1
|
Walton KL, Goney MP, Peppas Z, Stringer JM, Winship A, Hutt K, Goodchild G, Maskey S, Chan KL, Brûlé E, Bernard DJ, Stocker WA, Harrison CA. Inhibin Inactivation in Female Mice Leads to Elevated FSH Levels, Ovarian Overstimulation, and Pregnancy Loss. Endocrinology 2022; 163:6543938. [PMID: 35255139 PMCID: PMC9272799 DOI: 10.1210/endocr/bqac025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 11/21/2022]
Abstract
Inhibins are members of the transforming growth factor-β family, composed of a common α-subunit disulfide-linked to 1 of 2 β-subunits (βA in inhibin A or βB in inhibin B). Gonadal-derived inhibin A and B act in an endocrine manner to suppress the synthesis of follicle-stimulating hormone (FSH) by pituitary gonadotrope cells. Roles for inhibins beyond the pituitary, however, have proven difficult to delineate because deletion of the inhibin α-subunit gene (Inha) results in unconstrained expression of activin A and activin B (homodimers of inhibin β-subunits), which contribute to gonadal tumorigenesis and lethal cachectic wasting. Here, we generated mice with a single point mutation (Arg233Ala) in Inha that prevents proteolytic processing and the formation of bioactive inhibin. In vitro, this mutation blocked inhibin maturation and bioactivity, without perturbing activin production. Serum FSH levels were elevated 2- to 3-fold in InhaR233A/R233A mice due to the loss of negative feedback from inhibins, but no pathological increase in circulating activins was observed. While inactivation of inhibin A and B had no discernible effect on male reproduction, female InhaR233A/R233A mice had increased FSH-dependent follicle development and enhanced natural ovulation rates. Nevertheless, inhibin inactivation resulted in significant embryo-fetal resorptions and severe subfertility and was associated with disrupted maternal ovarian function. Intriguingly, heterozygous Inha+/R233A females had significantly enhanced fecundity, relative to wild-type littermates. These studies have revealed novel effects of inhibins in the establishment and maintenance of pregnancy and demonstrated that partial inactivation of inhibin A/B is an attractive approach for enhancing female fertility.
Collapse
Affiliation(s)
- Kelly L Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
- Correspondence: Kelly L Walton, PhD, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia 4072.
| | - Monica P Goney
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Zoe Peppas
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jessica M Stringer
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Amy Winship
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Karla Hutt
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Shreya Maskey
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Karen L Chan
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Emilie Brûlé
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Daniel J Bernard
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - William A Stocker
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Australia
| | - Craig A Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Correspondence: Craig A Harrison, PhD, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia 3168.
| |
Collapse
|
2
|
Houston BJ, O'Connor AE, Wang D, Goodchild G, Merriner DJ, Luan H, Conrad DF, Nagirnaja L, Aston KI, Kliesch S, Wyrwoll MJ, Friedrich C, Tüttelmann F, Harrison C, O'Bryan MK, Walton K. Human INHBB Gene Variant (c.1079T>C:p.Met360Thr) Alters Testis Germ Cell Content, but Does Not Impact Fertility in Mice. Endocrinology 2022; 163:6504015. [PMID: 35022746 DOI: 10.1210/endocr/bqab269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
Testicular-derived inhibin B (α/β B dimers) acts in an endocrine manner to suppress pituitary production of follicle-stimulating hormone (FSH), by blocking the actions of activins (β A/B/β A/B dimers). Previously, we identified a homozygous genetic variant (c.1079T>C:p.Met360Thr) arising from uniparental disomy of chromosome 2 in the INHBB gene (β B-subunit of inhibin B and activin B) in a man suffering from infertility (azoospermia). In this study, we aimed to test the causality of the p.Met360Thr variant in INHBB and testis function. Here, we used CRISPR/Cas9 technology to generate InhbbM364T/M364T mice, where mouse INHBB p.Met364 corresponds with human p.Met360. Surprisingly, we found that the testes of male InhbbM364T/M364T mutant mice were significantly larger compared with those of aged-matched wildtype littermates at 12 and 24 weeks of age. This was attributed to a significant increase in Sertoli cell and round spermatid number and, consequently, seminiferous tubule area in InhbbM364T/M364T males compared to wildtype males. Despite this testis phenotype, male InhbbM364T/M364T mutant mice retained normal fertility. Serum hormone analyses, however, indicated that the InhbbM364T variant resulted in reduced circulating levels of activin B but did not affect FSH production. We also examined the effect of this p.Met360Thr and an additional INHBB variant (c.314C>T: p.Thr105Met) found in another infertile man on inhibin B and activin B in vitro biosynthesis. We found that both INHBB variants resulted in a significant disruption to activin B in vitro biosynthesis. Together, this analysis supports that INHBB variants that limit activin B production have consequences for testis composition in males.
Collapse
Affiliation(s)
- Brendan J Houston
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Degang Wang
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- The Affiliated Zhongshan Boai Hospital of Southern Medical University, Guangdong, China
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - D Jo Merriner
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
| | - Haitong Luan
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Don F Conrad
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
| | - Liina Nagirnaja
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
| | - Kenneth I Aston
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
- Department of Surgery (Urology Division) University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Corinna Friedrich
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Craig Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Moira K O'Bryan
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Kelly Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
3
|
Ruohonen ST, Gaytan F, Usseglio Gaudi A, Velasco I, Kukoricza K, Perdices-Lopez C, Franssen D, Guler I, Mehmood A, Elo LL, Ohlsson C, Poutanen M, Tena-Sempere M. Selective loss of kisspeptin signaling in oocytes causes progressive premature ovulatory failure. Hum Reprod 2022; 37:806-821. [PMID: 35037941 PMCID: PMC8971646 DOI: 10.1093/humrep/deab287] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does direct kisspeptin signaling in the oocyte have a role in the control of follicular dynamics and ovulation? SUMMARY ANSWER Kisspeptin signaling in the oocyte plays a relevant physiological role in the direct control of ovulation; oocyte-specific ablation of kisspeptin receptor, Gpr54, induces a state of premature ovulatory failure in mice that recapitulates some features of premature ovarian insufficiency (POI). WHAT IS KNOWN ALREADY Kisspeptins, encoded by the Kiss1 gene, are essential for the control of ovulation and fertility, acting primarily on hypothalamic GnRH neurons to stimulate gonadotropin secretion. However, kisspeptins and their receptor, Gpr54, are also expressed in the ovary of different mammalian species, including humans, where their physiological roles remain contentious and poorly characterized. STUDY DESIGN, SIZE, DURATION A novel mouse line with conditional ablation of Gpr54 in oocytes, named OoGpr54−/−, was generated and studied in terms of follicular and ovulatory dynamics at different age-points of postnatal maturation. A total of 59 OoGpr54−/− mice and 47 corresponding controls were analyzed. In addition, direct RNA sequencing was applied to ovarian samples from 8 OoGpr54−/− and 7 control mice at 6 months of age, and gonadotropin priming for ovulatory induction was conducted in mice (N = 7) from both genotypes. PARTICIPANTS/MATERIALS, SETTING, METHODS Oocyte-selective ablation of Gpr54 in the oocyte was achieved in vivo by crossing a Gdf9-driven Cre-expressing transgenic mouse line with a Gpr54 LoxP mouse line. The resulting OoGpr54−/− mouse line was subjected to phenotypic, histological, hormonal and molecular analyses at different age-points of postnatal maturation (Day 45, and 2, 4, 6 and 10–11 months of age), in order to characterize the timing of puberty, ovarian follicular dynamics and ovulation, with particular attention to identification of features reminiscent of POI. The molecular signature of ovaries from OoGpr54−/− mice was defined by direct RNA sequencing. Ovulatory responses to gonadotropin priming were also assessed in OoGpr54−/− mice. MAIN RESULTS AND THE ROLE OF CHANCE Oocyte-specific ablation of Gpr54 caused premature ovulatory failure, with some POI-like features. OoGpr54−/− mice had preserved puberty onset, without signs of hypogonadism. However, already at 2 months of age, 40% of OoGpr54−/− females showed histological features reminiscent of ovarian failure and anovulation. Penetrance of the phenotype progressed with age, with >80% and 100% of OoGpr54−/− females displaying complete ovulatory failure by 6- and 10 months, respectively. This occurred despite unaltered hypothalamic Gpr54 expression and gonadotropin levels. Yet, OoGpr54−/− mice had decreased sex steroid levels. While the RNA signature of OoGpr54−/− ovaries was dominated by the anovulatory state, oocyte-specific ablation of Gpr54 significantly up- or downregulated of a set of 21 genes, including those encoding pituitary adenylate cyclase-activating polypeptide, Wnt-10B, matrix-metalloprotease-12, vitamin A-related factors and calcium-activated chloride channel-2, which might contribute to the POI-like state. Notably, the anovulatory state of young OoGpr54−/− mice could be rescued by gonadotropin priming. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Conditional ablation of Gpr54 in oocytes unambiguously caused premature ovulatory failure in mice; yet, the ultimate molecular mechanisms for such state of POI can be only inferred on the basis of RNAseq data and need further elucidation, since some of the molecular changes observed in OoGpr54−/− ovaries were secondary to the anovulatory state. Direct translation of mouse findings to human disease should be made with caution since, despite the conserved expression of Kiss1/kisspeptin and Gpr54 in rodents and humans, our mouse model does not recapitulate all features of common forms of POI. WIDER IMPLICATIONS OF THE FINDINGS Deregulation of kisspeptin signaling in the oocyte might be an underlying, and previously unnoticed, cause for some forms of POI in women. STUDY FUNDING/COMPETING INTEREST(S) This work was primarily supported by a grant to M.P. and M.T.-S. from the FiDiPro (Finnish Distinguished Professor) Program of the Academy of Finland. Additional financial support came from grant BFU2017-83934-P (M.T.-S.; Ministerio de Economía y Competitividad, Spain; co-funded with EU funds/FEDER Program), research funds from the IVIRMA International Award in Reproductive Medicine (M.T.-S.), and EFSD Albert Renold Fellowship Programme (S.T.R.). The authors have no conflicts of interest to declare in relation to the contents of this work. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Suvi T Ruohonen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, Turku, Finland
| | - Francisco Gaytan
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Andrea Usseglio Gaudi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Inmaculada Velasco
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Krisztina Kukoricza
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Cecilia Perdices-Lopez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Delphine Franssen
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Ipek Guler
- Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Arfa Mehmood
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, Turku, Finland.,Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Manuel Tena-Sempere
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, Turku, Finland.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain
| |
Collapse
|
4
|
Alarcón S, Esteban J, Roos R, Heikkinen P, Sánchez-Pérez I, Adamsson A, Toppari J, Koskela A, Finnilä MAJ, Tuukkanen J, Herlin M, Hamscher G, Leslie HA, Korkalainen M, Halldin K, Schrenk D, Håkansson H, Viluksela M. Endocrine, metabolic and apical effects of in utero and lactational exposure to non-dioxin-like 2,2',3,4,4',5,5'-heptachlorobiphenyl (PCB 180): A postnatal follow-up study in rats. Reprod Toxicol 2021; 102:109-127. [PMID: 33992733 DOI: 10.1016/j.reprotox.2021.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 12/19/2022]
Abstract
PCB 180 is a persistent and abundant non-dioxin-like PCB (NDL-PCB). We determined the developmental toxicity profile of ultrapure PCB 180 in developing offspring following in utero and lactational exposure with the focus on endocrine, metabolic and retinoid system alterations. Pregnant rats were given total doses of 0, 10, 30, 100, 300 or 1000 mg PCB 180/kg bw on gestational days 7-10 by oral gavage, and the offspring were sampled on postnatal days (PND) 7, 35 and 84. Decreased serum testosterone and triiodothyronine concentrations on PND 84, altered liver retinoid levels, increased liver weights and induced 7-pentoxyresorufin O-dealkylase (PROD) activity were the sensitive effects used for margin of exposure (MoE) calculations. Liver weights were increased together with induction of the metabolizing enzymes cytochrome P450 (CYP) 2B1, CYP3A1, and CYP1A1. Less sensitive effects included decreased serum estradiol and increased luteinizing hormone levels in females, decreased prostate and seminal vesicle weight and increased pituitary weight in males, increased cortical bone area and thickness of tibial diaphysis in females and decreased cortical bone mineral density in males. Developmental toxicity profiles were partly different in male and female offspring, males being more sensitive to increased liver weight, PROD induction and decreased thyroxine concentrations. MoE assessment indicated that the 95th percentile of current maternal PCB 180 concentrations do not exceed the estimated tolerable human lipid-based PCB 180 concentration. Although PCB 180 is much less potent than dioxin-like compounds, it shares several toxicological targets suggesting a potential for interactions.
