1
|
Zhang H, Liu TT, Ricke EA, Ricke WA. Prostatic androgen receptor signaling shows an age-related and lobe-specific alteration in mice. Sci Rep 2024; 14:30302. [PMID: 39638850 PMCID: PMC11621416 DOI: 10.1038/s41598-024-79879-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is an age-related disease that affects millions of aging males globally. While the pathogenesis of BPH remains incompletely understood, emerging evidence suggests a pivotal role for the androgen receptor (AR) in mediating prostate growth and function. Understanding age-related AR signaling alteration may inform novel BPH treatments. Here, we analyzed the prostatic protein expressions of AR, NKX3.1, and Ki-67 in young (2 months) and aged (24 months) mice. We also examined the potential mechanism of AR protein expression. Compared to young mice, decreased AR and NKX3.1 protein expression was observed in the anterior prostate (AP) and ventral prostate (VP) of aged mice, indicating reduced AR signaling in these prostate lobes. Additionally, we observed decreased protein expression of proliferation maker Ki-67 in aged AP, VP, and dorsal-lateral prostate (DLP), with no difference in apoptosis as compared to young counterparts. We conclude that prostatic androgen receptor signaling shows an age-related and lobe-specific alteration in mice.
Collapse
Affiliation(s)
- Han Zhang
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Teresa T Liu
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Emily A Ricke
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - William A Ricke
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- George M. O'Brien Urology Research Center of Excellence, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
2
|
Proffitt MR, Smith GT. Species variation in steroid hormone-related gene expression contributes to species diversity in sexually dimorphic communication in electric fishes. Horm Behav 2024; 164:105576. [PMID: 38852479 PMCID: PMC11330740 DOI: 10.1016/j.yhbeh.2024.105576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Sexually dimorphic behaviors are often regulated by gonadal steroid hormones. Species diversity in behavioral sex differences may arise as expression of genes mediating steroid action in brain regions controlling these behaviors evolves. The electric communication signals of apteronotid knifefishes are an excellent model for comparatively studying neuroendocrine regulation of sexually dimorphic behavior. These fish produce and detect weak electric organ discharges (EODs) for electrolocation and communication. EOD frequency (EODf), controlled by the medullary pacemaker nucleus (Pn), is sexually dimorphic and regulated by androgens and estrogens in some species, but is sexually monomorphic and unaffected by hormones in other species. We quantified expression of genes for steroid receptors, metabolizing enzymes, and cofactors in the Pn of two species with sexually dimorphic EODf (Apteronotus albifrons and Apteronotus leptorhynchus) and two species with sexually monomorphic EODf ("Apteronotus" bonapartii and Parapteronotus hasemani). The "A." bonapartii Pn expressed lower levels of androgen receptor (AR) genes than the Pn of species with sexually dimorphic EODf. In contrast, the P. hasemani Pn robustly expressed AR genes, but expressed lower levels of genes for 5α-reductases, which convert androgens to more potent metabolites, and higher levels of genes for 17β-hydroxysteroid dehydrogenases that oxidize androgens and estrogens to less potent forms. These findings suggest that sexual monomorphism of EODf arose convergently via two different mechanisms. In "A." bonapartii, reduced Pn expression of ARs likely results in insensitivity of EODf to androgens, whereas in P. hasemani, gonadal steroids may be metabolically inactivated in the Pn, reducing their potential to influence EODf.
Collapse
Affiliation(s)
- Melissa R Proffitt
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA
| | - G Troy Smith
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA.
| |
Collapse
|
3
|
Kumar R, Fatima F, Yadav G, Singh S, Haldar S, Alexiou A, Ashraf GM. Epigenetic Modifications by Estrogen and Androgen in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:6-17. [PMID: 35232367 DOI: 10.2174/1871527321666220225110501] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
For the development and maintenance of neuron networks in the brain, epigenetic mechanisms are necessary, as indicated by recent findings. This includes some of the high-order brain processes, such as behavior and cognitive functions. Epigenetic mechanisms could influence the pathophysiology or etiology of some neuronal diseases, altering disease susceptibility and therapy responses. Recent studies support epigenetic dysfunctions in neurodegenerative and psychiatric conditions, such as Alzheimer's disease (AD). These dysfunctions in epigenetic mechanisms also play crucial roles in the transgenerational effects of the environment on the brain and subsequently in the inheritance of pathologies. The possible role of gonadal steroids in the etiology and progression of neurodegenerative diseases, including Alzheimer's disease, has become the subject of a growing body of research over the last 20 years. Recent scientific findings suggest that epigenetic changes, driven by estrogen and androgens, play a vital role in brain functioning. Therefore, exploring the role of estrogen and androgen-based epigenetic changes in the brain is critical for the deeper understanding of AD. This review highlights the epigenetic modifications caused by these two gonadal steroids and the possible therapeutic strategies for AD.
Collapse
Affiliation(s)
- Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Faiza Fatima
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Garima Yadav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Simran Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Subhagata Haldar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, Hebersham, 2770 NSW, Australia, and AFNP Med Austria, 1010 Wien, Austria
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Ono K, Yano A, Ishimura K. Effect of peripherally derived steroid hormones on the expression of steroidogenic enzymes in the rat choroid plexus. THE JOURNAL OF MEDICAL INVESTIGATION 2021; 68:238-243. [PMID: 34759137 DOI: 10.2152/jmi.68.238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Peripherally derived steroids affect steroid production in the brain via the blood-brain barrier. However, steroid concentrations are lower in the cerebrospinal fluid than those in the blood, indicating restricted influx of steroids because of their metabolization by choroid plexus (CP) epithelial cells. Here, we analyzed the gene expression of steroidogenic enzymes [cholesterol side-chain cleavage enzyme (P450scc), 17α-hydroxylase/C17-C20 lyase (P450c17), 3β-hydroxysteroid dehydrogenase (3β-HSD), 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1), aromatase (Cyp19a1), and 5α-reductase type 1 (5α-R1)]. These genes were expressed to a lesser extent in the CP than in the testis and to a similar extent in the cerebral cortex. However, P450scc levels were higher in the CP than in the cerebral cortex, whereas Cyp19a1 levels showed the opposite trend. We also evaluated the effects of orchiectomy and testosterone on the expression of these genes. P450c17 and 5α-R1 levels were unaffected by orchiectomy, whereas P450scc and 3β-HSD levels were increased and decreased, respectively. Cyp19a1 expression increased upon testosterone treatment, whereas that of 17β-HSD decreased upon orchiectomy or administration of testosterone. Immunohistochemistry analysis revealed that 17β-HSD was expressed in the cytoplasm of CP epithelial cells. These results indicate that CP epithelial cells synthesize and convert the certain types of steroids to contribute to the homeostasis of steroids in the brain. J. Med. Invest. 68 : 238-243, August, 2021.
Collapse
Affiliation(s)
- Koji Ono
- Department of Anatomy and Cell Biology, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan.,Department of Anatomy, Kawasaki Medical School, Okayama, Japan
| | - Akihiko Yano
- Department of Anatomy and Cell Biology, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan.,Student Lab, Tokushima University Faculty of Medicine, Tokushima Japan.,Clinical Training Center, University of Yamanashi Hospital, Yamanashi, Japan
| | - Kazunori Ishimura
- Department of Anatomy and Cell Biology, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan.,Department of Nutrition and Health Promotion, Hiroshima Jogakuin University Faculty of Human Life Studies, Hiroshima, Japan
| |
Collapse
|
5
|
Otsuka K, Matsubara S, Shiraishi A, Takei N, Satoh Y, Terao M, Takada S, Kotani T, Satake H, Kimura AP. A Testis-Specific Long Noncoding RNA, Start, Is a Regulator of Steroidogenesis in Mouse Leydig Cells. Front Endocrinol (Lausanne) 2021; 12:665874. [PMID: 33897623 PMCID: PMC8061315 DOI: 10.3389/fendo.2021.665874] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
The testis expresses many long noncoding RNAs (lncRNAs), but their functions and overview of lncRNA variety are not well understood. The mouse Prss/Tessp locus contains six serine protease genes and two lncRNAs that have been suggested to play important roles in spermatogenesis. Here, we found a novel testis-specific lncRNA, Start (Steroidogenesis activating lncRNA in testis), in this locus. Start is 1822 nucleotides in length and was found to be localized mostly in the cytosol of germ cells and Leydig cells, although nuclear localization was also observed. Start-knockout (KO) mice generated by the CRISPR/Cas9 system were fertile and showed no morphological abnormality in adults. However, in adult Start-KO testes, RNA-seq and qRT-PCR analyses revealed an increase in the expression of steroidogenic genes such as Star and Hsd3b1, while ELISA analysis revealed that the testosterone levels in serum and testis were significantly low. Interestingly, at 8 days postpartum, both steroidogenic gene expression and testosterone level were decreased in Start-KO mice. Since overexpression of Start in two Leydig-derived cell lines resulted in elevation of the expression of steroidogenic genes including Star and Hsd3b1, Start is likely to be involved in their upregulation. The increase in expression of steroidogenic genes in adult Start-KO testes might be caused by a secondary effect via the androgen receptor autocrine pathway or the hypothalamus-pituitary-gonadal axis. Additionally, we observed a reduced number of Leydig cells at 8 days postpartum. Collectively, our results strongly suggest that Start is a regulator of steroidogenesis in Leydig cells. The current study provides an insight into the overall picture of the function of testis lncRNAs.
