201
|
Ramalingam S, Ramamurthy VP, Njar VCO. Dissecting major signaling pathways in prostate cancer development and progression: Mechanisms and novel therapeutic targets. J Steroid Biochem Mol Biol 2017; 166:16-27. [PMID: 27481707 PMCID: PMC7371258 DOI: 10.1016/j.jsbmb.2016.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed non-cutaneous malignancy and leading cause of cancer mortality in men. At the initial stages, prostate cancer is dependent upon androgens for their growth and hence effectively combated by androgen deprivation therapy (ADT). However, most patients eventually recur with an androgen deprivation-resistant phenotype, referred to as castration-resistant prostate cancer (CRPC), a more aggressive form for which there is no effective therapy presently available. The current review is an attempt to cover and establish an understanding of some major signaling pathways implicated in prostate cancer development and castration-resistance, besides addressing therapeutic strategies that targets the key signaling mechanisms.
Collapse
Affiliation(s)
- Senthilmurugan Ramalingam
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA.
| |
Collapse
|
202
|
Abstract
Prostate cancer is the second leading cause of cancer deaths in the USA. The challenge in managing castration-resistant prostate cancer (CRPC) stems not from the lack of therapeutic options but from the limited duration of clinical and survival benefit offered by treatments in this setting due to primary and acquired resistance. The remarkable molecular heterogeneity and tumor adaptability in advanced prostate cancer necessitate optimization of such treatment strategies. While the future of CRPC management will involve newer targeted therapies in deliberately biomarker-selected patients, interventions using current approaches may exhibit improved clinical benefit if employed in the context of optimal sequencing and combinations. This review outlines our current understanding of mechanisms of therapeutic resistance in progression to and after the development of castration resistance, highlighting targetable and reversible mechanisms of resistance.
Collapse
Affiliation(s)
- Mary Nakazawa
- Departments of Urology, Molecular and Cellular Biochemistry, Pathology and Toxicology and Cancer Biology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536, USA
| | - Channing Paller
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Natasha Kyprianou
- Departments of Urology, Molecular and Cellular Biochemistry, Pathology and Toxicology and Cancer Biology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536, USA.
| |
Collapse
|
203
|
AR-Signaling in Human Malignancies: Prostate Cancer and Beyond. Cancers (Basel) 2017; 9:cancers9010007. [PMID: 28085048 PMCID: PMC5295778 DOI: 10.3390/cancers9010007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
In the 1940s Charles Huggins reported remarkable palliative benefits following surgical castration in men with advanced prostate cancer, and since then the androgen receptor (AR) has remained the main therapeutic target in this disease. Over the past couple of decades, our understanding of AR-signaling biology has dramatically improved, and it has become apparent that the AR can modulate a number of other well-described oncogenic signaling pathways. Not surprisingly, mounting preclinical and epidemiologic data now supports a role for AR-signaling in promoting the growth and progression of several cancers other than prostate, and early phase clinical trials have documented preliminary signs of efficacy when AR-signaling inhibitors are used in several of these malignancies. In this article, we provide an overview of the evidence supporting the use of AR-directed therapies in prostate as well as other cancers, with an emphasis on the rationale for targeting AR-signaling across tumor types.
Collapse
|
204
|
|
205
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017; 97:135-187. [PMID: 27807202 PMCID: PMC5539371 DOI: 10.1152/physrev.00033.2015] [Citation(s) in RCA: 484] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
206
|
Geng Q, Ni LW, Ouyang B, Hu YH, Zhao Y, Guo J. Alanine and arginine rich domain containing protein, Aard, is directly regulated by androgen receptor in mouse Sertoli cells. Mol Med Rep 2016; 15:352-358. [PMID: 27959439 DOI: 10.3892/mmr.2016.6028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/28/2016] [Indexed: 11/06/2022] Open
Abstract
Alanine and arginine rich domain containing protein (Aard) is specifically expressed in Sertoli cells (SCs) of mouse testis and the expression increases in an age‑dependent manner. A number of previous studies have indicated that androgen and androgen receptor (AR) signaling pathways are particularly important for spermatogenesis in mouse SCs, however, the association between Aard and AR remain to be elucidated. The present study identified Aard as a gene that is directly regulated by AR in mouse SCs, which is important in spermatogenesis. The expression of AARD was significantly downregulated in the testes of Sertoli cell‑selective AR knockout mice compared with wild‑type mice as analyzed by western blotting and immunofluorescence analyses. Quantitative polymerase chain reaction and western blotting indicated that AARD was predominantly expressed in adult mouse testis and its expression was increased in an age-dependent manner. In addition, AARD expression was upregulated by testosterone in primary SCs in vitro, which was confirmed by bioinformatics analysis and a dual‑luciferase reporter assay. Finally, chromatin immunoprecipitation and electrophoretic mobility shift assays indicated that the ligand‑bound AR activated Aard transcription via directly binding to the androgen‑responsive element of the Aard promoter. To the best of our knowledge, the present study is the first to document that Aard is directly regulated by AR in mouse Sertoli cells.
Collapse
Affiliation(s)
- Qiang Geng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Li-Wei Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Bin Ouyang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yan-Hua Hu
- Union Stem Cell & Gene Engineering Co., Ltd, Tianjin 300384, P.R. China
| | - Yu Zhao
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Jun Guo
- Department of Andrology, Xiyuan Hospital of China Academy of Chinese Medical Science, Beijing 100091, P.R. China
| |
Collapse
|
207
|
Hornig NC, Ukat M, Schweikert HU, Hiort O, Werner R, Drop SLS, Cools M, Hughes IA, Audi L, Ahmed SF, Demiri J, Rodens P, Worch L, Wehner G, Kulle AE, Dunstheimer D, Müller-Roßberg E, Reinehr T, Hadidi AT, Eckstein AK, van der Horst C, Seif C, Siebert R, Ammerpohl O, Holterhus PM. Identification of an AR Mutation-Negative Class of Androgen Insensitivity by Determining Endogenous AR Activity. J Clin Endocrinol Metab 2016; 101:4468-4477. [PMID: 27583472 PMCID: PMC5095254 DOI: 10.1210/jc.2016-1990] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CONTEXT Only approximately 85% of patients with a clinical diagnosis complete androgen insensitivity syndrome and less than 30% with partial androgen insensitivity syndrome can be explained by inactivating mutations in the androgen receptor (AR) gene. OBJECTIVE The objective of the study was to clarify this discrepancy by in vitro determination of AR transcriptional activity in individuals with disorders of sex development (DSD) and male controls. DESIGN Quantification of DHT-dependent transcriptional induction of the AR target gene apolipoprotein D (APOD) in cultured genital fibroblasts (GFs) (APOD assay) and next-generation sequencing of the complete coding and noncoding AR locus. SETTING The study was conducted at a university hospital endocrine research laboratory. PATIENTS GFs from 169 individuals were studied encompassing control males (n = 68), molecular defined DSD other than androgen insensitivity syndrome (AIS; n = 18), AR mutation-positive AIS (n = 37), and previously undiagnosed DSD including patients with a clinical suspicion of AIS (n = 46). INTERVENTION(S) There were no interventions. MAIN OUTCOME MEASURE(S) DHT-dependent APOD expression in cultured GF and AR mutation status in 169 individuals was measured. RESULTS The APOD assay clearly separated control individuals (healthy males and molecular defined DSD patients other than AIS) from genetically proven AIS (cutoff < 2.3-fold APOD-induction; 100% sensitivity, 93.3% specificity, P < .0001). Of 46 DSD individuals with no AR mutation, 17 (37%) fell below the cutoff, indicating disrupted androgen signaling. CONCLUSIONS AR mutation-positive AIS can be reliably identified by the APOD assay. Its combination with next-generation sequencing of the AR locus uncovered an AR mutation-negative, new class of androgen resistance, which we propose to name AIS type II. Our data support the existence of cellular components outside the AR affecting androgen signaling during sexual differentiation with high clinical relevance.
Collapse
Affiliation(s)
- N C Hornig
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - M Ukat
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - H U Schweikert
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - O Hiort
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - R Werner
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - S L S Drop
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - M Cools
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - I A Hughes
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - L Audi
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - S F Ahmed
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - J Demiri
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - P Rodens
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - L Worch
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - G Wehner
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - A E Kulle
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - D Dunstheimer
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - E Müller-Roßberg
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - T Reinehr
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - A T Hadidi
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - A K Eckstein
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - C van der Horst
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - C Seif
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - R Siebert
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - O Ammerpohl
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| | - P-M Holterhus
- Department of Pediatrics (N.C.H., M.U., J.D., P.R., A.E.K., P.-M.H.), Division of Pediatric Endocrinology and Diabetes, and Institute of Human Genetics (L.W., R.S., O.A.), Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Schwanenweg 20, 24105 Kiel, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, Department of Medicine III (H.U.S., G.W.), Institute for Biochemistry and Molecular Biology, Nussallee 11, 53115 Bonn, Germany; Department of Pediatrics (O.H., R.W.), Division of Experimental Pediatric Endocrinology, University of Luebeck, 23538 Luebeck, Germany; Department of Pediatrics (S.L.S.D.), Division of Pediatric Endocrinology, Sophia Childreńs Hospital, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; Department of Pediatric Endocrinology (Medical Center), Ghent University Hospital, Ghent University, 9000 Ghent, Belgium; Department of Pediatrics (I.A.H.), University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Pediatric Endocrinology Research Unit (L.A.), Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Passeig Vall d'Hebron 119, 08035 Barcelona, Spain; Developmental Endocrinology Research Group (S.F.A.), School of Medicine, University of Glasgow, Yorkhill Glasgow G3 8SJ, United Kingdom; Kinderklinik (D.D.), Klinikum Augsburg, 86156 Augsburg, Germany; Klinikum Esslingen (E.M.-R.), 73730 Esslingen, Germany; Department of Pediatrics (T.R.), Division of Pediatric Endocrinology, Diabetes, and Nutrition, University Witten/Herdecke, 45711 Datteln, Germany; Hypospadiezentrum (A.T.H.), 63500 Seligenstadt, Germany; Gemeinschaftspraxis für Kinderchirurgie (A.K.E.), 24119 Kronshagen, Germany; Urologische Gemeinschaftspraxis (C.v.d.H), and UROLOGIE Zentrum Kiel (C.S.), 24103 Kiel, Germany; and Institute of Human Genetics (R.S.), University of Ulm and University Hospital of Ulm, 89081 Ulm, Germany
| |
Collapse
|
208
|
Leach DA, Powell SM, Bevan CL. WOMEN IN CANCER THEMATIC REVIEW: New roles for nuclear receptors in prostate cancer. Endocr Relat Cancer 2016; 23:T85-T108. [PMID: 27645052 DOI: 10.1530/erc-16-0319] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 12/20/2022]
Abstract
Prostate cancer has, for decades, been treated by inhibiting androgen signalling. This is effective in the majority of patients, but inevitably resistance develops and patients progress to life-threatening metastatic disease - hence the quest for new effective therapies for 'castrate-resistant' prostate cancer (CRPC). Studies into what pathways can drive tumour recurrence under these conditions has identified several other nuclear receptor signalling pathways as potential drivers or modulators of CRPC.The nuclear receptors constitute a large (48 members) superfamily of transcription factors sharing a common modular functional structure. Many of them are activated by the binding of small lipophilic molecules, making them potentially druggable. Even those for which no ligand exists or has yet been identified may be tractable to activity modulation by small molecules. Moreover, genomic studies have shown that in models of CRPC, other nuclear receptors can potentially drive similar transcriptional responses to the androgen receptor, while analysis of expression and sequencing databases shows disproportionately high mutation and copy number variation rates among the superfamily. Hence, the nuclear receptor superfamily is of intense interest in the drive to understand how prostate cancer recurs and how we may best treat such recurrent disease. This review aims to provide a snapshot of the current knowledge of the roles of different nuclear receptors in prostate cancer - a rapidly evolving field of research.