Collapse
Affiliation(s)
- Sonia Alarcón
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche (Alicante), Spain; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Javier Esteban
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche (Alicante), Spain.
| | - Robert Roos
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Päivi Heikkinen
- Environmental Health Unit, Finnish Institute for Health and Welfare (THL), P.O. Box 95, Kuopio, FI-70701, Finland
| | - Ismael Sánchez-Pérez
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche (Alicante), Spain
| | - Annika Adamsson
- Research Center for Integrative Physiology and Pharmacology and Centre for Population Health Research, Institute of Biomedicine, University of Turku, Department of Paediatrics, Turku University Hospital, Turku, FI-20520, Finland
| | - Jorma Toppari
- Research Center for Integrative Physiology and Pharmacology and Centre for Population Health Research, Institute of Biomedicine, University of Turku, Department of Paediatrics, Turku University Hospital, Turku, FI-20520, Finland
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Mikko A J Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Maria Herlin
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gerd Hamscher
- Institute of Food Chemistry and Food Biotechnology, Justus Liebig University, Giessen, D-35392, Germany
| | - Heather A Leslie
- Department of Environment and Health, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, NL-1081 HZ, The Netherlands
| | - Merja Korkalainen
- Environmental Health Unit, Finnish Institute for Health and Welfare (THL), P.O. Box 95, Kuopio, FI-70701, Finland
| | - Krister Halldin
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dieter Schrenk
- Food Chemistry and Toxicology, University of Kaiserslautern, Kaiserslautern, D-67663, Germany
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matti Viluksela
- School of Pharmacy (Toxicology), Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
5
|
Oduwole OO, Poliandri A, Okolo A, Rawson P, Doroszko M, Chrusciel M, Rahman NA, Serrano de Almeida G, Bevan CL, Koechling W, Huhtaniemi IT. Follicle-stimulating hormone promotes growth of human prostate cancer cell line-derived tumor xenografts. FASEB J 2021; 35:e21464. [PMID: 33724574 DOI: 10.1096/fj.202002168rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/11/2022]
Abstract
Chemical castration in prostate cancer can be achieved with gonadotropin-releasing hormone (GnRH) agonists or antagonists. Their effects differ by the initial flare of gonadotropin and testosterone secretion with agonists and the immediate pituitary-testicular suppression by antagonists. While both suppress luteinizing hormone (LH) and follicle-stimulating hormone (FSH) initially, a rebound in FSH levels occurs during agonist treatment. This rebound is potentially harmful, taken the expression of FSH receptors (R) in prostate cancer tissue. We herein assessed the role of FSH in promoting the growth of androgen-independent (PC-3, DU145) and androgen-dependent (VCaP) human prostate cancer cell line xenografts in nude mice. Gonadotropins were suppressed with the GnRH antagonist degarelix, and effects of add-back human recombinant FSH were assessed on tumor growth. All tumors expressed GnRHR and FSHR, and degarelix treatment suppressed their growth. FSH supplementation reversed the degarelix-evoked suppression of PC-3 tumors, both in preventive (degarelix and FSH treatment started upon cell inoculation) and therapeutic (treatments initiated 3 weeks after cell inoculation) setting. A less marked, though significant FSH effect occurred in DU145, but not in VCaP xenografts. FSHR expression in the xenografts supports direct FSH stimulation of tumor growth. Testosterone supplementation, to maintain the VCaP xenografts, apparently masked the FSH effect on their growth. Treatment with the LH analogue hCG did not affect PC-3 tumor growth despite their expression of luteinizing hormone/choriongonadotropin receptor. In conclusion, FSH, but not LH, may directly stimulate the growth of androgen-independent prostate cancer, suggesting that persistent FSH suppression upon GnRH antagonist treatment offers a therapeutic advantage over agonist.
Collapse
Affiliation(s)
- Olayiwola O Oduwole
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Ariel Poliandri
- Department of Molecular and Clinical Sciences, St. George's University of London, London, UK
| | - Anthony Okolo
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Phil Rawson
- Central Biomedical Services, Imperial College London, London, UK
| | - Milena Doroszko
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Nafis A Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | | | - Charlotte L Bevan
- Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Wolfgang Koechling
- Non-Clinical Development, Ferring Pharmaceuticals A/S, Copenhagen, Denmark
| | - Ilpo T Huhtaniemi
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
6
|
Mifepristone Treatment Promotes Testicular Leydig Cell Tumor Progression in Transgenic Mice. Cancers (Basel) 2020; 12:cancers12113263. [PMID: 33158280 PMCID: PMC7694279 DOI: 10.3390/cancers12113263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Recently, the antiprogestin activity of selective progesterone receptor (PR) modulator mifepristone (MF) has proven unsuccessful as a potential anti-cancer agent in various clinical trials. Herein, we analyzed the effects of MF treatment on Leydig cell tumor (LCT) progression in a transgenic mouse model (inhibin-α promoter-driven SV40 T-antigen), as well as on the proliferation of two Leydig tumor cell lines. MF significantly stimulated the proliferation of LCT in vitro. Similarly, a 1-mo MF or P4 treatment stimulated LCT tumor growth in vivo. Only the abundant membrane Pgrmc1 expression was found in LCTs, but no other classical Pgr or nonclassical membrane PRs. Functional analysis showed that PGRMC1 is required for MF and P4 to stimulate the proliferation and invasiveness of LCTs. Our findings provide novel information that the use of MF as an anti-cancer agent should be considered with caution due to its potential PGRMC1 tumor-promoting pathway activation in cancers. Abstract The selective progesterone receptor modulator mifepristone (MF) may act as a potent antiproliferative agent in different steroid-dependent cancers due to its strong antagonistic effect on the nuclear progesterone receptor (PGR). Hereby, we analyzed the effects of MF treatment on Leydig cell tumor (LCT) progression in a transgenic mouse model (inhibin-α promoter-driven SV40 T-antigen), as well as on LCT (BLTK-1 and mLTC-1) cell proliferation. MF significantly stimulated the proliferation of LCT in vitro. Similarly, a 1-mo MF or P4 treatment stimulated LCT tumor growth in vivo. Traceable/absent classical Pgr or nonclassical membrane PRs α, β, γ and Pgrmc2, but abundant membrane Pgrmc1 expression, was found in LCTs. MF did not activate glucocorticoid or androgen receptors in LCTs. Functional analysis showed that PGRMC1 is required for MF and P4 to stimulate the proliferation and invasiveness of LCTs. Accordingly, MF and P4 induced PGRMC1 translocation into the nucleus and thereby stimulated the release of TGFβ1 in LCT cells. MF and P4 treatments upregulated Tgfbr1, Tgfbr2, and Alk1 expression and stimulated TGFβ1 release in LCT cells. Our findings provide novel mechanistic insights into the action of MF as a membrane PR agonist that promotes LCT growth through PGRMC1 and the alternative TGFβ1 signaling pathway.
Collapse
|
7
|
Ohlsson C, Gustafsson KL, Farman HH, Henning P, Lionikaite V, Movérare-Skrtic S, Sjögren K, Törnqvist AE, Andersson A, Islander U, Bernardi AI, Poutanen M, Chambon P, Lagerquist MK. Phosphorylation site S122 in estrogen receptor α has a tissue-dependent role in female mice. FASEB J 2020; 34:15991-16002. [PMID: 33067917 DOI: 10.1096/fj.201901376rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Estrogen treatment increases bone mass and reduces fat mass but is associated with adverse effects in postmenopausal women. Knowledge regarding tissue-specific estrogen signaling is important to aid the development of new tissue-specific treatments. We hypothesized that the posttranslational modification phosphorylation in estrogen receptor alpha (ERα) may modulate ERα activity in a tissue-dependent manner. Phosphorylation of site S122 in ERα has been shown in vitro to affect ERα activity, but the tissue-specific role in vivo is unknown. We herein developed and phenotyped a novel mouse model with a point mutation at the phosphorylation site 122 in ERα (S122A). Female S122A mice had increased fat mass and serum insulin levels but unchanged serum sex steroid levels, uterus weight, bone mass, thymus weight, and lymphocyte maturation compared to WT mice. In conclusion, phosphorylation site S122 in ERα has a tissue-dependent role with an impact specifically on fat mass in female mice. This study is the first to demonstrate in vivo that a phosphorylation site in a transactivation domain in a nuclear steroid receptor modulates the receptor activity in a tissue-dependent manner.
Collapse
Affiliation(s)
- Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin L Gustafsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Helen H Farman
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Klara Sjögren
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna E Törnqvist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Annica Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Islander
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Angelina I Bernardi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique, National de la Sante et de la Recherche Medicale, ULP, Collège de France, Illkirch-Strasbourg, France
| | - Marie K Lagerquist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Sipilä P, Junnila A, Hakkarainen J, Huhtaniemi R, Mairinoja L, Zhang FP, Strauss L, Ohlsson C, Kotaja N, Huhtaniemi I, Poutanen M. The lack of HSD17B3 in male mice results in disturbed Leydig cell maturation and endocrine imbalance akin to humans with HSD17B3 deficiency. FASEB J 2020; 34:6111-6128. [PMID: 32190925 DOI: 10.1096/fj.201902384r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/31/2020] [Accepted: 02/13/2020] [Indexed: 11/11/2022]
Abstract
Hydroxysteroid (17β) dehydrogenase type 3 (HSD17B3) deficiency causes a disorder of sex development in humans, where affected males are born with female-appearing external genitalia, but are virilized during puberty. The hormonal disturbances observed in the Hsd17b3 knockout mice (HSD17B3KO), generated in the present study, mimic those found in patients with HSD17B3 mutations. Identical to affected humans, serum T in the adult HSD17B3KO mice was within the normal range, while a striking increase was detected in serum A-dione concentration. This resulted in a marked reduction of the serum T/A-dione ratio, a diagnostic hallmark for the patients with HSD17B3 deficiency. However, unlike humans, male HSD17B3KO mice were born with normally virilized phenotype, but presenting with delayed puberty. In contrast to the current belief, data from HSD17B3KO mice show that the circulating T largely originates from the testes, indicating a strong compensatory mechanism in the absence of HSD17B3. The lack of testicular malignancies in HSD17B3KO mice supports the view that testis tumors in human patients are due to associated cryptorchidism. The HSD17B3KO mice presented also with impaired Leydig cell maturation and signs of undermasculinization in adulthood. The identical hormonal disturbances between HSD17B3 deficient knockout mice and human patients make the current mouse model valuable for understanding the mechanism of the patient phenotypes, as well as endocrinopathies and compensatory steroidogenic mechanisms in HSD17B3 deficiency.
Collapse
Affiliation(s)
- P Sipilä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - A Junnila
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - J Hakkarainen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - R Huhtaniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - L Mairinoja
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - F P Zhang
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - L Strauss
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - C Ohlsson
- Institute of Medicine, the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - N Kotaja
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - I Huhtaniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - M Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland.,Institute of Medicine, the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
9
|
Colldén H, Landin A, Wallenius V, Elebring E, Fändriks L, Nilsson ME, Ryberg H, Poutanen M, Sjögren K, Vandenput L, Ohlsson C. The gut microbiota is a major regulator of androgen metabolism in intestinal contents. Am J Physiol Endocrinol Metab 2019; 317:E1182-E1192. [PMID: 31689143 PMCID: PMC6962501 DOI: 10.1152/ajpendo.00338.2019] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Androgens exert important effects both in androgen-responsive tissues and in the intestinal tract. To determine the impact of the gut microbiota (GM) on intestinal androgen metabolism, we measured unconjugated (free) and glucuronidated androgen levels in intestinal contents from the small intestine, with a low bacterial density, and from cecum and colon, with a high bacterial density. Using a specific, sensitive gas chromatography-tandem mass spectrometry method, we detected high levels of glucuronidated testosterone (T) and dihydrotestosterone (DHT) in small intestinal content of mice of both sexes, whereas in the distal intestine we observed remarkably high levels of free DHT, exceeding serum levels by >20-fold. Similarly, in young adult men high levels of unconjugated DHT, >70-fold higher than in serum, were detected in feces. In contrast to mice with a normal GM composition, germ-free mice had high levels of glucuronidated T and DHT, but very low free DHT levels, in the distal intestine. These findings demonstrate that the GM is involved in intestinal metabolism and deglucuronidation of DHT and T, resulting in extremely high free levels of the most potent androgen, DHT, in the colonic content of young and healthy mice and men.
Collapse
Affiliation(s)
- Hannah Colldén
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Landin
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Elebring
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Fändriks
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria E Nilsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Ryberg
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Klara Sjögren
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Molecular mechanisms underlying mifepristone's agonistic action on ovarian cancer progression. EBioMedicine 2019; 47:170-183. [PMID: 31466918 PMCID: PMC6796594 DOI: 10.1016/j.ebiom.2019.08.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background Recent clinical trials on ovarian cancer with mifepristone (MF) have failed, despite in vitro findings on its strong progesterone (P4) antagonist function. Methods Ovarian cancer human and murine cell lines, cultured high-grade human primary epithelial ovarian cancer (HG-hOEC) cells and their explants; as well as in vivo transgenic mice possessing ovarian cancer were used to assess the molecular mechanism underlying mifepristone (MF) agonistic actions in ovarian cancer progression. Findings Herein, we show that ovarian cancer cells express traceable/no nuclear P4 receptor (PGR), but abundantly P4 receptor membrane component 1 (PGRMC1). MF significantly stimulated ovarian cancer cell migration, proliferation and growth in vivo, and the translocation of PGRMC1 into the nucleus of cancer cells; the effects inhibited by PGRMC1 inhibitor. The beneficial antitumor effect of high-doses MF could not be achieved in human cancer tissue, and the low tissue concentrations achieved with the therapeutic doses only promoted the growth of ovarian cancers. Interpretation Our results indicate that treatment of ovarian cancer with MF and P4 may induce similar adverse agonistic effects in the absence of classical nuclear PGRs in ovarian cancer. The blockage of PGRMC1 activity may provide a novel treatment strategy for ovarian cancer. Fund This work was supported by grants from the National Science Centre, Poland (2013/09/N/NZ5/01831 to DP-T; 2012/05/B/NZ5/01867 to MC), Academy of Finland (254366 to NAR), Moikoinen Cancer Research Foundation (to NAR) and EU PARP Cluster grant (UDA-POIG.05.01.00-005/12-00/NCREMFP to SW).
Collapse
|
11
|
Rotgers E, Cisneros-Montalvo S, Nurmio M, Toppari J. Retinoblastoma protein represses E2F3 to maintain Sertoli cell quiescence in mouse testis. J Cell Sci 2019; 132:132/14/jcs229849. [PMID: 31308245 DOI: 10.1242/jcs.229849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/11/2019] [Indexed: 01/04/2023] Open
Abstract
Maintenance of the differentiated state and cell cycle exit in adult Sertoli cells depends on tumor suppressor retinoblastoma protein (RB, also known as RB1). We have previously shown that RB interacts with transcription factor E2F3 in the mouse testis. Here, we investigated how E2f3 contributes to adult Sertoli cell proliferation in a mouse model of Sertoli cell-specific knockout of Rb by crossing these mice with an E2f3 knockout mouse line. In the presence of intact RB, E2f3 was redundant in Sertoli cells. However, in the absence of RB, E2f3 is a key driver for cell cycle re-entry and loss of function in adult Sertoli cells. Knockout of E2f3 in Sertoli cells rescued the breakdown of Sertoli cell function associated with Rb loss, prevented proliferation of adult Sertoli cells and restored fertility of the mice. In summary, our results show that RB-mediated repression of E2F3 is critical for the maintenance of cell cycle exit and terminal differentiation in adult mouse Sertoli cells.