Collapse
Affiliation(s)
- Kai Otsuka
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Akira Shiraishi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Natsumi Takei
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Yui Satoh
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Miho Terao
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of NCCHD Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoya Kotani
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Atsushi P. Kimura
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
- *Correspondence: Atsushi P. Kimura,
| |
Collapse
|
6
|
Low KL, Tomm RJ, Ma C, Tobiansky DJ, Floresco SB, Soma KK. Effects of aging on testosterone and androgen receptors in the mesocorticolimbic system of male rats. Horm Behav 2020; 120:104689. [PMID: 31954104 DOI: 10.1016/j.yhbeh.2020.104689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/23/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
Abstract
As males age, systemic testosterone (T) levels decline. T regulates executive function, a collection of cognitive processes that are mediated by the mesocorticolimbic system. Here, we examined young adult (5 months) and aged (22 months) male Fischer 344 × Brown Norway rats, and measured systemic T levels in serum and local T levels in microdissected nodes of the mesocorticolimbic system (ventral tegmental area (VTA), nucleus accumbens (NAc), medial prefrontal cortex (mPFC), and orbitofrontal cortex (OFC)). We also measured androgen receptor (AR) immunoreactivity (-ir) in the mesocorticolimbic system. As expected, systemic T levels decreased with age. Local T levels in mesocorticolimbic regions - except the VTA - also decreased with age. Mesocorticolimbic T levels were higher than serum T levels at both ages. AR-ir was present in the VTA, NAc, mPFC, and OFC and decreased with age in the mPFC. Taken together with previous results, the data suggest that changes in androgen signaling may contribute to changes in executive function during aging.
Collapse
Affiliation(s)
- Katelyn L Low
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Ryan J Tomm
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Chunqi Ma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Tobiansky
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Zoology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
7
|
Holland S, Prescott M, Pankhurst M, Campbell RE. The influence of maternal androgen excess on the male reproductive axis. Sci Rep 2019; 9:18908. [PMID: 31827225 PMCID: PMC6906411 DOI: 10.1038/s41598-019-55436-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
Prenatal androgen excess is suspected to contribute to the development of polycystic ovary syndrome (PCOS) in women. Evidence from preclinical female animal models links maternal androgen excess with the development of PCOS-like features and associated alterations in the neuronal network regulating the reproductive axis. There is some evidence suggesting that maternal androgen excess leads to similar reproductive axis disruptions in men, despite the critical role that androgens play in normal sexual differentiation. Here, the specific impact of maternal androgen excess on the male hypothalamic-pituitary-gonadal axis was investigated using a prenatal androgenization protocol in mice shown to model PCOS-like features in females. Reproductive phenotyping of prenatally androgenised male (PNAM) mice revealed no discernible impact of maternal androgen excess at any level of the reproductive axis. Luteinising hormone pulse characteristics, daily sperm production, plasma testosterone and anti-Müllerian hormone levels were not different in the male offspring of dams administered dihydrotestosterone (DHT) during late gestation compared to controls. Androgen receptor expression was quantified through the hypothalamus and identified as unchanged. Confocal imaging of gonadotropin-releasing hormone (GnRH) neurons revealed that in contrast with prenatally androgenised female mice, PNAM mice exhibited no differences in the density of putative GABAergic innervation compared to controls. These data indicate that a maternal androgen environment capable of inducing reproductive dysfunction in female offspring has no evident impact on the reproductive axis of male littermates in adulthood.
Collapse
Affiliation(s)
- Sarah Holland
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand
| | - Michael Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
8
|
Cobb AR, Josephs RA, Lancaster CL, Lee HJ, Telch MJ. Cortisol, Testosterone, and Prospective Risk for War-zone Stress-Evoked Depression. Mil Med 2019; 183:e535-e545. [PMID: 29718455 DOI: 10.1093/milmed/usy065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Indexed: 01/08/2023] Open
Abstract
Introduction The major challenges of efforts to reveal biological risk factors and biomarkers of depression include the complexity of underlying systems, interactions with other systems, and contextual factors governing their expression. Altered endocrine function is believed to be a central contributor to depressive illness, but across studies, evidence for a link between endocrine markers and depression has been mixed, inconclusive, or conditional in nature. In the present study, we evaluated basal testosterone (T), cortisol (C), and CO2 inhalation-stress-reactivity measures of these hormones (TR, CR) as pre-deployment moderators of the later impact of war-zone stressors on depression symptoms in-theater. Materials and Methods At pre-deployment, U.S. soldiers (N = 120) completed demographic, clinical and hormone measures, and during deployment, they completed monthly, web-based assessments of war-zone stressors and depression symptoms (N = 533 observations). Mixed effects models estimated the effects of the pre-deployment hormone profiles in moderating war-zone stressors' impact on in-theater depression. Models also tested whether hormonally linked risk for later stress-evoked depression depends on pre-existing depression. Results Controlling for pre-deployment depression, high T was protective; whereas TR had depressogenic effects that were amplified by pre-deployment depression. Further, high C was protective, but heightened CR was depressogenic, but only among those with elevated pre-deployment depression. Conclusions Findings highlight the importance of examining basal and reactivity measures of endocrine function, and use of prospective, longitudinal models to test hypothesized causal pathways associated with depression vulnerability in the war-zone. Results also suggest that pre-existing depression and cortisol may work in tandem to increase vulnerability for later stress-evoked depression in the war-zone.
Collapse
Affiliation(s)
- Adam R Cobb
- Department of Psychology, The University of Texas at Austin, 1 University Station, A8000, Austin, TX
| | - Robert A Josephs
- Department of Psychology, The University of Texas at Austin, 1 University Station, A8000, Austin, TX
| | - Cynthia L Lancaster
- Department of Psychology, The University of Texas at Austin, 1 University Station, A8000, Austin, TX
| | - Han-Joo Lee
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI
| | - Michael J Telch
- Department of Psychology, The University of Texas at Austin, 1 University Station, A8000, Austin, TX
| |
Collapse
|
9
|
Wu Y, Peterson KE, Sánchez BN, Dolinoy DC, Mercado-Garcia A, Téllez-Rojo MM, Goodrich JM. Association of blood leukocyte DNA methylation at LINE-1 and growth-related candidate genes with pubertal onset and progression. Epigenetics 2018; 13:1222-1233. [PMID: 30582410 PMCID: PMC6986794 DOI: 10.1080/15592294.2018.1556198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/01/2018] [Accepted: 11/21/2018] [Indexed: 01/15/2023] Open
Abstract
Puberty is a developmentally plastic phase. Variations in pubertal tempo have implications for the risk of later adult diseases. Influences on pubertal tempo have been widely discussed, but the underlying biological mechanisms remain unclear. Epigenetic modifications are known to regulate development processes; they could play an important role in affecting pubertal outcomes. We conducted a population-based analysis to investigate the association of peripubertal blood DNA methylation at LINE-1 and growth-related candidate genes with pubertal onset and progression in healthy adolescents. The analytic sample included 114 males and 129 females aged 10 to 18 years. DNA methylation at growth-related candidate loci IGF2, H19, HSD11B2, as well as LINE-1 repetitive elements were quantified. Cox survival and ordinal regression models were used to examine sex- and locus-specific associations of epigenetic markers with pubertal development using physician-assessed Tanner stages and self-reported menarche, adjusted for covariates. Among boys, DNA methylation at H19 was associated with later pubarche. HSD11B2 methylation was associated with earlier onset of pubic hair and genitalia development and slower pubertal progression. IGF2 was associated with later onset of genital development. Among girls, LINE-1 methylation was associated with later onset of breast development. For each percent increase of methylation at H19, there was 5% increased odds in the earlier onset of breast development. DNA methylation of IGF2 was associated with earlier onset of pubic hair. DNA methylation at genes known to influence early-life growth may also influence pubertal outcomes.
