular atrophy (Kennedy's disease).
Collapse
|
4
|
Likos E, Bhattarai A, Weyman CM, Shukla GC. The androgen receptor messenger RNA: what do we know? RNA Biol 2022; 19:819-828. [PMID: 35704670 PMCID: PMC9225383 DOI: 10.1080/15476286.2022.2084839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The Androgen Receptor (AR), transcriptionally activated by its ligands, testosterone and dihydrotestosterone (DHT), is widely expressed in cells and tissues, influencing normal biology and disease states. The protein product of the AR gene is involved in the regulation of numerous biological functions, including the development and maintenance of the normal prostate gland and of the cardiovascular, musculoskeletal and immune systems. Androgen signalling, mediated by AR protein, plays a crucial role in the development of prostate cancer (PCa), and is presumed to be involved in other cancers including those of the breast, bladder, liver and kidney. Significant research and reviews have focused on AR protein function; however, inadequate research and literature exist to define the function of AR mRNA in normal and cancer cells. The AR mRNA transcript is nearly 11 Kb long and contains a long 3’ untranslated region (UTR), suggesting its biological role in post-transcriptional regulation, consequently affecting the overall functions of both normal and cancer cells. Research has demonstrated that many biological activities, including RNA stability, translation, cellular trafficking and localization, are associated with the 3’ UTRs of mRNAs. In this review, we describe the potential role of the AR 3’ UTR and summarize RNA-binding proteins (RBPs) that interact with the AR mRNA to regulate post-transcriptional metabolism. We highlight the importance of AR mRNA as a critical modulator of carcinogenesis and its important role in developing therapy-resistant prostate cancer.
Collapse
Affiliation(s)
- Eviania Likos
- Department of Biological, Geo. and Evs. Sciences, Cleveland State University, Cleveland, OH, USA
| | - Asmita Bhattarai
- Department of Biological, Geo. and Evs. Sciences, Cleveland State University, Cleveland, OH, USA
| | - Crystal M Weyman
- Department of Biological, Geo. and Evs. Sciences, Cleveland State University, Cleveland, OH, USA.,Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| | - Girish C Shukla
- Department of Biological, Geo. and Evs. Sciences, Cleveland State University, Cleveland, OH, USA.,Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| |
Collapse
|
5
|
Wang X, Yang D. The regulation of RNA metabolism in hormone signaling and breast cancer. Mol Cell Endocrinol 2021; 529:111221. [PMID: 33711334 PMCID: PMC8262629 DOI: 10.1016/j.mce.2021.111221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 02/02/2023]
Abstract
As the most frequent women's cancer, breast cancer causes the second most cancer-related death in women worldwide. Majority of the breast cancers are hormone receptor-positive and commonly treated by hormone therapy. Thus, the expression levels of hormone receptors signaling pathways are pivotal in the development and therapy of breast cancer. The expression of hormone receptors signaling pathways is not only regulated at the transcription level but also at the post-transcription level by both proteins and RNAs. In addition to that, the function of hormone receptors can also be regulated by RNAs. In this review, we summarize the roles of RNAs in hormone receptor-positive breast cancer. We introduce how mRNA stability and protein function of genes in hormone receptors signaling pathways are regulated by RNA-binding proteins, miRNAs, and lncRNAs. We believe these proteins and RNAs can be potential therapeutic targets of breast cancer.
Collapse
Affiliation(s)
- Xiaofei Wang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, PA, 15261, USA
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, PA, 15261, USA; UPMC Hillman Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
6
|
Multifocal Signal Modulation Therapy by Celecoxib: A Strategy for Managing Castration-Resistant Prostate Cancer. Int J Mol Sci 2019; 20:ijms20236091. [PMID: 31816863 PMCID: PMC6929142 DOI: 10.3390/ijms20236091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a significant health concern throughout the world. Standard therapy for advanced disease consists of anti-androgens, however, almost all prostate tumors become castration resistant (CRPC). Progression from androgen-sensitive PCa to CRPC is promoted by inflammatory signaling through cyclooxygenase-2 (COX-2) expression and ErbB family receptors/AKT activation, compensating androgen receptor inactivity. METHODS Making use of CRPC cell lines, we investigated the effects of the anti-inflammatory drug celecoxib. Biochemical data obtained using immunoblotting, enzyme-linked immunosorbent assay (ELISA), invasion, and xenografts were further integrated by bioinformatic analyses. RESULTS Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation, further amplified the inhibition of androgen signaling. Celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mice xenografts when administered in association with the anti-EGFR receptor antibody cetuximab. Bioinformatic analyses on human prostate cancer datasets support the relevance of these pathways in PCa progression. CONCLUSIONS Signaling nodes at the intersection of pathways implicated in PCa progression are simultaneously modulated by celecoxib treatment. In combination therapies with cetuximab, celecoxib could represent a novel therapeutic strategy to curb signal transduction during CRPC progression.
Collapse
|
7
|
Xu W, Morford J, Mauvais-Jarvis F. Emerging role of testosterone in pancreatic β-cell function and insulin secretion. J Endocrinol 2019; 240:JOE-18-0573.R1. [PMID: 30601759 PMCID: PMC6602868 DOI: 10.1530/joe-18-0573] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/24/2018] [Indexed: 12/16/2022]
Abstract
One of the most sexually dimorphic aspects of metabolic regulation is the bidirectional modulation of glucose homeostasis by testosterone in male and females. Severe testosterone deficiency predisposes men to type 2 diabetes (T2D), while in contrast, androgen excess predisposes women to hyperglycemia. The role of androgen deficiency and excess in promoting visceral obesity and insulin resistance in men and women respectively is well established. However, although it is established that hyperglycemia requires β cell dysfunction to develop, the role of testosterone in β cell function is less understood. This review discusses recent evidence that the androgen receptor (AR) is present in male and female β cells. In males, testosterone action on AR in β cells enhances glucose-stimulated insulin secretion by potentiating the insulinotropic action of glucagon-like peptide-1. In females, excess testosterone action via AR in β cells promotes insulin hypersecretion leading to oxidative injury, which in turn predisposes to T2D.
Collapse
Affiliation(s)
- Weiwei Xu
- W Xu, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Jamie Morford
- J Morford, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Franck Mauvais-Jarvis
- F Mauvais-Jarvis, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| |
Collapse
|
8
|
The ErbB family and androgen receptor signaling are targets of Celecoxib in prostate cancer. Cancer Lett 2017; 400:9-17. [PMID: 28450158 DOI: 10.1016/j.canlet.2017.04.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/06/2017] [Accepted: 04/14/2017] [Indexed: 01/23/2023]
|
9
|
Ebron JS, Shukla GC. Molecular characterization of a novel androgen receptor transgene responsive to MicroRNA mediated post-transcriptional control exerted via 3'-untranslated region. Prostate 2016; 76:834-44. [PMID: 26988939 DOI: 10.1002/pros.23174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/16/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Androgen Receptor (AR) gene is associated with Prostate cancer (PCa) and hence targeting androgen-and AR-signaling axis remains the most promising primary therapeutic option to treat the disease. The AR mRNA has a 6.8 kb long 3'-untranslated region (UTR) which harbors several experimentally validated and numerous predicted miRNA binding sites. AR 3'-UTR is likely to positively or negatively regulate AR expression by interacting with miRNAs and possibly other trans-acting auxiliary factors including 3'-UTR RNA binding proteins. In this context, systematic understanding of the regulatory role of AR 3'-UTR in intrinsic post-transcriptional control of AR gene expression is of significance to understand AR related diseases including PCa. METHODS In this study, we have constructed a heterologous reporter system in which Firefly luciferase and AR expression is experimentally influenced by the presence of AR 3'-UTR and its interactions with ectopically expressing miRNA. RESULTS The expression of AR 3'-UTR containing reporters, including the Firefly luciferase and the AR open reading frame (ORF) were repressed by the overexpression of miR-488* mimics. In addition, the AR expressed from 3'-UTR containing expression vectors was fully functional in its transactivation function as determined by a prostate specific antigen (PSA) reporter assay. Further, by using confocal microscopy we also demonstrate that AR can translocate to the nucleus upon DHT activation confirming the functional ability of AR. CONCLUSIONS AR transgenes with AR 3'-UTR fragments closely resemble the endogenous AR expression than any other previously characterized AR expression constructs. The 3'-UTR containing AR expression system is amiable to post-transcriptional manipulations including miRNA mediated repression of AR expression. This AR reporter system has the potential to be used in determining specificity of AR targeting miRNAs and their role in AR functional regulatory networks. Prostate 76:834-844, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jey Sabith Ebron
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
- Department of Biological Sciences, Cleveland State University, Cleveland, Ohio
| | - Girish C Shukla
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
- Department of Biological Sciences, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
10
|
Fleyshman D, Cheney P, Ströse A, Mudambi S, Safina A, Commane M, Purmal A, Morgan K, Wang NJ, Gray J, Spellman PT, Issaeva N, Gurova K. ARTIK-52 induces replication-dependent DNA damage and p53 activation exclusively in cells of prostate and breast cancer origin. Cell Cycle 2015; 15:455-70. [PMID: 26694952 DOI: 10.1080/15384101.2015.1127478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The realization, that the androgen receptor (AR) is essential for prostate cancer (PC) even after relapse following androgen deprivation therapy motivated the search for novel types of AR inhibitors. We proposed that targeting AR expression versus its function would work in cells having either wild type or mutant AR as well as be independent of androgen synthesis pathways. Previously, using a phenotypic screen in androgen-independent PC cells we identified a small molecule inhibitor of AR, ARTIK-52. Treatment with ARTIK-52 caused the loss of AR protein and death of AR-positive, but not AR-negative, PC cells. Here we present data that ARTIK-52 induces degradation of AR mRNA through a mechanism that we were unable to establish. However, we found that ARTIK-52 is toxic to breast cancer (BC) cells expressing AR, although they were not sensitive to AR knockdown, suggesting an AR-independent mechanism of toxicity. Using different approaches we detected that ARTIK-52 induces replication-dependent double strand DNA breaks exclusively in cancer cells of prostate and breast origin, while not causing DNA damage, or any toxicity, in normal cells, as well as in non-PC and non-BC tumor cells, independent of their proliferation status. This amazing specificity, combined with such a basic mechanism of toxicity, makes ARTIK-52 a potentially useful tool to discover novel attractive targets for the treatment of BC and PC. Thus, phenotypic screening allowed us to identify a compound, whose properties cannot be predicted based on existing knowledge and moreover, uncover a barely known link between AR and DNA damage response in PC and BC epithelial cells.
