1
|
González-Flores O, Domínguez-Ordóñez R, Delgado-Macuil RJ, Tlachi-López JL, Luna-Hernández A, Montes-Narváez O, Pfaus JG, García-Juárez M. Participation of kisspeptin, progesterone, and GnRH receptors on lordosis behavior induced by kisspeptin. Physiol Behav 2024; 283:114609. [PMID: 38851441 DOI: 10.1016/j.physbeh.2024.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
The neuropeptide kisspeptin (Kiss) is crucial in regulating the hypothalamic-pituitary-gonadal axis. It is produced by two main groups of neurons in the hypothalamus: the rostral periventricular region around the third ventricle and the arcuate nucleus. Kiss is the peptide product of the KiSS-1 gene and serves as the endogenous agonist for the GPR54 receptor. The Kiss/GPR54 system functions as a critical regulator of the reproductive system. Thus, we examined the effect of intracerebroventricular administration of 3 μg of Kiss to the right lateral ventricle of ovariectomized rats primed with a dose of 5 μg subcutaneous (sc) of estradiol benzoate (EB). Kiss treatment increased the lordosis quotient at all times tested. However, the lordosis reflex score was comparatively lower yet still significant compared to the control group. To investigate receptor specificity and downstream mechanisms on lordosis, we infused 10 μg of GPR54 receptor antagonist, Kiss-234, 5 μg of the progestin receptor antagonist, RU486, or 3 μg of antide, a gonadotropin-releasing hormone-1 (GnRH-1) receptor antagonist, to the right lateral ventricle 30 min before an infusion of 3 μg of Kiss. Results demonstrated a significant reduction in the facilitation of lordosis behavior by Kiss at 60 and 120 min when Kiss-234, RU486, or antide were administered. These findings suggest that Kiss stimulates lordosis expression by activating GPR54 receptors on GnRH neurons and that Kiss/GPR54 system is an essential intermediary by which progesterone activates GnRH.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Raymundo Domínguez-Ordóñez
- Licenciatura en Ingeniería Agronómica y Zootecnia, CRC, Benemérita Universidad Autónoma de, Puebla, México
| | - Raul Jacobo Delgado-Macuil
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional, Santa Inés, Tecuexcomac, Tlaxcala, México
| | | | - Ailyn Luna-Hernández
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Omar Montes-Narváez
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Charles University, Prague, Czech Republic
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México.
| |
Collapse
|
2
|
Impact of Vitamin B12 Insufficiency on the Incidence of Sarcopenia in Korean Community-Dwelling Older Adults: A Two-Year Longitudinal Study. Nutrients 2023; 15:nu15040936. [PMID: 36839293 PMCID: PMC9967932 DOI: 10.3390/nu15040936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The longitudinal effect of B12 insufficiency on sarcopenia has not yet been investigated in older adults. We aimed to study the impact of B12 levels on alterations in muscle mass, function and strength over two years. Non-sarcopenic older adults (n = 926) aged 70-84 were included. Using the Korean Frailty and Aging Cohort Study, this two-year longitudinal study used data across South Korea. The tools used for assessing muscle criteria were based on the Asian Working Group for Sarcopenia guidelines. Participants were divided into the insufficiency (initial serum B12 concentration < 350 pg/mL) and sufficiency groups (≥350 pg/mL). Logistic regression analyses were performed to evaluate the effect of initial B12 concentration on sarcopenia parameters over a two-year period. In women, multivariate analysis showed that the B12 insufficiency group had a significantly higher incidence of low SPPB scores (odds ratio [OR] = 3.28, 95% confidence interval [CI] = 1.59-6.76) and sarcopenia (OR = 3.72, 95% CI = 1.10-12.62). However, the B12 insufficiency group did not have a greater incidence of sarcopenia or other parameters in men. Our findings suggest B12 insufficiency negatively impacts physical performance and increases the incidence of sarcopenia only in women.