Collapse
Affiliation(s)
- Yue Wu
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karen E. Peterson
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
| | - Brisa N. Sánchez
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C. Dolinoy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Adriana Mercado-Garcia
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Martha M. Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Understanding how Age-Related Decline in Testosterone Affects Male Sexual Behavior: Neurosteroids as the Missing Piece. CURRENT SEXUAL HEALTH REPORTS 2018. [DOI: 10.1007/s11930-018-0175-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
11
|
Giatti S, Garcia-Segura LM, Barreto GE, Melcangi RC. Neuroactive steroids, neurosteroidogenesis and sex. Prog Neurobiol 2018; 176:1-17. [PMID: 29981391 DOI: 10.1016/j.pneurobio.2018.06.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/25/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022]
Abstract
The nervous system is a target and a source of steroids. Neuroactive steroids are steroids that target neurons and glial cells. They include hormonal steroids originated in the peripheral glands, steroids locally synthesized by the neurons and glial cells (neurosteroids) and synthetic steroids, some of them used in clinical practice. Here we review the mechanisms of synthesis, metabolism and action of neuroactive steroids, including the role of epigenetic modifications and the mitochondria in their sex specific actions. We examine sex differences in neuroactive steroid levels under physiological conditions and their role in the establishment of sex dimorphic structures in the nervous system and sex differences in its function. In addition, particular attention is paid to neuroactive steroids under pathological conditions, analyzing how pathology alters their levels and their role as neuroprotective factors, considering the influence of sex in both cases.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
12
|
Hunter I, Hay CW, Esswein B, Watt K, McEwan IJ. Tissue control of androgen action: The ups and downs of androgen receptor expression. Mol Cell Endocrinol 2018; 465:27-35. [PMID: 28789969 DOI: 10.1016/j.mce.2017.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022]
Abstract
The hormone testosterone plays crucial roles during male development and puberty and throughout life, as an anabolic regulator of muscle and bone structure and function. The actions of testosterone are mediated, primarily, through the androgen receptor, a member of the nuclear receptor superfamily. The androgen receptor gene is located on the X-chromosome and receptor levels are tightly controlled both at the level of transcription of the gene and post-translationally at the protein level. Sp1 has emerged as the major driver of expression of the androgen receptor gene, while auto-regulation by androgens is associated with both positive and negative regulation in a possible cell-selective manner. Research into the networks of positive and negative regulators of the androgen receptor gene are vital in order to understand the temporal and spatial control of receptor levels and the consequences for healthy aging and disease. A clear understanding of the multiple transcription factors participating in regulation of the androgen receptor gene will likely aid in the development and application of hormone therapies to boast or curb receptor activity.
Collapse
Affiliation(s)
- Irene Hunter
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Colin W Hay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Bianca Esswein
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK; Friedrich-Schiller-Universitat, Jena, Germany
| | - Kate Watt
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Iain J McEwan
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
13
|
Walters KA, Edwards MC, Tesic D, Caldwell ASL, Jimenez M, Smith JT, Handelsman DJ. The Role of Central Androgen Receptor Actions in Regulating the Hypothalamic-Pituitary-Ovarian Axis. Neuroendocrinology 2018; 106:389-400. [PMID: 29635226 DOI: 10.1159/000487762] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/16/2018] [Indexed: 12/20/2022]
Abstract
The androgen receptor (AR) is expressed throughout the hypothalamic-pituitary-gonadal (HPG) axis, and findings from female global AR knockout mice confirm that AR-mediated androgen actions play important roles in regulating female reproductive function. We generated neuron-specific AR knockout mice (NeurARKO) to investigate the functional role of neuronal AR-mediated androgen action in regulating the female HPG axis and fertility. Relative to control females, NeurARKO females exhibited elevated luteinizing hormone (LH) levels at diestrus (p < 0.05) and a compromised serum LH response to ovariectomy and E2 priming (p < 0.01). Furthermore, NeurARKO females displayed reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus at diestrus (p < 0.05) and proestrus (p < 0.05), but elevated Kiss1 (p < 0.05) and neurokinin B (Tac2, p < 0.05) mRNA expression in the arcuate nucleus at proestrus compared to WT controls. Ovarian follicle dynamics were also altered in NeurARKO ovaries at 3 months of age, with a significant reduction in large antral follicle numbers at the proestrus stage compared to control WT ovaries (p < 0.05). Increased follicular atresia was evident in NeurARKO ovaries with a 4-fold increase in unhealthy large preantral follicles (p < 0.01). Despite the findings of aberrant neuroendocrine and ovarian characteristics in the NeurARKO females, estrous cyclicity and overall fertility were comparable between NeurARKO and WT females. In conclusion, our findings revealed that selective loss of neuronal AR actions impacts the kisspeptin/GnRH/LH cascade leading to compromised ovarian follicle dynamics.
Collapse
Affiliation(s)
- Kirsty A Walters
- School of Women's & Children's Health, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Melissa C Edwards
- School of Women's & Children's Health, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Dijana Tesic
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Washington, Australia
| | - Aimee S L Caldwell
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Mark Jimenez
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Jeremy T Smith
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Washington, Australia
| | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Ning P, Zhong JG, Jiang F, Zhang Y, Zhao J, Tian F, Li W. Role of protein S in castration-resistant prostate cancer-like cells. Endocr Relat Cancer 2016; 23:595-607. [PMID: 27342144 DOI: 10.1530/erc-16-0126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 01/13/2023]
Abstract
Understanding how castration-resistant prostate cancer (CRPC) cells survive the androgen-deprivation condition is crucial for treatment of this advanced prostate cancer (PCa). Here, we reported for the first time the up-regulation of protein S (PROS), an anticoagulant plasma glycoprotein with multiple biological functions, in androgen-insensitive PCa cells and in experimentally induced castration-resistant PCa cells. Overexpression of exogenous PROS in LNCaP cells reduced androgen deprivation-induced apoptosis and enhanced anchorage-dependent clonogenic ability under androgen deprivation condition. Reciprocally, PROS1 knockdown inhibited cell invasiveness and migration, caused the growth inhibition of castration-resistant tumor xenograft under androgen-depleted conditions, and potentiated Taxol (a widely prescribed anti-neoplastic agent)-mediated cell death in PC3 cells. Furthermore, PROS overexpression significantly stimulated AKT activation but failed to evoke oxidative stress in LNCaP cells under normal condition, suggesting that the malignance-promoting effects of the above-mentioned pathway may occur in the order of oxidative stress/PROS/AKT. The potential mechanism may be due to control of oxidative stress-elicited activation of PI3K-AKT-mTOR pathway. Taken together, our gain-of-function, loss-of-function analyses suggest that PROS may facilitate cell proliferation and promote castration resistance in human castration-resistant PCa-like cells via its apoptosis-regulating property. Future study emphasizing on delineating how PROS regulate cellular processes controlling transformation during the development of castration resistance should open new doors for the development of novel therapeutic targets for CRPC.
Collapse
Affiliation(s)
- Peng Ning
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Jia-Guo Zhong
- Section 2 of Department of Surgery42nd Hospital of PLA, Jiajiang County Leshan City, Sichuan, China
| | - Fan Jiang
- Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Yi Zhang
- Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Jie Zhao
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China
| | - Feng Tian
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Li
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Göppert C, Harris RM, Theis A, Boila A, Hohl S, Rüegg A, Hofmann HA, Salzburger W, Böhne A. Inhibition of Aromatase Induces Partial Sex Change in a Cichlid Fish: Distinct Functions for Sex Steroids in Brains and Gonads. Sex Dev 2016; 10:97-110. [DOI: 10.1159/000445463] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2016] [Indexed: 11/19/2022] Open
|
16
|
Pradhan A, Olsson PE. Zebrafish sexual behavior: role of sex steroid hormones and prostaglandins. Behav Brain Funct 2015; 11:23. [PMID: 26385780 PMCID: PMC4575480 DOI: 10.1186/s12993-015-0068-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 06/25/2015] [Indexed: 12/11/2022] Open
Abstract
Background Mating behavior differ between sexes and involves gonadal hormones and possibly sexually dimorphic gene expression in the brain. Sex steroids and prostaglandin E2 (PGE2) have been shown to regulate mammalian sexual behavior. The present study was aimed at determining whether exposure to sex steroids and prostaglandins could alter zebrafish sexual mating behavior. Methods Mating behavior and successful spawning was recorded following exposure to 17β-estradiol (E2), 11-ketotestosterone (11-KT), prostaglandin D2 (PGD2) and PGE2 via the water. qRT-PCR was used to analyze transcript levels in the forebrain, midbrain, and hindbrain of male and female zebrafish and compared to animals exposed to E2 via the water. Results Exposure of zebrafish to sex hormones resulted in alterations in behavior and spawning when male fish were exposed to E2 and female fish were exposed to 11-KT. Exposure to PGD2, and PGE2 did not alter mating behavior or spawning success. Determination of gene expression patterns of selected genes from three brain regions using qRT-PCR analysis demonstrated that the three brain regions differed in gene expression pattern and that there were differences between the sexes. In addition, E2 exposure also resulted in altered gene transcription profiles of several genes. Conclusions Exposure to sex hormones, but not prostaglandins altered mating behavior in zebrafish. The expression patterns of the studied genes indicate that there are large regional and gender-based differences in gene expression and that E2 treatment alter the gene expression pattern in all regions of the brain. Electronic supplementary material The online version of this article (doi:10.1186/s12993-015-0068-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, 701 82, Örebro, Sweden
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, 701 82, Örebro, Sweden.