Collapse
Affiliation(s)
- Damien A Leach
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| | - Sue M Powell
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
209
|
Howe CG, Liu X, Hall MN, Ilievski V, Caudill MA, Malysheva O, Lomax-Luu AM, Parvez F, Siddique AB, Shahriar H, Uddin MN, Islam T, Graziano JH, Costa M, Gamble MV. Sex-Specific Associations between One-Carbon Metabolism Indices and Posttranslational Histone Modifications in Arsenic-Exposed Bangladeshi Adults. Cancer Epidemiol Biomarkers Prev 2016; 26:261-269. [PMID: 27765800 DOI: 10.1158/1055-9965.epi-16-0202] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 09/09/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Posttranslational histone modifications (PTHMs) are altered by arsenic, an environmental carcinogen. PTHMs are also influenced by nutritional methyl donors involved in one-carbon metabolism (OCM), which may protect against epigenetic dysregulation. METHODS We measured global levels of three PTHMs, which are dysregulated in cancers (H3K36me2, H3K36me3, H3K79me2), in peripheral blood mononuclear cells (PBMC) from 324 participants enrolled in the Folic Acid and Creatine Trial, a randomized trial in arsenic-exposed Bangladeshi adults. Sex-specific associations between several blood OCM indices (folate, vitamin B12, choline, betaine, homocysteine) and PTHMs were examined at baseline using regression models, adjusted for multiple tests by controlling for the false discovery rate (PFDR). We also evaluated the effects of folic acid supplementation (400 μg/d for 12 weeks), compared with placebo, on PTHMs. RESULTS Associations between choline and H3K36me2 and between vitamin B12 and H3K79me2 differed significantly by sex (Pdiff < 0.01 and <0.05, respectively). Among men, plasma choline was positively associated with H3K36me2 (PFDR < 0.05), and among women, plasma vitamin B12 was positively associated with H3K79me2 (PFDR < 0.01). Folic acid supplementation did not alter any of the PTHMs examined (PFDR = 0.80). CONCLUSIONS OCM indices may influence PTHMs in a sex-dependent manner, and folic acid supplementation, at this dose and duration, does not alter PTHMs in PBMCs. IMPACT This is the first study to examine the influences of OCM indices on PTHMs in a population that may have increased susceptibility to cancer development due to widespread exposure to arsenic-contaminated drinking water and a high prevalence of hyperhomocysteinemia. Cancer Epidemiol Biomarkers Prev; 26(2); 261-9. ©2016 AACR.
Collapse
Affiliation(s)
- Caitlin G Howe
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| | - Xinhua Liu
- Department of Biostatistics, Mailman School of Public Health, New York
| | - Megan N Hall
- Department of Epidemiology, Mailman School of Public Health, New York
| | - Vesna Ilievski
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Olga Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Angela M Lomax-Luu
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| | - Abu B Siddique
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Hasan Shahriar
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Mohammad N Uddin
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Tariqul Islam
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Joseph H Graziano
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| | - Max Costa
- Department of Environmental Medicine, NYU Langone Medical Center, New York University, New York
| | - Mary V Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, New York
| |
Collapse
|
210
|
Global analysis of transcription in castration-resistant prostate cancer cells uncovers active enhancers and direct androgen receptor targets. Sci Rep 2016; 6:33510. [PMID: 27641228 PMCID: PMC5027586 DOI: 10.1038/srep33510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022] Open
Abstract
Androgen receptor (AR) is a male sex steroid-activated transcription factor (TF) that plays a critical role in prostate cancers, including castration-resistant prostate cancers (CRPC) that typically express amplified levels of the AR. CRPC-derived VCaP cells display an excessive number of chromatin AR-binding sites (ARBs) most of which localize to distal inter- or intragenic regions. Here, we analyzed direct transcription programs of the AR in VCaP cells using global nuclear run-on sequencing (GRO-seq) and integrated the GRO-seq data with the ARB and VCaP cell-specific TF-binding data. Androgen immediately activated transcription of hundreds of protein-coding genes, including IGF-1 receptor and EGF receptor. Androgen also simultaneously repressed transcription of a large number of genes, including MYC. As functional enhancers have been postulated to produce enhancer-templated non-coding RNAs (eRNAs), we also analyzed the eRNAs, which revealed that only a fraction of the ARBs reside at functional enhancers. Activation of these enhancers was most pronounced at the sites that also bound PIAS1, ERG and HDAC3, whereas binding of HDAC3 and PIAS1 decreased at androgen-repressed enhancers. In summary, our genome-wide data of androgen-regulated enhancers and primary target genes provide new insights how the AR can directly regulate cellular growth and control signaling pathways in CPRC cells.
Collapse
|
211
|
Aleskandarany MA, Abduljabbar R, Ashankyty I, Elmouna A, Jerjees D, Ali S, Buluwela L, Diez-Rodriguez M, Caldas C, Green AR, Ellis IO, Rakha EA. Prognostic significance of androgen receptor expression in invasive breast cancer: transcriptomic and protein expression analysis. Breast Cancer Res Treat 2016; 159:215-27. [PMID: 27514395 DOI: 10.1007/s10549-016-3934-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023]
Abstract
Differential prognostic roles of Androgen Receptor (AR) have been proposed in breast cancer (BC) depending on tumour oestrogen receptor (ER) status. This study aimed to evaluate the prognostic and/or predictive significance of AR expression in invasive BC. In this study AR expression was studied on a large (n = 1141) consecutive series of early-stage (I-III) BC using tissue microarray and immunohistochemistry (IHC). AR mRNA expression was assessed in a subset of cases. The prognostic impact of AR mRNA expression was externally validated using the online BC gene expression data sets (n = 25 data sets, 4078 patients). Nuclear AR IHC expression was significantly associated with features of good prognosis including older age, smaller tumour size, lower grade and lobular histology particularly in the ER-positive tumours. AR was associated with ER-related markers GATA3, FOXa1, RERG and BEX1. Negative association was observed with HER2, p53, Ki67, TK1, CD71 and AGTR1. AR Overexpression was associated with longer survival (p < 0.001), independent of tumour size, grade, stage [p = 0.033, hazard ratio (HR) = 0.80 95 % CI = 0.64-0.98]. Similar associations were maintained in ER+ tumours in univariate and multivariate analysis (p < 0.01) both in patients with and without adjuvant endocrine or chemotherapy. AR mRNA expression showed significant association with tumour grade, molecular subtypes, and longer 10 and 15 years survival in luminal BC. In the external validation cohorts, AR gene expression data were associated with improved patients' outcome (p < 0.001, HR = 0.84, 95 % CI 0.79-0.90). AR is not only an independent prognostic factor in ER-positive luminal BC but is also expressed in ER-negative tumours. AR could act as a molecular target in patients with ER-positive disease predicting response to adjuvant therapy.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Cell Nucleus
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasm Grading
- Prognosis
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Estrogen/metabolism
- Retrospective Studies
- Survival Analysis
- Tissue Array Analysis
- Tumor Burden
Collapse
Affiliation(s)
- Mohammad A Aleskandarany
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK.
- Department of Pathology, Faculty of Medicine, Menoufia University, Al Minufiyah, Egypt.
| | - Rezvan Abduljabbar
- Department of Oncology, Azadi Teaching Hospital, Duhok, Kurdistan, 1014 AM, Iraq
| | - Ibraheem Ashankyty
- Molecular Diagnostics and Personalised Therapeutics Unit, University of Ha'il, Ha'il, Saudi Arabia
| | - Ahmed Elmouna
- Molecular Diagnostics and Personalised Therapeutics Unit, University of Ha'il, Ha'il, Saudi Arabia
| | - Dena Jerjees
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Laki Buluwela
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Maria Diez-Rodriguez
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Carlos Caldas
- Centre for the Cambridge Experimental Cancer Medicine Centre (ECMC) and Cambridge Breast Cancer Research Unit, Cambridge, UK
| | - Andrew R Green
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Ian O Ellis
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, Molecular Pathology Research Unit, School of Medicine, University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, NG5 1PB, UK
- Department of Pathology, Faculty of Medicine, Menoufia University, Al Minufiyah, Egypt
| |
Collapse
|
212
|
Howe CG, Gamble MV. Influence of Arsenic on Global Levels of Histone Posttranslational Modifications: a Review of the Literature and Challenges in the Field. Curr Environ Health Rep 2016; 3:225-37. [PMID: 27352015 PMCID: PMC4967376 DOI: 10.1007/s40572-016-0104-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Arsenic is a human carcinogen and also increases the risk for non-cancer outcomes. Arsenic-induced epigenetic dysregulation may contribute to arsenic toxicity. Although there are several reviews on arsenic and epigenetics, these have largely focused on DNA methylation. Here, we review investigations of the effects of arsenic on global levels of histone posttranslational modifications (PTMs). Multiple studies have observed that arsenic induces higher levels of H3 lysine 9 dimethylation (H3K9me2) and also higher levels of H3 serine 10 phosphorylation (H3S10ph), which regulate chromosome segregation. In contrast, arsenic causes a global loss of H4K16ac, a histone PTM that is a hallmark of human cancers. Although the findings for other histone PTMs have not been entirely consistent across studies, we discuss biological factors which may contribute to these inconsistencies, including differences in the dose, duration, and type of arsenic species examined; the tissue or cell line evaluated; differences by sex; and exposure timing. We also discuss two important considerations for the measurement of histone PTMs: proteolytic cleavage of histones and arsenic-induced alterations in histone expression.