Collapse
Affiliation(s)
- Emmi Rotgers
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku 20520, Finland.,Department of Pediatrics, Turku University Hospital, Turku 20520, Finland
| | - Sheyla Cisneros-Montalvo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku 20520, Finland.,Department of Pediatrics, Turku University Hospital, Turku 20520, Finland
| | - Mirja Nurmio
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku 20520, Finland.,Department of Pediatrics, Turku University Hospital, Turku 20520, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku 20520, Finland .,Department of Pediatrics, Turku University Hospital, Turku 20520, Finland
| |
Collapse
|
12
|
Lack of androgen receptor SUMOylation results in male infertility due to epididymal dysfunction. Nat Commun 2019; 10:777. [PMID: 30770815 PMCID: PMC6377611 DOI: 10.1038/s41467-019-08730-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
Androgen receptor (AR) is regulated by SUMOylation at its transactivation domain. In vitro, the SUMOylation is linked to transcriptional repression and/or target gene-selective regulation. Here, we generated a mouse model (ArKI) in which the conserved SUMO acceptor lysines of AR are permanently abolished (ArK381R, K500R). ArKI males develop normally, without apparent defects in their systemic androgen action in reproductive tissues. However, the ArKI males are infertile. Their spermatogenesis appears unaffected, but their epididymal sperm maturation is defective, shown by severely compromised motility and fertilization capacity of the sperm. Fittingly, their epididymal AR chromatin-binding and gene expression associated with sperm maturation and function are misregulated. AR is SUMOylated in the wild-type epididymis but not in the testis, which could explain the tissue-specific response to the lack of AR SUMOylation. Our studies thus indicate that epididymal AR SUMOylation is essential for the post-testicular sperm maturation and normal reproductive capability of male mice. SUMOylation is known to regulate androgen receptor (AR) activity in cultured cells. Here, using SUMOylation-deficient AR knock-in mice, the authors demonstrate that SUMOylation is required for AR-related gene expression specifically in the epididymal tissues, but not the testis.
Collapse
|
13
|
Dou L, Zheng Y, Li L, Gui X, Chen Y, Yu M, Guo Y. The effect of cinnamon on polycystic ovary syndrome in a mouse model. Reprod Biol Endocrinol 2018; 16:99. [PMID: 30340496 PMCID: PMC6194596 DOI: 10.1186/s12958-018-0418-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most prevalent cause of anovulatory infertility and hyperandrogenism. Evidence favors insulin resistance and compensatory hyperinsulinemia as the predominant, perhaps primary, defects in PCOS. The use of insulin-sensitizing drugs has been shown to improve both the reproductive and the metabolic aspects of PCOS. Cinnamon has been found to have insulin sensitizing effect and improve menstrual cyclicity in women with PCOS. The aim of this study was to determine the effect and mechanism of cinnamon on PCOS using a dehydroepiandrosterone (DHEA) induced PCOS mouse model. METHODS Prepubertal C57BL/6 mice (age 25 days) were raised to developed into control group, DHEA group and DHEA plus cinnamon group for 20 days. The stages of the estrous cycle were determined based on vaginal cytology; metabolic characteristics were examined by intraperitoneal glucose tolerance test and insulin tolerance test, the serum levels of hormones (testosterone, insulin, LH, FSH, IGF-1, IGFBP-1) were checked using enzyme-linked immunosorbent assay (ELISA) method, the ovarian morphology was observed by stained with hematoxylin and eosin. IGF-1 and IGFBP-1 expression in ovary were detected by immunohistochemical stain. RESULTS Cinnamon restores the cyclicity and ovary morphology in PCOS mice model induced by DHEA. There are significant differences of serum level of total testosterone (0.033 ± 0.009 ng/ml), among control group, DHEA and cinnamon group (0.052 ± 0.011 ng/ml), and DHEA group (0.079 ± 0.015 ng/ml); There was an increasing tendency of serum FSH level from DHEA group (5.02 ± 0.31 ng/ml), DHEA and cinnamon group (5.81 ± 0.51 ng/ml), to control group (7.13 ± 0.74 ng/ml); and there was a decreasing trend of serum LH level from DHEA group (3.75 ± 0.57 ng/ml), DHEA and cinnamon group (1.35 ± 0.61 ng/ml), or control group (0.69 ± 0.34 ng/ml); serum insulin level is significantly higher in DHEA treated mice (1.61 ± 0.31 ng/ml) than control group (0.93 ± 0.19 ng/ml), or DHEA and cinnamon effect (1.27 ± 0.23 ng/ml) (p < 0.05). The DHEA group also has a higher serum IGF-1 level (0.35 ± 0.06 ng/ml) than control group (0.17 ± 0.04 ng/ml) or DHEA and cinnamon group (0.21 ± 0.05 ng/ml) (p < 0.05). While DHEA group has a lower IGFBP-1 level (5.5 ± 1.6 ng/ml) than control group (15.8 ± 2.1 ng/ml) or DHEA and cinnamon group (10.3 ± 2.5 ng/ml) (p < 0.05). Cinnamon also attenuates DHEA induced a higher IGF-1 and lower IGFBP-1 expression in ovary by immunohistochemistry. CONCLUSIONS These preliminary data suggest that cinnamon supplementation improves insulin resistance and may be a potential therapeutic agent for the treatment of PCOS.
Collapse
Affiliation(s)
- Lei Dou
- grid.412636.4Department of Obstetrics and gynecology, First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Yahong Zheng
- grid.412636.4Department of Obstetrics and gynecology, First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Lu Li
- grid.412636.4Department of Obstetrics and gynecology, First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Xiaowei Gui
- grid.412636.4Department of Obstetrics and gynecology, First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Yajuan Chen
- Department of Obstetrics and gynecology, Anshan Branch of First Affiliated Hospital of China Medical University, Anshan, China
| | - Meng Yu
- 0000 0000 9678 1884grid.412449.eDepartment of Reproductive Biology and Transgenic Animals, China Medical University, Shenyang, China
| | - Yi Guo
- grid.412636.4Department of Obstetrics and gynecology, First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
14
|
Camlin NJ, Jarnicki AG, Vanders RL, Walters KA, Hansbro PM, McLaughlin EA, Holt JE. Grandmaternal smoke exposure reduces female fertility in a murine model, with great-grandmaternal smoke exposure unlikely to have an effect. Hum Reprod 2018; 32:1270-1281. [PMID: 28402417 DOI: 10.1093/humrep/dex073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/28/2017] [Indexed: 01/22/2023] Open
Abstract
STUDY QUESTION What effect does multigenerational (F2) and transgenerational (F3) cigarette smoke exposure have on female fertility in mice? SUMMARY ANSWER Cigarette smoking has a multigenerational effect on female fertility. WHAT IS KNOWN ALREADY It has been well established that cigarette smoking decreases female fertility. Furthermore, a growing body of evidence suggests that smoking during pregnancy decreases the fertility of daughters and increases cancer and asthma incidence in grandchildren and great-grandchildren. STUDY DESIGN, SIZE, DURATION Six-week-old C57BL/6 female mice were exposed nasally to cigarette smoke or room air (controls) for 5 weeks prior to being housed with males. Females continued to be exposed to smoke throughout pregnancy and lactation until pups were weaned. A subset of F1 female pups born to these smoke and non-smoke exposed females were bred to create the F2 grandmaternal exposed generation (multigenerational). Finally, a subset of F2 females were bred to create the F3 great-grandmaternal exposed generation (transgenerational). The reproductive health of F2 and F3 females was examined at 8 weeks and 9 months. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian and oocyte quality was examined in smoke exposed and control animals. A small-scale fertility trial was performed before ovarian changes were examined using ovarian histology and immunofluorescence and/or immunoblotting analysis of markers of apoptosis (TUNEL) and proliferation (proliferating cell nuclear antigen (PCNA) and anti-Mullerian hormone (AMH)). Oocyte quality was examined using immunocytochemistry to analyze the metaphase II spindle and ploidy status. Parthenogenetic activation of oocytes was used to investigate meiosis II timing and preimplantation embryo development. Finally, diestrus hormone serum levels (FSH and LH) were quantified. MAIN RESULTS AND THE ROLE OF CHANCE F2 smoke exposed females had no detectable change in ovarian follicle quality at 8 weeks, although by 9 months ovarian somatic cell proliferation was reduced (P = 0.0197) compared with non-smoke exposed control. Further investigation revealed changes between control and smoke exposed F2 oocyte quality, including altered meiosis II timing at 8 weeks (P = 0.0337) and decreased spindle pole to pole length at 9 months (P = 0.0109). However, no change in preimplantation embryo development was observed following parthenogenetic activation. The most noticeable effect of cigarette smoke exposure was related to the subfertility of F2 females; F2 smoke exposed females displayed significantly increased time to conception (P = 0.0042) and significantly increased lag time between pregnancies (P = 0.0274) compared with non-smoke exposed F2 females. Conversely, F3 smoke exposed females displayed negligible oocyte and follicle changes up to 9 months of age, and normal preimplantation embryo development. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION This study focused solely on a mouse model of cigarette smoke exposure to simulate human exposure. WIDER IMPLICATIONS OF THE FINDINGS Our results demonstrate that grandmaternal cigarette smoke exposure reduces female fertility in mice, highlighting the clinical need to promote cessation of cigarette smoking in pregnant women. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the Australian Research Council, National Health and Medical Research Council, Hunter Medical Research Institute, Newcastle Permanent Building Society Charitable Trust, and the University of Newcastle Priory Research Centers in Chemical Biology, Healthy Lungs and Grow Up Well. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- N J Camlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia
| | - A G Jarnicki
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan, NSW 2308, Australia
| | - R L Vanders
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan, NSW 2308, Australia
| | - K A Walters
- School of Women's & Children's Health, University of New South Wales, Sydney, NSW2052, Australia
| | - P M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan, NSW 2308, Australia
| | - E A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.,School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - J E Holt
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
15
|
Adam M, Heikelä H, Sobolewski C, Portius D, Mäki-Jouppila J, Mehmood A, Adhikari P, Esposito I, Elo LL, Zhang FP, Ruohonen ST, Strauss L, Foti M, Poutanen M. Hydroxysteroid (17β) dehydrogenase 13 deficiency triggers hepatic steatosis and inflammation in mice. FASEB J 2018; 32:3434-3447. [PMID: 29401633 DOI: 10.1096/fj.201700914r] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hydroxysteroid (17β) dehydrogenases (HSD17Bs) form an enzyme family characterized by their ability to catalyze reactions in steroid and lipid metabolism. In the present study, we characterized the phenotype of HSD17B13-knockout (HSD17B13KO) mice deficient in Hsd17b13. In these studies, hepatic steatosis was detected in HSD17B13KO male mice, indicated by histologic analysis and by the increased triglyceride concentration in the liver, whereas reproductive performance and serum steroid concentrations were normal in HSD17B13KO mice. In line with these changes, the expression of key proteins in fatty acid synthesis, such as FAS, acetyl-CoA carboxylase 1, and SCD1, was increased in the HSD17B13KO liver. Furthermore, the knockout liver showed an increase in 2 acylcarnitines, suggesting impaired mitochondrial β-oxidation in the presence of unaltered malonyl CoA and AMPK expression. The glucose tolerance did not differ between wild-type and HSD17B13KO mice in the presence of lower levels of glucose 6-phosphatase in HSD17B13KO liver compared with wild-type liver. Furthermore, microgranulomas and increased portal inflammation together with up-regulation of immune response genes were observed in HSD17B13KO mice. Our data indicate that disruption of Hsd17b13 impairs hepatic-lipid metabolism in mice, resulting in liver steatosis and inflammation, but the enzyme does not play a major role in the regulation of reproductive functions.-Adam, M., Heikelä, H., Sobolewski, C., Portius, D., Mäki-Jouppila, J., Mehmood, A., Adhikari, P., Esposito, I., Elo, L. L., Zhang, F.-P., Ruohonen, S. T., Strauss, L., Foti, M., Poutanen, M. Hydroxysteroid (17β) dehydrogenase 13 deficiency triggers hepatic steatosis and inflammation in mice.
Collapse
Affiliation(s)
- Marion Adam
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Hanna Heikelä
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Dorothea Portius
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Jenni Mäki-Jouppila
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arfa Mehmood
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Prem Adhikari
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Irene Esposito
- Institute of Pathology, Technische Universität München, Munich, Germany; and
| | - Laura L Elo
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Fu-Ping Zhang
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Leena Strauss
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Internal Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
16
|
Hakkarainen J, Zhang FP, Jokela H, Mayerhofer A, Behr R, Cisneros-Montalvo S, Nurmio M, Toppari J, Ohlsson C, Kotaja N, Sipilä P, Poutanen M. Hydroxysteroid (17β) dehydrogenase 1 expressed by Sertoli cells contributes to steroid synthesis and is required for male fertility. FASEB J 2018; 32:3229-3241. [PMID: 29401623 DOI: 10.1096/fj.201700921r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The pituitary gonadotrophins and testosterone are the main hormonal regulators of spermatogenesis, but estradiol is also known to play a role in the process. The hormonal responses in the testis are partially mediated by somatic Sertoli cells that provide nutritional and physical support for differentiating male germ cells. Hydroxysteroid (17β) dehydrogenase 1 (HSD17B1) is a steroidogenic enzyme that especially catalyzes the conversion of low potent 17keto-steroids to highly potent 17β-hydroxysteroids. In this study, we show that Hsd17b1 is highly expressed in Sertoli cells of fetal and newborn mice, and HSD17B1 knockout males present with disrupted spermatogenesis with major defects, particularly in the head shape of elongating spermatids. The cell-cell junctions between Sertoli cells and germ cells were disrupted in the HSD17B1 knockout mice. This resulted in complications in the orientation of elongating spermatids in the seminiferous epithelium, reduced sperm production, and morphologically abnormal spermatozoa. We also showed that the Sertoli cell-expressed HSD17B1 participates in testicular steroid synthesis, evidenced by a compensatory up-regulation of HSD17B3 in Leydig cells. These results revealed a novel role for HSD17B1 in the control of spermatogenesis and male fertility, and that Sertoli cells significantly contribute to steroid synthesis in the testis.-Hakkarainen, J., Zhang, F.-P., Jokela, H., Mayerhofer, A., Behr, R., Cisneros-Montalvo, S., Nurmio, M., Toppari, J., Ohlsson, C., Kotaja, N., Sipilä, P., Poutanen, M. Hydroxysteroid (17β) dehydrogenase 1 expressed by Sertoli cells contributes to steroid synthesis and is required for male fertility.