Collapse
|
3
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
4
|
Ysrraelit MC, Correale J. Impact of Andropause on Multiple Sclerosis. Front Neurol 2021; 12:766308. [PMID: 34803897 PMCID: PMC8602357 DOI: 10.3389/fneur.2021.766308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Andropause results from the natural decrease in testosterone levels that occurs with age. In contrast to menopause, which is a universal, well-characterized process associated with absolute gonadal failure, andropause ensues after gradual decline of both hypothalamic-pituitary-gonadal axis activity, as well as of testicular function, a process which usually develops over a period of many years. Increasing evidence on greater risk of Multiple sclerosis (MS) associated with lower testosterone levels is being reported. Likewise, epidemiological studies have shown a later age of onset of MS in men, relative to women, which could perhaps respond to the decline in protective testosterone levels. In this review, we will discuss the role of androgens in the development and function of the innate and adaptive immune response, as well as in neuroprotective mechanisms relevant to MS. Testosterone effects observed in different animal models and in epidemiological studies in humans will be discussed, as well as their correlation with physical disability and cognitive function levels. Finally, published and ongoing clinical trials exploring the role of androgens, particularly at key stages of sexual maturation, will be reviewed.
Collapse
Affiliation(s)
- Maria C Ysrraelit
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Jorge Correale
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
5
|
Bianchi VE, Bresciani E, Meanti R, Rizzi L, Omeljaniuk RJ, Torsello A. The role of androgens in women's health and wellbeing. Pharmacol Res 2021; 171:105758. [PMID: 34242799 DOI: 10.1016/j.phrs.2021.105758] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022]
Abstract
Androgens in women, as well as in men, are intrinsic to maintenance of (i) reproductive competency, (ii) cardiac health, (iii) appropriate bone remodeling and mass retention, (iii) muscle tone and mass, and (iv) brain function, in part, through their mitigation of neurodegenerative disease effects. In recognition of the pluripotency of endogenous androgens, exogenous androgens, and selected congeners, have been prescribed off-label for several decades to treat low libido and sexual dysfunction in menopausal women, as well as, to improve physical performance. However, long-term safety and efficacy of androgen administration has yet to be fully elucidated. Side effects often observed include (i) hirsutism, (ii) acne, (iii) deepening of the voice, and (iv) weight gain but are associated most frequently with supra-physiological doses. By contrast, short-term clinical trials suggest that the use of low-dose testosterone therapy in women appears to be effective, safe and economical. There are, however, few clinical studies, which have focused on effects of androgen therapy on pre- and post-menopausal women; moreover, androgen mechanisms of action have not yet been thoroughly explained in these subjects. This review considers clinical effects of androgens on women's health in order to prevent chronic diseases and reduce cancer risk in gynecological tissues.
Collapse
Affiliation(s)
- Vittorio E Bianchi
- Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta 42, Falciano 47891, San Marino.
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Robert J Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario P7B 5E1, Canada.
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| |
Collapse
|
6
|
Ali AA, Cui X, Pertile RAN, Li X, Medley G, Alexander SA, Whitehouse AJO, McGrath JJ, Eyles DW. Developmental vitamin D deficiency increases foetal exposure to testosterone. Mol Autism 2020; 11:96. [PMID: 33298169 PMCID: PMC7727109 DOI: 10.1186/s13229-020-00399-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders which are more common in males. The ‘prenatal sex steroid’ hypothesis links excessive sex-steroid exposure during foetal life with the behavioural differences observed in ASD. However, the reason why sex steroid exposure may be excessive remains unclear. Epidemiological studies have identified several environmental risk factors associated with ASD, including developmental vitamin D (DVD) deficiency. We have demonstrated in an animal model that DVD-deficiency is associated with a hyper-inflammatory response in placentas from male but not female foetuses. Vitamin D also regulates the expression of several steroidogenic enzymes in vitro. Therefore using this animal model, we have examined whether DVD-deficiency leads to increased sex-steroid levels in both the maternal and foetal compartments. Methods Female rats are fed a vitamin D deficient diet from 6 weeks before mating until tissue collection at embryonic day 18. We examined the levels of testosterone, androstenedione and corticosterone in maternal plasma, foetal brains and amniotic fluid. We further examined gene expressions of steroidogenic enzymes and DNA methylation of aromatase promoters in foetal brains as a potential molecular mechanism regulating testosterone expression. Results We show that DVD-deficiency increases testosterone levels in maternal blood. We also show elevated levels of testosterone and androstenedione in the amniotic fluid of female but not male DVD-deficient foetuses. Testosterone levels were also elevated in DVD-deficient male brains. Vitamin D, like other steroid-related hormones, regulates gene expression via methylation. Therefore we examined whether the significant elevation in testosterone in male brains was due to such a potential gene-silencing mechanism. We show that the promoter of aromatase was hyper-methylated compared to male controls. Limitations A reduction in aromatase, in addition to causing excessive testosterone, could also lead to a reduction in estradiol which was not examined here. Conclusions This study is the first to show how an epidemiologically established environmental risk factor for ASD may selectively elevate testosterone in male embryonic brains. These findings provide further mechanistic support for the prenatal sex steroid theory of ASD.