Collapse
Affiliation(s)
- Caitlin G. Howe
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. Address: 11 Floor, 722 W. 168 Street, New York, New York, 10032. . Phone: 212-305-1205. Fax: 212-305-3857
| | - Mary V. Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. Address: 11 Floor, 722 W. 168 Street, New York, New York, 10032. . Phone: 212-305-7949. Fax: 212-305-3857
| |
Collapse
|
213
|
Wang Q, Zhou JL, Wang H, Ju Q, Ding Z, Zhou XL, Ge X, Shi QM, Pan C, Zhang JP, Zhang MR, Yu HM, Xu LC. Inhibition effect of cypermethrin mediated by co-regulators SRC-1 and SMRT in interleukin-6-induced androgen receptor activation. CHEMOSPHERE 2016; 158:24-29. [PMID: 27239967 DOI: 10.1016/j.chemosphere.2016.05.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/21/2016] [Accepted: 05/17/2016] [Indexed: 06/05/2023]
Abstract
It is hypothesized that the pesticide cypermethrin may induce androgen receptor (AR) antagonism via ligand-independent mechanisms. The Real-Time Cell Analysis (RTCA) iCELLigence system was used to investigate the inhibitory effect of cypermethrin on interleukin-6 (IL-6)-induced ligand-independent LNCaP cell growth. Then, the mammalian two-hybrid assays were applied to clarify whether the mechanism of IL-6-induced AR antagonism of cypermethrin was associated with the interactions of the AR and co-activator steroid receptor co-activator-1 (SRC-1) and co-repressor silencing mediator for retinoid and thyroid hormone receptors (SMRT). Cypermethrin inhibited the LNCaP cell growth induced by IL-6. The interactions of AR-SRC-1 and AR-SMRT mediated by IL-6 were suppressed by cypermethrin. The results indicate that the IL-6-mediated AR antagonism induced by cypermethrin is related to repress the recruitment of co-regulators SRC-1 and SMRT to the AR in a ligand-independent manner. Inhibition of the interactions of AR-SRC-1 and AR-SMRT mediated by IL-6 contributes to the AR antagonism induced by cypermethrin.
Collapse
Affiliation(s)
- Qi Wang
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Ji-Long Zhou
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Hui Wang
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Qiang Ju
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Zhen Ding
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Xiao-Long Zhou
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Xing Ge
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Qiao-Mei Shi
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Chen Pan
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Jin-Peng Zhang
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Mei-Rong Zhang
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Hong-Min Yu
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China
| | - Li-Chun Xu
- School of Public Health, Xuzhou Medical College, 209 Tong-Shan Road, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
214
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
215
|
Zouboulis CC, Picardo M, Ju Q, Kurokawa I, Törőcsik D, Bíró T, Schneider MR. Beyond acne: Current aspects of sebaceous gland biology and function. Rev Endocr Metab Disord 2016; 17:319-334. [PMID: 27726049 DOI: 10.1007/s11154-016-9389-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sebaceous gland is most commonly found in association with a hair follicle. Its traditional function is the holocrine production of sebum, a complex mixture of lipids, cell debris, and other rather poorly characterized substances. Due to the gland's central role in acne pathogenesis, early research had focused on its lipogenic activity. Less studied aspects of the sebaceous gland, such as stem cell biology, the regulation of cellular differentiation by transcription factors, the significance of specific lipid fractions, the endocrine and specially the neuroendocrine role of the sebaceous gland, and its contribution to the innate immunity, the detoxification of the skin, and skin aging have only recently attracted the attention of researchers from different disciplines. Here, we summarize recent multidisciplinary progress in sebaceous gland research and discuss how sebaceous gland research may stimulate the development of novel therapeutic strategies targeting specific molecular pathways of the pathogenesis of skin diseases.
Collapse
Affiliation(s)
- Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Auenweg 38, 06847, Dessau, Germany.
| | - Mauro Picardo
- San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Qiang Ju
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Ichiro Kurokawa
- Department of Dermatology, Meiwa Hospital, Nishinomiya, Japan
| | - Dániel Törőcsik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Marlon R Schneider
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| |
Collapse
|
216
|
Sipilä P, Björkgren I. Segment-specific regulation of epididymal gene expression. Reproduction 2016; 152:R91-9. [DOI: 10.1530/rep-15-0533] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/23/2016] [Indexed: 01/24/2023]
Abstract
The epididymis is necessary for post-testicular sperm maturation. During their epididymal transit, spermatozoa gain ability for progressive movement and fertilization. The epididymis is composed of several segments that have distinct gene expression profiles that enable the establishment of the changing luminal environment required for sperm maturation. The epididymal gene expression is regulated by endocrine, lumicrine, and paracrine factors in a segment-specific manner. Thus, in addition to its importance for male fertility, the epididymis is a valuable model tissue for studying the regulation of gene expression. This review concentrates on recent advances in understanding the androgen, small RNA, and epigenetically mediated regulation of segment-specific gene expression in the epididymis.
Collapse
|
217
|
Armstrong CM, Gao AC. Adaptive pathways and emerging strategies overcoming treatment resistance in castration resistant prostate cancer. Asian J Urol 2016. [PMID: 28642838 PMCID: PMC5477778 DOI: 10.1016/j.ajur.2016.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The therapies available for prostate cancer patients whom progress from hormone-sensitive to castration resistant prostate cancer include both systemic drugs, including docetaxel and cabazitaxel, and drugs that inhibit androgen signaling such as enzalutamide and abiraterone. Unfortunately, it is estimated that up to 30% of patients have primary resistance to these treatments and over time even those who initially respond to therapy will eventually develop resistance and their disease will continue to progress regardless of the presence of the drug. Determining the mechanisms involved in the development of resistance to these therapies has been the area of intense study and several adaptive pathways have been uncovered. Androgen receptor (AR) mutations, expression of AR-V7 (or other constitutively active androgen receptor variants), intracrine androgen production and overexpression of androgen synthesis enzymes such as Aldo-Keto Reductase Family 1, Member C3 (AKR1C3) are among the many mechanisms associated with resistance to anti-androgens. In regards to the taxanes, one of the key contributors to drug resistance is increased drug efflux through ATP Binding Cassette Subfamily B Member 1 (ABCB1). Targeting these resistance mechanisms using different strategies has led to various levels of success in overcoming resistance to current therapies. For instance, targeting AR-V7 with niclosamide or AKR1C3 with indomethacin can improve enzalutamide and abiraterone treatment. ABCB1 transport activity can be inhibited by the dietary constituent apigenin and antiandrogens such as bicalutamide which in turn improves response to docetaxel. A more thorough understanding of how drug resistance develops will lead to improved treatment strategies. This review will cover the current knowledge of resistance mechanisms to castration resistant prostate cancer therapies and methods that have been identified which may improve treatment response.
Collapse
Affiliation(s)
| | - Allen C Gao
- Department of Urology, University of California, Davis, Sacramento, CA, USA.,Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA.,VA Northern California Health Care System, Sacramento, CA, USA
| |
Collapse
|
218
|
Mou L, Gui Y. A novel variant of androgen receptor is associated with idiopathic azoospermia. Mol Med Rep 2016; 14:2915-20. [PMID: 27498682 PMCID: PMC5042743 DOI: 10.3892/mmr.2016.5587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/07/2016] [Indexed: 12/13/2022] Open
Abstract
A variety of genetic variants can lead to abnormal human spermatogenesis. The androgen receptor (AR) is an important steroid hormone receptor that is critical for male sexual differentiation and the maintenance of normal spermatogenesis. In the present study, each exon of AR in 776 patients diagnosed with idiopathic azoospermia (IA) and 709 proven fertile men were sequenced using use panel re‑sequencing methods to examine whether AR is involved in the pathogenesis of IA. Two synonymous variants and seven missense variants were detected. Of the missense variants, a luciferase assay demonstrated that the R630W variant reduced the transcriptional regulatory function of AR. This novel variant (p. R630W) of AR is the first to be identified in association with IA, thereby highlighting the importance of AR during spermatogenesis.
Collapse
Affiliation(s)
- Lisha Mou
- Shenzhen Domesticated Organ Medical Engineering Research and Development Center, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Biomedical Research Institute, Shenzhen PKU‑HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
219
|
Fialova B, Luzna P, Gursky J, Langova K, Kolar Z, Trtkova KS. Epigenetic modulation of AR gene expression in prostate cancer DU145 cells with the combination of sodium butyrate and 5′-Aza-2′-deoxycytidine. Oncol Rep 2016; 36:2365-74. [DOI: 10.3892/or.2016.5000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/24/2016] [Indexed: 11/05/2022] Open
|
220
|
Howe CG, Liu X, Hall MN, Slavkovich V, Ilievski V, Parvez F, Siddique AB, Shahriar H, Uddin MN, Islam T, Graziano JH, Costa M, Gamble MV. Associations between Blood and Urine Arsenic Concentrations and Global Levels of Post-Translational Histone Modifications in Bangladeshi Men and Women. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1234-40. [PMID: 26967670 PMCID: PMC4977054 DOI: 10.1289/ehp.1510412] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/21/2015] [Accepted: 02/22/2016] [Indexed: 05/23/2023]
Abstract
BACKGROUND Exposure to inorganic arsenic is associated with numerous adverse health outcomes, with susceptibility differing by sex. Although evidence from in vitro studies suggests that arsenic alters post-translational histone modifications (PTHMs), evidence in humans is limited. OBJECTIVES The objectives were to determine: a) if arsenic exposure is associated with global (percent) levels of PTHMs H3K36me2, H3K36me3, and H3K79me2 in a sex-dependent manner, and b) if %PTHMs are stable when arsenic exposure is reduced. METHODS We examined associations between arsenic, measured in blood and urine, and %PTHMs in peripheral blood mononuclear cells from 317 participants enrolled in the Bangladesh Folic Acid and Creatine Trial (FACT). We also examined the stability of %PTHMs after the use of arsenic-removal water filters (n = 60). RESULTS Associations between natural log-transformed (ln) urinary arsenic, adjusted for creatinine (uAsCr), and %H3K36me2 differed significantly between men and women (p = 0.01). ln(uAsCr) was positively associated with %H3K36me2 in men [β = 0.12; 95% confidence interval (CI): 0.01, 0.23, p = 0.03] but was negatively associated with %H3K36me2 in women (β = -0.05; 95% CI: -0.12, 0.02, p = 0.19). The patterns of associations with blood arsenic were similar. On average, water filter use was also associated with reductions in %H3K36me2 (p < 0.01), but this did not differ significantly by sex. Arsenic was not significantly associated with %H3K36me3 or %H3K79me2 in men or women. CONCLUSIONS Arsenic exposure was associated with %H3K36me2 in a sex-specific manner but was not associated with %H3K36me3 or %H3K79me2. Additional studies are needed to assess changes in %H3K36me2 after arsenic removal. CITATION Howe CG, Liu X, Hall MN, Slavkovich V, Ilievski V, Parvez F, Siddique AB, Shahriar H, Uddin MN, Islam T, Graziano JH, Costa M, Gamble MV. 2016. Associations between blood and urine arsenic concentrations and global levels of post-translational histone modifications in Bangladeshi men and women. Environ Health Perspect 124:1234-1240; http://dx.doi.org/10.1289/ehp.1510412.