Collapse
Affiliation(s)
- Daria Fleyshman
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Peter Cheney
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Anda Ströse
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Shaila Mudambi
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Alfiya Safina
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Mairead Commane
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Andrei Purmal
- b Department of Chemistry , Cleveland BioLabs , Buffalo , NY , USA
| | - Kelsey Morgan
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Nicholas J Wang
- c Collaborative Life Sciences Building (CLSB), Oregon Health & Science University , Portland , OR , USA
| | - Joe Gray
- c Collaborative Life Sciences Building (CLSB), Oregon Health & Science University , Portland , OR , USA
| | - Paul T Spellman
- c Collaborative Life Sciences Building (CLSB), Oregon Health & Science University , Portland , OR , USA
| | - Natalia Issaeva
- d Department of Surgery , Otolaryngology and Yale Cancer Center, Yale University School of Medicine , New Haven , CT , USA
| | - Katerina Gurova
- a Department of Cell Stress Biology , Roswell Park Cancer Institute , Buffalo , NY , USA
| |
Collapse
|
11
|
Park KS, Shin SW, Choi JW, Um SH. Specific protein markers for stem cell cross-talk with neighboring cells in the environment. Int J Stem Cells 2014; 6:75-86. [PMID: 24386551 DOI: 10.15283/ijsc.2013.6.2.75] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 01/04/2023] Open
Abstract
A stem cell interacts with the neighboring cells in its environment. To maintain a living organism's metabolism, either cell-cell or cell-environment interactions may be significant. Usually, these cells communicate with each other through biological signaling by interactive behaviors of primary proteins or complementary chemicals. The signaling intermediates offer the stem cell's functionality on its metabolism. With the rapid advent of omics technologies, various specific markers by which stem cells cooperate with their surroundings have been discovered and established. In this article, we review several stem cell markers used to communicate with either cancer or immune cells in the human body.
Collapse
Affiliation(s)
- Kyung Soo Park
- Department of Chemical and Biomolecular Engineering and Sogang University, Seoul, Korea
| | - Seung Won Shin
- School of Chemical Engineering and Sungkyunkwan University, Suwon
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering and Sogang University, Seoul, Korea ; Graduate School of Management of Technology, Sogang University, Seoul, Korea
| | - Soong Ho Um
- School of Chemical Engineering and Sungkyunkwan University, Suwon ; SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon
| |
Collapse
|
12
|
Köhler A, Demir Ü, Kickstein E, Krauss S, Aigner J, Aranda-Orgillés B, Karagiannidis AI, Achmüller C, Bu H, Wunderlich A, Schweiger MR, Schaefer G, Schweiger S, Klocker H, Schneider R. A hormone-dependent feedback-loop controls androgen receptor levels by limiting MID1, a novel translation enhancer and promoter of oncogenic signaling. Mol Cancer 2014; 13:146. [PMID: 24913494 PMCID: PMC4074869 DOI: 10.1186/1476-4598-13-146] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/30/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND High androgen receptor (AR) level in primary tumour predicts increased prostate cancer (PCa)-specific mortality. Furthermore, activations of the AR, PI3K, mTOR, NFκB and Hedgehog (Hh) signaling pathways are involved in the fatal development of castration-resistant prostate cancer during androgen ablation therapy. MID1, a negative regulator of the tumor-suppressor PP2A, is known to promote PI3K, mTOR, NFκB and Hh signaling. Here we investigate the interaction of MID1 and AR. METHODS AR and MID1 mRNA and protein levels were measured by qPCR, Western blot and immunohistochemistry. Co-immunoprecipitation followed by PCR and RNA-pull-down followed by Western blot was used to investigate protein-mRNA interaction, chromatin-immunoprecipitation followed by next-generation sequencing for identification of AR chromatin binding sites. AR transcriptional activity and activity of promoter binding sites for AR were analyzed by reporter gene assays. For knockdown or overexpression of proteins of interest prostate cancer cells were transfected with siRNA or expression plasmids, respectively. RESULTS The microtubule-associated MID1 protein complex associates with AR mRNA via purine-rich trinucleotide repeats, expansions of which are known to correlate with ataxia and cancer. The level of MID1 directly correlates with the AR protein level in PCa cells. Overexpression of MID1 results in a several fold increase in AR protein and activity without major changes in mRNA-levels, whereas siRNA-triggered knockdown of MID1 mRNA reduces AR-protein levels significantly. Upregulation of AR protein by MID1 occurs via increased translation as no major changes in AR protein stability could be observed. AR on the other hand, regulates MID1 via several functional AR binding sites in the MID1 gene, and, in the presence of androgens, exerts a negative feedback loop on MID1 transcription. Thus, androgen withdrawal increases MID1 and concomitantly AR-protein levels. In line with this, MID1 is significantly over-expressed in PCa in a stage-dependent manner. CONCLUSION Promotion of AR, in addition to enhancement of the Akt-, NFκB-, and Hh-pathways by sustained MID1-upregulation during androgen deprivation therapy provides a powerful proliferative scenario for PCa progression into castration resistance. Thus MID1 represents a novel, multi-faceted player in PCa and a promising target to treat castration resistant prostate cancer.
Collapse
Affiliation(s)
- Andrea Köhler
- Institute of Biochemistry, Center of Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Ümmühan Demir
- Department of Urology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Eva Kickstein
- Max-Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sybille Krauss
- Max-Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Present address: German Center for Neurodegenerative Diseases (DZNE), Biomedical Center (BMZ1), Building 344, 53127 Bonn, Germany
| | - Johanna Aigner
- Max-Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | | | - Antonios I Karagiannidis
- Institute of Biochemistry, Center of Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Clemens Achmüller
- Institute of Biochemistry, Center of Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Huajie Bu
- Department of Urology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Andrea Wunderlich
- Institute of Vertebrate Genetics, Max-Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Michal-Ruth Schweiger
- Institute of Vertebrate Genetics, Max-Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Georg Schaefer
- Department of Urology, Innsbruck Medical University, 6020 Innsbruck, Austria
- Department of Pathology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Susann Schweiger
- Universitätsmedizin Mainz, Institute for Human Genetics, 55131 Mainz, Germany
- Division of Medical Sciences, Medical School, DD1 9SY Dundee, UK
| | - Helmut Klocker
- Department of Urology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Rainer Schneider
- Institute of Biochemistry, Center of Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
13
|
Yeap BB. Hormonal changes and their impact on cognition and mental health of ageing men. Maturitas 2014; 79:227-35. [PMID: 24953176 DOI: 10.1016/j.maturitas.2014.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 01/25/2023]
Abstract
Demographic changes resulting in ageing of the world's population have major implications for health. As men grow older, circulating levels of the principal androgen or male sex hormone testosterone (T) decline, while the prevalence of ill-health increases. Observational studies in middle-aged and older men have shown associations between lower levels of T and poorer mental health in older men, including worse cognitive performance, dementia and presence of depressive symptoms. The role of T metabolites, the more potent androgen dihydrotestosterone (DHT) and the oestrogen receptor ligand estradiol (E2) in the pathophysiology of cognitive decline are unclear. Studies of men undergoing androgen deprivation therapy in the setting of prostate cancer have shown subtle detrimental effects of reduced T levels on cognitive performance. Randomised trials of T supplementation in older men have been limited in size and produced variable results, with some studies showing improvement in specific tests of cognitive function. Interventional data from trials of T therapy in men with dementia are limited. Lower levels of T have also been associated with depressive symptoms in older men. Some studies have reported an effect of T therapy to improve mood and depressive symptoms in men with low or low-normal T levels. T supplementation should be considered in men with a diagnosis of androgen deficiency. Beyond this clinical indication, further research is needed to establish the benefits of T supplementation in older men at risk of deteriorating cognition and mental health.