| |
Collapse
|
17
|
Doi H, Nishitani S, Shinohara K. Sex difference in the relationship between salivary testosterone and inter-temporal choice. Horm Behav 2015; 69:50-8. [PMID: 25530487 DOI: 10.1016/j.yhbeh.2014.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 11/25/2022]
Abstract
Humans often prefer a small immediate reward to large reward in the future. This myopic tendency in inter-temporal choice is termed delay discounting, and has been the focus of intensive research in the past decades. Recent studies indicate that the neural regions underlying delay discounting are influenced by the gonadal steroids. However, the specific relationship between the testosterone levels and delay discounting is unclear at this point, especially in females. The present study investigated the relationship between salivary testosterone concentrations and discounting rates in delay- and probability-discounting tasks with healthy males and females. The results revealed a positive correlation between testosterone concentrations and delay-discounting rates in females and a negative correlation in males. Testosterone concentrations were unrelated to probability-discounting rates. Although causal effects of testosterone cannot be certain in this correlational study, if testosterone directly influenced this behavior, observed sex differences in delay discounting may be evidence of a curvilinear effect of testosterone. Alternatively, the findings may reflect inverse pattern of responsiveness to testosterone between male and female neural systems, or basic sex-difference in the neural mechanism underlying delay-discounting independent of testosterone itself.
Collapse
Affiliation(s)
- Hirokazu Doi
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shota Nishitani
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuyuki Shinohara
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
18
|
Age- and Sex-Dependent Changes in Androgen Receptor Expression in the Developing Mouse Cortex and Hippocampus. NEUROSCIENCE JOURNAL 2015; 2015:525369. [PMID: 26317111 PMCID: PMC4437260 DOI: 10.1155/2015/525369] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 01/02/2015] [Accepted: 01/02/2015] [Indexed: 02/06/2023]
Abstract
During the perinatal period, male mice are exposed to higher levels of testosterone (T) than females, which promotes sexual dimorphism in their brain structures and behaviors. In addition to acting via estrogen receptors after being locally converted into estradiol by aromatase, T also acts directly through androgen receptor (AR) in the brain. Therefore, we hypothesized that AR expression in the developing mouse cortex and hippocampus was sexually dimorphic. To test our hypothesis, we measured and determined AR mRNA and protein levels in mouse cortex/hippocampus collected on the day of birth (PN0) and 7 (PN7), 14 (PN14), and 21 (PN21) days after birth. We demonstrated that, as age advanced, AR mRNA levels increased in the cortex/hippocampus of both sexes but showed no sex difference. Two AR proteins, the full-length (110 kDa) and a smaller isoform (70 kDa), were detected in the developing mouse cortex/hippocampus with an age-dependent increase in protein levels of both AR isoforms at PN21 and a transient masculine increase in expression of the full-length AR protein on PN7. Thus, we conclude that the postnatal age and sex differences in AR protein expression in combination with the sex differences in circulating T may cause sexual differentiation of the mouse cortex/hippocampus.
Collapse
|
19
|
Environmental Health Factors and Sexually Dimorphic Differences in Behavioral Disruptions. Curr Environ Health Rep 2014; 1:287-301. [PMID: 25705580 DOI: 10.1007/s40572-014-0027-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mounting evidence suggests that environmental factors-in particular, those that we are exposed to during perinatal life-can dramatically shape the organism's risk for later diseases, including neurobehavioral disorders. However, depending on the environmental insult, one sex may demonstrate greater vulnerability than the other sex. Herein, we focus on two well-defined extrinsic environmental factors that lead to sexually dimorphic behavioral differences in animal models and linkage in human epidemiological studies. These include maternal or psychosocial stress (such as social stress) and exposure to endocrine-disrupting compounds (such as one of the most prevalent, bisphenol A [BPA]). In general, the evidence suggests that early environmental exposures, such as BPA and stress, lead to more pronounced behavioral deficits in males than in females, whereas female neurobehavioral patterns are more vulnerable to later in life stress. These findings highlight the importance of considering sex differences and developmental timing when examining the effects of environmental factors on later neurobehavioral outcomes.
Collapse
|
20
|
Testosterone down regulates the expression of Fmr-1 gene in the cerebral cortex of gonadectomized old male mice. Biogerontology 2014; 15:503-15. [DOI: 10.1007/s10522-014-9521-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/17/2014] [Indexed: 10/25/2022]
|
21
|
Jašarević E, Geary DC, Rosenfeld CS. Sexually selected traits: a fundamental framework for studies on behavioral epigenetics. ILAR J 2014; 53:253-69. [PMID: 23744965 DOI: 10.1093/ilar.53.3-4.253] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that epigenetic-based mechanisms contribute to various aspects of sex differences in brain and behavior. The major obstacle in establishing and fully understanding this linkage is identifying the traits that are most susceptible to epigenetic modification. We have proposed that sexual selection provides a conceptual framework for identifying such traits. These are traits involved in intrasexual competition for mates and intersexual choice of mating partners and generally entail a combination of male-male competition and female choice. These behaviors are programmed during early embryonic and postnatal development, particularly during the transition from the juvenile to adult periods, by exposure of the brain to steroid hormones, including estradiol and testosterone. We evaluate the evidence that endocrine-disrupting compounds, including bisphenol A, can interfere with the vital epigenetic and gene expression pathways and with the elaboration of sexually selected traits with epigenetic mechanisms presumably governing the expression of these traits. Finally, we review the evidence to suggest that these steroid hormones can induce a variety of epigenetic changes in the brain, including the extent of DNA methylation, histone protein alterations, and even alterations of noncoding RNA, and that many of the changes differ between males and females. Although much previous attention has focused on primary sex differences in reproductive behaviors, such as male mounting and female lordosis, we outline why secondary sex differences related to competition and mate choice might also trace their origins back to steroid-induced epigenetic programming in disparate regions of the brain.
Collapse
Affiliation(s)
- Eldin Jašarević
- Department of Psychological Sciences, the Interdisciplinary Neuroscience Program, and the Bond Life Sciences Center, University of Missouri, Columbia 65211, USA
| | | | | |
Collapse
|
22
|
Sinclair D, Purves-Tyson TD, Allen KM, Weickert CS. Impacts of stress and sex hormones on dopamine neurotransmission in the adolescent brain. Psychopharmacology (Berl) 2014; 231:1581-99. [PMID: 24481565 PMCID: PMC3967083 DOI: 10.1007/s00213-013-3415-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
Abstract
RATIONALE Adolescence is a developmental period of complex neurobiological change and heightened vulnerability to psychiatric illness. As a result, understanding factors such as sex and stress hormones which drive brain changes in adolescence, and how these factors may influence key neurotransmitter systems implicated in psychiatric illness, is paramount. OBJECTIVES In this review, we outline the impact of sex and stress hormones at adolescence on dopamine neurotransmission, a signaling pathway which is critical to healthy brain function and has been implicated in psychiatric illness. We review normative developmental changes in dopamine, sex hormone, and stress hormone signaling during adolescence and throughout postnatal life, then highlight the interaction of sex and stress hormones and review their impacts on dopamine neurotransmission in the adolescent brain. RESULTS AND CONCLUSIONS Adolescence is a time of increased responsiveness to sex and stress hormones, during which the maturing dopaminergic neural circuitry is profoundly influenced by these factors. Testosterone, estrogen, and glucocorticoids interact with each other and have distinct, brain region-specific impacts on dopamine neurotransmission in the adolescent brain, shaping brain maturation and cognitive function in adolescence and adulthood. Some effects of stress/sex hormones on cortical and subcortical dopamine parameters bear similarities with dopaminergic abnormalities seen in schizophrenia, suggesting a possible role for sex/stress hormones at adolescence in influencing risk for psychiatric illness via modulation of dopamine neurotransmission. Stress and sex hormones may prove useful targets in future strategies for modifying risk for psychiatric illness.