Collapse
Affiliation(s)
| | - Fu-Ping Zhang
- Institute of Biomedicine, University of Turku, Turku, Finland.,Cell Biology-Anatomy III, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Heli Jokela
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Artur Mayerhofer
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Rüdiger Behr
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | | | - Mirja Nurmio
- Institute of Biomedicine, University of Turku, Turku, Finland.,Institute of Medicine, the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Jorma Toppari
- Institute of Biomedicine, University of Turku, Turku, Finland.,Institute of Medicine, the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Claes Ohlsson
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Noora Kotaja
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Petra Sipilä
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Institute of Biomedicine, University of Turku, Turku, Finland.,Cell Biology-Anatomy III, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Martinsried, Germany.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Ahmed K, LaPierre MP, Gasser E, Denzler R, Yang Y, Rülicke T, Kero J, Latreille M, Stoffel M. Loss of microRNA-7a2 induces hypogonadotropic hypogonadism and infertility. J Clin Invest 2017; 127:1061-1074. [PMID: 28218624 DOI: 10.1172/jci90031] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/15/2016] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are negative modulators of gene expression that fine-tune numerous biological processes. miRNA loss-of-function rarely results in highly penetrant phenotypes, but rather, influences cellular responses to physiologic and pathophysiologic stresses. Here, we have reported that a single member of the evolutionarily conserved miR-7 family, miR-7a2, is essential for normal pituitary development and hypothalamic-pituitary-gonadal (HPG) function in adulthood. Genetic deletion of mir-7a2 causes infertility, with low levels of gonadotropic and sex steroid hormones, small testes or ovaries, impaired spermatogenesis, and lack of ovulation in male and female mice, respectively. We found that miR-7a2 is highly expressed in the pituitary, where it suppresses golgi glycoprotein 1 (GLG1) expression and downstream bone morphogenetic protein 4 (BMP4) signaling and also reduces expression of the prostaglandin F2a receptor negative regulator (PTGFRN), an inhibitor of prostaglandin signaling and follicle-stimulating hormone (FSH) and luteinizing hormone (LH) secretion. Our results reveal that miR-7a2 critically regulates sexual maturation and reproductive function by interconnecting miR-7 genomic circuits that regulate FSH and LH synthesis and secretion through their effects on pituitary prostaglandin and BMP4 signaling.
Collapse
|
18
|
Sankar A, Kooistra SM, Gonzalez JM, Ohlsson C, Poutanen M, Helin K. Maternal expression of the JMJD2A/KDM4A histone demethylase is critical for pre-implantation development. Development 2017; 144:3264-3277. [DOI: 10.1242/dev.155473] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022]
Abstract
Regulation of chromatin composition through post-translational modifications of histones contributes to transcriptional regulation and is essential for many cellular processes, including differentiation and development. JMJD2A/KDM4A is a lysine demethylase with specificity towards di- and tri-methylated lysine 9 and lysine 36 of histone H3 (H3K9me2/me3 and H3K36me2/me3). Here, we report that Kdm4a as a maternal factor plays a key role in embryo survival and is vital for female fertility. Kdm4a−/- female mice ovulate normally with comparable fertilization but poor implantation rates, and cannot support healthy transplanted embryos to term. This is due to a role for Kdm4a in uterine function, where its loss causes reduced expression of key genes involved in ion transport, nutrient supply and cytokine signalling, that impact embryo survival. In addition, a significant proportion of Kdm4a deficient oocytes displays a poor intrinsic ability to develop into blastocysts. These embryos cannot compete with healthy embryos for implantation in vivo, highlighting Kdm4a as a maternal effect gene. Thus, our study dissects an important dual role for maternal Kdm4a in determining faithful early embryonic development and the implantation process.
Collapse
Affiliation(s)
- Aditya Sankar
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Present Address: Centre for Chromosome Stability, Institute of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Susanne Marije Kooistra
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- Present Address: Department of Neuroscience, University Medical Centre, Groningen, University of Groningen, Groningen, The Netherlands
| | - Javier Martin Gonzalez
- Core Facility for Transgenic Mice, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Claes Ohlsson
- Department of Physiology Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Physiology Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kristian Helin
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
19
|
Zaleska M, Waclawik A, Bodek G, Zezula-Szpyra A, Li X, Janowski T, Hansel WH, Rahman NA, Ziecik AJ. Growth Repression in Diethylstilbestrol/Dimethylbenz[a]anthracene–Induced Rat Mammary Gland Tumor Using Hecate-CGβ Conjugate. Exp Biol Med (Maywood) 2016; 229:335-44. [PMID: 15044717 DOI: 10.1177/153537020422900408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Recently, we have shown that Hecate-CGβ conjugate, which is a fusion of the lytic peptide Hecate and a 15–amino acid fragment of the β-chain of chorionic gonadotropin (CGβ), selectively destroys mammary gland carcinoma cells that possess luteinizing hormone receptors (LHR) in vitro. We induced mammary gland tumors using combined prenatal exposure to synthetic diethylstilbestrol (DES) and additional postnatal exposure to dimethylbenz[a]anthracene (DMBA). Rats with tumors were equally randomized (10/group) and treated with either sham (control) or 12 mg/kg body wt of either Hecate or Hecate-CGβ once a week for 3 weeks by tail vein injections. One week after the last injection, rats were kilted. Reverse-transcription–nested polymerase chain reaction/Southern blotting revealed alternatively spliced mRNA for LHR in tumor tissues of 5 of 30 females, which was further confirmed by Western blot analysis. The percentage of tumor volume increase was lowest in the group treated with Hecate-CGβ (45.3 ± 27.6), compared with Hecate- and shamtreated, control group (324.8 ± 78.1 and 309.9 ± 51.2, respectively; P < 0.001). Hecate-CGβ induced a significant decrease in tumor burden compared with controls (9.5 ± 2.1 mg/g body wt vs. 21.6 ± 2.9; P < 0.01). A smaller reduction in tumor burden was also observed in Hecate-treated females (17.6 ± 1.6 mg/g body wt vs. 21.6 ± 2.9; P < 0.05). Our results prove the principle that Hecate-CGβ conjugate is able to repress mammary gland tumor growth, even in tumor tissues that lack or have very low levels of LHR. The mechanism of Hecate-CGβ conjugate action in repression of DES/DMBA-induced tumor growth needs to be further analyzed to clarify the molecular mechanisms of Hecate-CGβ conjugate action in vivo.
Collapse
Affiliation(s)
- Monika Zaleska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kemiläinen H, Adam M, Mäki-Jouppila J, Damdimopoulou P, Damdimopoulos AE, Kere J, Hovatta O, Laajala TD, Aittokallio T, Adamski J, Ryberg H, Ohlsson C, Strauss L, Poutanen M. The Hydroxysteroid (17β) Dehydrogenase Family Gene HSD17B12 Is Involved in the Prostaglandin Synthesis Pathway, the Ovarian Function, and Regulation of Fertility. Endocrinology 2016; 157:3719-3730. [PMID: 27490311 DOI: 10.1210/en.2016-1252] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The hydroxysteroid (17beta) dehydrogenase (HSD17B)12 gene belongs to the hydroxysteroid (17β) dehydrogenase superfamily, and it has been implicated in the conversion of estrone to estradiol as well as in the synthesis of arachidonic acid (AA). AA is a precursor of prostaglandins, which are involved in the regulation of female reproduction, prompting us to study the role of HSD17B12 enzyme in the ovarian function. We found a broad expression of HSD17B12 enzyme in both human and mouse ovaries. The enzyme was localized in the theca interna, corpus luteum, granulosa cells, oocytes, and surface epithelium. Interestingly, haploinsufficiency of the HSD17B12 gene in female mice resulted in subfertility, indicating an important role for HSD17B12 enzyme in the ovarian function. In line with significantly increased length of the diestrous phase, the HSD17B+/- females gave birth less frequently than wild-type females, and the litter size of HSD17B12+/- females was significantly reduced. Interestingly, we observed meiotic spindle formation in immature follicles, suggesting defective meiotic arrest in HSD17B12+/- ovaries. The finding was further supported by transcriptome analysis showing differential expression of several genes related to the meiosis. In addition, polyovular follicles and oocytes trapped inside the corpus luteum were observed, indicating a failure in the oogenesis and ovulation, respectively. Intraovarian concentrations of steroid hormones were normal in HSD17B12+/- females, whereas the levels of AA and its metabolites (6-keto prostaglandin F1alpha, prostaglandin D2, prostaglandin E2, prostaglandin F2α, and thromboxane B2) were decreased. In conclusion, our study demonstrates that HSD17B12 enzyme plays an important role in female fertility through its role in AA metabolism.
Collapse
Affiliation(s)
- Heidi Kemiläinen
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Marion Adam
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Jenni Mäki-Jouppila
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Pauliina Damdimopoulou
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Anastasios E Damdimopoulos
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Juha Kere
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Outi Hovatta
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Teemu D Laajala
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Tero Aittokallio
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Jerzy Adamski
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Henrik Ryberg
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Leena Strauss
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Matti Poutanen
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| |
Collapse
|
21
|
Walton KL, Kelly EK, Johnson KE, Robertson DM, Stanton PG, Harrison CA. A Novel, More Efficient Approach to Generate Bioactive Inhibins. Endocrinology 2016; 157:2799-809. [PMID: 27054553 DOI: 10.1210/en.2015-1963] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Gonadal-derived inhibins are essential factors in mammalian reproduction, negatively regulating pituitary production of FSH. Interestingly, declines in inhibin levels across the menopause transition correlate with not only an increase in FSH but also a rapid decrease in bone mass. Therefore, inhibins have been touted as potential therapeutics for osteoporosis in postmenopausal women. However, as heterodimeric proteins of α- and β- (βA or βB)-subunits, inhibins are difficult to produce recombinantly, are poorly processed to their mature bioactive forms, and their expression is always accompanied by production of activins (β-subunit homodimers), the proteins they antagonize. In this study, we developed the methodology to circumvent most of these issues. Initially, the cleavage sites between the pro- and mature domains of the α- and βA-subunits were modified to ensure complete processing. These modifications led to a marked increase (9-fold) in the levels of bioactive inhibin A and a striking decrease (12.5-fold) in mature activin A production. Next, a single point mutation (M418A) was incorporated into the βA-subunit, which reduced residual activin activity approximately 100-fold and, in so doing, increased inhibin bioactivity 8-fold. Finally, we showed that inhibin A noncovalently associated with its prodomain was more potent (∼20-fold) than mature inhibin A in specific in vitro bioassays, indicating an important role of the prodomain in inhibin bioactivity. In conclusion, the production of potent inhibin analogs in the virtual absence of activin activity will greatly facilitate the investigation of the therapeutic potential of these gonadal hormones on bone and other tissues.
Collapse
Affiliation(s)
- Kelly L Walton
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Emily K Kelly
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | - David M Robertson
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Peter G Stanton
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Craig A Harrison
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| |
Collapse
|
22
|
Rotgers E, Nurmio M, Pietilä E, Cisneros-Montalvo S, Toppari J. E2F1 controls germ cell apoptosis during the first wave of spermatogenesis. Andrology 2016; 3:1000-14. [PMID: 26311345 PMCID: PMC5042044 DOI: 10.1111/andr.12090] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 01/04/2023]
Abstract
Cell cycle control during spermatogenesis is a highly complex process owing to the control of the mitotic expansion of the spermatogonial cell population and following meiosis, induction of DNA breaks during meiosis and the high levels of physiological germ-cell apoptosis. We set out to study how E2F1, a key controller of cell cycle, apoptosis, and DNA damage responses, functions in the developing and adult testis. We first analyzed the expression pattern of E2f1 during post-natal testis development using RNA in situ hybridization, which showed a differential expression pattern of E2f1 in the adult and juvenile mouse testes. To study the function of E2f1, we took advantage of the E2F1(-/-) mouse line, which was back-crossed to C57Bl/6J genetic background. E2f1 loss led to a severe progressive testicular atrophy beginning at the age of 20 days. Spermatogonial apoptosis during the first wave of spermatogenesis was decreased. However, already in the first wave of spermatogenesis an extensive apoptosis of spermatocytes was observed. In the adult E2F1(-/-) testes, the atrophy due to loss of spermatocytes was further exacerbated by loss of spermatogonial stem cells. Surprisingly, only subtle changes in global gene expression array profiling were observed in E2F1(-/-) testis at PND20. To dissect the changes in each testicular cell type, an additional comparative analysis of the array data was performed making use of previously published data on transcriptomes of the individual testicular cell types. Taken together, our data indicate that E2F1 has a differential role during first wave of spermatogenesis and in the adult testis, which emphasizes the complex nature of cell cycle control in the developing testis.