Collapse
Affiliation(s)
- Bu B Yeap
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; Department of Endocrinology and Diabetes, Fremantle and Fiona Stanley Hospitals, Perth, Western Australia, Australia.
| |
Collapse
|
14
|
Tadokoro-Cuccaro R, Davies J, Mongan NP, Bunch T, Brown RS, Audi L, Watt K, McEwan IJ, Hughes IA. Promoter-Dependent Activity on Androgen Receptor N-Terminal Domain Mutations in Androgen Insensitivity Syndrome. Sex Dev 2014; 8:339-49. [DOI: 10.1159/000369266] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 11/19/2022] Open
|
15
|
Frank SB, Miranti CK. Disruption of prostate epithelial differentiation pathways and prostate cancer development. Front Oncol 2013; 3:273. [PMID: 24199173 PMCID: PMC3813973 DOI: 10.3389/fonc.2013.00273] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/18/2013] [Indexed: 12/14/2022] Open
Abstract
One of the foremost problems in the prostate cancer (PCa) field is the inability to distinguish aggressive from indolent disease, which leads to difficult prognoses and thousands of unnecessary surgeries. This limitation stems from the fact that the mechanisms of tumorigenesis in the prostate are poorly understood. Some genetic alterations are commonly reported in prostate tumors, including upregulation of Myc, fusion of Ets genes to androgen-regulated promoters, and loss of Pten. However, the specific roles of these aberrations in tumor initiation and progression are poorly understood. Likewise, the cell of origin for PCa remains controversial and may be linked to the aggressive potential of the tumor. One important clue is that prostate tumors co-express basal and luminal protein markers that are restricted to their distinct cell types in normal tissue. Prostate epithelium contains layer-specific stem cells as well as rare bipotent cells, which can differentiate into basal or luminal cells. We hypothesize that the primary oncogenic cell of origin is a transient-differentiating bipotent cell. Such a cell must maintain tight temporal and spatial control of differentiation pathways, thus increasing its susceptibility for oncogenic disruption. In support of this hypothesis, many of the pathways known to be involved in prostate differentiation can be linked to genes commonly altered in PCa. In this article, we review what is known about important differentiation pathways (Myc, p38MAPK, Notch, PI3K/Pten) in the prostate and how their misregulation could lead to oncogenesis. Better understanding of normal differentiation will offer new insights into tumor initiation and may help explain the functional significance of common genetic alterations seen in PCa. Additionally, this understanding could lead to new methods for classifying prostate tumors based on their differentiation status and may aid in identifying more aggressive tumors.
Collapse
Affiliation(s)
- Sander B Frank
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute , Grand Rapids, MI , USA ; Genetics Graduate Program, Michigan State University , East Lansing, MI , USA
| | | |
Collapse
|
16
|
Rhea JM, French D, Molinaro RJ. Direct total and free testosterone measurement by liquid chromatography tandem mass spectrometry across two different platforms. Clin Biochem 2013; 46:656-64. [DOI: 10.1016/j.clinbiochem.2013.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 01/11/2013] [Accepted: 01/12/2013] [Indexed: 10/27/2022]
|
17
|
Peters KM, Edwards SL, Nair SS, French JD, Bailey PJ, Salkield K, Stein S, Wagner S, Francis GD, Clark SJ, Brown MA. Androgen receptor expression predicts breast cancer survival: the role of genetic and epigenetic events. BMC Cancer 2012; 12:132. [PMID: 22471922 PMCID: PMC3349557 DOI: 10.1186/1471-2407-12-132] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 04/02/2012] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Breast cancer outcome, including response to therapy, risk of metastasis and survival, is difficult to predict using currently available methods, highlighting the urgent need for more informative biomarkers. Androgen receptor (AR) has been implicated in breast carcinogenesis however its potential to be an informative biomarker has yet to be fully explored. In this study, AR protein levels were determined in a cohort of 73 Grade III invasive breast ductal adenocarcinomas. METHODS The levels of Androgen receptor protein in a cohort of breast tumour samples was determined by immunohistochemistry and the results were compared with clinical characteristics, including survival. The role of defects in the regulation of Androgen receptor gene expression were examined by mutation and methylation screening of the 5' end of the gene, reporter assays of the 5' and 3' end of the AR gene, and searching for miRNAs that may regulate AR gene expression. RESULTS AR was expressed in 56% of tumours and expression was significantly inversely associated with 10-year survival (P = 0.004). An investigation into the mechanisms responsible for the loss of AR expression revealed that hypermethylation of the AR promoter is associated with loss of AR expression in breast cancer cells but not in primary breast tumours. In AR negative breast tumours, mutation screening identified the same mutation (T105A) in the 5'UTR of two AR negative breast cancer patients but not reported in the normal human population. Reporter assay analysis of this mutation however found no evidence for a negative impact on AR 5'UTR activity. The role of miR-124 in regulating AR expression was also investigated, however no evidence for this was found. CONCLUSION This study highlights the potential for AR expression to be an informative biomarker for breast cancer survival and sets the scene for a more comprehensive investigation of the molecular basis of this phenomenon.
Collapse
Affiliation(s)
- Kate M Peters
- School of Chemistry and Molecular Biosciences, The University of Queensland, St, Lucia 4072 Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Stope MB, Schubert T, Staar D, Rönnau C, Streitbörger A, Kroeger N, Kubisch C, Zimmermann U, Walther R, Burchardt M. Effect of the heat shock protein HSP27 on androgen receptor expression and function in prostate cancer cells. World J Urol 2012; 30:327-31. [PMID: 22362414 DOI: 10.1007/s00345-012-0843-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/10/2012] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Heat shock proteins (HSP) are involved in processes of folding, activation, trafficking and transcriptional activity of most steroid receptors including the androgen receptor (AR). Accumulating evidence links rising heat shock protein 27 (HSP27) levels with the development of castration-resistant prostate cancer. In order to study the functional relationship between HSP27 and the AR, we modulated the expression of the small heat shock protein HSP27 in human prostate cancer (PC) cell lines. METHODS HSP27 protein concentrations in LNCaP and PC-3 cells were modulated by over-expression or silencing of HSP27. The effects of HSP27 on AR protein and mRNA levels were monitored by Western blotting and quantitative RT-PCR. RESULTS Treatment for the AR-positive LNCaP with HSP27-specific siRNA resulted in a down-regulation of AR levels. This down-regulation of protein was paralleled by a decrease in AR mRNA. Most interestingly, over-expression of HSP27 in PC-3 cells led to a significant increase in AR mRNA although the cells were unable to produce functional AR protein. CONCLUSION The observation that HSP27 is involved in the regulation of AR mRNA by a yet unknown mechanism highlights the complexity of HSP27-AR signaling network.
Collapse
Affiliation(s)
- Matthias B Stope
- Department of Urology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ahtiainen JP, Lehti M, Hulmi JJ, Kraemer WJ, Alen M, Nyman K, Selänne H, Pakarinen A, Komulainen J, Kovanen V, Mero AA, Häkkinen K. Recovery after Heavy Resistance Exercise and Skeletal Muscle Androgen Receptor and Insulin-Like Growth Factor-I Isoform Expression in Strength Trained Men. J Strength Cond Res 2011; 25:767-77. [DOI: 10.1519/jsc.0b013e318202e449] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Wang HY, Hu Y, Wang SH, Shan QL, Li YC, Nie J, Yi LY, Bao DP, Xu CY, He ZH. Association of androgen receptor CAG repeat polymorphism with VO₂max response to hypoxic training in North China Han men. ACTA ACUST UNITED AC 2011; 33:794-9. [PMID: 20374306 DOI: 10.1111/j.1365-2605.2009.01027.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The purposes of this study were to investigate the distribution characteristics of CAG repeat polymorphism in exon 1 of androgen receptor gene and the association of CAG polymorphism with VO₂max response to hypoxic training in North China Han men. Sixty-five healthy young men completed a 30-day HiHiLo training (living high, exercise high and training low) in a simulated normobaric hypoxic environment. All subjects slept in 14.3-14.8% O₂ concentration, and participated in hypoxic training three times a week in 15.4% O₂ concentration. VO₂max (peak oxygen consumption) and body weight were measured before and after hypoxic training. A total of 15 repeat alleles were observed by CAG genotype analyze, in which (CAG)22 was most common. When using 21 and 22 alleles, respectively, as cut-off points, we found that the baseline body weight of two shorter genotype groups was significantly lower than that of longer ones, and that the ΔVO₂max and ΔrVO₂max (Δrelative value of VO₂max) of two shorter genotype groups were significantly higher than those of longer ones after hypoxic training. These findings indicated that AR CAG repeat polymorphism was associated with the exercise performance after simulated normobaric hypoxic HiHiLo training in North China Han men, and that the shorter genotypes had a better individual response to hypoxic training.