Collapse
Affiliation(s)
- Duncan Sinclair
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia ,Neuropsychiatric Signaling Program, Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Katherine M Allen
- Schizophrenia Research Institute, Sydney, Australia ,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, Australia ,Macquarie Group Chair of Schizophrenia Research, Neuroscience Research Australia, Barker Street, Randwick, NSW 2031 Australia ,School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
23
|
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: 3.1] [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
|
24
|
Zhang GL, Wang W, Kang YX, Xue Y, Yang H, Zhou CM, Shi GM. Chronic testosterone propionate supplement could activated the Nrf2-ARE pathway in the brain and ameliorated the behaviors of aged rats. Behav Brain Res 2013; 252:388-95. [PMID: 23756138 DOI: 10.1016/j.bbr.2013.05.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/27/2013] [Accepted: 05/31/2013] [Indexed: 10/26/2022]
Abstract
Aging is usually associated with a progressive disruption of the redox balance leading to recurrent damage resulting from oxidative stress. Oxidative stress resulting from excessive free-radical release is likely implicated in the initiation and progression of motor behavior disorders. Therefore, antioxidant therapies have received considerable attention in motor behavior defects treatment. The nuclear factor erythroid 2-related factor 2 (Nrf2) binds to antioxidant response element (ARE) to induce antioxidant and phase II detoxification enzymes under conditions of oxidative stress, which reduces oxidative stress and accumulation of toxic metabolites. Testosterone has many physiological and behavioral effects throughout the lifespan and shown to affect motor behavior in adult male rats and gonadectomized rats. However, whether Nrf2-ARE pathway is activated after testosterone administration has not been studied in aged rats. The tilting-plane test and the horizontal-wire test as well as the oxidative stress parameters, the expression of Nrf2, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO1) and the number of tyrosine hydroxylase immunoreactive (TH-ir) cells in brain were examined in aged rats following chronic subcutaneous injections of testosterone propionate (TP). Our study showed that chronic TP supplement significantly ameliorated the decline of balancing reactions and muscular strength associated with aging. Oxidative stress parameters were ameliorate, the expression of Nrf2, HO-1 and NQO1 at protein or gene levels and the number of TH-ir cells significantly increased in substantia nigra or caudate putamen after TP treatment in aged rats. Our findings demonstrated that chronic TP treatment activated Nrf2-ARE pathway may influence the maintenance of the balancing reactions and muscular strength and reduce TH-ir cells death in aged rats. Therefore, TP supplement have shown for therapeutic strategies in the treatment and modification of motor behavior disorders.
Collapse
Affiliation(s)
- Guo-liang Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | | | | | | | | | | | | |
Collapse
|
25
|
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: 6.5] [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
|
26
|
Menger Y, Bettscheider M, Murgatroyd C, Spengler D. Sex differences in brain epigenetics. Epigenomics 2012; 2:807-21. [PMID: 22122084 DOI: 10.2217/epi.10.60] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Sexual differentiation of the brain takes place during a perinatal-sensitive time window as a result of gonadal hormone-induced activational and organizational effects on neuronal substrates. Increasing evidence suggests that epigenetic mechanisms can contribute to the establishment and maintenance of some aspects of these processes, and that these epigenetic mechanisms may themselves be under the control of sex hormones. Epigenetic programming of neuroendocrine and behavioral phenotypes frequently occurs sex specifically, pointing to sex differences in brain epigenetics as a possible determinant. Understanding how sex-specific epigenomes and sex-biased responses to environmental cues contribute to the development of brain diseases might provide new insights for epigenetic therapy.
Collapse
Affiliation(s)
- Yannick Menger
- Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany
| | | | | | | |
Collapse
|
27
|
Lenz B, Müller CP, Stoessel C, Sperling W, Biermann T, Hillemacher T, Bleich S, Kornhuber J. Sex hormone activity in alcohol addiction: integrating organizational and activational effects. Prog Neurobiol 2011; 96:136-63. [PMID: 22115850 DOI: 10.1016/j.pneurobio.2011.11.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 11/03/2011] [Accepted: 11/08/2011] [Indexed: 01/06/2023]
Abstract
There are well-known sex differences in the epidemiology and etiopathology of alcohol dependence. Male gender is a crucial risk factor for the onset of alcohol addiction. A directly modifying role of testosterone in alcohol addiction-related behavior is well established. Sex hormones exert both permanent (organizational) and transient (activational) effects on the human brain. The sensitive period for these effects lasts throughout life. In this article, we present a novel early sex hormone activity model of alcohol addiction. We propose that early exposure to sex hormones triggers structural (organizational) neuroadaptations. These neuroadaptations affect cellular and behavioral responses to adult sex hormones, sensitize the brain's reward system to the reinforcing properties of alcohol and modulate alcohol addictive behavior later in life. This review outlines clinical findings related to the early sex hormone activity model of alcohol addiction (handedness, the second-to-fourth-finger length ratio, and the androgen receptor and aromatase) and includes clinical and preclinical literature regarding the activational effects of sex hormones in alcohol drinking behavior. Furthermore, we discuss the role of the hypothalamic-pituitary-adrenal and -gonadal axes and the opioid system in mediating the relationship between sex hormone activity and alcohol dependence. We conclude that a combination of exposure to sex hormones in utero and during early development contributes to the risk of alcohol addiction later in life. The early sex hormone activity model of alcohol addiction may prove to be a valuable tool in the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Bernd Lenz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Marshall KM. Introduction to the interaction between gonadal steroids and the central nervous system. Curr Top Behav Neurosci 2011; 8:1-13. [PMID: 21644052 DOI: 10.1007/7854_2011_136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The sex steroids are frequently referred to as the gonadal steroids and are erroneously assumed to be exclusively linked to the ovaries in women or the testes in men and the functions of the reproductive tract. This chapter will provide an overview of some of the extragonadal effects of these hormones, focusing on the central nervous system, and the mechanisms of hormone action. Hormone synthesis and metabolism within the CNS will be discussed with particular focus on the role of aromatase. Sex steroids exert many of their effects via intracellular receptors and these genomic responses tend to be slow in onset, however, some responses to steroids occur more quickly and are mediated via membrane receptors and involve interactions with many different transduction pathways to produce a diverse array of responses. These complexities do pose challenges but also offer opportunity for novel approaches for therapeutic exploitation as the pharmacological tools with which to modulate systems become increasingly available.
Collapse
Affiliation(s)
- Kay M Marshall
- School of Pharmacy, University of Bradford, Bradford, BD7 1DP, UK.
| |
Collapse
|
29
|
Moraes AN, Gouvêa SA, Gonçalves WLS, Romero WG, Moyses MR, Bissoli NS, Pires JGP, Abreu GR. Raloxifene reduces blood pressure in hypertensive animals after ovarian hormone deprivation. Basic Clin Pharmacol Toxicol 2011; 109:334-8. [PMID: 21624057 DOI: 10.1111/j.1742-7843.2011.00734.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Raloxifene is a selective oestrogen receptor modulator that has been approved for the prevention and treatment of osteoporosis in post-menopausal women. Studies have revealed several effects of raloxifene on the cardiovascular system, which might contribute to the blood pressure regulatory mechanisms, particularly in the systemic arterial hypertension. Therefore, the aim of this study was to investigate the effects of raloxifene on the blood pressure, renal excretion of water and Na(+) and plasma nitrite/nitrate levels in 2-kidney-1-clip (2K1C) hypertensive female rats. The groups were as follows: hypertensive (2K1C), hypertensive ovariectomized (2K1C + OVX) and hypertensive ovariectomized treated with raloxifene (2K1C + OVX + R). Seven days after the surgery that produced menopause, 2K1C hypertension was produced in anaesthetized animals. Seven days after the clip application, the rats were put into metabolic cages to allow for the measurement of water ingestion and diuresis, and raloxifene was administered (2 mg/kg/day i.p., for 7 more days). We found a large reduction (p < 0.01) in mean arterial pressure (197 ± 6 to 164 ± 2 mmHg), an increase in renal excretion of sodium and water (p < 0.05) and an increase in plasma levels of nitrite/nitrate in 2K1C + OVX + R animals, when compared with the 2K1C (23.4 ± 1 versus 14 ± 0.5 nmol/mL; p < 0.01, respectively). These findings suggest that raloxifene exerted its antihypertensive effect, at least in part, by improving the renal excretion of sodium and water.