Collapse
Affiliation(s)
- E Rotgers
- Department of Physiology, University of Turku, Turku, Finland.,Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - M Nurmio
- Department of Physiology, University of Turku, Turku, Finland.,Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - E Pietilä
- Department of Physiology, University of Turku, Turku, Finland
| | - S Cisneros-Montalvo
- Department of Physiology, University of Turku, Turku, Finland.,Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - J Toppari
- Department of Physiology, University of Turku, Turku, Finland.,Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
23
|
Kettunen KM, Karikoski R, Hämäläinen RH, Toivonen TT, Antonenkov VD, Kulesskaya N, Voikar V, Hölttä-Vuori M, Ikonen E, Sainio K, Jalanko A, Karlberg S, Karlberg N, Lipsanen-Nyman M, Toppari J, Jauhiainen M, Hiltunen JK, Jalanko H, Lehesjoki AE. Trim37-deficient mice recapitulate several features of the multi-organ disorder Mulibrey nanism. Biol Open 2016; 5:584-95. [PMID: 27044324 PMCID: PMC4874348 DOI: 10.1242/bio.016246] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mulibrey nanism (MUL) is a rare autosomal recessive multi-organ disorder characterized by severe prenatal-onset growth failure, infertility, cardiopathy, risk for tumors, fatty liver, and type 2 diabetes. MUL is caused by loss-of-function mutations in TRIM37, which encodes an E3 ubiquitin ligase belonging to the tripartite motif (TRIM) protein family and having both peroxisomal and nuclear localization. We describe a congenic Trim37 knock-out mouse (Trim37−/−) model for MUL. Trim37−/− mice were viable and had normal weight development until approximately 12 months of age, after which they started to manifest increasing problems in wellbeing and weight loss. Assessment of skeletal parameters with computer tomography revealed significantly smaller skull size, but no difference in the lengths of long bones in Trim37−/− mice as compared with wild-type. Both male and female Trim37−/− mice were infertile, the gonads showing germ cell aplasia, hilus and Leydig cell hyperplasia and accumulation of lipids in and around Leydig cells. Male Trim37−/− mice had elevated levels of follicle-stimulating and luteinizing hormones, but maintained normal levels of testosterone. Six-month-old Trim37−/− mice had elevated fasting blood glucose and low fasting serum insulin levels. At 1.5 years Trim37−/− mice showed non-compaction cardiomyopathy, hepatomegaly, fatty liver and various tumors. The amount and morphology of liver peroxisomes seemed normal in Trim37−/− mice. The most consistently seen phenotypes in Trim37−/− mice were infertility and the associated hormonal findings, whereas there was more variability in the other phenotypes observed. Trim37−/− mice recapitulate several features of the human MUL disease and thus provide a good model to study disease pathogenesis related to TRIM37 deficiency, including infertility, non-alcoholic fatty liver disease, cardiomyopathy and tumorigenesis. Summary: A congenic Trim37-deficient mouse model recapitulates several features of the human disorder Mulibrey nanism, and thus provides a good model to study disease pathogenesis related to TRIM37 deficiency.
Collapse
Affiliation(s)
- Kaisa M Kettunen
- Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland Research Programs Unit, Molecular Neurology, University of Helsinki, FI-00290 Helsinki, Finland Neuroscience Center, University of Helsinki, FI-00790 Helsinki, Finland Institute for Molecular Medicine Finland FIMM, University of Helsinki, FI-00290 Helsinki, Finland
| | - Riitta Karikoski
- Department of Pathology, Central Hospital of Tavastia, FI-13530 Hämeenlinna, Finland
| | - Riikka H Hämäläinen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio FI-70211, Finland
| | | | - Vasily D Antonenkov
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90220 Oulu, Finland
| | | | - Vootele Voikar
- Neuroscience Center, University of Helsinki, FI-00790 Helsinki, Finland
| | - Maarit Hölttä-Vuori
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland Minerva Foundation Institute for Medical Research, FI-00290 Helsinki, Finland
| | - Elina Ikonen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland Minerva Foundation Institute for Medical Research, FI-00290 Helsinki, Finland
| | - Kirsi Sainio
- Biochemistry and Developmental Biology, Institute of Biomedicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - Anu Jalanko
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, FI-00290 Helsinki, Finland
| | - Susann Karlberg
- Department of Endocrinology, Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland
| | - Niklas Karlberg
- Department of Endocrinology, Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland
| | - Marita Lipsanen-Nyman
- Department of Endocrinology, Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland
| | - Jorma Toppari
- Departments of Physiology and Pediatrics, University of Turku, FI-20520 Turku, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, FI-00290 Helsinki, Finland
| | - J Kalervo Hiltunen
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90220 Oulu, Finland
| | - Hannu Jalanko
- Department of Nephrology and Transplantation, Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland
| | - Anna-Elina Lehesjoki
- Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland Research Programs Unit, Molecular Neurology, University of Helsinki, FI-00290 Helsinki, Finland Neuroscience Center, University of Helsinki, FI-00790 Helsinki, Finland
| |
Collapse
|
24
|
Hakkarainen J, Jokela H, Pakarinen P, Heikelä H, Kätkänaho L, Vandenput L, Ohlsson C, Zhang FP, Poutanen M. Hydroxysteroid (17β)-dehydrogenase 1–deficient female mice present with normal puberty onset but are severely subfertile due to a defect in luteinization and progesterone production. FASEB J 2015; 29:3806-16. [DOI: 10.1096/fj.14-269035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/18/2015] [Indexed: 11/11/2022]
|
25
|
Folliculogenesis Is Not Fully Inhibited during GnRH Analogues Treatment in Mice Challenging Their Efficiency to Preserve the Ovarian Reserve during Chemotherapy in This Model. PLoS One 2015; 10:e0137164. [PMID: 26325271 PMCID: PMC4556658 DOI: 10.1371/journal.pone.0137164] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/13/2015] [Indexed: 11/27/2022] Open
Abstract
As many chemotherapy regimens induce follicular depletion, fertility preservation became a major concern in young cancer patients. By maintaining follicles at the resting stage, gonadotropin-releasing hormone analogues (GnRHa) were proposed as an ovarian-protective option during chemotherapy. However, their efficacy and mechanisms of action remain to be elucidated. Mice were dosed with cyclophosphamide (Cy, 100–500mg/kg i.p) to quantify follicular depletion and evaluate apoptosis at different times. We observed a dose-dependent depletion of the follicular reserve within 24 hours after Cy injection with a mean follicular loss of 45% at the dose of 200mg/kg. Apoptosis occurs in the granulosa cells of growing follicles within 12 hours after Cy treatment, while no apoptosis was detected in resting follicles suggesting that chemotherapy acutely affects both resting and growing follicles through different mechanisms. We further tested the ability of both GnRH agonist and antagonist to inhibit oestrus cycles, follicular growth and FSH secretion in mice and to protect ovarian reserve against chemotherapy. Although GnRHa were efficient to disrupt oestrus cycles, they failed to inhibit follicular development, irrespective of the doses and injection sites (sc or im). Around 20% of healthy growing follicles were still observed during GnRHa treatment and serum FSH levels were not reduced either by antagonist or agonist. GnRHa had no effect on Cy-induced follicular damages. Thus, we showed that GnRHa were not as efficient at inhibiting the pituitary-gonadal axis in mice as in human. Furthermore, the acute depletion of primordial follicles observed after chemotherapy does not support the hypothesis that the ovary may be protected by gonadotropin suppression.
Collapse
|
26
|
Björkgren I, Gylling H, Turunen H, Huhtaniemi I, Strauss L, Poutanen M, Sipilä P. Imbalanced lipid homeostasis in the conditional Dicer1 knockout mouse epididymis causes instability of the sperm membrane. FASEB J 2014; 29:433-42. [DOI: 10.1096/fj.14-259382] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ida Björkgren
- Department of PhysiologyInstitute of BiomedicineUniversity of TurkuTurkuFinland
- Turku Doctoral Programme of Biomedical SciencesTurkuFinland
| | - Helena Gylling
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
- Department of MedicineDivision of Internal MedicineUniversity of HelsinkiHelsinkiFinland
| | - Heikki Turunen
- Department of PhysiologyInstitute of BiomedicineUniversity of TurkuTurkuFinland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith CampusLondonUnited Kingdom
| | - Leena Strauss
- Department of PhysiologyInstitute of BiomedicineUniversity of TurkuTurkuFinland
- Turku Center for Disease ModelingUniversity of TurkuTurkuFinland
| | - Matti Poutanen
- Department of PhysiologyInstitute of BiomedicineUniversity of TurkuTurkuFinland
- Turku Center for Disease ModelingUniversity of TurkuTurkuFinland
- Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Petra Sipilä
- Department of PhysiologyInstitute of BiomedicineUniversity of HelsinkiHelsinkiFinland
- Laboratory Animal CentreUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
27
|
Nurmio M, Kallio J, Adam M, Mayerhofer A, Toppari J, Jahnukainen K. Peritubular myoid cells have a role in postnatal testicular growth. SPERMATOGENESIS 2014; 2:79-87. [PMID: 22670217 PMCID: PMC3364795 DOI: 10.4161/spmg.20067] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
FSH stimulates testicular growth by increasing Sertoli cell proliferation and elongation of seminiferous cords. Little is known about the peritubular myoid cells in testicular development. In order to investigate the role of peritubular myoid cells in early testicular growth in rodents, two traditional models to induce testicular growth were used: FSH treatment and hemicastration. In order to affect proliferation of peritubular myoid cells, both treatments were combined with imatinib, a tyrosine kinase inhibitor. In addition, effects of imatinib on human testicular peritubular cell proliferation were investigated. Testicular weight, diameter and length of seminiferous cords, numbers of germ, Sertoli and BrdU-positive cells and FSH-levels were measured. FSH treatment and hemicastration increased length of the seminiferous cords and testicular weight by increasing first the early proliferation of peritubular myoid cells and later also the proliferation of the Sertoli cells. Imatinib blocked the FSH and hemicastration -induced testicular hypertrophy and decreased the proliferation of PDGF-stimulated human testicular peritubular cells in vitro. Present results provide new evidence that peritubular myoid cells have an important role in postnatal testicular growth.
Collapse
|
28
|
Viluksela M, Heikkinen P, van der Ven LTM, Rendel F, Roos R, Esteban J, Korkalainen M, Lensu S, Miettinen HM, Savolainen K, Sankari S, Lilienthal H, Adamsson A, Toppari J, Herlin M, Finnilä M, Tuukkanen J, Leslie HA, Hamers T, Hamscher G, Al-Anati L, Stenius U, Dervola KS, Bogen IL, Fonnum F, Andersson PL, Schrenk D, Halldin K, Håkansson H. Toxicological profile of ultrapure 2,2',3,4,4',5,5'-heptachlorbiphenyl (PCB 180) in adult rats. PLoS One 2014; 9:e104639. [PMID: 25137063 PMCID: PMC4138103 DOI: 10.1371/journal.pone.0104639] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 07/10/2014] [Indexed: 11/19/2022] Open
Abstract
PCB 180 is a persistent non-dioxin-like polychlorinated biphenyl (NDL-PCB) abundantly present in food and the environment. Risk characterization of NDL-PCBs is confounded by the presence of highly potent dioxin-like impurities. We used ultrapure PCB 180 to characterize its toxicity profile in a 28-day repeat dose toxicity study in young adult rats extended to cover endocrine and behavioral effects. Using a loading dose/maintenance dose regimen, groups of 5 males and 5 females were given total doses of 0, 3, 10, 30, 100, 300, 1000 or 1700 mg PCB 180/kg body weight by gavage. Dose-responses were analyzed using benchmark dose modeling based on dose and adipose tissue PCB concentrations. Body weight gain was retarded at 1700 mg/kg during loading dosing, but recovered thereafter. The most sensitive endpoint of toxicity that was used for risk characterization was altered open field behavior in females; i.e. increased activity and distance moved in the inner zone of an open field suggesting altered emotional responses to unfamiliar environment and impaired behavioral inhibition. Other dose-dependent changes included decreased serum thyroid hormones with associated histopathological changes, altered tissue retinoid levels, decreased hematocrit and hemoglobin, decreased follicle stimulating hormone and luteinizing hormone levels in males and increased expression of DNA damage markers in liver of females. Dose-dependent hypertrophy of zona fasciculata cells was observed in adrenals suggesting activation of cortex. There were gender differences in sensitivity and toxicity profiles were partly different in males and females. PCB 180 adipose tissue concentrations were clearly above the general human population levels, but close to the levels in highly exposed populations. The results demonstrate a distinct toxicological profile of PCB 180 with lack of dioxin-like properties required for assignment of WHO toxic equivalency factor. However, PCB 180 shares several toxicological targets with dioxin-like compounds emphasizing the potential for interactions.
Collapse
Affiliation(s)
- Matti Viluksela
- Department of Environmental Health, National Institute for Health and Welfare, Kuopio, Finland
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
- * E-mail:
| | - Päivi Heikkinen
- Department of Environmental Health, National Institute for Health and Welfare, Kuopio, Finland
| | - Leo T. M. van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Filip Rendel
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert Roos
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Javier Esteban
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche (Alicante), Spain
| | - Merja Korkalainen
- Department of Environmental Health, National Institute for Health and Welfare, Kuopio, Finland
| | - Sanna Lensu
- Department of Environmental Health, National Institute for Health and Welfare, Kuopio, Finland
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Hanna M. Miettinen
- Department of Environmental Health, National Institute for Health and Welfare, Kuopio, Finland
| | | | - Satu Sankari
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Hellmuth Lilienthal
- Center of Toxicology, IPA – Institute for Prevention and Occupational Medicine, German Social Accident Insurance, Ruhr University of Bochum, Bochum, Germany
| | - Annika Adamsson
- Department of Physiology, University of Turku, Turku, Finland
| | - Jorma Toppari
- Department of Physiology, University of Turku, Turku, Finland
- Department of Paediatrics, University of Turku, Turku, Finland
| | - Maria Herlin
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mikko Finnilä
- Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland
| | - Heather A. Leslie
- Institute for Environmental Studies, VU University Amsterdam, Amsterdam, The Netherlands
| | - Timo Hamers
- Institute for Environmental Studies, VU University Amsterdam, Amsterdam, The Netherlands
| | - Gerd Hamscher
- Institute of Food Chemistry and Food Biotechnology, Justus-Liebig University, Giessen, Germany
| | - Lauy Al-Anati
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulla Stenius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kine-Susann Dervola
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Inger-Lise Bogen
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Frode Fonnum
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Dieter Schrenk
- Food Chemistry and Toxicology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Krister Halldin
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Caldwell ASL, Middleton LJ, Jimenez M, Desai R, McMahon AC, Allan CM, Handelsman DJ, Walters KA. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinology 2014; 155:3146-59. [PMID: 24877633 DOI: 10.1210/en.2014-1196] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Polycystic ovary syndrome (PCOS) affects 5-10% of women of reproductive age, causing a range of reproductive, metabolic and endocrine defects including anovulation, infertility, hyperandrogenism, obesity, hyperinsulinism, and an increased risk of type 2 diabetes and cardiovascular disease. Hyperandrogenism is the most consistent feature of PCOS, but its etiology remains unknown, and ethical and logistic constraints limit definitive experimentation in humans to determine mechanisms involved. In this study, we provide the first comprehensive characterization of reproductive, endocrine, and metabolic PCOS traits in 4 distinct murine models of hyperandrogenism, comprising prenatal dihydrotestosterone (DHT, potent nonaromatizable androgen) treatment during days 16-18 of gestation, or long-term treatment (90 days from 21 days of age) with DHT, dehydroepiandrosterone (DHEA), or letrozole (aromatase inhibitor). Prenatal DHT-treated mature mice exhibited irregular estrous cycles, oligo-ovulation, reduced preantral follicle health, hepatic steatosis, and adipocyte hypertrophy, but lacked overall changes in body-fat composition. Long-term DHT treatment induced polycystic ovaries displaying unhealthy antral follicles (degenerate oocyte and/or > 10% pyknotic granulosa cells), as well as anovulation and acyclicity in mature (16-week-old) females. Long-term DHT also increased body and fat pad weights and induced adipocyte hypertrophy and hypercholesterolemia. Long-term letrozole-treated mice exhibited absent or irregular cycles, oligo-ovulation, polycystic ovaries containing hemorrhagic cysts atypical of PCOS, and displayed no metabolic features of PCOS. Long-term dehydroepiandrosterone treatment produced no PCOS features in mature mice. Our findings reveal that long-term DHT treatment replicated a breadth of ovarian, endocrine, and metabolic features of human PCOS and provides the best mouse model for experimental studies of PCOS pathogenesis.