Collapse
Affiliation(s)
| | | | - Megan N. Hall
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | | | | | | | - Abu B. Siddique
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Hasan Shahriar
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Mohammad N. Uddin
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | - Tariqul Islam
- Columbia University Arsenic Project in Bangladesh, Dhaka, Bangladesh
| | | | - Max Costa
- Department of Environmental Medicine, Langone Medical Center, New York University, New York, New York, USA
| | - Mary V. Gamble
- Department of Environmental Health Sciences,
- Address correspondence to M.V. Gamble, Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 11th Floor, 722 W. 168th St., New York, NY 10032 USA. Telephone: (212) 305-7949. E-mail:
| |
Collapse
|
221
|
Li G, Huo Y, Sun K, Wang X, Li H, Gao L, Ma B. 2D:4D indicates phimosis risk: A study on digit ratio and early foreskin development. Early Hum Dev 2016; 99:21-5. [PMID: 27390108 DOI: 10.1016/j.earlhumdev.2016.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/12/2016] [Accepted: 05/24/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Many researchers have adopted 2D:4D (second to fourth finger length ratio) as a noninvasive retrospective biomarker for prenatal androgen exposure in recent years. It is thought to be related to diverse traits including behavioral phenotypes, disease susceptibility, and development of urogenital system. OBJECTIVE To examine the relationship between 2D:4D and early foreskin development. METHODS We analyzed the digit ratio and foreskin condition in 176 cases (range 0-6years). The boys were divided into four groups according to their ages: group 1, neonates (below 28days, n=13); group 2, infants (1-12months, n=45); group 3, toddlers (1-2years old, n=42); group 4, preschool children (3-6years old, n=76). We measured the lengths of the second and fourth digits of the left and right hands. The foreskin status was classified into 4 types. Type I (phimosis), type II (partial phimosis), type III (adhesion of prepuce), type IV (normal). RESULTS The phimosis rate was 92.3%, 82.2%, 45.2%, and 38.7% in group 1 to group 4. In contrast, the proportion of normal foreskin increased from 0% in neonates to 13.2% in preschool children. The percentage of higher level of foreskin development shows a downward trend with the increase of digits ratio, and as the age grows, the percentage of normal foreskin cases also increases. CONCLUSIONS These results suggest that a higher R2D:4D (right hand 2D:4D) is a risk factor for phimosis in the early human development. Age is also a significant influence factor of foreskin conditions. Additional research is required to identify pathophysiologic mechanisms and to determine clinical significance.
Collapse
Affiliation(s)
- Guanjian Li
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China
| | - Ying Huo
- School of Public Health, Peking University, Beijing, China
| | - Ke Sun
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China; XinJiang Medical University, XinJiang, China
| | - Xiaodong Wang
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China; XinJiang Medical University, XinJiang, China
| | - Hao Li
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China; XinJiang Medical University, XinJiang, China
| | - Le Gao
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China
| | - Bin Ma
- The second Affiliated Hospital of XinJiang Medical University, XinJiang, China.
| |
Collapse
|
222
|
Seleit I, Bakry OA, El Repey HS, Ali R. Intrinsic versus Extrinsic Aging: A Histopathological, Morphometric and Immunohistochemical Study of Estrogen Receptor β and Androgen Receptor. Skin Pharmacol Physiol 2016; 29:178-89. [PMID: 27399919 DOI: 10.1159/000446662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/28/2016] [Indexed: 11/19/2022]
Abstract
Skin is a target organ of sex steroids which play important roles in skin health and disease. The aim of this study is to investigate the expression of estrogen receptor β (ERβ) and androgen receptor (AR) in human skin from different age groups for a better understanding of the hormonal regulation of skin aging. Using standard immunohistochemical techniques, biopsies of sun-unprotected and sun-protected skin were taken from 60 normal subjects. Sun-protected skin showed significantly higher immunoreactivity for ERβ and AR compared to sun-unprotected skin of all age groups. Significantly higher ERβ H score and percent of expression were associated with the 20-35 years age group compared to the groups that were 35-50 years and >50 years old (p < 0.02, p = 0.03, respectively) in sun-unprotected and sun-protected skin (p < 0.001, p = 0.01, respectively). AR H score showed a negative correlation with age (p = 0.04) with no significant difference in immunoreactivity in different age groups, either in sun-unprotected or sun-protected skin. There was also a significant correlation between ERβ H score and epidermal thickness in sun-unprotected (p = 0.04) and sun-protected skin (p = 0.04) in studied subjects regardless of age. The same relationships did not reach significance with AR expression. However, a significant positive correlation was detected between H scores and percent of expression of ERβ and AR in sun-unprotected (p = 0.01, p = 0.02, respectively) and sun-protected skin (p = 0.005, p = 0.02, respectively) regardless of age. In conclusion, both ERβ and AR decline gradually with intrinsic and extrinsic aging. This decline is more obvious with extrinsic aging. Further large-scaled studies are recommended to expand, validate and translate current findings to clinically significant, diagnostic and therapeutic applications. Molecular studies to investigate the probable ligand-independent action of both receptors are warranted. In addition, their gene expression patterns and associated signaling and metabolic pathways can also be tackled to provide a basis for further interventions in pathological processes that involve their dysregulation.
Collapse
Affiliation(s)
- Iman Seleit
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufiya University, Shibin El Koom, Egypt
| | | | | | | |
Collapse
|
223
|
Li YC, Luo ML, Guo H, Wang TT, Lin SR, Chen JB, Ma Q, Gu YL, Jiang ZM, Gui YT. Identification of NR0B1 as a novel androgen receptor co-repressor in mouse Sertoli cells. Int J Mol Med 2016; 38:853-60. [PMID: 27431683 DOI: 10.3892/ijmm.2016.2672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/22/2016] [Indexed: 11/06/2022] Open
Abstract
Nuclear receptor subfamily 0 group B member 1 (Nr0b1) is an atypical member of the nuclear receptor family that is predominantly expressed in mouse Sertoli cells (SCs). Mutations of NR0B1 in humans cause adrenal failure and hypogonadotropic hypogonadism. The targeted mutagenesis of Nr0b1 in mice has revealed a primary gonadal defect characterized by the overexpression of aromatase and cellular obstruction of the seminiferous tubules and efferent ductules, leading to germ cell death and infertility. The transgenic expression of Nr0b1 under the control of the Müllerian-inhibiting substance promoter (MIS-Nr0b1), which is selectively expressed in SCs, improves fertility. Testicular androgen receptor (AR) was also expressed in SCs. Many genes are directly regulated by androgen and its AR, which are involved in spermatogenesis and male infertility. As the association between NR0B1 and AR remains unclear in mouse SCs, we decided to further explore the relationship between them. In the present study, we have identified NR0B1 as a novel AR co-repressor in mouse SCs. Using RT‑qPCR and immunofluorescence, we determined that NR0B1 was mainly expressed in mouse SCs in an age-dependent manner from 2-8 weeks of age postnatally. The inhibition of the effects of AR on AR target genes by NR0B1, in an androgen‑dependent manner, was further demonstrated by western blot analysis and RT-qPCR in TM4 cells, a mouse Sertoli cell line. Finally, in vitro luciferase and co-immunoprecipitation assays validated that NR0B1, as an AR co-repressor, significantly inhibited the transcriptional activation of its target genes. These results suggest that novel inhibitory mechanisms underlie the effects of NR0B1 in modulating androgen-dependent gene transcription in mouse SCs.
Collapse
Affiliation(s)
- Yu-Chi Li
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Man-Ling Luo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Huan Guo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Tian-Tian Wang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Shou-Ren Lin
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Jian-Bo Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Qian Ma
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Yan-Li Gu
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Zhi-Mao Jiang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Yao-Ting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
224
|
Sideris S, Aoun F, Martinez CN, Latifyan S, Awada A, Costante G, Gil T. Role of corticosteroids in prostate cancer progression: implications for treatment strategy in metastatic castration-resistant patients. J Endocrinol Invest 2016; 39:729-38. [PMID: 26786788 DOI: 10.1007/s40618-016-0430-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/27/2022]
Abstract
Corticosteroid agents (CA) are widely used in the treatment of metastatic castration-resistant prostate cancer (mCRPC) either as concomitant treatment with active agents such as docetaxel, cabazitaxel and abiraterone or in a palliative setting, predominantly due to their anti-inflammatory activity. However, the chronic use of CA has numerous side effects, especially in case of steroid-induced adrenal insufficiency. Furthermore, the latest clinical and preclinical data demonstrate that CA themselves are likely to promote tumour progression in certain populations of patients with mCRPC. Therefore, the role of CA in advanced disease should be carefully weighed for each patient and their withdrawal should be considered in some patients. This is necessary, especially in clinical trials that need good performance status patients to evaluate the activity and the safety of emerging drugs in mCRPC that do not require the concurrent use of CA. In oncology, there is no consensus on an algorithm of gradual steroid tapering and frequently the approach to this procedure is empirical. An algorithm is presented in this article based on clinical observations. Prospective studies are necessary to evaluate the efficacy and safety of the above-proposed algorithm in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- S Sideris
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - F Aoun
- Urology Department, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - C N Martinez
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - S Latifyan
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - A Awada
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - G Costante
- Endocrinology Department, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium.
| | - T Gil
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| |
Collapse
|
225
|
Hsiao JJ, Smits MM, Ng BH, Lee J, Wright ME. Discovery Proteomics Identifies a Molecular Link between the Coatomer Protein Complex I and Androgen Receptor-dependent Transcription. J Biol Chem 2016; 291:18818-42. [PMID: 27365400 PMCID: PMC5009256 DOI: 10.1074/jbc.m116.732313] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
Aberrant androgen receptor (AR)-dependent transcription is a hallmark of human prostate cancers. At the molecular level, ligand-mediated AR activation is coordinated through spatial and temporal protein-protein interactions involving AR-interacting proteins, which we designate the “AR-interactome.” Despite many years of research, the ligand-sensitive protein complexes involved in ligand-mediated AR activation in prostate tumor cells have not been clearly defined. Here, we describe the development, characterization, and utilization of a novel human LNCaP prostate tumor cell line, N-AR, which stably expresses wild-type AR tagged at its N terminus with the streptavidin-binding peptide epitope (streptavidin-binding peptide-tagged wild-type androgen receptor; SBP-AR). A bioanalytical workflow involving streptavidin chromatography and label-free quantitative mass spectrometry was used to identify SBP-AR and associated ligand-sensitive cytosolic proteins/protein complexes linked to AR activation in prostate tumor cells. Functional studies verified that ligand-sensitive proteins identified in the proteomic screen encoded modulators of AR-mediated transcription, suggesting that these novel proteins were putative SBP-AR-interacting proteins in N-AR cells. This was supported by biochemical associations between recombinant SBP-AR and the ligand-sensitive coatomer protein complex I (COPI) retrograde trafficking complex in vitro. Extensive biochemical and molecular experiments showed that the COPI retrograde complex regulates ligand-mediated AR transcriptional activation, which correlated with the mobilization of the Golgi-localized ARA160 coactivator to the nuclear compartment of prostate tumor cells. Collectively, this study provides a bioanalytical strategy to validate the AR-interactome and define novel AR-interacting proteins involved in ligand-mediated AR activation in prostate tumor cells. Moreover, we describe a cellular system to study how compartment-specific AR-interacting proteins influence AR activation and contribute to aberrant AR-dependent transcription that underlies the majority of human prostate cancers.