Collapse
Affiliation(s)
- Adriana Nunes Moraes
- Department of Health Sciences, CEUNES, Federal University of Espirito Santo, São Mateus, ES, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Campagna R, Brunelli L, Airoldi L, Fanelli R, Hakansson H, Heimeier RA, De Boever P, Boix J, Llansola M, Felipo V, Pastorelli R. Cerebellum Proteomics Addressing the Cognitive Deficit of Rats Perinatally Exposed to the Food-Relevant Polychlorinated Biphenyl 138. Toxicol Sci 2011; 123:170-9. [DOI: 10.1093/toxsci/kfr156] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Okuliarova M, Kostal L, Zeman M. Effects of divergent selection for yolk testosterone content on growth characteristics of Japanese quail. Comp Biochem Physiol A Mol Integr Physiol 2011; 160:81-6. [PMID: 21627997 DOI: 10.1016/j.cbpa.2011.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/13/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
Effects of yolk androgens on postnatal growth of offspring have been widely studied but their physiological role in the growth control is not fully understood due to an inconsistency in obtained results. We investigated androgen-mediated maternal effects on postnatal growth in relation to endocrine control mechanisms using two lines of Japanese quail divergently selected for high (HET) and low (LET) egg testosterone (T) content. Embryonic growth did not differ between the lines. During the growth period HET quail were heavier and displayed longer tarsi as compared with LET quail, with more pronounced line differences in males than females. HET males were heavier than LET males from the age of 2 weeks, reached the age of maximum growth rate earlier, and displayed higher asymptotic body weight than LET males. Accelerated growth in HET males was not accompanied by increased postembryonic plasma T concentrations. Plasma triiodothyronine levels did not differ between lines while plasma thyroxine levels were decreased in HET as compared with LET female chicks. Line differences in body weight disappeared in adult quail suggesting that yolk androgens, increased in a physiological way, resulted in stimulation of juvenile growth rate in precocial Japanese quail under stable social and environmental conditions.
Collapse
Affiliation(s)
- Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic.
| | | | | |
Collapse
|
32
|
Li Z, Kroll KJ, Jensen KM, Villeneuve DL, Ankley GT, Brian JV, Sepúlveda MS, Orlando EF, Lazorchak JM, Kostich M, Armstrong B, Denslow ND, Watanabe KH. A computational model of the hypothalamic: pituitary: gonadal axis in female fathead minnows (Pimephales promelas) exposed to 17α-ethynylestradiol and 17β-trenbolone. BMC SYSTEMS BIOLOGY 2011; 5:63. [PMID: 21545743 PMCID: PMC3118352 DOI: 10.1186/1752-0509-5-63] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 05/05/2011] [Indexed: 11/10/2022]
Abstract
Background Endocrine disrupting chemicals (e.g., estrogens, androgens and their mimics) are known to affect reproduction in fish. 17α-ethynylestradiol is a synthetic estrogen used in birth control pills. 17β-trenbolone is a relatively stable metabolite of trenbolone acetate, a synthetic androgen used as a growth promoter in livestock. Both 17α-ethynylestradiol and 17β-trenbolone have been found in the aquatic environment and affect fish reproduction. In this study, we developed a physiologically-based computational model for female fathead minnows (FHM, Pimephales promelas), a small fish species used in ecotoxicology, to simulate how estrogens (i.e., 17α-ethynylestradiol) or androgens (i.e., 17β-trenbolone) affect reproductive endpoints such as plasma concentrations of steroid hormones (e.g., 17β-estradiol and testosterone) and vitellogenin (a precursor to egg yolk proteins). Results Using Markov Chain Monte Carlo simulations, the model was calibrated with data from unexposed, 17α-ethynylestradiol-exposed, and 17β-trenbolone-exposed FHMs. Four Markov chains were simulated, and the chains for each calibrated model parameter (26 in total) converged within 20,000 iterations. With the converged parameter values, we evaluated the model's predictive ability by simulating a variety of independent experimental data. The model predictions agreed with the experimental data well. Conclusions The physiologically-based computational model represents the hypothalamic-pituitary-gonadal axis in adult female FHM robustly. The model is useful to estimate how estrogens (e.g., 17α-ethynylestradiol) or androgens (e.g., 17β-trenbolone) affect plasma concentrations of 17β-estradiol, testosterone and vitellogenin, which are important determinants of fecundity in fish.
Collapse
Affiliation(s)
- Zhenhong Li
- Division of Environmental and Biomolecular Systems, Oregon Health & Science University, Beaverton, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Imamura T. Epigenetic setting for long-term expression of estrogen receptor α and androgen receptor in cells. Horm Behav 2011; 59:345-52. [PMID: 20619266 DOI: 10.1016/j.yhbeh.2010.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 05/20/2010] [Accepted: 05/22/2010] [Indexed: 12/15/2022]
Abstract
Epigenetic regulation of the nuclear estrogen and androgen receptors, ER and AR, constitutes the molecular basis for the long-lasting effects of sex steroids on gene expression in cells. The effects prevail at hundreds of gene loci in the proximity of estrogen- and androgen-responsive elements and many more such loci through intra- and even inter-chromosomal level regulation. Such a memory system should be active in a flexible manner during the early development of vertebrates, and later replaced to establish more stable marks on genomic DNA. In mammals, DNA methylation is utilized as a very stable mark for silencing of the ERα and AR isoform expression during cancer cell and normal brain development. The factors affecting the DNA methylation of the ERα and AR genes in cells include estrogen and androgen. Since testosterone induces brain masculinization through its aromatization to estradiol in a narrow time window of the perinatal stage in rodents, the autoregulation of estrogen receptors, especially the predominant form of ERα, at the level of DNA methylation to set up the "cell memory" affecting the sexually differentiated status of brain function has been attracting increasing attention. The alternative usage of the androgen-AR system for brain masculinization and estrogenic regulation of AR expression in some species imply that the DNA methylation pattern of the AR gene can be established by closely related but different systems for sex steroid-induced phenomena, including brain masculinization.
Collapse
Affiliation(s)
- Takuya Imamura
- Laboratory for Biodiversity, Global COE Program, Division of Biological Science, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto 606-8502, Japan.
| |
Collapse
|
34
|
Liu P, Meng XH, Wang H, Ji YL, Zhao M, Zhao XF, Xu ZM, Chen YH, Zhang C, Xu DX. Effects of pubertal fenvalerate exposure on testosterone and estradiol synthesis and the expression of androgen and estrogen receptors in the developing brain. Toxicol Lett 2011; 201:181-9. [PMID: 21232584 DOI: 10.1016/j.toxlet.2010.12.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/23/2010] [Accepted: 12/27/2010] [Indexed: 10/18/2022]
Abstract
Fenvalerate is a potential endocrine disruptor. Several studies have demonstrated that fenvalerate disrupts testosterone (T) synthesis in testes. T and estradiol (E(2)) are de novo synthesized in the developing brain. Thus, the aim of the present study was to investigate the effects of pubertal fenvalerate exposure on the synthesis of T and E(2) and the expression of androgen receptor (AR) and estrogen receptors (ERs) in cerebral cortex. CD-1 mice were orally administered daily with either vehicle or fenvalerate (7.5 or 30 mg/kg) from postnatal day (PND) 28 to PND56. The level of T and E(2) in cerebral cortex was significantly decreased in males exposed to fenvalerate. In agreement with the decrease in T and E(2) syntheses, the expression of 17β-HSD, a key enzyme for T synthesis, was significantly reduced in cerebral cortex of fenvalerate-exposed males. Conversely, in females, the expression of 17β-HSD in cerebral cortex was mildly up-regulated by fenvalerate and the level of T and E(2) was mildly increased. Pubertal fenvalerate exposure had no effect on the expression of StAR, P450(17α) and P450scc, the key enzymes for T synthesis, and P450 aromatase, the key enzyme for E(2) synthesis, in cerebral cortex of males and females. Interestingly, the expression of AR in cerebral cortex was up-regulated in male and female mice exposed to fenvalerate, whereas pubertal fenvalerate exposure did not affect the level of ERα and ERβ in cerebral cortex. Taken together, these results suggest that pubertal fenvalerate exposure disrupts T and E(2) synthesis and the expression of AR in cerebral cortex. These changes of steroid status in the developing brain might be deleterious for neurobehavioral development.