Collapse
Affiliation(s)
- H Y Wang
- Science and Research Center of Beijing Sports University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ahtiainen JP, Hulmi JJ, Kraemer WJ, Lehti M, Nyman K, Selänne H, Alen M, Pakarinen A, Komulainen J, Kovanen V, Mero AA, Häkkinen K. Heavy resistance exercise training and skeletal muscle androgen receptor expression in younger and older men. Steroids 2011; 76:183-92. [PMID: 21070797 DOI: 10.1016/j.steroids.2010.10.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/15/2010] [Accepted: 10/29/2010] [Indexed: 11/26/2022]
Abstract
Effects of heavy resistance exercise on serum testosterone and skeletal muscle androgen receptor (AR) concentrations were examined before and after a 21-week resistance training period. Seven healthy untrained young adult men (YT) and ten controls (YC) as well as ten older men (OT) and eight controls (OC) volunteered as subjects. Heavy resistance exercise bouts (5 × 10 RM leg presses) were performed before and after the training period. Muscle biopsies were obtained before and 1h and 48 h after the resistance exercise bouts from m.vastus lateralis (VL) to determine cross-sectional area of muscle fibers (fCSA) and AR mRNA expression and protein concentrations. No changes were observed in YC and OC while resistance training led to significant increases in maximal strength of leg extensors (1 RM), fCSA and lean body mass in YT and OT. Acute increases occurred in serum testosterone concentrations due to resistance exercises but basal testosterone remained unaltered. Mean AR mRNA expression and protein concentration remained unchanged after heavy resistance exercise bouts compared to pre-values. The individual pre- to post-training changes in resting (pre-exercise) AR protein concentration correlated with the changes in fCSA and lean body mass in the combined group of YT and OT. Similarly, it correlated with the changes in 1 RM in YT. Although mean AR expression did not changed due to the resistance exercise training, the present findings suggest that the individual changes of AR protein concentration in skeletal muscle following resistance training may have an impact on training-induced muscular adaptations in both younger and older men.
Collapse
Affiliation(s)
- Juha P Ahtiainen
- Department of Biology of Physical Activity and Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Agarwal M, Naghi J, Philip K, Phan A, Willix RD, Schwarz ER. Growth hormone and testosterone in heart failure therapy. Expert Opin Pharmacother 2010; 11:1835-44. [PMID: 20486829 DOI: 10.1517/14656566.2010.485615] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Heart failure is a progressive disease affecting millions of people worldwide. The disease carries a significantly high morbidity and mortality risk. There are multiple pharmaceutical options to decrease this risk and prolong survival; however, despite optimization of medical management, several patients still await heart transplant, the only definitive cure for heart failure. To slow the progression of disease preventing need for transplantation, improve clinical symptoms, and improve heart failure outcomes, there is a persistent need to discover new therapeutic strategies. Of interest, low growth hormone and testosterone levels have been associated with a worsening degree of heart failure. Many studies have begun to show a clinical improvement in heart failure symptoms when these levels are corrected with hormonal therapy. These findings, although mixed, are promising and indicate that both testosterone and growth hormone therapy should be considered as adjunctive therapy in advanced heart failure patients. AREAS COVERED IN THIS REVIEW This review discusses the physiology of both of these natural hormones, their therapeutic effects in heart failure and data from the published literature on studies using growth hormone or testosterone in patients with chronic heart failure. An extensive search of PubMed was conducted for topics on heart failure, growth hormone, insulin-like growth factor, testosterone, their physiology and pathophysiology, and trials in which they have been used as therapeutic interventions between 1989 and 2009. WHAT THE READER WILL GAIN The reader will gain an understanding of the intricate balance of both of these hormones in the disease state of heart failure. In addition, the trials conducted using these hormones in pharmacotherapy for heart failure are discussed along with proposed theories for interstudy variability. TAKE HOME MESSAGE Testosterone deficiency and growth hormone resistance are positively associated with a poor state of heart failure. Treatment of deficiency improves outcomes in heart failure; however, there is a significant paucity of data with regard to testosterone and heart failure as well as a significant amount of study variability with growth hormone and heart failure.
Collapse
Affiliation(s)
- Megha Agarwal
- Cedars Sinai Medical Center Los Angeles, Cedars Sinai Heart Institute, 8700 Beverly Boulevard, Suite 6215, Los Angeles, California 90048, USA
| | | | | | | | | | | |
Collapse
|
23
|
Direct measurement of serum free testosterone by ultrafiltration followed by liquid chromatography tandem mass spectrometry. Clin Biochem 2010; 43:490-6. [DOI: 10.1016/j.clinbiochem.2009.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 11/13/2009] [Accepted: 12/06/2009] [Indexed: 11/17/2022]
|
24
|
Zhou H, Mazan-Mamczarz K, Martindale JL, Barker A, Liu Z, Gorospe M, Leedman PJ, Gartenhaus RB, Hamburger AW, Zhang Y. Post-transcriptional regulation of androgen receptor mRNA by an ErbB3 binding protein 1 in prostate cancer. Nucleic Acids Res 2010; 38:3619-31. [PMID: 20159994 PMCID: PMC2887957 DOI: 10.1093/nar/gkq084] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Androgen receptor (AR)-mediated pathways play a critical role in the development and progression of prostate cancer. However, little is known about the regulation of AR mRNA stability and translation, two central processes that control AR expression. The ErbB3 binding protein 1 (EBP1), an AR corepressor, negatively regulates crosstalk between ErbB3 ligand heregulin (HRG)-triggered signaling and the AR axis, affecting biological properties of prostate cancer cells. EBP1 protein expression is also decreased in clinical prostate cancer. We previously demonstrated that EBP1 overexpression results in decreased AR protein levels by affecting AR promoter activity. However, EBP1 has recently been demonstrated to be an RNA binding protein. We therefore examined the ability of EBP1 to regulate AR post-transcriptionally. Here we show that EBP1 promoted AR mRNA decay through physical interaction with a conserved UC-rich motif within the 3'-UTR of AR. The ability of EBP1 to accelerate AR mRNA decay was further enhanced by HRG treatment. EBP1 also bound to a CAG-formed stem-loop in the 5' coding region of AR mRNA and was able to inhibit AR translation. Thus, decreases of EBP1 in prostate cancer could be important for the post-transcriptional up-regulation of AR contributing to aberrant AR expression and disease progression.
Collapse
Affiliation(s)
- Hua Zhou
- Greenebaum Cancer Center, Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Horne AW, King AE, Shaw E, McDonald SE, Williams ARW, Saunders PT, Critchley HOD. Attenuated sex steroid receptor expression in fallopian tube of women with ectopic pregnancy. J Clin Endocrinol Metab 2009; 94:5146-54. [PMID: 19864448 PMCID: PMC2989877 DOI: 10.1210/jc.2009-1476] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Sex steroid hormone receptor (SHR) dynamics are well-documented in human endometrium but have not been comprehensively studied in Fallopian tube (FT). OBJECTIVE The aim of the study was to compare expression patterns and hormonal regulation of SHR in FT with that described in endometrium and to determine whether SHR expression is altered in FT of women with ectopic pregnancy (EP). DESIGN Tissue was analyzed and cultured. PATIENTS OR OTHER PARTICIPANTS Women undergoing surgery for benign gynecological conditions (n = 14) and EP (n = 6) participated in the study. INTERVENTIONS Quantitative RT-PCR and immunohistochemistry were used to determine SHR mRNA expression and protein localization, respectively. SHR levels were measured in tubal explant cultures stimulated with estrogen and progestogen. RESULTS ERalpha and ERbeta mRNAs were constitutively expressed in FT during the menstrual cycle. PR-AB and PR-B mRNAs were decreased in midluteal phase compared to follicular phase. ERalpha, PR-AB, and PR-B mRNAs were down-regulated in human FT in vitro by treatment with progestogen. ERalpha, ERbeta1, ERbeta2, PR, and AR proteins localized to cell nuclei of epithelium, stroma, and smooth muscle of nonpregnant FT. In FT from women with EP, PR-B mRNA was decreased when compared to midluteal FT, and ERalpha protein was not detected. CONCLUSIONS SHR expression in FT is different from that observed in endometrium recovered at similar stages of the menstrual cycle, and expression in FT from women with EP is also altered compared with normal FT. These data are an important benchmark for furthering the understanding of normal human FT physiology, changes in expression of SHR in FT in response to progesterone, and disorders of FT function, such as EP.
Collapse
MESH Headings
- Adult
- DNA Primers
- Endometrium/metabolism
- Fallopian Tubes/metabolism
- Female
- Gene Expression/physiology
- Gonadal Steroid Hormones/metabolism
- Humans
- Immunohistochemistry
- Menstrual Cycle/metabolism
- Pregnancy
- Pregnancy, Ectopic/metabolism
- RNA, Messenger/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Andrew W Horne
- Division of Reproductive and Developmental Sciences The University of Edinburgh
| | - Anne E King
- Division of Reproductive and Developmental Sciences The University of Edinburgh
| | - Edward Shaw
- Division of Reproductive and Developmental Sciences The University of Edinburgh
| | - Sarah E McDonald
- Division of Reproductive and Developmental Sciences The University of Edinburgh
| | | | | | - Hilary OD Critchley
- Division of Reproductive and Developmental Sciences The University of Edinburgh
- Corresponding author and requests for reprints: Professor Hilary Critchley Division of Reproductive and Developmental Sciences Centre for Reproductive Biology University of Edinburgh The Queen's Medical Research Institute 47 Little France Crescent Edinburgh EH16 4TJ United Kingdom Tel: 0131 242 6858 Fax: 0131 242 6441
| |
Collapse
|
26
|
Jaworski T. Degradation and beyond: control of androgen receptor activity by the proteasome system. Cell Mol Biol Lett 2009; 11:109-31. [PMID: 16847754 PMCID: PMC6275697 DOI: 10.2478/s11658-006-0011-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 01/31/2006] [Indexed: 12/29/2022] Open
Abstract
The androgen receptor (AR) is a transcription factor belonging to the family of nuclear receptors which mediates the action of androgens in the development of urogenital structures. AR expression is regulated post-translationally by the ubiquitin/proteasome system. This regulation involves more complex mechanisms than typical degradation. The ubiquitin/proteasome system may regulate AR via mechanisms that do not engage in receptor turnover. Given the critical role of AR in sexual development, this complex regulation is especially important. Deregulation of AR signalling may be a causal factor in prostate cancer development. AR is the main target in prostate cancer therapies. Due to the critical role of the ubiquitin/proteasome system in AR regulation, current research suggests that targeting AR degradation is a promising approach.