Collapse
Affiliation(s)
- Jordy J Hsiao
- From the Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, Iowa 52242
| | - Melinda M Smits
- From the Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, Iowa 52242
| | - Brandon H Ng
- From the Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, Iowa 52242
| | - Jinhee Lee
- From the Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, Iowa 52242
| | - Michael E Wright
- From the Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
226
|
Sakkiah S, Ng HW, Tong W, Hong H. Structures of androgen receptor bound with ligands: advancing understanding of biological functions and drug discovery. Expert Opin Ther Targets 2016; 20:1267-82. [PMID: 27195510 DOI: 10.1080/14728222.2016.1192131] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Androgen receptor (AR) is a ligand-dependent transcription factor and a member of the nuclear receptor superfamily. It plays a vital role in male sexual development and regulates gene expression in various tissues, including prostate. Androgens are compounds that exert their biological effects via interaction with AR. Binding of androgens to AR initiates conformational changes in AR that affect binding of co-regulator proteins and DNA. AR agonists and antagonists are widely used in a variety of clinical applications (i.e. hypogonadism and prostate cancer therapy). AREAS COVERED This review provides a close look at structures of AR-ligand complexes and mutations in the receptor that have been revealed, discusses current challenges in the field, and sheds light on future directions. EXPERT OPINION AR is one of the primary targets for the treatment of prostate cancer, as AR antagonists inhibit prostate cancer growth. However, these drugs are not effective for long-term treatment and lead to castration-resistant prostate cancer. The structures of AR-ligand complexes are an invaluable scientific asset that enhances our understanding of biological functions and mechanisms of androgenic and anti-androgenic chemicals as well as promotes the discovery of superior drug candidates.
Collapse
Affiliation(s)
- Sugunadevi Sakkiah
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Hui Wen Ng
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Weida Tong
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Huixiao Hong
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| |
Collapse
|
227
|
Sun S, Zhong X, Wang C, Sun H, Wang S, Zhou T, Zou R, Lin L, Sun N, Sun G, Wu Y, Wang B, Song X, Cao L, Zhao Y. BAP18 coactivates androgen receptor action and promotes prostate cancer progression. Nucleic Acids Res 2016; 44:8112-28. [PMID: 27226492 PMCID: PMC5041452 DOI: 10.1093/nar/gkw472] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/14/2016] [Indexed: 01/28/2023] Open
Abstract
BPTF associated protein of 18 kDa (BAP18) has been reported as a component of MLL1-WDR5 complex. However, BAP18 is an uncharacterized protein. The detailed biological functions of BAP18 and underlying mechanisms have not been defined. Androgen receptor (AR), a member of transcription factor, plays an essential role in prostate cancer (PCa) and castration-resistant prostate cancer (CRPC) progression. Here, we demonstrate that BAP18 is identified as a coactivator of AR in Drosophilar experimental system and mammalian cells. BAP18 facilitates the recruitment of MLL1 subcomplex and AR to androgen-response element (ARE) of AR target genes, subsequently increasing histone H3K4 trimethylation and H4K16 acetylation. Knockdown of BAP18 attenuates cell growth and proliferation of PCa cells. Moreover, BAP18 depletion results in inhibition of xenograft tumor growth in mice even under androgen-depletion conditions. In addition, our data show that BAP18 expression in clinical PCa samples is higher than that in benign prostatic hyperplasia (BPH). Our data suggest that BAP18 as an epigenetic modifier regulates AR-induced transactivation and the function of BAP18 might be targeted in human PCa to promote tumor growth and progression to castration-resistance.
Collapse
Affiliation(s)
- Shiying Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Xinping Zhong
- Department of General Surgery, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Chunyu Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongmiao Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Shengli Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Renlong Zou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Lin Lin
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Ning Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Ge Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yi Wu
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Botao Wang
- School of Computer Science and Engineering, Northeastern University, Shenyang, Liaoning 110004, China
| | - Xiaoyu Song
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Liu Cao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yue Zhao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
228
|
Petkov PI, Schultz TW, Donner EM, Honma M, Morita T, Hamada S, Wakata A, Mishima M, Maniwa J, Todorov M, Kaloyanova E, Kotov S, Mekenyan OG. Integrated approach to testing and assessment for predicting rodent genotoxic carcinogenicity. J Appl Toxicol 2016; 36:1536-1550. [DOI: 10.1002/jat.3338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Petko I. Petkov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Terry W. Schultz
- College of Veterinary Medicine; The University of Tennessee; Knoxville TN 37996-4500 USA
| | - E. Maria Donner
- DuPont Haskell Global Centers for Health and Environmental Sciences, Newark; DE USA
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis; National Institute of Health Sciences; Tokyo Japan
| | - Takeshi Morita
- Division of Risk Assessment; National Institute of Health Sciences; Tokyo Japan
| | | | | | - Masayuki Mishima
- Chugai Pharmaceutical Co., Ltd., Fuji Gotemba Research Labs; Shizuoka Japan
| | - Jiro Maniwa
- Clinical Science Division, Research & Development AstraZeneca KK; Osaka Japan
| | - Milen Todorov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Elena Kaloyanova
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Stefan Kotov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Ovanes G. Mekenyan
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| |
Collapse
|
229
|
Yoo S, Choi SY, You D, Kim CS. New drugs in prostate cancer. Prostate Int 2016; 4:37-42. [PMID: 27358841 PMCID: PMC4916061 DOI: 10.1016/j.prnil.2016.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 01/21/2023] Open
Abstract
The standard primary treatment for advanced prostate cancer has been hormonal therapy since the 1940s. However, prostate cancer inevitably progresses to castration-resistant prostate cancer (CRPC) after a median duration of 18 months of androgen deprivation therapy. In patients with CRPC, docetaxel has been regarded as the standard treatment. However, survival advantages of docetaxel over other treatments are slim, and the need for new agents persists. In recent years, novel agents, including abiraterone, enzalutamide, cabazitaxel, radium-223, and sipuleucel-T, have been approved for the treatment of CRPC, and more such agents based on diverse mechanisms are under investigation or evaluation. In this article, the authors reviewed the current literature on recent advances in medical treatment of prostate cancer, especially CRPC. In addition, the authors elaborated on novel drugs for prostate cancer currently undergoing investigation and their mechanisms.
Collapse
Affiliation(s)
| | | | | | - Choung-Soo Kim
- Corresponding author. Department of Urology, Asan Medical Center, 388-1 Pungnap 2 dong, Songpa-gu, Seoul 138-736, South Korea.Department of UrologyAsan Medical Center388-1 Pungnap 2 dongSongpa-guSeoul138-736South Korea
| |
Collapse
|
230
|
Zarif JC, Miranti CK. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal 2016; 28:348-356. [PMID: 26829214 PMCID: PMC4788534 DOI: 10.1016/j.cellsig.2016.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.
Collapse
Affiliation(s)
- Jelani C Zarif
- The James Buchanan Brady Urological Institute at The Johns Hopkins University School of Medicine Baltimore, MD 21287, United States
| | - Cindy K Miranti
- Lab of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, United States.
| |
Collapse
|
231
|
Godoy G, Gakis G, Smith CL, Fahmy O. Effects of Androgen and Estrogen Receptor Signaling Pathways on Bladder Cancer Initiation and Progression. Bladder Cancer 2016; 2:127-137. [PMID: 27376135 PMCID: PMC4927898 DOI: 10.3233/blc-160052] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epidemiologic studies have long demonstrated clear differences in incidence and progression of bladder cancer between genders suggesting that the mechanisms of development and progression in these tumors have a strong association with steroid hormonal pathways. Such observations led to preclinical studies investigating the role of androgen and estrogen receptors, as well as their cognate hormones in bladder cancer initiation and progression. Using various in vitro cell line assays and in vivo mouse models, studies have elucidated different mechanisms and signaling pathways through which these steroid receptors may participate in this disease. More recently, RNA expression data from multiple studies revealed a luminal subtype of bladder cancer that exhibited an estrogen receptor signaling pathway, making it a strong candidate for further consideration of targeted therapies in the future. Despite the promising preclinical data demonstrating potential roles for both antiandrogen and antiestrogen strategies targeting these pathways in different stages of bladder cancer, only two clinical trials are currently active and accruing patients for such clinical studies. Targeted therapies in bladder cancer are a large unmet need and have the potential to change treatment paradigms and improve oncological outcomes of patients with bladder cancer.
Collapse
Affiliation(s)
- Guilherme Godoy
- Scott Department of Urology, Baylor College of Medicine , Houston, TX, USA
| | - Georgios Gakis
- Department of Urology, Eberhard-Karls University , Tuebingen, Germany
| | - Carolyn L Smith
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Omar Fahmy
- Department of Urology, Eberhard-Karls University , Tuebingen, Germany
| |
Collapse
|
232
|
Caffo O, Maines F, Veccia A, Kinspergher S, Galligioni E. Splice Variants of Androgen Receptor and Prostate Cancer. Oncol Rev 2016; 10:297. [PMID: 27471583 PMCID: PMC4943095 DOI: 10.4081/oncol.2016.297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/13/2016] [Indexed: 11/23/2022] Open
Abstract
Over the last ten years, two new-generation hormonal drugs and two chemotherapeutic agents have been approved for the treatment of metastatic castration-resistant prostate cancer. Unfortunately, some patients have primary resistance to them and the others eventually develop secondary resistance. It has recently been suggested that the presence of androgen receptor splice variants plays a leading role in the primary and secondary resistance to the new hormonal drugs, whereas their presence seem to have only a partial effect on the activity of the chemotherapeutic agents. The aim of this paper is to review the published data concerning the role of androgen receptor splice variants in prostate cancer biology, and their potential use as biomarkers when making therapeutic decisions.
Collapse
Affiliation(s)
- Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital, Trento, Italy
| | | | | | | | | |
Collapse
|
233
|
Esmaeili M, Jennek S, Ludwig S, Klitzsch A, Kraft F, Melle C, Baniahmad A. The tumor suppressor ING1b is a novel corepressor for the androgen receptor and induces cellular senescence in prostate cancer cells. J Mol Cell Biol 2016; 8:207-20. [PMID: 26993046 DOI: 10.1093/jmcb/mjw007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/10/2015] [Indexed: 12/28/2022] Open
Abstract
The androgen receptor (AR) signaling is critical for prostate cancer (PCa) progression to the castration-resistant stage with poor clinical outcome. Altered function of AR-interacting factors may contribute to castration-resistant PCa (CRPCa). Inhibitor of growth 1 (ING1) is a tumor suppressor that regulates various cellular processes including cell proliferation. Interestingly, ING1 expression is upregulated in senescent primary human prostate cells; however, its role in AR signaling in PCa was unknown. Using a proteomic approach by surface-enhanced laser desorption ionization-mass spectrometry (SELDI-MS) combined with immunological techniques, we provide here evidence that ING1b interacts in vivo with the AR. The interaction was confirmed by co-immunoprecipitation, in vitro GST-pull-down, and quantitative intracellular colocalization analyses. Functionally, ING1b inhibits AR-responsive promoters and endogenous key AR target genes in the human PCa LNCaP cells. Conversely, ING1b knockout (KO) mouse embryonic fibroblasts (MEFs) exhibit enhanced AR activity, suggesting that the interaction with ING1b represses the AR-mediated transcription. Also, data suggest that ING1b expression is downregulated in CRPCa cells compared with androgen-dependent LNCaP cells. Interestingly, its ectopic expression induces cellular senescence and reduces cell migration in both androgen-dependent and CRPCa cells. Intriguingly, ING1b can also inhibit androgen-induced growth in LNCaP cells in a similar manner as AR antagonists. Moreover, ING1b upregulates different cell cycle inhibitors including p27(KIP1), which is a novel target for ING1b. Taken together, our findings reveal a novel corepressor function of ING1b on various AR functions, thereby inhibiting PCa cell growth.