Collapse
Affiliation(s)
- Ping Liu
- Anhui Provincial Key Laboratory of Population Health & Aristogenics, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Amyloid Precursor Protein (APP) mRNA Level is Higher in the Old Mouse Cerebral Cortex and is Regulated by Sex Steroids. J Mol Neurosci 2010; 43:235-40. [DOI: 10.1007/s12031-010-9402-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 05/31/2010] [Indexed: 11/26/2022]
|
36
|
Feng Y, Weijdegård B, Wang T, Egecioglu E, Fernandez-Rodriguez J, Huhtaniemi I, Stener-Victorin E, Billig H, Shao R. Spatiotemporal expression of androgen receptors in the female rat brain during the oestrous cycle and the impact of exogenous androgen administration: a comparison with gonadally intact males. Mol Cell Endocrinol 2010; 321:161-74. [PMID: 20197080 DOI: 10.1016/j.mce.2010.02.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 02/18/2010] [Accepted: 02/22/2010] [Indexed: 01/16/2023]
Abstract
Little is known about the regulation and cellular distribution of androgen receptors (ARs) in female rodent brains at various stages of the oestrous cycle. This information is critical for further studies of androgen signalling in the regulation of brain function under physiological and pathophysiological conditions. In this report, we show that the distribution of AR immunoreactivity in the female rat brain is consistent with reported AR mRNA hybridisation signals in the male brain, except for the dentate gyrus of the hippocampus. Immunohistochemical and Western blot analyses performed herein revealed that the onset of region-specific changes in AR proteins was strongly correlated with circulating and ovarian levels of estradiol and testosterone across the oestrous cycle. During the metestrus and diestrus stages, however, the highest levels of AR expression were abolished by chronic dihydrotestosterone (DHT) treatment. This demonstrates that fluctuations in endogenous androgens are required for the regulation of AR expression in the female rat brain. Colocalisation studies revealed that: (1) anatomical variations in AR protein localisation existed between female and male brains, (2) AR immunoreactivity was both neuronal and non-neuronal, and (3) AR protein expression was lower in female rat brains at all stages of the oestrous cycle compared to age-matched males. Our results indicate the presence of regional sex differences in AR expression and changes in the proportion of AR between different subcellular compartments. Furthermore, DHT was found to down-regulate the level of AR in the subcellular compartment in females in a region-specific manner. As a whole, the present study provides the first step toward understanding the dynamics of AR expression and regulation in the brain during normal physiological conditions and for differences in neuronal androgen effects based on sex.
Collapse
Affiliation(s)
- Yi Feng
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Walters KA, Simanainen U, Handelsman DJ. Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Hum Reprod Update 2010; 16:543-58. [DOI: 10.1093/humupd/dmq003] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
38
|
Meyer RP, Gehlhaus M. A role for CYP in the drug–hormone crosstalk of the brain. Expert Opin Drug Metab Toxicol 2010; 6:675-87. [DOI: 10.1517/17425251003680791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Cunningham RL, Giuffrida A, Roberts JL. Androgens induce dopaminergic neurotoxicity via caspase-3-dependent activation of protein kinase Cdelta. Endocrinology 2009; 150:5539-48. [PMID: 19837873 PMCID: PMC2795716 DOI: 10.1210/en.2009-0640] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aged men have a greater incidence of Parkinson's disease (PD) than women. PD is a neurodegenerative condition associated with the loss of dopamine neurons in the nigrostriatal pathway. This study examined the neurotoxic effects of androgens in a dopaminergic cell line (N27 cells) and the downstream signaling pathways activated by androgens. Treatment of N27 cells with testosterone- and dihydrotestosterone-induced mitochondrial dysfunction, protein kinase C (PKC)-delta cleavage, and apoptosis in dopaminergic neuronal cells. Inhibition of caspase-3 prevented the cleavage of PKCdelta from the full-length element to the catalytic fragment and apoptosis in N27 cells, suggesting that androgen-induced apoptosis is mediated by caspase-3-dependent activation of PKCdelta. Androgen-induced apoptosis may be specific to dopamine neurons as evidenced by a lack of testosterone-induced apoptosis in GnRH neurons. These results support a neurotoxic consequence of testosterone on dopaminergic neurons and may provide insight into the gender bias found in PD.
Collapse
Affiliation(s)
- Rebecca L Cunningham
- Department of Pharmacology and the Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78229, USA.
| | | | | |
Collapse
|
40
|
Walters KA, McTavish KJ, Seneviratne MG, Jimenez M, McMahon AC, Allan CM, Salamonsen LA, Handelsman DJ. Subfertile female androgen receptor knockout mice exhibit defects in neuroendocrine signaling, intraovarian function, and uterine development but not uterine function. Endocrinology 2009; 150:3274-82. [PMID: 19359383 PMCID: PMC2703552 DOI: 10.1210/en.2008-1750] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Female androgen receptor (AR) knockout mice (AR(-/-)) generated by an in-frame Ar exon 3 deletion are subfertile, but the mechanism is not clearly defined. To distinguish between extra- and intraovarian defects, reciprocal ovarian transplants were undertaken. Ovariectomized AR(-/-) hosts with wild-type (AR(+/+)) ovary transplants displayed abnormal estrus cycles, with longer cycles (50%, P < 0.05), and 66% were infertile (P < 0.05), whereas AR(+/+) hosts with either AR(-/-) or surgical control AR(+/+) ovary transplants displayed normal estrus cycles and fertility. These data imply a neuroendocrine defect, which is further supported by increased FSH (P <0.05) and estradiol (P <0.05), and greater LH suppressibility by estradiol in AR(-/-) females at estrus (P <0.05). Additional intraovarian defects were observed by the finding that both experimental transplant groups exhibited significantly reduced pups per litter (P < 0.05) and corpora lutea numbers (P < 0.05) compared with surgical controls. All groups exhibited normal uterine and lactation functions. AR(-/-) uteri were morphologically different from AR(+/+) with an increase in horn length (P < 0.01) but a reduction in uterine diameter (P < 0.05), total uterine area (P < 0.05), endometrial area (P < 0.05), and myometrial area (P < 0.01) at diestrus, indicating a role for AR in uterine growth and development. Both experimental transplant groups displayed a significant reduction in uterine diameter (P < 0.01) compared with transplanted wild-type controls, indicating a role for both AR-mediated intraovarian and intrauterine influences on uterine physiology. In conclusion, these data provide direct evidence that extraovarian neuroendocrine, but not uterine effects, as well as local intraovarian AR-mediated actions are important in maintaining female fertility, and a disruption of AR signaling leads to altered uterine development.
Collapse
Affiliation(s)
- K A Walters
- Andrology Laboratory, ANZAC Research Institute, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Meyer RP, Gehlhaus M, Schwab R, Bürck C, Knoth R, Hagemeyer CE. Concordant up-regulation of cytochrome P450 Cyp3a11, testosterone oxidation and androgen receptor expression in mouse brain after xenobiotic treatment. J Neurochem 2009; 109:670-81. [DOI: 10.1111/j.1471-4159.2009.05994.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Movérare-Skrtic S, Mellström D, Vandenput L, Ehrich M, Ohlsson C. Peripheral blood leukocyte distribution and body mass index are associated with the methylation pattern of the androgen receptor promoter. Endocrine 2009; 35:204-10. [PMID: 19199084 DOI: 10.1007/s12020-009-9153-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 11/24/2008] [Accepted: 11/26/2008] [Indexed: 12/31/2022]
Abstract
Methylation of CpG sites in the promoter region can affect gene transcription. DNA derived from peripheral blood leukocytes (PBL) from the well-characterized clinical cohorts might be useful to study the influence of environmental factors on DNA methylation. However, these studies could be confounded by the heterogeneous nature of PBL. The aims of this study were to determine the impact of PBL distribution on methylation status of the androgen receptor (AR) promoter, and determine the associations between PBL distribution-adjusted methylation status of the AR promoter and AR-related phenotypes. PBL differential count analyses were performed at the time of blood sampling for DNA preparation in 170 elderly men. The DNA was bisulfite treated, and the methylation status of five CpG units in the AR promoter was analyzed using a high-throughput technique based on MALDI-TOF mass spectrometry. The degree of methylation of all the five investigated CpG units was strongly positively associated with the percent of lymphocytes in the PBL (r (s) = 0.17-0.49, P < 0.05). Furthermore, the PBL distribution-adjusted methylation status of a specific CpG unit in the AR promoter was significantly associated with body mass index (r (s) = 0.24) and other measures reflecting fat mass in elderly men. In conclusion, adjustment for PBL distribution needs to be done to be able to use DNA from whole blood for methylation analysis of the AR promoter and most likely also when investigating other promoters.