Collapse
Affiliation(s)
- Tomasz Jaworski
- Department of Cellular Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
27
|
Cai C, Portnoy DC, Wang H, Jiang X, Chen S, Balk SP. Androgen receptor expression in prostate cancer cells is suppressed by activation of epidermal growth factor receptor and ErbB2. Cancer Res 2009; 69:5202-9. [PMID: 19491261 DOI: 10.1158/0008-5472.can-09-0026] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancers (PCa) that relapse after androgen deprivation therapies [castration-resistant PCa (CRPC)] express high levels of androgen receptor (AR) and androgen-regulated genes, and evidence from several groups indicates that ErbB family receptor tyrosine kinases [epidermal growth factor (EGF) receptor (EGFR) and ErbB2] may contribute to enhancing this AR activity. We found that activation of these kinases with EGF and heregulin-beta1 rapidly (within 8 hours) decreased expression of endogenous AR and androgen-regulated PSA in LNCaP PCa cells. AR expression was similarly decreased in LAPC4 and C4-2 cells, but not in the CWR22Rv1 PCa cell line. The rapid decrease in AR was not due to increased AR protein degradation and was not blocked by phosphatidylinositol 3-kinase (LY294002) or MEK (UO126) inhibitors. Significantly, AR mRNA levels in LNCaP cells were markedly decreased by EGF and heregulin-beta1, and experiments with actinomycin D to block new mRNA synthesis showed that AR mRNA degradation was increased. AR mRNA levels were still markedly decreased by EGF and heregulin-beta1 in LNCaP cells adapted to growth in androgen-depleted medium, although AR protein levels did not decline due to increased AR protein stability. These findings show that EGFR and ErbB2 can negatively regulate AR mRNA and may provide an approach to suppress AR expression in CRPC.
Collapse
Affiliation(s)
- Changmeng Cai
- Cancer Biology Program/Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
28
|
Testosterone and ill-health in aging men. ACTA ACUST UNITED AC 2009; 5:113-21. [DOI: 10.1038/ncpendmet1050] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Accepted: 11/20/2008] [Indexed: 12/12/2022]
|
29
|
Influence of surgical trauma on the mRNA expression of sex hormone receptors in PBMCs in male and female patients. Langenbecks Arch Surg 2008; 393:871-6. [PMID: 18297304 DOI: 10.1007/s00423-008-0304-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 01/31/2008] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Gender-specific immune responses have been found after trauma-hemorrhage. Male and female sex hormones seem to be responsible for this gender dimorphism. Alterations in sex hormone receptor expression in mice appear to contribute to the immunomodulatory effect of sex hormones after blood loss. The effect of surgical trauma on the expression of sex hormone receptors in peripheral blood mononuclear cells (PBMCs) from patients, however, remains unknown. MATERIALS AND METHODS PBMCs were obtained from 14 patients (7 men and 7 women) undergoing major abdominal surgery preoperatively and 2 h postoperatively. The expression of the androgen and the estrogen alpha- and beta- receptors were determined by reverse transcriptase polymerase chain reaction (RT-PCR). beta-Actin was used as housekeeping gene. RESULTS The results indicate that surgical trauma has no influence on the expression of the androgen receptor and the estrogen receptors alpha and beta in male and female patients. DISCUSSION The data demonstrate that, in contrast to mice, no alterations in the expression of androgen and estrogen hormone receptors were evident after surgery in patients. Thus, differences in the expression of sex hormone receptors do not appear to be responsible for the gender-specific immune response after surgery.
Collapse
|
30
|
Ing NH, Massuto DA, Jaeger LA. Estradiol up-regulates AUF1p45 binding to stabilizing regions within the 3'-untranslated region of estrogen receptor alpha mRNA. J Biol Chem 2007; 283:1764-1772. [PMID: 18029355 DOI: 10.1074/jbc.m704745200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Estradiol up-regulates expression of the estrogen receptor alpha gene in the uterus by stabilizing estrogen receptor alpha mRNA. Previously, we defined two discrete minimal estradiol-modulated stability sequences (MEMSS) within the extensive 3'-untranslated region of estrogen receptor alpha mRNA with an in vitro stability assay using cytosolic extracts from sheep uterus. We report here that excess MEMSS RNA inhibited the enhanced stability of estrogen receptor alpha mRNA in extracts from estradiol-treated ewes compared with those from control ewes. Several estradiol-induced MEMSS-binding proteins were characterized by UV cross-linking in uterine extracts from ewes in a time course study (0, 8, 16, and 24 h after estradiol injection). The pattern of binding proteins changed at 16 h post-injection, concurrent with enhanced estrogen receptor alpha mRNA stability and the highest rate of accumulation of estrogen receptor alpha mRNA. The predominant MEMSS-binding protein induced by estradiol treatment was identified as AUF1 (A + U-rich RNA-binding factor 1) protein isoform p45 (a product of the heterogeneous nuclear ribonucleoprotein D gene). Immunoblot analysis indicated that only two of four AUF1 protein isoforms were present in the uterine cytosolic extracts and that estradiol treatment strongly increased the ratio of AUF1 isoforms p45 to p37. Nonphosphorylated recombinant AUF1p45 protected estrogen receptor alpha mRNA in vitro in a dose-dependent manner. These studies describe estrogenic induction of AUF1p45 binding to the estrogen receptor alpha mRNA as a molecular mechanism for post-transcriptional up-regulation of gene expression.
Collapse
Affiliation(s)
- Nancy H Ing
- Department of Animal Science, Texas A & M University, College Station, Texas 77843; Veterinary Integrative Biosciences, Texas A & M University, College Station, Texas 77843.
| | - Dana A Massuto
- Veterinary Integrative Biosciences, Texas A & M University, College Station, Texas 77843
| | - Laurie A Jaeger
- Veterinary Integrative Biosciences, Texas A & M University, College Station, Texas 77843
| |
Collapse
|
31
|
Androgen receptor gene polymorphism may affect the risk of urothelial carcinoma. J Biomed Sci 2007; 15:261-9. [DOI: 10.1007/s11373-007-9215-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 09/20/2007] [Indexed: 01/02/2023] Open
|
32
|
Tsezou A, Tzetis M, Gennatas C, Giannatou E, Pampanos A, Malamis G, Kanavakis E, Kitsiou S. Association of repeat polymorphisms in the estrogen receptors alpha, beta (ESR1, ESR2) and androgen receptor (AR) genes with the occurrence of breast cancer. Breast 2007; 17:159-66. [PMID: 17904846 DOI: 10.1016/j.breast.2007.08.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 08/10/2007] [Accepted: 08/20/2007] [Indexed: 10/22/2022] Open
Abstract
Genetic variation in genes involved in estrogen biosynthesis, metabolism and signal transduction have been suggested to play a role in breast cancer. To determine the possible contribution of genetic variation in the ESR1 (ER-alpha), ESR2 (ER-beta) and AR genes in breast cancer risk the -1174(TA)(7-27), c. 1092+3607(CA)(10-26) and c. 172(CAG)(6-40) repeat variants were studied in a case-control study of 79 women with sporadic breast cancer and 155 controls. No significant difference was observed in the frequency distribution of -1174(TA)(7-27) in the ESR1 gene between patients and controls, while a significant difference was observed for repeat polymorphisms c. 1092+3607(CA)(10-26) in the ESR2 gene and c. 172(CAG)(6-40) in the AR gene (p0.0001). A significantly decreased odds ratio (OR) for breast cancer risk was observed in individuals having the LL and the SL genotypes for both the ESR2 (OR=0.010, 95% CI 0.003-0.036, p<0.001; OR=0.013, 95% CI 0.004-0.040, p<0.0001, respectively) and the AR gene (OR=0.040, 95% CI 0.011-0.138, p<0.0001; OR=0.189, 95% CI 0.10-0.359, p<0.0001, respectively), compared to SS genotype. The protective effect of these genotypes remained evident even after adjustment for various risk factors (BMI, age, age at menarche and menopause, family history). In conclusion, an association for breast cancer risk between short (SS) alleles for the repeat variants of the ESR2 and AR genes was found in women of Greek descent.