Collapse
Affiliation(s)
- Asad Amanat Ali
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | | | - Xiang Li
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Gregory Medley
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Suzanne Adele Alexander
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - Andrew J O Whitehouse
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia
| | - John Joseph McGrath
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia.,NCRR-National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Aarhus C, Denmark
| | - Darryl Walter Eyles
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia. .,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia.
| |
Collapse
|
7
|
Fink JE, Hackney AC, Matsumoto M, Maekawa T, Horie S. Mobility and Biomechanical Functions in the Aging Male: Testosterone and the Locomotive Syndrome. Aging Male 2020; 23:403-410. [PMID: 30269622 DOI: 10.1080/13685538.2018.1504914] [Citation(s) in RCA: 10] [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] [Indexed: 12/22/2022] Open
Abstract
In the current aging society, the occurrence of the locomotive syndrome, a condition in which the locomotive organs are impaired, is increasing. The locomotive system includes support (bones), mobility and impact absorption (joints and intervertebral disks), drive and control (muscles, nerves), and network (blood vessels). The impairment of any of those systems can lead to a major decrease in quality of life. In recent years, several studies on methods to improve and prevent conditions impairing the locomotive syndrome have been conducted. Almost in parallel with the structure supporting mobility and body functions, testosterone levels decrease with age. Testosterone is a hormone-regulating several pathways affecting each aspect of the locomotive syndrome. Testosterone is regulated by the pituitary gland triggering several processes in the body through genomic and non-genomic pathways, affecting muscles, bones, nerves, joints, intervertebral discs, and blood vessels. The purpose of this review is to investigate the role of testosterone in each of the systems involved in the locomotive syndrome.
Collapse
Affiliation(s)
- Julius E Fink
- Graduate School of Medicine, Department of Metabolism and Endocrinology, Juntendo University, Tokyo, JAPAN
| | - Anthony C Hackney
- Department of Exercise & Sport Science; Department of Nutrition - School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Masahito Matsumoto
- Advanced Diabetic Therapeutics, Department of Metabolic Endocrinology, Juntendo University,Tokyo, Japan
| | - Takahiro Maekawa
- Department of Rehabilitation for the Movement Functions Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Shigeo Horie
- Graduate School of Medicine, Department of Urology, Juntendo University, Tokyo, Japan
| |
Collapse
|
8
|
McLeod VM, Chiam MDF, Lau CL, Rupasinghe TW, Boon WC, Turner BJ. Dysregulation of Steroid Hormone Receptors in Motor Neurons and Glia Associates with Disease Progression in ALS Mice. Endocrinology 2020; 161:5867502. [PMID: 32621747 DOI: 10.1210/endocr/bqaa113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease targeting motor neurons which shows sexual dimorphism in its incidence, age of onset, and progression rate. All steroid hormones, including androgens, estrogens, and progestogens, have been implicated in modulating ALS. Increasing evidence suggests that steroid hormones provide neuroprotective and neurotrophic support to motor neurons, either directly or via surrounding glial cell interactions, by activating their respective nuclear hormone receptors and initiating transcriptional regulatory responses. The SOD1G93A transgenic mouse also shows sex-specific differences in age of onset and progression, and remains the most widely used model in ALS research. To provide a more comprehensive understanding of the influences of steroid hormone signaling in ALS, we systemically characterized sex hormone receptor expression at transcript and protein levels, cellular localization, and the impact of disease course in lumbar spinal cords of male and female SOD1G93A mice. We found that spinal motor neurons highly express nuclear androgen receptor (AR), estrogen receptor (ER)α, ERβ, and progesterone receptor with variations in glial cell expression. AR showed the most robust sex-specific difference in expression and was downregulated in male SOD1G93A mouse spinal cord, in association with depletion in 5α-reductase type 2 isoform, which primarily metabolizes testosterone to 5α-dihydrotestosterone. ERα was highly enriched in reactive astrocytes of SOD1G93A mice and ERβ was strongly upregulated. The 5α-reductase type 1 isoform was upregulated with disease progression and may influence local spinal cord hormone levels. In conclusion, steroid hormone receptor expression is dynamic and cell-type specific in SOD1G93A mice which may provide targets to modulate progression in ALS.