Collapse
Affiliation(s)
- A S L Caldwell
- Andrology Laboratory (A.S.L.C., L.J.M., M.J., R.D., C.M.A.,D.J.H., K.A.W.) and Biogerontology Laboratory (A.C.M.), ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Retinoblastoma protein (RB) interacts with E2F3 to control terminal differentiation of Sertoli cells. Cell Death Dis 2014; 5:e1274. [PMID: 24901045 PMCID: PMC4611710 DOI: 10.1038/cddis.2014.232] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 11/09/2022]
Abstract
The retinoblastoma protein (RB) is essential for normal cell cycle control. RB function depends, at least in part, on interactions with the E2F family of DNA-binding transcription factors (E2Fs). To study the role of RB in the adult testis, a Sertoli cell (SC)-specific Rb knockout mouse line (SC-RbKO) was generated using the Cre/loxP recombination system. SC-RbKO mice exhibited an age-dependent testicular atrophy, impaired fertility, severe SC dysfunction, and spermatogenic defects. Removal of Rb in SC induced aberrant SC cycling, dedifferentiation, and apoptosis. Here we show that E2F3 is the only E2F expressed in mouse SCs and that RB interacts with E2F3 during mouse testicular development. In the absence of RB, the other retinoblastoma family members p107 and p130 began interacting with E2F3 in the adult testes. In vivo silencing of E2F3 partially restored the SC maturation and survival as well as spermatogenesis in the SC-RbKO mice. These results point to RB as a key regulator of SC function in adult mice and that the RB/E2F3 pathway directs SC maturation, cell cycle quiescence, and RB protects SC from apoptosis.
Collapse
|
31
|
Oduwole OO, Vydra N, Wood NEM, Samanta L, Owen L, Keevil B, Donaldson M, Naresh K, Huhtaniemi IT. Overlapping dose responses of spermatogenic and extragonadal testosterone actions jeopardize the principle of hormonal male contraception. FASEB J 2014; 28:2566-76. [PMID: 24599970 PMCID: PMC4376501 DOI: 10.1096/fj.13-249219] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Testosterone (T), alone or in combination with progestin, provides a promising approach to hormonal male contraception. Its principle relies on enhanced negative feedback of exogenous T to suppress gonadotropins, thereby blocking the testicular T production needed for spermatogenesis, while simultaneously maintaining the extragonadal androgen actions, such as potency and libido, to avoid hypogonadism. A serious drawback of the treatment is that a significant proportion of men do not reach azoospermia or severe oligozoospermia, commensurate with contraceptive efficacy. We tested here, using hypogonadal luteinizing hormone/choriongonadotropin receptor (LHCGR) knockout (LHR−/−) mice, the basic principle of the T-based male contraceptive method, that a specific T dose could maintain extragonadal androgen actions without simultaneously activating spermatogenesis. LHR−/− mice were treated with increasing T doses, and the responses of their spermatogenesis and extragonadal androgen actions (including gonadotropin suppression and sexual behavior) were assessed. Conspicuously, all dose responses to T were practically superimposable, and no dose of T could be defined that would maintain sexual function and suppress gonadotropins without simultaneously activating spermatogenesis. This finding, never addressed in clinical contraceptive trials, is not unexpected in light of the same androgen receptor mediating androgen actions in all organs. When extrapolated to humans, our findings may jeopardize the current approach to hormonal male contraception and call for more effective means of inhibiting intratesticular T production or action, to achieve consistent spermatogenic suppression.—Oduwole, O. O., Vydra, N., Wood, N. E. M., Samanta, L., Owen, L., Keevil, B., Donaldson, M., Naresh, K., Huhtaniemi, I. T. Overlapping dose responses of spermatogenic and extragonadal testosterone actions jeopardize the principle of hormonal male contraception.
Collapse
Affiliation(s)
- Olayiwola O Oduwole
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, and
| | - Natalia Vydra
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, and Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Nicholas E M Wood
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, and
| | - Luna Samanta
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, and Biochemistry Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, India
| | - Laura Owen
- Biochemistry Department, University Hospital of South Manchester, Manchester, UK; and
| | - Brian Keevil
- Biochemistry Department, University Hospital of South Manchester, Manchester, UK; and
| | - Mandy Donaldson
- Department of Clinical Biochemistry, Imperial College Healthcare National Health Service Trust, Charing Cross Hospital, London, UK
| | - Kikkeri Naresh
- Department of Histopathology, Imperial College Healthcare National Health Service Trust, Imperial College London, Hammersmith Campus, London, UK
| | - Ilpo T Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, and
| |
Collapse
|
32
|
Haverfield JT, Meachem SJ, Nicholls PK, Rainczuk KE, Simpson ER, Stanton PG. Differential permeability of the blood-testis barrier during reinitiation of spermatogenesis in adult male rats. Endocrinology 2014; 155:1131-44. [PMID: 24424039 DOI: 10.1210/en.2013-1878] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The blood-testis barrier (BTB) sequesters meiotic spermatocytes and differentiating spermatids away from the vascular environment. We aimed to assess whether meiosis and postmeiotic differentiation could occur when the BTB is permeable. Using a model of meiotic suppression and reinitiation, BTB function was assessed using permeability tracers of small, medium, and large (0.6-, 70-, and 150-kDa) sizes to emulate blood- and lymphatic-borne factors that could cross the BTB. Adult rats (n = 9/group) received the GnRH antagonist acyline (10 wk) to suppress gonadotropins, followed by testosterone (24cm Silastic implant), for 2, 4, 7, 10, 15, and 35 days. In acyline-suppressed testes, all tracers permeated the seminiferous epithelium. As spermatocytes up to diplotene stage XIII reappeared, both the 0.6- and 70-kDa tracers, but not 150 kDa, permeated around these cells. Intriguingly, the 0.6- and 70-kDa tracers were excluded from pachytene spermatocytes at stages VII and VIII but not in subsequent stages. The BTB became progressively impermeable to the 0.6- and 70-kDa tracers as stages IV-VII round spermatids reappeared in the epithelium. This coincided with the appearance of the tight junction protein, claudin-12, in Sertoli cells and at the BTB. We conclude that meiosis can occur when the BTB is permeable to factors up to 70 kDa during the reinitiation of spermatogenesis. Moreover, BTB closure corresponds with the presence of particular pachytene spermatocytes and round spermatids. This research has implications for understanding the effects of BTB dynamics in normal spermatogenesis and also potentially in states where spermatogenesis is suppressed, such as male hormonal contraception or infertility.
Collapse
Affiliation(s)
- Jenna T Haverfield
- Prince Henry's Institute (J.T.H., S.J.M., P.K.N., K.E.R., E.R.S., P.G.S.), Monash Medical Centre, Clayton, VIC 3168, Australia; and Departments of Anatomy and Developmental Biology (J.T.H., S.J.M.) and Biochemistry and Molecular Biology (P.K.N., E.R.S., P.G.S.), Monash University, Clayton, VIC 3800, Australia
| | | | | | | | | | | |
Collapse
|
33
|
Gerrits H, Paradé MCBC, Koonen-Reemst AMCB, Bakker NEC, Timmer-Hellings L, Sollewijn Gelpke MD, Gossen JA. Reversible infertility in a liver receptor homologue-1 (LRH-1)-knockdown mouse model. Reprod Fertil Dev 2014; 26:293-306. [DOI: 10.1071/rd12131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 12/22/2012] [Indexed: 12/15/2022] Open
Abstract
Liver receptor homologue-1 (LRH-1) is an orphan nuclear receptor that has been implicated in steroid hormone biosynthesis and fertility. Herein we describe a transgenic inducible short hairpin (sh) RNA mouse model that was used to study the effect of transient LRH-1 knockdown in vivo. Induction of expression of the shRNA directed against LRH-1 for 2–6 weeks resulted in 80% knockdown of LRH-1 protein in the ovary and complete infertility. Gonadotropin hyperstimulation could not rescue the observed defects in ovulation and corpus luteum formation in LRH-1-knockdown mice. The infertility phenotype was fully reversible because LRH-1-knockdown females became pregnant and delivered normal size litters and healthy pups after cessation of LRH-1 shRNA expression. Timed ovarian microarray analysis showed that, in line with the observed decrease in plasma progesterone levels, key steroid biosynthesis genes, namely Star, Cyp11a1, Hsd3b and Scarb1, were downregulated in LRH-1-knockdown ovaries. In contrast with what has been described previously, no clear effect was observed on oestrogenic activity in LRH-1-knockdown mice. Only Sult1e1 and, surprisingly, Hsd17b7 expression was modulated with potentially opposite effects on oestradiol bioavailability. In conclusion, the fully reversible infertility phenotype of LRH-1-knockdown mice shows the feasibility of an LRH-1 antagonist as new contraceptive therapy with a mechanism of action that most prominently affects cholesterol availability and progesterone production.
Collapse
|
34
|
Abel MH, Charlton HM, Huhtaniemi I, Pakarinen P, Kumar TR, Christian HC. An investigation into pituitary gonadotrophic hormone synthesis, secretion, subunit gene expression and cell structure in normal and mutant male mice. J Neuroendocrinol 2013; 25:863-75. [PMID: 23895394 PMCID: PMC5599115 DOI: 10.1111/jne.12081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 07/16/2013] [Accepted: 07/24/2013] [Indexed: 01/30/2023]
Abstract
To investigate brain-pituitary-gonadal inter-relationships, we have compared the effects of mutations that perturb the hypothalamic-pituitary-gonadal axis in male mice. Specifically, serum and pituitary gonadotrophin concentrations, gonadotrophin gene expression, and gonadotroph structure and number were measured. Follicle-stimulating hormone (FSH)β knockout (FSHβKO), FSH receptor knockout (FSHRKO), luteinising hormone (LH) receptor knockout (LuRKO), hypogonadal (hpg), testicular feminised (tfm) and gonadectomised mice were compared with control wild-type mice or heterozygotes. Serum levels of LH were similar in FSHβKO, FSHRKO and heterozygote males despite decreased androgen production in KO males. As expected, there was no detectable FSH in the serum or pituitary and an absence of expression of the FSHβ subunit gene in FSHβKO mice. However, there was a significant increase in expression of the common α and LHβ subunit genes in FSHRKO males. The morphology of FSHβKO and FSHRKO gonadotrophs was not significantly different from controls, except that the subpopulation of granules consisting of an electron-dense core and electron-lucent 'halo' was not observed in FSHβKO gonadotrophs and the granules were smaller in diameter. In the gonadotrophin-releasing hormone deficient hpg mouse, gonadotrophin mRNA and hormone levels were significantly lower compared to control mice and gonadotrophs were correspondingly smaller, with less abundant endoplasmic reticulum and reduced secretory granules. In LuRKO, tfm and gonadectomised mice, hyperstimulation of LHβ and FSHβ mRNA and serum protein concentrations was reflected by subcellular changes in gonadotroph morphology, including more dilated rough endoplasmic reticulum and more secretory granules distributed adjacent to the plasma membrane. In summary, major differences in pituitary content and serum concentrations of the gonadotrophins LH and FSH have been found between normal and mutant male mice. These changes are associated with changes in transcriptional activity of the gonadotrophin subunit genes and are reflected by changes in the cellular structure and secretory granule architecture within the gonadotroph cells.
Collapse
Affiliation(s)
- M. H. Abel
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - H. M. Charlton
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - I. Huhtaniemi
- Department of Physiology, University of Turku, Turku, Finland
| | - P. Pakarinen
- Department of Physiology, University of Turku, Turku, Finland
| | - T. R. Kumar
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center,, Kansas City, KS, USA
| | - H. C. Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Cheng XB, Jimenez M, Desai R, Middleton LJ, Joseph SR, Ning G, Allan CM, Smith JT, Handelsman DJ, Walters KA. Characterizing the neuroendocrine and ovarian defects of androgen receptor-knockout female mice. Am J Physiol Endocrinol Metab 2013; 305:E717-26. [PMID: 23880317 DOI: 10.1152/ajpendo.00263.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Homozygous androgen receptor (AR)-knockout (ARKO) female mice are subfertile due to both intra- and extraovarian (neuroendocrine) defects as defined by ovary transplantation. Using ARKO mice, this study set out to reveal the precise AR-regulated pathways required for optimal androgen-regulated ovulation and fertility. ARKO females exhibit deficient neuroendocrine negative feedback, with a reduced serum luteinizing hormone (LH) response to ovariectomy (OVX) (P < 0.01). Positive feedback is also altered as intact ARKO females, at late proestrus, exhibit an often mistimed endogenous ovulatory LH surge. Furthermore, at late proestrus, intact ARKO females display diminished preovulatory serum estradiol (E2; P < 0.01) and LH (P < 0.05) surge levels and reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus (P < 0.01) compared with controls. However, this reduced ovulatory LH response in intact ARKO females can be rescued by OVX and E2 priming or treatment with endogenous GnRH. These findings reveal that AR regulates the negative feedback response to E2, E2-positive feedback is compromised in ARKO mice, and AR-regulated negative and positive steroidal feedback pathways impact on intrahypothalamic control of the kisspeptin/GnRH/LH cascade. In addition, intraovarian AR-regulated pathways controlling antral to preovulatory follicle dynamics are disrupted because adult ARKO ovaries collected at proestrus have small antral follicles with reduced oocyte/follicle diameter ratios (P < 0.01) and increased proportions of unhealthy large antral follicles (P < 0.05) compared with controls. As a consequence of aberrant follicular growth patterns, proestrus ARKO ovaries also exhibit fewer preovulatory follicle (P < 0.05) and corpora lutea numbers (P < 0.01). However, embryo development to the blastocyst stage is unchanged in ARKO females, and hence, the subfertility is a consequence of reduced ovulations and not altered embryo quality. These findings reveal that the AR has a functional role in neuroendocrine regulation and timing of the ovulatory LH surge as well as antral/preovulatory follicle development.