Collapse
Affiliation(s)
- A Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Medical School, University of Thessalia, University Hospital of Larissa, Larissa, Greece
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Jeske YWA, McGown IN, Cowley DM, Oley C, Thomsett MJ, Choong CSY, Cotterill AM. Androgen receptor genotyping in a large Australasian cohort with androgen insensitivity syndrome; identification of four novel mutations. J Pediatr Endocrinol Metab 2007; 20:893-908. [PMID: 17937062 DOI: 10.1515/jpem.2007.20.8.893] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We genotyped the androgen receptor (AR) gene in 31 Australasian patients with androgen insensitivity syndrome (AIS). The entire coding region of AR was examined including analysis of polymorphic CAG and GGN repeats in all patients. AR defects were found in 66.7% (6/9) of patients with complete AIS (CAIS) and 13.6% (3/22) of patients with partial AIS (PAIS). A novel deletion (N858delG) leading to a premature stop codon was found in CAIS patient P1. CAIS patient P2 has a novel deletion (N2676delGAGT) resulting in a stop at codon 787. These mutations would result in inactivation of AR protein. A novel insertion of a cysteine residue in the first zinc finger of the AR DNA-binding domain (N2045_2047dupCTG) was found in CAIS patient P3. PAIS patient P4 has a novel amino acid substitution (Arg760Ser) in the AR ligand binding domain, which may impair ligand binding. Five patients were found to have previously reported AR mutations and no mutations were identified in the remaining patients.
Collapse
|
34
|
Sircar K, Gottlieb B, Alvarado C, Aprikian A, Beitel LK, Alam-Fahmy M, Begin L, Trifiro M. Androgen receptor CAG repeat length contraction in diseased and non-diseased prostatic tissues. Prostate Cancer Prostatic Dis 2007; 10:360-8. [PMID: 17440439 DOI: 10.1038/sj.pcan.4500967] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To investigate contraction of CAG repeats within the androgen receptor gene (AR) as shorter CAG repeats have been implicated as a possible risk factor in prostate cancer (PCa). AR CAG repeat lengths were analyzed in DNA from microdissected diseased prostates, leukocytes from matched peripheral blood, and control non-diseased prostates. Consistently, all prostatic tissues, whether from benign or cancerous areas of diseased prostates, or from control prostates, showed multiple AR CAG repeat contractions. Germline DNA from blood leukocytes had single CAG repeat lengths in the normal range. AR CAG repeat length contraction may be involved in prostate carcinogenesis and may precede the pathological process.
Collapse
Affiliation(s)
- K Sircar
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Hernandez ME, Soto-Cid A, Aranda-Abreu GE, Díaz R, Rojas F, Garcia LI, Toledo R, Manzo J. A study of the prostate, androgens and sexual activity of male rats. Reprod Biol Endocrinol 2007; 5:11. [PMID: 17367532 PMCID: PMC1845164 DOI: 10.1186/1477-7827-5-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 03/16/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prostate is a sexual gland that produces important substances for the potency of sperm to fertilize eggs within the female reproductive tract, and is under complex endocrine control. Taking advantage of the peculiar behavioral pattern of copulating male rats, we developed experimental paradigms to determine the influence of sexual behavior on the level of serum testosterone, prostate androgen receptors, and mRNA for androgen receptors in male rats displaying up to four consecutive ejaculations. METHODS The effect of four consecutive ejaculations was investigated by determining levels of (i) testosterone in serum by solid phase RIA, (ii) androgen receptors at the ventral prostate with Western Blots, and (iii) androgen receptors-mRNA with RT-PCR. Data were analyzed with a one-way ANOVA followed by a post hoc application of Dunnett's test if required. RESULTS The constant execution of sexual behavior did not produce any change in the weight of the ventral prostate. Serum testosterone increased after the second ejaculation, and remained elevated even after four ejaculations. The androgen receptor at the ventral prostate was higher after the first to third ejaculations, but returned suddenly to baseline levels after the fourth ejaculation. The level of mRNA increased after the first ejaculation, continued to increase after the second, and reached the highest peak after the third ejaculation; however, it returned suddenly to baseline levels after the fourth ejaculation. CONCLUSION Four consecutive ejaculations by sexually experienced male rats had important effects on the physiological responses of the ventral prostate. Fast responses were induced as a result of sexual behavior that involved an increase and decrease in androgen receptors after one and four ejaculations, respectively. However, a progressive response was observed in the elevation of mRNA for androgen receptors, which also showed a fast decrease after four ejaculations. All of these changes with the prostate gland occurred in the presence of a sustained elevation of testosterone in the serum that started after two ejaculations. A consideration of these fast-induced changes suggests that the nerve supply plays a key role in prostate physiology during the sexual behavior of male rats.
Collapse
Affiliation(s)
| | - Abraham Soto-Cid
- Facultad de Quimica Farmaceutica Biologica, Universidad Veracruzana, Xalapa, Ver., Mexico
| | | | - Rosaura Díaz
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico
| | - Fausto Rojas
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico
| | - Luis I Garcia
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico
| | - Rebeca Toledo
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico
| | - Jorge Manzo
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico
| |
Collapse
|
36
|
Cox DG, Blanché H, Pearce CL, Calle EE, Colditz GA, Pike MC, Albanes D, Allen NE, Amiano P, Berglund G, Boeing H, Buring J, Burtt N, Canzian F, Chanock S, Clavel-Chapelon F, Feigelson HS, Freedman M, Haiman CA, Hankinson SE, Henderson BE, Hoover R, Hunter DJ, Kaaks R, Kolonel L, Kraft P, LeMarchand L, Lund E, Palli D, Peeters PHM, Riboli E, Stram DO, Thun M, Tjonneland A, Trichopoulos D, Yeager M. A comprehensive analysis of the androgen receptor gene and risk of breast cancer: results from the National Cancer Institute Breast and Prostate Cancer Cohort Consortium (BPC3). Breast Cancer Res 2007; 8:R54. [PMID: 16987421 PMCID: PMC1779488 DOI: 10.1186/bcr1602] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 08/24/2006] [Accepted: 09/20/2006] [Indexed: 01/12/2023] Open
Abstract
Introduction Androgens have been hypothesised to influence risk of breast cancer through several possible mechanisms, including their conversion to estradiol or their binding to the oestrogen receptor and/or androgen receptor (AR) in the breast. Here, we report on the results of a large and comprehensive study of the association between genetic variation in the AR gene and risk of breast cancer in the National Cancer Institute Breast and Prostate Cancer Cohort Consortium (BPC3). Methods The underlying genetic variation was determined by first sequencing the coding regions of the AR gene in a panel of 95 advanced breast cancer cases. Second, a dense set of markers from the public database was genotyped in a panel of 349 healthy women. The linkage disequilibrium relationships (blocks) across the gene were then identified, and haplotype-tagging single nucleotide polymorphisms (htSNPs) were selected to capture the common genetic variation across the locus. The htSNPs were then genotyped in the nested breast cancer cases and controls from the Cancer Prevention Study II, European Prospective Investigation into Cancer and Nutrition, Multiethnic Cohort, Nurses' Health Study, and Women's Health Study cohorts (5,603 breast cancer cases and 7,480 controls). Results We found no association between any genetic variation (SNP, haplotype, or the exon 1 CAG repeat) in the AR gene and risk of breast cancer, nor were any statistical interactions with known breast cancer risk factors observed. Conclusion Among postmenopausal Caucasian women, common variants of the AR gene are not associated with risk of breast cancer.