Collapse
Affiliation(s)
- Sofia Movérare-Skrtic
- Division of Endocrinology, Departments of Internal Medicine and Geriatrics, Center for Bone Research at the Sahlgrenska Academy (CBS), University of Gothenburg, Gröna Stråket 8, 413 45 Gothenburg, Sweden.
| | | | | | | | | |
Collapse
|
43
|
Vismara G, Simonini F, Onesto E, Bignamini M, Miceli V, Martini L, Poletti A. Androgens inhibit androgen receptor promoter activation in motor neurons. Neurobiol Dis 2009; 33:395-404. [DOI: 10.1016/j.nbd.2008.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 10/17/2008] [Accepted: 11/14/2008] [Indexed: 11/25/2022] Open
|
44
|
Androgen Receptor is Expressed in Murine Choroid Plexus and Downregulated by 5α-Dihydrotestosterone in Male and Female Mice. J Mol Neurosci 2008; 38:41-9. [DOI: 10.1007/s12031-008-9157-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 10/14/2008] [Indexed: 10/21/2022]
|
45
|
Age- and sex-related analysis of methylation of 5′-upstream sequences of Fmr-1 gene in mouse brain and modulation by sex steroid hormones. Biogerontology 2008; 9:455-65. [DOI: 10.1007/s10522-008-9178-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Accepted: 08/29/2008] [Indexed: 10/21/2022]
|
46
|
Ghosh S, Thakur MK. PS2 protein expression is upregulated by sex steroids in the cerebral cortex of aging mice. Neurochem Int 2008; 52:363-7. [PMID: 17728018 DOI: 10.1016/j.neuint.2007.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 07/20/2007] [Indexed: 01/21/2023]
Abstract
Mutations in presenilin (PS) genes cause majority of early onset Alzheimer's disease (AD), an age related neurodegenerative disorder. PS proteins undergo proteolytic cleavage to produce biologically active fragments, which constitute the catalytic core of the gamma-secretase enzyme. This enzyme cleaves beta-amyloid precursor protein (betaAPP) to generate Abeta peptides, which are influenced by sex steroids. Recently we have reported the downregulation of PS1 expression by sex steroids in the brain of adult mice. Here we have examined the effect of gonadectomy and subsequent administration of gonadal hormones 17beta-estradiol and testosterone on the level of PS2 C-terminal fragment (CTF) in the cerebral cortex of adult and old AKR strain mice of both sexes. PS2 expression was downregulated following gonadectomy, but upregulated by supplementation of gonadal steroids in both age groups and sexes. Thus these results demonstrate up-regulation of PS2 protein expression by sex steroids, which in turn may influence PS2 associated brain functions.
Collapse
Affiliation(s)
- Soumi Ghosh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | | |
Collapse
|
47
|
Vahter ME. Interactions between arsenic-induced toxicity and nutrition in early life. J Nutr 2007; 137:2798-804. [PMID: 18029502 DOI: 10.1093/jn/137.12.2798] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Exposure to arsenic through drinking water is a major public health problem affecting most countries, although the situation is particularly severe in low-income nations. The health consequences of chronic arsenic exposure include increased risk for various forms of cancer and numerous noncancer effects, including diabetes, skin diseases, chronic cough, and toxic effects on liver, kidney, cardiovascular system, and peripheral and central nervous systems. In recent years increasing reports of effects on fetal and child development have appeared. There seems to be a wide variation in susceptibility to arsenic toxicity, which is likely to be related to factors such as variation in arsenic metabolism, nutrition, host-related defense mechanisms, and genetic predisposition. The main mechanisms of arsenic-nutrition interactions include arsenic-induced oxidative stress, which requires nutrient-dependent defense systems, and arsenic metabolism (methylation) via 1-carbon metabolism, which requires methyl groups, folic acid, vitamin B-12, and betaine for the remethylation of homocysteine to methionine. An efficient first methylation step in combination with a slow second methylation step seems to be most critical from a toxicological point of view. A third mode of arsenic-nutrition interaction involves epigenetic effects and fetal programming via DNA methylation.
Collapse
Affiliation(s)
- Marie E Vahter
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
48
|
Abstract
Although androgens and the androgen receptor (AR) have defining roles in male reproductive development and function, previously no role in female reproductive physiology beyond testosterone (T) as the precursor in estradiol (E(2)) biosynthesis was firmly established. Understanding the role and specific mechanisms of androgen action via the AR in the ovary has been limited by confusion on how to interpret results from pharmacological studies, because many androgens can be metabolized in vivo and in vitro to steroids that can also exert actions via the estrogen receptor (ESR). Recent genetic studies using mouse models with specific disruption of the Ar gene have highlighted the role that AR-mediated actions play in maintaining female fertility through key roles in the regulation of follicle health, development, and ovulation. Furthermore, these genetic studies have revealed that AR-mediated effects influence age-related female fertility, possibly via mechanisms acting predominantly at the hypothalamic-pituitary axis in a dose-dependent manner. This review focuses on combining the findings from pharmacological studies and novel genetic mouse models to unravel the roles of ovarian androgen actions in relation to female fertility and ovarian aging, as well as creating new insights into the role of androgens in androgen-associated reproductive disorders such as polycystic ovarian syndrome.
Collapse
Affiliation(s)
- K A Walters
- Andrology Laboratory, ANZAC Research Institute, Concord Hospital, University of Sydney, New South Wales 2139, Australia
| | | | | |
Collapse
|
49
|
Walters KA, Allan CM, Jimenez M, Lim PR, Davey RA, Zajac JD, Illingworth P, Handelsman DJ. Female mice haploinsufficient for an inactivated androgen receptor (AR) exhibit age-dependent defects that resemble the AR null phenotype of dysfunctional late follicle development, ovulation, and fertility. Endocrinology 2007; 148:3674-84. [PMID: 17463055 DOI: 10.1210/en.2007-0248] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of classical genomic androgen receptor (AR) mediated actions in female reproductive physiology remains unclear. Female mice homozygous for an in-frame deletion of exon 3 of the Ar (AR(-/-)) were subfertile, exhibiting delayed production of their first litter (AR(+/+) = 22 d vs. AR(-/-) = 61 d, P < 0.05) and producing 60% fewer pups/litter (AR(+/+): 8.1 +/- 0.4 vs. AR(-/-): 3.2 +/- 0.9, P < 0.01). Heterozygous females (AR(+/-)) exhibited an age-dependent 55% reduction (P < 0.01) in pups per litter, evident from 6 months of age (P < 0.05), compared with AR(+/+), indicating a significant gene dosage effect on female fertility. Ovulation was defective with a significant reduction in corpora lutea numbers (48-79%, P < 0.01) in 10- to 12- and 26-wk-old AR(+/-) and AR(-/-) females and a 57% reduction in oocytes recovered from naturally mated AR(-/-) females (AR(+/+): 9.8 +/- 1.0 vs. AR(-/-): 4.2 +/- 1.2, P < 0.01); however, early embryo development to the two-cell stage was unaltered. The delay in first litter, reduction in natural ovulation rate, and aromatase expression in AR(+/-) and AR(-/-) ovaries, coupled with the restored ovulation rate by gonadotropin hyperstimulation in AR(-/-) females, suggest aberrant gonadotropin regulation. A 2.7-fold increase (AR(+/+): 35.4 +/- 13.4 vs. AR(-/-): 93.9 +/- 6.1, P < 0.01) in morphologically unhealthy antral follicles demonstrated deficiencies in late follicular development, although growing follicle populations and growth rates were unaltered. This novel model reveals that classical genomic AR action is critical for normal ovarian function, although not for follicle depletion and that haploinsufficiency for an inactivated AR may contribute to a premature reduction in female fecundity.
Collapse
Affiliation(s)
- K A Walters
- ANZAC Research Institute, Department of Andrology, Concord Hospital, University of Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Thakur MK, Kumar RC. 17Beta-estradiol modulates age-dependent binding of 40 kDa nuclear protein to androgen receptor promoter in mouse cerebral cortex. Biogerontology 2007; 8:575-82. [PMID: 17619166 DOI: 10.1007/s10522-007-9102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 05/15/2007] [Indexed: 11/24/2022]
Abstract
Androgen influences the function of central and peripheral nervous system and plays a crucial role in maintaining reproductive behaviors and neuroendocrine regulation. Such action is mediated by interaction of androgen receptor (AR) promoter with nuclear proteins, which are involved in transcriptional regulation of androgen responsive genes. We have analyzed the binding of AR core promoter to nuclear proteins from the cerebral cortex of adult and old mice of both sexes by electrophoretic mobility shift assay (EMSA) and characterized the bound protein by Southwestern blotting. EMSA showed that the binding of nuclear proteins declined in the cerebral cortex of intact old mice as compared to adult. Following gonadectomy, the binding was reduced in old male and adult female but increased in old female. In contrast, estradiol supplementation increased the binding in old male and adult female but decreased in old female. Southwestern blotting analysis revealed that a 40 kDa nuclear protein bound to the promoter and the binding pattern was similar to that observed in EMSA. Further characterization of this protein may help to explore the intricate mechanism that underlies the transcriptional regulation of androgen responsive genes during aging.
Collapse
Affiliation(s)
- Mahendra K Thakur
- Biochemistry & Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| | | |
Collapse
|