Collapse
Affiliation(s)
- Mohsen Esmaeili
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Susanne Jennek
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Susann Ludwig
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | | | - Florian Kraft
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Christian Melle
- Biomolecular Photonics Group, Jena University Hospital, Jena, Germany
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| |
Collapse
|
234
|
Shukla GC, Plaga AR, Shankar E, Gupta S. Androgen receptor-related diseases: what do we know? Andrology 2016; 4:366-81. [PMID: 26991422 DOI: 10.1111/andr.12167] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 01/09/2023]
Abstract
The androgen receptor (AR) and the androgen-AR signaling pathway play a significant role in male sexual differentiation and the development and function of male reproductive and non-reproductive organs. Because of AR's widely varied and important roles, its abnormalities have been identified in various diseases such as androgen insensitivity syndrome, spinal bulbar muscular atrophy, benign prostatic hyperplasia, and prostate cancer. This review provides an overview of the function of androgens and androgen-AR mediated diseases. In addition, the diseases delineated above are discussed with respect to their association with mutations and other post-transcriptional modifications in the AR. Finally, we present an introduction to the potential therapeutic application of most recent pharmaceuticals including miRNAs in prostate cancer that specifically target the transactivation function of the AR at post-transcriptional stages.
Collapse
Affiliation(s)
- G C Shukla
- Center of Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA.,Department of Biological Sciences, Cleveland State University, Cleveland, OH, USA
| | - A R Plaga
- Center of Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA.,Department of Biological Sciences, Cleveland State University, Cleveland, OH, USA
| | - E Shankar
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, OH, USA
| | - S Gupta
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, OH, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA.,Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, USA.,Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| |
Collapse
|
235
|
Udayakumar TS, Stoyanova R, Shareef MM, Mu Z, Philip S, Burnstein KL, Pollack A. Edelfosine Promotes Apoptosis in Androgen-Deprived Prostate Tumors by Increasing ATF3 and Inhibiting Androgen Receptor Activity. Mol Cancer Ther 2016; 15:1353-63. [PMID: 26944919 DOI: 10.1158/1535-7163.mct-15-0332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 02/15/2016] [Indexed: 12/31/2022]
Abstract
Edelfosine is a synthetic alkyl-lysophospholipid that possesses significant antitumor activity in several human tumor models. Here, we investigated the effects of edelfosine combined with androgen deprivation (AD) in LNCaP and VCaP human prostate cancer cells. This treatment regimen greatly decreased cell proliferation compared with single agent or AD alone, resulting in higher levels of apoptosis in LNCaP compared with VCaP cells. Edelfosine caused a dose-dependent decrease in AKT activity, but did not affect the expression of total AKT in either cell line. Furthermore, edelfosine treatment inhibited the expression of androgen receptor (AR) and was associated with an increase in activating transcription factor 3 (ATF3) expression levels, a stress response gene and a negative regulator of AR transactivation. ATF3 binds to AR after edelfosine + AD and represses the transcriptional activation of AR as demonstrated by PSA promoter studies. Knockdown of ATF3 using siRNA-ATF3 reversed the inhibition of PSA promoter activity, suggesting that the growth inhibition effect of edelfosine was ATF3 dependent. Moreover, expression of AR variant 7 (ARv7) and TMPRSS2-ERG fusion gene were greatly inhibited after combined treatment with AD and edelfosine in VCaP cells. In vivo experiments using an orthotopic LNCaP model confirmed the antitumor effects of edelfosine + AD over the individual treatments. A significant decrease in tumor volume and PSA levels was observed when edelfosine and AD were combined, compared with edelfosine alone. Edelfosine shows promise in combination with AD for the treatment of prostate cancer patients. Mol Cancer Ther; 15(6); 1353-63. ©2016 AACR.
Collapse
Affiliation(s)
- Thirupandiyur S Udayakumar
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Radka Stoyanova
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Mohammed M Shareef
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Zhaomei Mu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sakhi Philip
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kerry L Burnstein
- Department of Molecular and Cellular Pharmacology, University of Miami, Miami, Florida
| | - Alan Pollack
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
236
|
Wu M, Kim SH, Datta I, Levin A, Dyson G, Li J, Kaypee S, Swamy MM, Gupta N, Kwon HJ, Menon M, Kundu TK, Reddy GPV. Hydrazinobenzoylcurcumin inhibits androgen receptor activity and growth of castration-resistant prostate cancer in mice. Oncotarget 2016; 6:6136-50. [PMID: 25704883 PMCID: PMC4467427 DOI: 10.18632/oncotarget.3346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/20/2015] [Indexed: 01/09/2023] Open
Abstract
There is a critical need for therapeutic agents that can target the amino-terminal domain (NTD) of androgen receptor (AR) for the treatment of castration-resistant prostate cancer (CRPC). Calmodulin (CaM) binds to the AR NTD and regulates AR activity. We discovered that Hydrazinobenzoylcurcumin (HBC), which binds exclusively to CaM, inhibited AR activity. HBC abrogated AR interaction with CaM, suppressed phosphorylation of AR Serine81, and blocked the binding of AR to androgen-response elements. RNA-Seq analysis identified 57 androgen-regulated genes whose expression was significantly (p ≤ 0.002) altered in HBC treated cells as compared to controls. Oncomine analysis revealed that genes repressed by HBC are those that are usually overexpressed in prostate cancer (PCa) and genes stimulated by HBC are those that are often down-regulated in PCa, suggesting a reversing effect of HBC on androgen-regulated gene expression associated with PCa. Ingenuity Pathway Analysis revealed a role of HBC affected genes in cellular functions associated with proliferation and survival. HBC was readily absorbed into the systemic circulation and inhibited the growth of xenografted CRPC tumors in nude mice. These observations demonstrate that HBC inhibits AR activity by targeting the AR NTD and suggest potential usefulness of HBC for effective treatment of CRPC.
Collapse
Affiliation(s)
- Min Wu
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Sahn-Ho Kim
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Indrani Datta
- Bioinformatics Core, Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Albert Levin
- Bioinformatics Core, Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Gregory Dyson
- Biostatistics Core, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jing Li
- Pharmacology Core, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Stephanie Kaypee
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | - M Mahadeva Swamy
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | - Nilesh Gupta
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Ho Jeong Kwon
- Department of Biotechnology, Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Mani Menon
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | | |
Collapse
|
237
|
Karacosta LG, Kuroski LA, Hofmann WA, Azabdaftari G, Mastri M, Gocher AM, Dai S, Hoste AJ, Edelman AM. Nucleoporin 62 and Ca(2+)/calmodulin dependent kinase kinase 2 regulate androgen receptor activity in castrate resistant prostate cancer cells. Prostate 2016; 76:294-306. [PMID: 26552607 DOI: 10.1002/pros.23121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/14/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Re-activation of the transcriptional activity of the androgen receptor (AR) is an important factor mediating progression from androgen-responsive to castrate-resistant prostate cancer (CRPC). However, the mechanisms regulating AR activity in CRPC remain incompletely understood. Ca(2+) /calmodulin-dependent kinase kinase (CaMKK) 2 was previously shown to regulate AR activity in androgen-responsive prostate cancer cells. Our objective was to further explore the basis of this regulation in CRPC cells. METHODS The abundance of CaMKK2 in nuclear fractions of androgen-responsive prostate cancer and CRPC, cells were determined by subcellular fractionation and Western blotting. CaMKK2 association with nuclear pore complexes (NPCs) and nucleoporins (Nups) including Nup62, were imaged by structured illumination and super-resolution fluorescence microscopy and co-immunoprecipitation, respectively. The abundance and subcellular localization of CaMKK2 and Nup62 in human clinical specimens of prostate cancer was visualized by immunohistochemistry. The role of Nups in the growth and viability of CRPC cells was assessed by RNA interference and cell counting. The involvement of CaMKK2 and Nup62 in regulating AR transcriptional activity was addressed by RNA interference, chromatin immunoprecipitation, androgen response element reporter assay, and Western blotting. RESULTS CaMKK2 was expressed at higher levels in the nuclear fraction of CPRC C4-2 cells, than in that of androgen-responsive LNCaP cells. In C4-2 cells, CaMKK2 associated with NPCs of the nuclear envelope and physically interacted with Nup62. CaMKK2 and Nup62 demonstrated pronounced, and similar increases in both expression and perinuclear/nuclear localization in human clinical specimens of advanced prostate cancer relative to normal prostate. Knockdown of Nup62, but not of Nups, 98 or 88, reduced growth and viability of C4-2 cells. Knockdown of Nup62 produced a greater reduction of the growth and viability of C4-2 cells than of non-neoplastic RWPE-1 prostatic cells. Nup62, CaMKK2, and the AR were recruited to androgen response elements of the AR target genes, prostate specific antigen, and transmembrane protease, serine 2. Knockdown of CaMKK2 and Nup62 reduced prostate specific antigen expression and AR transcriptional activity driven by androgen response elements from the prostate-specific probasin gene promoter. CONCLUSION Nup62 and CaMKK2 are required for optimal AR transcriptional activity and a potential mechanism for AR re-activation in CRPC.