Collapse
Affiliation(s)
- Xiaobing B Cheng
- ANZAC Research Institute, Andrology Laboratory, Concord Hospital, University of Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Gold E, Marino FE, Harrison C, Makanji Y, Risbridger G. Activin-βcreduces reproductive tumour progression and abolishes cancer-associated cachexia in inhibin-deficient mice. J Pathol 2013. [DOI: 10.1002/path.4142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Elspeth Gold
- Department of Anatomy; University of Otago; Dunedin New Zealand
| | | | | | | | - Gail Risbridger
- Department of Anatomy and Developmental Biology; Monash University; Clayton Victoria Australia
| |
Collapse
|
37
|
Walters KA, Middleton LJ, Joseph SR, Hazra R, Jimenez M, Simanainen U, Allan CM, Handelsman DJ. Targeted loss of androgen receptor signaling in murine granulosa cells of preantral and antral follicles causes female subfertility. Biol Reprod 2012; 87:151. [PMID: 23115271 DOI: 10.1095/biolreprod.112.102012] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Ovarian granulosa cells display strong androgen receptor (AR) expression, suggesting a functional role for direct AR-mediated actions within developing mammalian follicles. By crossing AR-floxed and anti-Müllerian hormone (AMH)-Cre recombinase mice, we generated granulosa cell-specific androgen receptor knockout mice (GCARKO). Cre expression, assessed by lacZ activity, localized to 70%-100% of granulosa cells in most preantral to antral follicles, allowing for selected evaluation of granulosa cell AR-dependent actions during follicle development. Relative to wild-type (WT) females, GCARKO females were subfertile, producing a 24% reduction in the number of litters (P < 0.05) over 6 mo and an age-dependent decrease in total number of pups born, evident from 6 mo of age (P < 0.05). Follicle dynamics were altered in GCARKO ovaries at 3 mo of age, with a significant reduction in large preantral and small antral follicle numbers compared to WT ovaries (P < 0.05). Global premature follicle depletion was not observed, but increased follicular atresia was evident in GCARKO ovaries at 6 mo of age, with an 81% increase in unhealthy follicles and zona pellucida remnants (P < 0.01). Cumulus cell expansion was decreased (P < 0.01) and oocyte viability was diminished in GCARKO females, with a significant reduction in the percentage of oocytes fertilized after natural mating and, thus, in the rate of progression to the two-cell embryo stage (P < 0.05). In addition, compared with age-matched WT females, 6-mo-old GCARKO females exhibited significantly prolonged estrous cycles (P ≤ 0.05), suggesting altered hypothalamic-pituitary-gonadal feedback signaling. In conclusion, our findings revealed that selective loss of granulosa cell AR actions during preantral and antral stages of development leads to a premature reduction in female fecundity through reduced follicle health and oocyte viability.
Collapse
Affiliation(s)
- Kirsty A Walters
- ANZAC Research Institute, Department of Andrology, Concord Hospital, University of Sydney, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Nicholls PK, Stanton PG, Chen JL, Olcorn JS, Haverfield JT, Qian H, Walton KL, Gregorevic P, Harrison CA. Activin signaling regulates Sertoli cell differentiation and function. Endocrinology 2012; 153:6065-77. [PMID: 23117933 DOI: 10.1210/en.2012-1821] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Throughout development, activin A signaling stimulates proliferation and inhibits differentiation of testicular Sertoli cells. A decline in activin levels at puberty corresponds with the differentiation of Sertoli cells that is required to sustain spermatogenesis. In this study, we consider whether terminally differentiated Sertoli cells can revert to a functionally immature phenotype in response to activin A. To increase systemic activin levels, the right tibialis anterior muscle of 7-wk-old C57BL/6J mice was transduced with an adeno-associated virus (rAAV6) expressing activin A. We show that chronic activin signaling reduces testis mass by 23.5% compared with control animals and induces a hypospermatogenic phenotype, consistent with a failure of Sertoli cells to support spermatogenesis. We use permeability tracers and transepithelial electrical resistance measurements to demonstrate that activin potently disrupts blood-testis-barrier function in adult mice and ablates tight junction formation in differentiated primary Sertoli cells, respectively. Furthermore, increased activin signaling reinitiates a program of cellular proliferation in primary Sertoli cells as determined by 5-ethynyl-2'-deoxyuridine incorporation. Proliferative cells reexpress juvenile markers, including cytokeratin-18, and suppress mature markers, including claudin-11. Thus, activin A is the first identified factor capable of reprogramming Sertoli cells to an immature, dedifferentiated phenotype. This study indicates that activin signaling must be strictly controlled in the adult in order to maintain Sertoli cell function in spermatogenesis.
Collapse
Affiliation(s)
- Peter K Nicholls
- Prince Henry's Institute of Medical Research, Clayton 3168, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Simanainen U, Gao YR, Walters KA, Watson G, Desai R, Jimenez M, Handelsman DJ. Androgen resistance in female mice increases susceptibility to DMBA-induced mammary tumors. Discov Oncol 2012; 3:113-24. [PMID: 22370991 DOI: 10.1007/s12672-012-0107-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hormones, notably estrogens, are pivotal in the origins of breast cancer but androgenic effects, while supported by persistence of AR expression in breast cancers, remain controversial. This study determined the role of the androgen actions via androgen receptor (AR) in experimental mammary cancer. Androgen-resistant female and male mice (ARKO) were generated using Cre/loxP technique and featured a global AR inactivation. The effect of AR inactivation and influence of genetic background on 7,12-dimethylbenz[a]anthracene (DMBA)-induced tumorigenesis was confirmed using two separate ARKO models with different genetic backgrounds. The onset of palpable mammary tumors was significantly faster in ARKO females (median time 22 vs 34 weeks, respectively; (p = 0.0024; multivariate Cox regression) compared to WT and independent of the mouse genetic background. The cumulative incidence at 9 months was 81 ± 10% [mean ± SE] for ARKO compared to 50 ± 13% in WT females. The increased DMBA susceptibility of ARKO females was associated with a higher epithelial proliferation index but not with major structural or receptor (estrogen or progesterone) expression differences between the virgin WT or ARKO female mammary glands. AR inactivation allowed substantial ductal extension in ARKO males while WT males displayed only rudimentary epithelial branches or complete regression of epithelial structures. Yet, DMBA did not induce epithelial mammary tumors in WT or ARKO males, demonstrating that AR inactivation alone is insufficient to promote mammary tumors. These results demonstrate that AR inactivation accelerates mammary carcinogenesis in female mice exposed to the chemical carcinogen DMBA regardless of mouse genetic background but require prior exposure to endogenous ovarian hormones.
Collapse
Affiliation(s)
- Ulla Simanainen
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, NSW 2139, Australia
| | | | | | | | | | | | | |
Collapse
|
40
|
Turunen HT, Sipilä P, Strauss L, Björkgren I, Huhtaniemi I, Poutanen M. Loss of Bmyc results in increased apoptosis associated with upregulation of Myc expression in juvenile murine testis. Reproduction 2012; 144:495-503. [PMID: 22869780 DOI: 10.1530/rep-11-0444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bmyc is a member of the Myc family of transcriptional regulators in the mouse and the rat. It is predominantly expressed in hormonally controlled tissues, with highest level of expression in the epididymis. The BMYC protein has been shown to function as a transcription factor in vitro and to inhibit MYC. To study the significance of BMYC in vivo, a Bmyc knockout (KO) mouse model was generated by homologous recombination. The KO mice were viable and fertile and did not display gross morphological or histological changes compared to the WT mice. However, the testes and the epididymides of the KO mice were smaller than those of the WT mice. Correspondingly, a tendency for a lower sperm concentration in the cauda epididymides of the KO mice was detected. The testosterone produced/testis was significantly reduced, and accordingly, the LH levels were increased in the KO mice. Also, the expression levels of Myc and several of its target genes were elevated in the testes of prepubertal KO mice, whereas no differences in gene expression levels were detected in adult mice. Associated with the increased Myc expression, more apoptotic spermatogenic cells were detected in the seminiferous tubules of the KO mice. In conclusion, our data suggest that Bmyc is a regulator of Myc in vivo and that overexpression of Myc in the developing testis leads to increased apoptosis of spermatogenic cells.
Collapse
Affiliation(s)
- Heikki T Turunen
- Department of Physiology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | | | | | | | | | | |
Collapse
|
41
|
Ventelä S, Côme C, Mäkelä JA, Hobbs RM, Mannermaa L, Kallajoki M, Chan EK, Pandolfi PP, Toppari J, Westermarck J. CIP2A promotes proliferation of spermatogonial progenitor cells and spermatogenesis in mice. PLoS One 2012; 7:e33209. [PMID: 22461891 PMCID: PMC3312892 DOI: 10.1371/journal.pone.0033209] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 02/13/2012] [Indexed: 01/05/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a critical regulator of protein serine/threonine phosphorylation. However, the physiological and developmental roles of different PP2A complexes are very poorly understood. Here, we show that a newly characterized PP2A inhibitory protein CIP2A is co-expressed with ki-67 and with self-renewal protein PLZF in the spermatogonial progenitor cell (SPC) population in the testis. CIP2A and PLZF expression was shown also to correlate Ki-67 expression in human testicular spermatogonia. Functionally, CIP2A mutant mouse testes exhibited smaller number of PLZF-positive SPCs and reduced sperm counts. Moreover, seminiferous tubuli cells isolated from CIP2A mutant mice showed reduced expression of Plzf and other renewal genes Oct-4 and Nanog at mRNA level. However, PLZF-deficient testes did not show altered CIP2A expression. Importantly, spermatogonia-specific restoration of CIP2A expression rescued PLZF expression and sperm production defects observed in CIP2A mutant mice. Taken together, these results reveal first physiological function for an emerging human oncoprotein CIP2A, and provide insights into maintenance of PLZF-positive progenitors. Moreover, demonstration that CIP2A expression can be systematically inhibited without severe consequences to normal mouse development and viability may have clinical relevance regarding targeting of oncogenic CIP2A for future cancer therapies.
Collapse
Affiliation(s)
- Sami Ventelä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Physiology, University of Turku, Turku, Finland
- Department of Otorhinolaryngology, Turku University Hospital, Turku, Finland
| | - Christophe Côme
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Juho-Antti Mäkelä
- Department of Physiology, University of Turku, Turku, Finland
- Turku Graduate School of Biomedical Sciences, University of Turku, Turku, Finland
| | - Robin M. Hobbs
- Cancer Genetics Program, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leni Mannermaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Edward K. Chan
- Department of Oral Biology, University of Florida, Gainesville, Florida, United States of America
| | - Pier Paolo Pandolfi
- Cancer Genetics Program, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jorma Toppari
- Department of Physiology, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Pathology, University of Turku, Turku, Finland
- * E-mail:
| |
Collapse
|
42
|
Turunen HT, Sipilä P, Krutskikh A, Toivanen J, Mankonen H, Hämäläinen V, Björkgren I, Huhtaniemi I, Poutanen M. Loss of cysteine-rich secretory protein 4 (Crisp4) leads to deficiency in sperm-zona pellucida interaction in mice. Biol Reprod 2012; 86:1-8. [PMID: 21865554 DOI: 10.1095/biolreprod.111.092403] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Mammalian sperm gain their ability to fertilize the egg during transit through the epididymis and by interacting with proteins secreted by the epididymal epithelial cells. Certain members of the CRISP (cysteine-rich secretory protein) family form the major protein constituent of the luminal fluid in the mammalian epididymis. CRISP4 is the newest member of the CRISP family expressed predominantly in the epididymis. Its structure and expression pattern suggest a role in sperm maturation and/or sperm-egg interaction. To study the relevance of CRISP4 in reproduction, we have generated a Crisp4 iCre knock-in mouse model through insertion of the iCre recombinase coding cDNA into the Crisp4 locus. This allows using the mouse line both as a Crisp4 deficient model and as an epididymis-specific iCre-expressing mouse line applicable for the generation of conditional, epididymis-specific knockout mice. We show that the loss of CRISP4 leads to a deficiency of the spermatozoa to undergo progesterone-induced acrosome reaction and to a decreased fertilizing ability of the sperm in the in vitro fertilization conditions, although the mice remain fully fertile in normal mating. However, removal of the egg zona pellucida returned the fertilization potential of the CRISP4-deficient spermatozoa, and accordingly we detected a reduced number of Crisp4-deficient spermatozoa bound to oocytes as compared with the wild-type spermatozoa. We also demonstrate that iCre recombinase is expressed in a pattern similar to endogenous Crisp4 and is able to initiate the recombination event with its target sequences in vivo.