Collapse
Affiliation(s)
- Victoria M McLeod
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Mathew D F Chiam
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Chew L Lau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Thusitha W Rupasinghe
- Metabolomics Australia, School of BioSciences, University of Melbourne, VIC, Australia
| | - Wah C Boon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Perron Institute for Neurological and Translational Science, Queen Elizabeth Medical Centre, Nedlands, WA, Australia
| |
Collapse
|
9
|
Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen Therapy in Neurodegenerative Diseases. J Endocr Soc 2020; 4:bvaa120. [PMID: 33094209 PMCID: PMC7568521 DOI: 10.1210/jendso/bvaa120] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington disease, are characterized by the loss of neurons as well as neuronal function in multiple regions of the central and peripheral nervous systems. Several studies in animal models have shown that androgens have neuroprotective effects in the brain and stimulate axonal regeneration. The presence of neuronal androgen receptors in the peripheral and central nervous system suggests that androgen therapy might be useful in the treatment of neurodegenerative diseases. To illustrate, androgen therapy reduced inflammation, amyloid-β deposition, and cognitive impairment in patients with AD. As well, improvements in remyelination in MS have been reported; by comparison, only variable results are observed in androgen treatment of PD. In ALS, androgen administration stimulated motoneuron recovery from progressive damage and regenerated both axons and dendrites. Only a few clinical studies are available in human individuals despite the safety and low cost of androgen therapy. Clinical evaluations of the effects of androgen therapy on these devastating diseases using large populations of patients are strongly needed.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, Falciano, San Marino
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
10
|
Corona G, Guaraldi F, Rastrelli G, Sforza A, Maggi M. Testosterone Deficiency and Risk of Cognitive Disorders in Aging Males. World J Mens Health 2020; 39:9-18. [PMID: 32378366 PMCID: PMC7752509 DOI: 10.5534/wjmh.200017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cognitive impairment and dementia are predicted to undergo a dramatic increase in the following years with more than 131.5 million people being affected by 2030. Although vascular diseases play the most important role in the pathogenesis of memory impairment in aging men, some pre-clinical and clinical evidence has suggested a possible contribution of the age-dependent reduction of testosterone (T). In this paper we have summarized and discussed all the information derived from available animal and experimental studies. In addition, we meta-analyzed data rising from all randomized placebo controlled trials (RCTs) published so far. Only limited preclinical and clinical evidence can support a possible contribution of T in the pathogenesis of the age-dependent impairment of cognitive functions. In addition, our meta-analysis did not support the use of T replacement therapy for the improvement of several cognitive domains analyzed including attention/working memory, executive function, language, verbal memory, visual memory, visuomotor ability, and visuospatial ability. However, it is important to recognize that the vast majority of available RCTs included mixed populations of subjects with eugonadism and hypogonadism preventing any final conclusion being drawn on these issues.