Collapse
Affiliation(s)
- Loukia G Karacosta
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| | - Laura A Kuroski
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| | - Wilma A Hofmann
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York
| | - Gissou Azabdaftari
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Michalis Mastri
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, New York
| | - Angela M Gocher
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| | - Shuhang Dai
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| | - Allen J Hoste
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York
| | - Arthur M Edelman
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
238
|
DePriest AD, Fiandalo MV, Schlanger S, Heemers F, Mohler JL, Liu S, Heemers HV. Regulators of Androgen Action Resource: a one-stop shop for the comprehensive study of androgen receptor action. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:bav125. [PMID: 26876983 PMCID: PMC4752970 DOI: 10.1093/database/bav125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
Androgen receptor (AR) is a ligand-activated transcription factor that is the main target for treatment of non-organ-confined prostate cancer (CaP). Failure of life-prolonging AR-targeting androgen deprivation therapy is due to flexibility in steroidogenic pathways that control intracrine androgen levels and variability in the AR transcriptional output. Androgen biosynthesis enzymes, androgen transporters and AR-associated coregulators are attractive novel CaP treatment targets. These proteins, however, are characterized by multiple transcript variants and isoforms, are subject to genomic alterations, and are differentially expressed among CaPs. Determining their therapeutic potential requires evaluation of extensive, diverse datasets that are dispersed over multiple databases, websites and literature reports. Mining and integrating these datasets are cumbersome, time-consuming tasks and provide only snapshots of relevant information. To overcome this impediment to effective, efficient study of AR and potential drug targets, we developed the Regulators of Androgen Action Resource (RAAR), a non-redundant, curated and user-friendly searchable web interface. RAAR centralizes information on gene function, clinical relevance, and resources for 55 genes that encode proteins involved in biosynthesis, metabolism and transport of androgens and for 274 AR-associated coregulator genes. Data in RAAR are organized in two levels: (i) Information pertaining to production of androgens is contained in a ‘pre-receptor level’ database, and coregulator gene information is provided in a ‘post-receptor level’ database, and (ii) an ‘other resources’ database contains links to additional databases that are complementary to and useful to pursue further the information provided in RAAR. For each of its 329 entries, RAAR provides access to more than 20 well-curated publicly available databases, and thus, access to thousands of data points. Hyperlinks provide direct access to gene-specific entries in the respective database(s). RAAR is a novel, freely available resource that provides fast, reliable and easy access to integrated information that is needed to develop alternative CaP therapies. Database URL: http://www.lerner.ccf.org/cancerbio/heemers/RAAR/search/
Collapse
Affiliation(s)
| | | | - Simon Schlanger
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | | | - James L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hannelore V Heemers
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA Department of Urology Department of Hematology/Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
239
|
Wadosky KM, Koochekpour S. Therapeutic Rationales, Progresses, Failures, and Future Directions for Advanced Prostate Cancer. Int J Biol Sci 2016; 12:409-26. [PMID: 27019626 PMCID: PMC4807161 DOI: 10.7150/ijbs.14090] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/15/2015] [Indexed: 02/07/2023] Open
Abstract
Patients with localized prostate cancer (PCa) have several therapeutic options with good prognosis. However, survival of patients with high-risk, advanced PCa is significantly less than patients with early-stage, organ-confined disease. Testosterone and other androgens have been directly linked to PCa progression since 1941. In this review, we chronicle the discoveries that led to modern therapeutic strategies for PCa. Specifically highlighted is the biology of androgen receptor (AR), the nuclear receptor transcription factor largely responsible for androgen-stimulated and castrate-recurrent (CR) PCa. Current PCa treatment paradigms can be classified into three distinct but interrelated categories: targeting AR at pre-receptor, receptor, or post-receptor signaling. The continuing challenge of disease relapse as CR and/or metastatic tumors, destined to occur within three years of the initial treatment, is also discussed. We conclude that the success of PCa therapies in the future depends on targeting molecular mechanisms underlying tumor recurrence that still may affect AR at pre-receptor, receptor, and post-receptor levels.
Collapse
Affiliation(s)
| | - Shahriar Koochekpour
- ✉ Corresponding author: Dr. Shahriar Koochekpour, Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA, Telephone: 716-845-3345; Fax: 716-845-1698;
| |
Collapse
|
240
|
Matsushita S, Suzuki K, Ogino Y, Hino S, Sato T, Suyama M, Matsumoto T, Omori A, Inoue S, Yamada G. Androgen Regulates Mafb Expression Through its 3'UTR During Mouse Urethral Masculinization. Endocrinology 2016; 157:844-57. [PMID: 26636186 DOI: 10.1210/en.2015-1586] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
External genitalia are prominent organs showing hormone-dependent sexual differentiation. Androgen is an essential regulator of masculinization of the genital tubercle, which is the anlage of external genitalia. We have previously shown that v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB) is an androgen-inducible regulator of embryonic urethral masculinization in mice. However, it remains unclear how androgen regulates Mafb expression. The current study suggests that the Mafb 3' untranslated region (UTR) is an essential region for its regulation by androgen. We identified 2 functional androgen response elements (AREs) in Mafb 3'UTR. Androgen receptor is bound to such AREs in 3'UTR during urethral masculinization. In addition to 3'UTR, Mafb 5'UTR also showed androgen responsiveness. Moreover, we also demonstrated that β-catenin, one of genital tubercle masculinization factors, may be an additional regulator of Mafb expression during urethral masculinization. This study provides insights to elucidate mechanisms of gene regulation through AREs present in Mafb 3'UTR for a better understanding of the processes of urethral masculinization.
Collapse
Affiliation(s)
- Shoko Matsushita
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yukiko Ogino
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinjiro Hino
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tetsuya Sato
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Mikita Suyama
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takahiro Matsumoto
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Akiko Omori
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Satoshi Inoue
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Gen Yamada
- Department of Developmental Genetics (S.M., K.S., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Okazaki Institute for Integrative Bioscience (Y.O.), National Institute for Basic Biology, National Institutes of Natural Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8787, Japan; Department of Medical Cell Biology (S.H.), Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Institute of Biomedical Sciences (T.M.), University of Tokushima Graduate School, Tokushima 770-8503, Japan; Venetian Institute of Molecular Medicine (A.O.), 35129 Padua, Italy; and Department of Anti-Aging Medicine (S.I.), Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
241
|
Ketefian A, Jones MR, Krauss RM, Chen YDI, Legro RS, Azziz R, Goodarzi MO. Association study of androgen signaling pathway genes in polycystic ovary syndrome. Fertil Steril 2016; 105:467-73.e4. [PMID: 26493122 PMCID: PMC4744098 DOI: 10.1016/j.fertnstert.2015.09.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To evaluate genes involved in androgen receptor (AR) signaling as candidate genes for polycystic ovary syndrome (PCOS). DESIGN Two groups of women with PCOS and control women (discovery and replication cohorts), were genotyped for single-nucleotide polymorphisms (SNPs) in eight genes for AR chaperones and co-chaperones: HSPA1A, HSPA8, ST13, STIP1, PTGES3, FKBP4, BAG1, and STUB1. Single-nucleotide polymorphisms were tested for association with PCOS status and with androgenic and metabolic parameters. SETTING Tertiary referral center. PATIENT(S) Discovery cohort: 354 women with PCOS and 161 control women. Replication cohort: 397 women with PCOS and 306 control women. INTERVENTION(S) Phenotypic and genotypic assessment. MAIN OUTCOME MEASURE(S) Single-nucleotide polymorphism genotypes, association with PCOS status, and androgenic and metabolic parameters. RESULT(S) In the discovery cohort, FKBP4 SNPs rs2968909 and rs4409904 were associated with lower odds of PCOS. This finding was not confirmed in the replication cohort analysis; however, when combining the two cohorts, rs4409904 was associated with lower odds of PCOS. In subjects with PCOS in the replication cohort as well as in the combined cohort, rs2968909 was associated with lower body mass index. CONCLUSION(S) Single-nucleotide polymorphisms in FKBP4, which codes for the AR co-chaperone FKBP52, may be associated with PCOS and body mass index in patients with PCOS. The remaining genes studied do not seem to be major contributors to the development of PCOS. These findings warrant confirmation in future studies, and genes encoding other androgen pathway components remain to be studied.
Collapse
Affiliation(s)
- Aline Ketefian
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California; Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Michelle R Jones
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ronald M Krauss
- Children's Hospital Oakland Research Institute, Oakland, California
| | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Richard S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Ricardo Azziz
- Departments of Obstetrics and Gynecology and Medicine, Georgia Regents University, Augusta, Georgia
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
242
|
Chandrasekar T, Yang JC, Gao AC, Evans CP. Mechanisms of resistance in castration-resistant prostate cancer (CRPC). Transl Androl Urol 2016; 4:365-80. [PMID: 26814148 PMCID: PMC4708226 DOI: 10.3978/j.issn.2223-4683.2015.05.02] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite advances in prostate cancer diagnosis and management, morbidity from prostate cancer remains high. Approximately 20% of men present with advanced or metastatic disease, while 29,000 men continue to die of prostate cancer each year. Androgen deprivation therapy (ADT) has been the standard of care for initial management of advanced or metastatic prostate cancer since Huggins and Hodges first introduced the concept of androgen-dependence in 1972, but progression to castration-resistant prostate cancer (CRPC) occurs within 2-3 years of initiation of ADT. CRPC, previously defined as hormone-refractory prostate cancer, is now understood to still be androgen dependent. Multiple mechanisms of resistance help contribute to the progression to castration resistant disease, and the androgen receptor (AR) remains an important driver in this progression. These mechanisms include AR amplification and hypersensitivity, AR mutations leading to promiscuity, mutations in coactivators/corepressors, androgen-independent AR activation, and intratumoral and alternative androgen production. More recently, identification of AR variants (ARVs) has been established as another mechanism of progression to CRPC. Docetaxel chemotherapy has historically been the first-line treatment for CRPC, but in recent years, newer agents have been introduced that target some of these mechanisms of resistance, thereby providing additional survival benefit. These include AR signaling inhibitors such as enzalutamide (Xtandi, ENZA, MDV-3100) and CYP17A1 inhibitors such as abiraterone acetate (Zytiga). Ultimately, these agents will also fail to suppress CRPC. While some of the mechanisms by which these agents fail are unique, many share similarities to the mechanisms contributing to CRPC progression. Understanding these mechanisms of resistance to ADT and currently approved CRPC treatments will help guide future research into targeted therapies.
Collapse
Affiliation(s)
| | - Joy C Yang
- Department of Urology, University of California, Davis, CA, USA
| | - Allen C Gao
- Department of Urology, University of California, Davis, CA, USA
| | | |
Collapse
|
243
|
Harada N, Katsuki T, Takahashi Y, Masuda T, Yoshinaga M, Adachi T, Izawa T, Kuwamura M, Nakano Y, Yamaji R, Inui H. Androgen receptor silences thioredoxin-interacting protein and competitively inhibits glucocorticoid receptor-mediated apoptosis in pancreatic β-Cells. J Cell Biochem 2016; 116:998-1006. [PMID: 25639671 DOI: 10.1002/jcb.25054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/18/2014] [Indexed: 01/09/2023]
Abstract
Androgen receptor (AR) is known to bind to the same cis-element that glucocorticoid receptor (GR) binds to. However, the effects of androgen signaling on glucocorticoid signaling have not yet been elucidated. Here, we investigated the effects of testosterone on dexamethasone (DEX, a synthetic glucocorticoid)-induced apoptosis of pancreatic β-cells, which might be involved in the pathogenesis of type 2 diabetes mellitus in males. We used INS-1 #6 cells, which were isolated from the INS-1 pancreatic β-cell line and which express high levels of AR. Testosterone and dihydrotestosterone inhibited apoptosis induced by DEX in INS-1 #6 cells. AR knockdown and the AR antagonist hydroxyflutamide each diminished the anti-apoptotic effects of testosterone. AR was localized in the nucleus of both INS-1 #6 cells and pancreatic β-cells of male rats. Induction of thioredoxin-interacting protein (TXNIP) is known to cause pro-apoptotic effects in β-cells. Testosterone suppressed the DEX-induced increase of TXNIP at the transcriptional level. A Chromatin immunoprecipitation assays showed that both AR and GR competitively bound to the TXNIP promoter in ligand-dependent manners. Recombinant DNA-binding domain of AR bound to the same cis-element of the TXNIP promoter that GR binds to. Our results show that AR and GR competitively bind to the same cis-element of TXNIP promoter as a silencer and enhancer, respectively. These results indicate that androgen signaling functionally competes with glucocorticoid signaling in pancreatic β-cell apoptosis.