Collapse
Affiliation(s)
- David G Cox
- Program in Molecular and Genetic Epidemiology, Epidemiology Department, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
- Channing Laboratory, Harvard Medical School, 181 Longwood Ave., Boston, MA, USA
| | - Hélène Blanché
- CEPH, Fondation Jean Dausset, 27 rue Juliette Dodu, 75010 Paris, France
| | - Celeste L Pearce
- Keck School of Medicine, University of Southern California, East Lake Ave. Los Angeles, CA, 90089 USA
| | - Eugenia E Calle
- Epidemiology and Surveillance Research American Cancer Society, 1599 Clifton Rd. NE, Atlanta, GA, 30329 USA
| | - Graham A Colditz
- Channing Laboratory, Harvard Medical School, 181 Longwood Ave., Boston, MA, USA
| | - Malcolm C Pike
- Keck School of Medicine, University of Southern California, East Lake Ave. Los Angeles, CA, 90089 USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Executive Blvd Rockville, MD, 20852 USA
| | - Naomi E Allen
- Cancer Research UK Epidemiology Unit, University of Oxford, Richard Doll Building, Old Road Campus Oxford, UK OX3 7LF
| | - Pilar Amiano
- Molecular and Nutritional Epidemiology Unit, Scientific Institute of Tuscany, 50131 Florence, Italy
| | - Goran Berglund
- Department of Medicine, Lund University, 221 00 Lund, Sweden
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Julie Buring
- Division of Preventive Medicine, Brigham & Women's Hospital, Department of Medicine, Harvard Medical School, 900 Commonwealth Ave., Boston, MA 02215, USA
| | - Noel Burtt
- Broad Institute at Harvard and the Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Federico Canzian
- Genomic Epidemiology Group, Division of Molecular Genetic Epidemiology, German Cancer Research Center, 69121 Heidelberg, Germany
| | - Stephen Chanock
- Core Genotyping Facility, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20892, USA
| | | | - Heather Spencer Feigelson
- Epidemiology and Surveillance Research American Cancer Society, 1599 Clifton Rd. NE, Atlanta, GA, 30329 USA
| | - Matthew Freedman
- Cancer Research UK Epidemiology Unit, University of Oxford, Richard Doll Building, Old Road Campus Oxford, UK OX3 7LF
- Dana-Farber Cancer Institute, Department of Medical Oncology, 44 Binney St., Boston, MA 02115, USA
| | - Christopher A Haiman
- Keck School of Medicine, University of Southern California, East Lake Ave. Los Angeles, CA, 90089 USA
| | - Susan E Hankinson
- Channing Laboratory, Harvard Medical School, 181 Longwood Ave., Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, 677 Huntington Ave,. Boston, MA 02115, USA
| | - Brian E Henderson
- Keck School of Medicine, University of Southern California, East Lake Ave. Los Angeles, CA, 90089 USA
| | - Robert Hoover
- Core Genotyping Facility, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20892, USA
| | - David J Hunter
- Program in Molecular and Genetic Epidemiology, Epidemiology Department, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
- Channing Laboratory, Harvard Medical School, 181 Longwood Ave., Boston, MA, USA
| | - Rudolf Kaaks
- Nutrition and Hormones Group, International Agency for Research on Cancer,150 Cours Albert Thomas, 69008 Lyon, France
| | - Laurence Kolonel
- Epidemiology Program, Cancer Research Center, University of Hawaii, 1236 Lauhala St., Honolulu, HI 96813, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Epidemiology Department, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
- Channing Laboratory, Harvard Medical School, 181 Longwood Ave., Boston, MA, USA
| | - Loic LeMarchand
- Epidemiology Program, Cancer Research Center, University of Hawaii, 1236 Lauhala St., Honolulu, HI 96813, USA
| | - Eiliv Lund
- Institute of Community Medicine, University of Tromsø, 9037 Tromsø, Norway
| | - Domenico Palli
- Molecular and Nutritional Epidemiology Unit, Scientific Institute of Tuscany, 50131 Florence, Italy
| | - Petra HM Peeters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 Utrecht, The Netherlands
| | - Elio Riboli
- Faculty of Medicine, Division of Epidemiology, Public Health and Primary Care, Imperial College, W2 1PG London, UK
| | - Daniel O Stram
- Keck School of Medicine, University of Southern California, East Lake Ave. Los Angeles, CA, 90089 USA
| | - Michael Thun
- Epidemiology and Surveillance Research American Cancer Society, 1599 Clifton Rd. NE, Atlanta, GA, 30329 USA
| | - Anne Tjonneland
- Institute of Cancer Epidemiology, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Dimitrios Trichopoulos
- Department of Hygiene and Epidemiology, School of Medicine, University of Athens, 75 Mikras Asias Str., 11527 Goudi, Athens, Greece
| | - Meredith Yeager
- Core Genotyping Facility, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20892, USA
| | | |
Collapse
|
37
|
Molinaro RJ, Jha BK, Malathi K, Varambally S, Chinnaiyan AM, Silverman RH. Selection and cloning of poly(rC)-binding protein 2 and Raf kinase inhibitor protein RNA activators of 2',5'-oligoadenylate synthetase from prostate cancer cells. Nucleic Acids Res 2006; 34:6684-95. [PMID: 17145707 PMCID: PMC1751551 DOI: 10.1093/nar/gkl968] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The antiviral and antitumor functions of RNase L are enabled by binding to the allosteric effectors 5'-phosphorylated, 2',5'-linked oligoadenylates (2-5A). 2-5A is produced by interferon-inducible 2',5'-oligoadenylate synthetases (OAS) upon activation by viral double-stranded RNA (dsRNA). Because mutations in RNase L have been implicated as risk factors for prostate cancer, we sought to determine if OAS activators are present in prostate cancer cells. We show that prostate cancer cell lines (PC3, LNCaP and DU145), but not normal prostate epithelial cells (PrEC), contain RNA fractions capable of binding to and activating OAS. To identify the RNA activators, we developed a cDNA cloning strategy based on stringent affinity of RNAs for OAS. We thus identified mRNAs for Raf kinase inhibitor protein (RKIP) and poly(rC)-binding protein 2 (PCBP2) that bind and potently activate OAS. In addition, human endogenous retrovirus (hERV) envelope RNAs were present in PC3 cells that bind and activate OAS. Analysis of several gene expression profiling studies indicated that PCBP2 RNA was consistently elevated in metastatic prostate cancer. Results suggest that OAS activation may occur in prostate cancer cells in vivo stimulated by cellular mRNAs for RKIP and PCBP2.
Collapse
MESH Headings
- 2',5'-Oligoadenylate Synthetase/metabolism
- Adenine Nucleotides/chemistry
- Cell Line, Tumor
- Cloning, Molecular
- Endogenous Retroviruses/genetics
- Enzyme Activation
- Gene Products, env/genetics
- Humans
- Male
- Oligoribonucleotides/chemistry
- Phosphatidylethanolamine Binding Protein/genetics
- Phosphatidylethanolamine Binding Protein/metabolism
- Prostate/metabolism
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/isolation & purification
- RNA, Neoplasm/metabolism
- RNA, Viral/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Ross J. Molinaro
- Department of Chemistry, Cleveland State UniversityEuclid Avenue at East 24th Street, Cleveland, OH 44115, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Babal Kant Jha
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Krishnamurthy Malathi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Michigan Medical School1400 E Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Medical School1400 E Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan Medical School1400 E Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Medical School1400 E Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Robert H. Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic9500 Euclid Avenue, Cleveland, OH 44195, USA
- To whom correspondence should be addressed. Tel: +1 216 445 9650; Fax: +1 216 445 6269;
| |
Collapse
|
38
|
Seidlová-Wuttke D, Pitzel L, Thelen P, Wuttke W. Inhibition of 5α-reductase in the rat prostate by Cimicifuga racemosa. Maturitas 2006. [DOI: 10.1016/j.maturitas.2006.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
Medland SE, Duffy DL, Spurdle AB, Wright MJ, Geffen GM, Montgomery GW, Martin NG. Opposite effects of androgen receptor CAG repeat length on increased risk of left-handedness in males and females. Behav Genet 2006; 35:735-44. [PMID: 16273319 DOI: 10.1007/s10519-005-6187-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 06/01/2005] [Indexed: 11/24/2022]
Abstract
Prenatal exposure to testosterone has been hypothesised to effect lateralization by influencing cell death in the foetal brain. Testosterone binds to the X chromosome linked androgen receptor, which contains a polymorphic polyglutamine CAG repeat, the length of which is positively correlated with testosterone levels in males, and negatively correlated in females. To determine whether the length of the androgen receptor mediates the effects of testosterone on laterality, we examined the association between the number of CAG repeats in the androgen receptor gene and handedness for writing. Association was tested by adding regression terms for the length of the androgen receptor alleles to a multi-factorial-threshold model of liability to left-handedness. In females we found the risk of left-handedness was greater in those with a greater number of repeats (p=0.04), this finding was replicated in a second independent sample of female twins (p=0.014). The length of the androgen receptor explained 6% of the total variance and 24% of the genetic variance in females. In males the risk of left-handedness was greater in those with fewer repeats (p=0.02), with variation in receptor length explaining 10% of the total variance and 24% of the genetic variance. Thus, consistent with Witelson's theory of testosterone action, in all three samples the likelihood of left handedness increased in those individuals with variants of the androgen receptor associated with lower testosterone levels.
Collapse
Affiliation(s)
- Sarah E Medland
- Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Brisbane 4029, Australia.
| | | | | | | | | | | | | |
Collapse
|
40
|
Suzuki T, Miki Y, Moriya T, Akahira JI, Ishida T, Hirakawa H, Yamaguchi Y, Hayashi SI, Sasano H. 5α-Reductase type 1 and aromatase in breast carcinoma as regulators ofin situ androgen production. Int J Cancer 2006; 120:285-91. [PMID: 17066438 DOI: 10.1002/ijc.22317] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previous in vitro studies demonstrated that bioactive androgen 5alpha-dihydrotestosterone (DHT) exerted antiproliferative effects through an interaction with androgen receptor (AR) in breast carcinoma cells. However, AR status has not been examined in association with DHT concentration in breast carcinoma tissues, and significance of androgenic actions remains unclear in breast carcinomas. Therefore, in our study, we first examined intratumoral DHT concentrations in 38 breast carcinoma tissues using liquid chromatography/electrospray tandem mass spectrometry. Intratumoral DHT concentration was positively associated with 5alpha-reductase type 1 (5alphaRed1), and negatively correlated with aromatase. We then examined clinical significance of AR and 5alphaRed1 status in 115 breast carcinoma tissues by immunohistochemistry. Breast carcinomas positive for both AR and 5alphaRed1 were inversely associated with tumor size or Ki-67. These patients showed significant associations with a decreased risk of recurrence and improved prognosis for overall survival, and the AR / 5alphaRed1 status was demonstrated an independent prognostic factor. Moreover, we examined possible regulation of DHT production by aromatase in in vitro studies. DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. These results suggest that intratumoral DHT concentration is mainly determined by 5alphaRed1 and aromatase in breast carcinoma tissues, and antiproliferative effect of DHT may primarily occur in the cases positive for both AR and 5alphaRed1. Aromatase inhibitors may be more effective in these patients, possibly due to increasing local DHT concentration with estrogen deprivation.