Collapse
Affiliation(s)
- Heikki T Turunen
- Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
It has generally been assumed that bone mass is controlled by endocrine mechanisms and the local bone environment. Recent findings demonstrate that central pathways are involved in the regulation of bone mass. Estrogen is involved in the regulation of bone homeostasis and the CNS is also a target for estrogen actions. The aim of this study was to investigate in vivo the role of central estrogen receptor-α (ERα) expression for bone mass. Nestin-Cre mice were crossed with ERα(flox) mice to generate mice lacking ERα expression specifically in nervous tissue (nestin-ERα(-/-)). Bone mineral density was increased in both the trabecular and cortical bone compartments in nestin-ERα(-/-) mice compared with controls. Femoral bone strength was increased in nestin-ERα(-/-) mice, as demonstrated by increased stiffness and maximal load of failure. The high bone mass phenotype in nestin-ERα(-/-) mice was mainly caused by increased bone formation. Serum leptin levels were elevated as a result of increased leptin expression in white adipose tissue (WAT) and slightly increased amount of WAT in nestin-ERα(-/-) mice. Leptin receptor mRNA levels were reduced in the hypothalamus but not in bone. In conclusion, inactivation of central ERα signaling results in increased bone mass, demonstrating that the balance between peripheral stimulatory and central inhibitory ERα actions is important for the regulation of bone mass. We propose that the increased bone mass in nestin-ERα(-/-) mice is mediated via decreased central leptin sensitivity and thereby increased secretion of leptin from WAT, which, in turn, results in increased peripheral leptin-induced bone formation.
Collapse
|
44
|
van de Lagemaat R, van Koppen CJ, Krajnc-Franken MAM, Folmer BJB, van Diepen HA, Mulders SM, Timmers CM. Contraception by induction of luteinized unruptured follicles with short-acting low molecular weight FSH receptor agonists in female animal models. Reproduction 2011; 142:893-905. [DOI: 10.1530/rep-11-0234] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
During recent decades minor innovative drugs have been developed for the female contraceptive market and they all contain steroidal progestagens (and estrogens) that act centrally and have side effects that can be attributed to this central action. In this study, we present an innovative tissue-specific approach for female contraception by low molecular weight (LMW) FSH receptor (FSHR) agonists, which interact with the FSHR that is dominantly expressed in the granulosa cells. The oral administration of LMW FSHR agonists with a short circulation time, induced formation of luteinized unruptured follicles (LUFs) from the Graafian follicles, thereby preventing the release of the oocyte. The short-acting LMW FSHR compounds were fully agonistic to FSHR (EC50=4–5 nM). In an isolated mouse follicle culture, a short incubation period (2 h) resulted in inhibition of follicular rupture, where continuous incubation induced follicle growth. Pharmacokinetics after oral administration showed a surge-like exposure in rats and monkeys. Oral administration of short-acting LMW FSHR agonists inhibited ovulation at 10 mg/kg in rats and guinea pigs by generating LUFs without affecting cyclicity. Also, inhibition of follicular rupture was shown to be reversible within one cycle. Finally, LUFs were induced without affecting the hormonal cyclicity in cynomolgus monkeys, a mono-ovulatory species. In healthy women LUF formation occurs naturally, with a LUF acting as corpus luteum that produces enough progesterone to ensure normal menstrual cyclicity. Together with the presented data this indicates that the innovative approach with short-acting LMW FSHR agonists could lead to oral contraception for females at the ovarian level.
Collapse
|
45
|
Gerrits H, Bakker NE, van de Ven-de Laat CJ, Bourgondien FG, Peddemors C, Litjens RH, Kok HJ, Vogel GM, Krajnc-Franken MA, Gossen JA. Gender-specific increase of bone mass by CART peptide treatment is ovary-dependent. J Bone Miner Res 2011; 26:2886-98. [PMID: 21887702 DOI: 10.1002/jbmr.500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) has emerged as a neurotransmitter and hormone that has been implicated in many processes including food intake, maintenance of body weight, and reward, but also in the regulation of bone mass. CART-deficient mice are characterized by an osteoporotic phenotype, whereas female transgenic mice overexpressing CART display an increase in bone mass. Here we describe experiments that show that peripheral subcutaneous sustained release of different CART peptide isoforms for a period up to 60 days increased bone mass by 80% in intact mice. CART peptides increased trabecular bone mass, but not cortical bone mass, and the increase was caused by reduced osteoclast activity in combination with normal osteoblast activity. The observed effect on bone was gender-specific, because male mice did not respond to treatment with CART peptides. In addition, male transgenic CART overexpressing mice did not display increased bone mass. Ovariectomy (OVX) completely abolished the increase of bone mass by CART peptides, both in CART peptide-treated wild-type mice and in CART transgenic mice. The effect of CART peptide treatment on trabecular bone was not mediated by 17β-estradiol (E(2)) because supplementation of OVX mice with E(2) could not rescue the effect of CART peptides on bone. Together, these results indicate that sustained release of CART peptides increases bone mass in a gender-specific way via a yet unknown mechanism that requires the presence of the ovary.
Collapse
Affiliation(s)
- Han Gerrits
- Merck Sharp & Dohme Research Laboratories, Women's Health Department, Oss, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
van Diepen HA, Pansier J, Oude Wesselink P, van Drie A, van Duin M, Mulders S. Non-invasive translational Cynomolgus model for studying folliculogenesis and ovulation using color Doppler ultrasonography. J Med Primatol 2011; 41:18-23. [PMID: 22084982 DOI: 10.1111/j.1600-0684.2011.00514.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND In women, different events of folliculogenesis can be measured and evaluated using ultrasound (US) technology. The availability of a non-invasive translational non-human primate model to study these processes would represent a major contribution to further advance R&D efforts toward novel therapies in assisted reproduction. METHODS In our study, follicular growth and ovulation was measured in six cyclic Cynomolgus monkeys using abdominal Doppler US. RESULTS The mean follicular diameter on cycle day -6 (cycle day 0=day of ovulation) was 3.7mm that increased to 6.8mm on cycle day -1. After ovulation, the mean diameter decreased to 4.6mm, confirming ovulation. The mean percentage of follicular size reduction after ovulation was 31%. CONCLUSION Ultrasonography in combination with color-flow Doppler imaging was shown to be a useful, non-invasive translational method to measure ovarian follicular growth and occurrence and timing of follicular rupture in Cynomolgus monkeys.
Collapse
Affiliation(s)
- Harry A van Diepen
- Women's Health Department, Merck Research Laboratories, Oss, The Netherlands.
| | | | | | | | | | | |
Collapse
|
47
|
Rahman NA, Bennink HJTC, Chrusciel M, Sharp V, Zimmerman Y, Dina R, Li X, Ellonen A, Rivero-Müller A, Dilworth S, Ghaem-Maghami S, Vainio O, Huhtaniemi I. A novel treatment strategy for ovarian cancer based on immunization against zona pellucida protein (ZP) 3. FASEB J 2011; 26:324-33. [PMID: 21974931 DOI: 10.1096/fj.11-192468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We tested the principle of treating malignant ovarian tumors by vaccination against their ectopically expressed protein, zona pellucida glycoprotein (ZP) 3, using as the experimental model the granulosa cell tumors that develop in transgenic mice expressing the simian virus 40 T-antigen under the inhibin-α promoter (inhα/Tag). We found high ZP3 expression in granulosa cell tumors of the transgenic mice, in human surface ovarian cancer and granulosa cell lines, and in human granulosa cell tumors and their metastases. Early preventive immunization (between 2 and 5.5 mo of age) of transgenic mice with recombinant human (rh) ZP3 prevented ovarian tumorigenesis, and delayed therapeutic immunization (between 4.5 and 7 mo) reduced weights of existing tumors by 86 and 75%, respectively (P<0.001), compared to vehicle-treated control mice. No objective side effects of the immunizations were observed. Liver metastases were found in nontreated/vehicle-treated controls (n=7/39), but none following active rhZP3 immunizations (n=0/36; P<0.05). Immunization with rhZP3 was highly effective, as demonstrated by the induction of anti-ZP3 antibodies, as well as proliferative responses to the ZP3 antigen. These results signal rhZP3 immunization as a novel strategy to be developed for the immunotherapy of ovarian granulosa cell tumors, as well as for that of other malignancies that may express ZP3.
Collapse
Affiliation(s)
- Nafis A Rahman
- Department of Physiology, University of Turku, Turku, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Willems A, De Gendt K, Deboel L, Swinnen JV, Verhoeven G. The development of an inducible androgen receptor knockout model in mouse to study the postmeiotic effects of androgens on germ cell development. SPERMATOGENESIS 2011; 1:341-353. [PMID: 22332118 DOI: 10.4161/spmg.1.4.18740] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/09/2011] [Accepted: 11/10/2011] [Indexed: 12/22/2022]
Abstract
A mouse model with a Sertoli cell (SC)-selective ablation of the androgen receptor (AR)-the SCARKO mouse-demonstrated that the effects of androgens on spermatogenesis depend on the presence of an active AR in SC. This model has been extremely valuable in the study of the effects of androgens on the initiation of spermatogenesis. However, due to the early (prenatal) inactivation of the AR SCARKO mice develop a complete block in meiosis, making it impossible to study the effects of androgens on postmeiotic steps of germ cell development. It would therefore be of interest to develop a model in which the AR can be ablated at any chosen time point. Here we used a mouse line ubiquitously expressing a tamoxifen (TM)-inducible Cre recombinase to develop an inducible AR knockout model (iARKO). It is shown that treatment with TM (3 mg/day) for five consecutive days efficiently inactivates the AR in testicular tissue and decreases the expression of known AR-target genes in SC (Rhox5, Spinlw1) without markedly affecting testicular cell composition one day after the final injection. TM treatment did, however, decrease serum gonadotropin levels and the expression of several Leydig cell genes (StAR, Cyp17a1, Insl3), resulting in decreased testosterone levels. Nevertheless, the intratesticular testosterone concentration still far exceeds the estimated concentrations required to saturate the AR. It may be concluded that the study of androgen-responsive postmeiotic genes in SC may be feasible using a TM-inducible AR knockout model provided that appropriate controls are included correcting for off-target effects of TM.
Collapse
Affiliation(s)
- Ariane Willems
- Laboratory for Experimental Medicine and Endocrinology; Catholic University Leuven; Leuven, Belgium
| | | | | | | | | |
Collapse
|
49
|
Attila M, Oksala R, Agmo A. Sexual incentive motivation in male rats requires both androgens and estrogens. Horm Behav 2010; 58:341-51. [PMID: 19769979 DOI: 10.1016/j.yhbeh.2009.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 08/27/2009] [Accepted: 08/28/2009] [Indexed: 10/20/2022]
Abstract
In Experiment 1 castrated male rats were implanted with a Silastic capsule containing either E or cholesterol (CHOL) 35 days after castration. They were then tested for sexual incentive motivation and copulatory behaviors every 5th day for 3 weeks. None of the treatments affected sexual incentive motivation. After the last test, all subjects were implanted with DHT-containing Silastic capsules, and tests continued for another 3 weeks. While E+DHT enhanced sexual incentive motivation and copulatory behavior, DHT alone failed to do so. In Experiment 2 the aromatase inhibitor fadrozole (F) was combined with testosterone (T). T restored all behaviors to the level seen in intact rats, and F significantly reduced these effects. In fact, T+F was not different from DHT. T and DHT restored the weight of the prostate and seminal vesicles to levels close to those of intact rats. In Experiment 3 a lower dose of E was employed. Also this dose of E failed to affect sexual incentive motivation while E+DHT restored it to the level of intact animals. Castration enhanced the serum concentrations of LH and FSH. E alone caused a marked reduction, and E+DHT brought both gonadotropins back to the level of intact animals. It was concluded that the doses of E and DHT employed in these experiments were within or close to the physiological range, and that such doses of E completely fail to enhance sexual incentive motivation in castrated animals. DHT has small or no effects. It appears that sexual incentive motivation and copulation require simultaneous stimulation of androgen and estrogen receptors.
Collapse
Affiliation(s)
- Martti Attila
- Orion Pharma, Department of Oncology and Critical Care Research, Turku, Finland
| | | | | |
Collapse
|
50
|
Allan CM, Couse JF, Simanainen U, Spaliviero J, Jimenez M, Rodriguez K, Korach KS, Handelsman DJ. Estradiol induction of spermatogenesis is mediated via an estrogen receptor-{alpha} mechanism involving neuroendocrine activation of follicle-stimulating hormone secretion. Endocrinology 2010; 151:2800-10. [PMID: 20410197 PMCID: PMC2875821 DOI: 10.1210/en.2009-1477] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both testosterone and its nonaromatizable metabolite dihydrotestosterone (DHT) induce spermatogenesis in gonadotropin-deficient hpg mice. Surprisingly, because aromatization is not required, estradiol (E2) also induces spermatogenesis and increases circulating FSH in hpg mice, but the mechanism remains unclear. We studied E2-induced spermatogenesis in hpg mice on an estrogen receptor (ER)-alpha (hpg/alphaERKO) or ERbeta (hpg/betaERKO) knockout or wild-type ER (hpg/WT) background treated with subdermal E2 or DHT implants for 6 wk. In hpg/WT and hpg/betaERKO, but not hpg/alphaERKO mice, E2 increased testis and epididymal weight, whereas DHT-induced increases were unaffected by ERalpha or ERbeta inactivation. E2 but not DHT treatment increased serum FSH (but not LH) in hpg/WT and hpg/betaERKO but not hpg/alphaERKO hpg mice. DHT or E2 alone increased (premeiotic) spermatogonia and (meiotic) spermatocytes without significant change in Sertoli cell numbers. DHT alone increased postmeiotic spermatids, regardless of ER presence, compared with variable ERalpha-dependent E2 postmeiotic responses. An ERalpha-mediated effect was confirmed by treating hpg mice for 6 wk by subdermal selective ER-alpha (16alpha-LE(2)) or ERbeta (8beta-VE(2)) agonist implants. ERalpha (but not ERbeta) agonist increased testis and epididymal weight, Sertoli cell, spermatogonia, meiotic, and postmeiotic germ cell numbers. Only ERalpha agonist markedly increased serum FSH, whereas either agonist induced small rises in serum LH. Administration of ERalpha agonist or E2 in the presence of functional ERalpha induced prominent gene expression of specific Sertoli (Eppin, Rhox5) and Leydig cell (Cyp11a1, Hsd3b1) markers. We conclude that E2-induced spermatogenesis in hpg mice involves an ERalpha-dependent neuroendocrine mechanism increasing blood FSH and Sertoli cell function.
Collapse
Affiliation(s)
- Charles M Allan
- ANZAC Research Institute, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | | | |
Collapse
|