Collapse
Affiliation(s)
- Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, 5998531, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Joseph JF, Parr MK. Synthetic androgens as designer supplements. Curr Neuropharmacol 2016; 13:89-100. [PMID: 26074745 PMCID: PMC4462045 DOI: 10.2174/1570159x13666141210224756] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/25/2014] [Accepted: 10/25/2014] [Indexed: 01/02/2023] Open
Abstract
Anabolic androgenic steroids (AAS) are some of the most common performance
enhancing drugs (PED) among society. Despite the broad spectrum of adverse effects and legal
consequences, AAS are illicitly marketed and distributed in many countries. To circumvent existing
laws, the chemical structure of AAS is modified and these designer steroids are sold as nutritional
supplements mainly over the Internet. Several side effects are linked with AAS abuse. Only little is
known about the pharmacological effects and metabolism of unapproved steroids due to the absence
of clinical studies. The large number of designer steroid findings in dietary supplements and the
detection of new compounds combined with legal loopholes for their distribution in many countries
show that stricter regulations and better information policy are needed.
Collapse
Affiliation(s)
- Jan Felix Joseph
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Maria Kristina Parr
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| |
Collapse
|
245
|
|
246
|
Luo M, Li Y, Guo H, Lin S, Chen J, Ma Q, Gu Y, Jiang Z, Gui Y. Protein Arginine Methyltransferase 6 Involved in Germ Cell Viability during Spermatogenesis and Down-Regulated by the Androgen Receptor. Int J Mol Sci 2015; 16:29467-81. [PMID: 26690413 PMCID: PMC4691129 DOI: 10.3390/ijms161226186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 02/05/2023] Open
Abstract
Androgens and the androgen receptor (AR) are of great importance to spermatogenesis and male fertility. AR knockout (ARKO) mice display a complete insensitivity to androgens and male infertility; however, the exact molecular mechanism for this effect remains unclear. In this study, we found that the expression levels of Prmt6 mRNA and protein were significantly up-regulated in the testes of ARKO mice compared to wild type (WT) mice. PRMT6 was principally localized to the nucleus of spermatogonia and spermatocytes by immunofluorescence staining. Furthermore, luciferase assay data showed that AR together with testosterone treatment suppressed Prmt6 transcription via binding to the androgen-responsive element (ARE) of the Prmt6 promoter. Moreover, knockdown of Prmt6 suppressed germ cells migration and promoted apoptosis. In addition, both of these cellular activities could not be enhanced by testosterone treatment. Taken together, these data indicate that PRMT6, which was down-regulated by AR and influenced cell migration and apoptosis of germ cells, could play a potentially important role in spermatogenesis.
Collapse
Affiliation(s)
- Manling Luo
- Department of Physiology, Shantou University Medical College, Shantou 515041, China.
| | - Yuchi Li
- Department of Physiology, Shantou University Medical College, Shantou 515041, China.
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| | - Huan Guo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
- Department of Surgery, Guangzhou Medical University, Guangzhou 510182, China.
| | - Shouren Lin
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| | - Jianbo Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
- Department of Surgery, Anhui Medical University, Hefei 230032, China.
| | - Qian Ma
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| | - Yanli Gu
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| | - Zhimao Jiang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen 518036, China.
| |
Collapse
|
247
|
Assimon VA, Southworth DR, Gestwicki JE. Specific Binding of Tetratricopeptide Repeat Proteins to Heat Shock Protein 70 (Hsp70) and Heat Shock Protein 90 (Hsp90) Is Regulated by Affinity and Phosphorylation. Biochemistry 2015; 54:7120-31. [PMID: 26565746 PMCID: PMC4714923 DOI: 10.1021/acs.biochem.5b00801] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heat shock protein 70 (Hsp70) and heat shock protein 90 (Hsp90) require the help of tetratricopeptide repeat (TPR) domain-containing cochaperones for many of their functions. Each monomer of Hsp70 or Hsp90 can interact with only a single TPR cochaperone at a time, and each member of the TPR cochaperone family brings distinct functions to the complex. Thus, competition for TPR binding sites on Hsp70 and Hsp90 appears to shape chaperone activity. Recent structural and biophysical efforts have improved our understanding of chaperone-TPR contacts, focusing on the C-terminal EEVD motif that is present in both chaperones. To better understand these important protein-protein interactions on a wider scale, we measured the affinity of five TPR cochaperones, CHIP, Hop, DnaJC7, FKBP51, and FKBP52, for the C-termini of four members of the chaperone family, Hsc70, Hsp72, Hsp90α, and Hsp90β, in vitro. These studies identified some surprising selectivity among the chaperone-TPR pairs, including the selective binding of FKBP51/52 to Hsp90α/β. These results also revealed that other TPR cochaperones are only able to weakly discriminate between the chaperones or between their paralogs. We also explored whether mimicking phosphorylation of serine and threonine residues near the EEVD motif might impact affinity and found that pseudophosphorylation had selective effects on binding to CHIP but not other cochaperones. Together, these findings suggest that both intrinsic affinity and post-translational modifications tune the interactions between the Hsp70 and Hsp90 proteins and the TPR cochaperones.
Collapse
Affiliation(s)
| | | | - Jason E. Gestwicki
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, CA 94158
| |
Collapse
|
248
|
Yang Y, Jia D, Kim H, Abd Elmageed ZY, Datta A, Davis R, Srivastav S, Moroz K, Crawford BE, Moparty K, Thomas R, Hudson RS, Ambs S, Abdel-Mageed AB. Dysregulation of miR-212 Promotes Castration Resistance through hnRNPH1-Mediated Regulation of AR and AR-V7: Implications for Racial Disparity of Prostate Cancer. Clin Cancer Res 2015; 22:1744-56. [PMID: 26553749 DOI: 10.1158/1078-0432.ccr-15-1606] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/11/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE The causes of disproportionate incidence and mortality of prostate cancer among African Americans (AA) remain elusive. The purpose of this study was to investigate the mechanistic role and assess clinical utility of the splicing factor heterogeneous nuclear ribonucleoprotein H1 (hnRNP H1) in prostate cancer progression among AA men. EXPERIMENTAL DESIGN We employed an unbiased functional genomics approach coupled with suppressive subtractive hybridization (SSH) and custom cDNA microarrays to identify differentially expressed genes in microdissected tumors procured from age- and tumor grade-matched AA and Caucasian American (CA) men. Validation analysis was performed in independent cohorts and tissue microarrays. The underlying mechanisms of hnRNPH1 regulation and its impact on androgen receptor (AR) expression and tumor progression were explored. RESULTS Aberrant coexpression of AR and hnRNPH1 and downregulation of miR-212 were detected in prostate tumors and correlate with disease progression in AA men compared with CA men. Ectopic expression of miR-212 mimics downregulated hnRNPH1 transcripts, which in turn reduced expression of AR and its splice variant AR-V7 (or AR3) in prostate cancer cells. hnRNPH1 physically interacts with AR and steroid receptor coactivator-3 (SRC-3) and primes activation of androgen-regulated genes in a ligand-dependent and independent manner. siRNA silencing of hnRNPH1 sensitized prostate cancer cells to bicalutamide and inhibited prostate tumorigenesis in vivo CONCLUSIONS Our findings define novel roles for hnRNPH1 as a putative oncogene, splicing factor, and an auxiliary AR coregulator. Targeted disruption of the hnRNPH1-AR axis may have therapeutic implications to improve clinical outcomes in patients with advanced prostate cancer, especially among AA men.
Collapse
Affiliation(s)
- Yijun Yang
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Dingwu Jia
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Hogyoung Kim
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Amrita Datta
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Rodney Davis
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sudesh Srivastav
- Department of Biostatistics, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Krzysztof Moroz
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana. Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Byron E Crawford
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Krishnarao Moparty
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana. Division of Urology, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana
| | - Raju Thomas
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana. Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Robert S Hudson
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana. Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana. Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
249
|
The Molecular Taxonomy of Primary Prostate Cancer. Cell 2015; 163:1011-25. [PMID: 26544944 PMCID: PMC4695400 DOI: 10.1016/j.cell.2015.10.025] [Citation(s) in RCA: 2192] [Impact Index Per Article: 243.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/14/2015] [Accepted: 10/06/2015] [Indexed: 12/12/2022]
Abstract
There is substantial heterogeneity among primary prostate cancers, evident in the spectrum of molecular abnormalities and its variable clinical course. As part of The Cancer Genome Atlas (TCGA), we present a comprehensive molecular analysis of 333 primary prostate carcinomas. Our results revealed a molecular taxonomy in which 74% of these tumors fell into one of seven subtypes defined by specific gene fusions (ERG, ETV1/4, and FLI1) or mutations (SPOP, FOXA1, and IDH1). Epigenetic profiles showed substantial heterogeneity, including an IDH1 mutant subset with a methylator phenotype. Androgen receptor (AR) activity varied widely and in a subtype-specific manner, with SPOP and FOXA1 mutant tumors having the highest levels of AR-induced transcripts. 25% of the prostate cancers had a presumed actionable lesion in the PI3K or MAPK signaling pathways, and DNA repair genes were inactivated in 19%. Our analysis reveals molecular heterogeneity among primary prostate cancers, as well as potentially actionable molecular defects.
Collapse
|
250
|
Wang Z, Kim J, Teng Y, Ding HF, Zhang J, Hai T, Cowell JK, Yan C. Loss of ATF3 promotes hormone-induced prostate carcinogenesis and the emergence of CK5(+)CK8(+) epithelial cells. Oncogene 2015; 35:3555-64. [PMID: 26522727 PMCID: PMC4853303 DOI: 10.1038/onc.2015.417] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/28/2015] [Accepted: 10/05/2015] [Indexed: 12/25/2022]
Abstract
Steroid sex hormones can induce prostate carcinogenesis, and are thought to contribute to the development of prostate cancer during aging. However, the mechanism for hormone-induced prostate carcinogenesis remains elusive. Here we report that activating transcription factor 3 (ATF3) – a broad stress sensor – suppressed hormone-induced prostate carcinogenesis in mice. While implantation of testosterone and estradiol (T+E2) pellets for 2 months in wild-type mice rarely induced prostatic intraepithelial neoplasia (PIN) in dorsal prostates (1 out of 8 mice), loss of ATF3 led to the appearance of not only PIN but also invasive lesions in almost all examined animals. The enhanced carcinogenic effects of hormones on ATF3-deficient prostates did not appear to be caused by a change in estrogen signaling, but were more likely a consequence of elevated androgen signaling that stimulated differentiation of prostatic basal cells into transformation-preferable luminal cells. Indeed, we found that hormone-induced lesions in ATF3-knockout mice often contained cells with both basal and luminal characteristics, such as p63+ cells (a basal cell marker) showing luminal-like morphology, or cells double-stained with basal (CK5+) and luminal (CK8+) markers. Consistent with these findings, low ATF3 expression was found to be a poor prognostic marker for prostate cancer in a cohort of 245 patients. Our results thus support that ATF3 is a tumor suppressor in prostate cancer.
Collapse
Affiliation(s)
- Z Wang
- GRU Cancer Center, Georgia Regents University, Augusta, GA, USA.,Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | - J Kim
- Department of Statistics, Sungkyunkwan University, Seoul, South Korea
| | - Y Teng
- GRU Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - H-F Ding
- GRU Cancer Center, Georgia Regents University, Augusta, GA, USA.,Department of Pathology, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| | - J Zhang
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - T Hai
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH, USA
| | - J K Cowell
- GRU Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - C Yan
- GRU Cancer Center, Georgia Regents University, Augusta, GA, USA.,Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|