Collapse
MESH Headings
- 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/analysis
- 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism
- Adult
- Aged
- Aged, 80 and over
- Androgens/analysis
- Androgens/metabolism
- Aromatase/analysis
- Aromatase/drug effects
- Aromatase/metabolism
- Aromatase Inhibitors/pharmacology
- Breast Neoplasms/chemistry
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/chemistry
- Carcinoma, Ductal, Breast/enzymology
- Carcinoma, Ductal, Breast/pathology
- Chromatography, Liquid
- Coculture Techniques
- Dihydrotestosterone/analysis
- Dihydrotestosterone/metabolism
- Female
- Humans
- Immunohistochemistry
- Ki-67 Antigen/analysis
- Middle Aged
- Receptors, Androgen/analysis
- Receptors, Androgen/metabolism
- Tandem Mass Spectrometry
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
McGrath M, Lee IM, Hankinson SE, Kraft P, Hunter DJ, Buring J, De Vivo I. Androgen receptor polymorphisms and endometrial cancer risk. Int J Cancer 2005; 118:1261-8. [PMID: 16161040 DOI: 10.1002/ijc.21436] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The androgen receptor (AR) gene is a transcription factor responsible for mediating the physiological effects of androgens. Evidence suggests that androgens and the androgen receptor are involved in uterine cell proliferation. A polymorphic CAG repeat in exon 1 of the AR gene encodes a polyglutamine tract that is inversely correlated with the transcriptional activity of this gene. We assessed the association between the functional CAG repeat polymorphism and AR haplotypes and the risk of endometrial cancer in two nested case-control studies within the Nurses' Health Study (n = 222 cases, 666 controls) and the Women's Health Study (n = 137 cases, 411 controls) using conditional and unconditional logistic regression. Associations between AR CAG repeat polymorphism and endometrial cancer risk were similar in the 2 case-control studies. In the pooled analysis, women with an average repeat allele > or =22 repeats compared to <22 repeats were at a statistically significant decreased risk of endometrial cancer (odds ratio (OR) = 0.76; 95% confidence interval (CI), 0.59-0.98). Women with one or two long alleles (> or =27 repeats) compared to both alleles <22 repeats were also at a statistically significant decreased risk (OR = 0.60; 95% CI, 0.36-0.99). We observed a modest yet statistically significant association for each one unit increase in the average repeat length and endometrial cancer risk (OR = 0.94; 95% CI, 0.88-1.00). Associations for the AR CAG average repeat length and endometrial cancer risk differed by menopausal status (p = 0.02). No significant associations between the AR haplotypes and endometrial cancer risk were observed. Our findings suggest that an increasing number of functional CAG repeats may be associated with endometrial carcinogenesis because of AR's reduced ability to recruit coregulators and other transcriptional components. (supplementary material for this article can be found on the International Journal of Cancer website at http://www.interscience.wiley.com/jpages/0020-7136/suppmat/index.html).
Collapse
Affiliation(s)
- Monica McGrath
- Department of Epidemiology, Harvard School of Public Health, 181 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Liu X, Steffensen KR, Sanna A, Arru G, Fois ML, Rosati G, Sotgiu S, Link H, Gustafsson JA, Huang YM. Anti-inflammatory nuclear receptor superfamily in multiple sclerosis patients from Sardinia and Sweden. Neurobiol Dis 2005; 20:961-8. [PMID: 16023861 DOI: 10.1016/j.nbd.2005.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2005] [Revised: 04/17/2005] [Accepted: 06/06/2005] [Indexed: 01/05/2023] Open
Abstract
Several nuclear hormone receptors have been associated with inflammatory reactions. Particularly, liver X receptors (LXRs) have recently been identified as key transcriptional regulators of genes involved in lipid homeostasis and inflammation. LXRs are negative regulators of macrophage inflammatory gene expression. Multiple sclerosis (MS), a demyelinating disease of the central nervous system of unknown cause, is characterized by recurrent inflammation involving macrophages and their inflammatory mediators. Sweden belongs to the countries with a high MS incidence. In Italy, the MS incidence is lower, except on the island of Sardinia where the incidence is even higher than in Sweden. Subjects from Sardinia are ethnically more homogeneous, and differ from Swedes also regarding genetic background and environment. We studied mRNA expression of several nuclear hormone receptors in blood mononuclear cells (MNC) from female patients with untreated relapsing-remitting MS from Sassari, Sardinia, and Stockholm, Sweden. Sex- and age-matched healthy controls (HC) were from both areas. mRNA expression was evaluated by quantitative real-time PCR. We found altered mRNA expression of LXRs, estrogen receptors (ERs), and androgen receptor (AR) in MS. mRNA expression of both LXRalpha and LXRbeta is lower in MS from Stockholm but not from Sassari. In particular, LXRalpha mRNA expression was significantly lower in MS from Stockholm as compared with all groups in the study including MS from Sassari. Low levels of ERalpha mRNA are seen in MS from both Stockholm and Sassari. The splice variant ERbetacx showed significantly higher mRNA expression in MS from Sassari and Stockholm as compared with corresponding HC. In particular, ERbetacx mRNA in MS from Sassari was remarkably higher as compared with all other groups in the study. Higher levels of AR mRNA are present in HC from Sassari. The findings indicate that the expression levels of anti-inflammatory nuclear receptor superfamily genes in MS appear to reflect both ethnic and environmental influences.
Collapse
Affiliation(s)
- Xuan Liu
- Neurotec Department, Division of Neuroimmunology, Karolinska Institute, 141 86 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sader MA, McGrath KCY, Hill MD, Bradstock KF, Jimenez M, Handelsman DJ, Celermajer DS, Death AK. Androgen receptor gene expression in leucocytes is hormonally regulated: implications for gender differences in disease pathogenesis. Clin Endocrinol (Oxf) 2005; 62:56-63. [PMID: 15638871 DOI: 10.1111/j.1365-2265.2004.02173.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE There is evidence that male sex hormones influence the rate of progression of inflammatory and cardiovascular diseases. We have previously shown that human leucocytes and arterial cells isolated from male donors express more androgen receptor (AR) than those from female cells, with potentially pro-atherogenic effects. We now investigate whether the gender difference in AR expression is due to genetic or hormonal regulation. DESIGN AND PATIENTS The influence of hormones on AR expression were studied in hpg mice (a mouse model of androgen deficiency) treated with testosterone, oestradiol or dihydrotestosterone (DHT). Blood samples were obtained for leucocyte AR expression and hormone levels from 53 subjects, grouped into: 12 male [six young adult (27-45 years), six elderly (71-79 years)] and six female (young adult 25-45 years) healthy controls; six male-to-female transsexuals (M2F; 20-50 years) receiving stable pharmacological oral oestrogen treatment; six female-to-male transsexuals (F2M; 31-51 years) receiving stable androgen replacement therapy; five younger men (18-56 years) who had been receiving long-term androgen replacement therapy for hypogonadal disease; six elderly men (72-88 years) who had undergone medical castration for prostate cancer treatment; and 12 male bone marrow transplant recipients (BMT; 23-65 years) from either male or female donors. MEASUREMENTS Serum testosterone and oestradiol concentrations were measured by established immunoflurometric assays from unextracted human serum. AR mRNA levels were measured by RT-PCR and AR protein levels by western blot (cell culture) or immunohistochemistry (mouse arteries). RESULTS We found that AR mRNA levels were significantly down-regulated in the leucocytes of hpg mice that were treated with exogenous testosterone, oestradiol or DHT. AR protein levels were also lower in aortic tissue from the same mice. In humans, we found AR expression was significantly down-regulated by exogenous treatment with testosterone in F2M (31 +/- 13%, compared with control) or oestradiol in M2F (22 +/- 5%) but was significantly up-regulated by endogenous testosterone in BMT (128 +/- 17%). Low androgen levels measured in castrated older men were associated with markedly increased AR expression (207 +/- 26%, P < 0.05) compared with age-matched older male controls (100 +/- 2%). CONCLUSIONS Our results indicate a regulated ability of vascular cells to respond to sex hormones, with the effects of exogenous therapies differing markedly from those due to endogenous sex hormones. We conclude that the gender difference in AR expression in vascular cells is hormonally, rather than genetically, controlled.
Collapse
Affiliation(s)
- Mark A Sader
- Department of Cardiology, Royal Prince Alfred Hospital, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Mammalian spermatogenesis is a complex hormone-dependent developmental program in which a myriad of events must take place to ensure that germ cells reach their proper stage of development at the proper time. Many of these events are controlled by cell type- and stage-specific transcription factors. The regulatory mechanisms involved provide an intriguing paradigm for the field of developmental biology and may lead to the development of new contraceptives an and innovative routs to treat male infertility. In this review, we address three aspects of the genetic regulatory mechanism that drive spermatogenesis. First, we detail what is known about how steroid hormones (both androgens and estrogens) and their cognate receptors initiate and maintain mammalian spermatogenesis. Steroids act through three mechanistic routes: (i) direct activation of genes through hormone-dependent promoter elements, (ii) secondary transcriptional responses through activation of hormone-dependent transcription factors, and (iii) rapid, transcription-independent (nonclassical) events induced by steroid hormones. Second, we provide a survey of transcription factors that function in mammalian spermatogenesis, including homeobox, zinc-finger, heat-shock, and cAMP-response family members. Our survey is not intended to cover all examples but to give a flavor for the gamut of biological roles conferred by transcription factors in the testis, particularly those defined in knockout mice. Third, we address how testis-specific transcription is achieved. In particular, we cover the evidence for and against the idea that some testis-specific genes are transcriptionally silent in somatic tissues as a result of DNA methylation.
Collapse
Affiliation(s)
- James A Maclean
- Department of Immunology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | |
Collapse
|