Collapse
Affiliation(s)
- Giovanni Corona
- Endocrinology Unit, Medical Department, Azienda Usl, Maggiore-Bellaria Hospital, Bologna, Italy.
| | - Federica Guaraldi
- Pituitary Unit, IRCCS Institute of Neurological Science of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giulia Rastrelli
- Andrology, Female Endocrinology and Gender Incongruence Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - Alessandra Sforza
- Endocrinology Unit, Medical Department, Azienda Usl, Maggiore-Bellaria Hospital, Bologna, Italy
| | - Mario Maggi
- Endocrinology Unit, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
11
|
Salimi M, Alishah Z, Khazali H, Mahmoudi F. Orexin Decreases Aromatase Gene Expression in The Hypothalamus of Androgenized Female Rats. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2016; 10:190-5. [PMID: 27441052 PMCID: PMC4948071 DOI: 10.22074/ijfs.2016.4909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 07/11/2015] [Indexed: 11/04/2022]
Abstract
BACKGROUND Orexin is a hypothalamic orexigenic neuropeptide, which third cerebral injection of it mainly exerts inhibitory effects on reproductive functions. It increases significantly the Aromatase (Cyp19) gene expression in the hypothalamus of male rats. Aromatase is an enzyme which converts androgens to estradiol in the hypothalamus of rats. Prenatal or neonatal exposure of females to testosterone masculinizes the pattern of Cyp19 mRNA levels in adulthood. In the present study the effects of central injections of orexin-A on hypothalamic Cyp19 gene expression of adult female rats were investigated, while they had been androgenized on third day of postnatal life. MATERIALS AND METHODS In this experimental study, twenty female Wistar rats received subcutaneous injections of testosterone propionate (50 µg/100 µl) on their third day of postnatal life. Adult androgenized rats weighing 180-220 g, received either 3 µl saline or one of 2, 4 or 8 µg/3 µl concentration of orexin via third cerebral ventricle. Five non-androgenized rats, as control group, received intra cerebral ventricle (ICV) injection of 3 µl saline. The hypothalamuses were dissected out and mean Cyp19 mRNA levels were determined by semi-quantitative real time-polymerase chain reaction (PCR) method. Data were analyzed by unpaired t test and one-way ANOVA using SPSS software, version 16. RESULTS Mean relative Cyp19 mRNA level was significantly increased in the hypothalamus of androgenized compared to non-androgenized female rats. Central injec- tions of 2, 4 or 8 µg/3 µl orexin decreased significantly the hypothalamic Cyp19 mRNA level of androgenized rats compared to androgenized-control groups. CONCLUSION The results suggested that the orexin may exert inhibitory effects on the gene expression of Cyp19 in the hypothalamus of neonatal androgenized female rats in adulthood.
Collapse
Affiliation(s)
- Maliheh Salimi
- Department of Physiology, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Zahra Alishah
- Department of Physiology, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Homayoun Khazali
- Department of Physiology, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Fariba Mahmoudi
- Department of Biology, Facualty of Basic Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| |
Collapse
|
12
|
Reddy RC, Amodei R, Estill CT, Stormshak F, Meaker M, Roselli CE. Effect of Testosterone on Neuronal Morphology and Neuritic Growth of Fetal Lamb Hypothalamus-Preoptic Area and Cerebral Cortex in Primary Culture. PLoS One 2015; 10:e0129521. [PMID: 26053052 PMCID: PMC4460015 DOI: 10.1371/journal.pone.0129521] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/08/2015] [Indexed: 12/22/2022] Open
Abstract
Testosterone plays an essential role in sexual differentiation of the male sheep brain. The ovine sexually dimorphic nucleus (oSDN), is 2 to 3 times larger in males than in females, and this sex difference is under the control of testosterone. The effect of testosterone on oSDN volume may result from enhanced expansion of soma areas and/or dendritic fields. To test this hypothesis, cells derived from the hypothalamus-preoptic area (HPOA) and cerebral cortex (CTX) of lamb fetuses were grown in primary culture to examine the direct morphological effects of testosterone on these cellular components. We found that within two days of plating, neurons derived from both the HPOA and CTX extend neuritic processes and express androgen receptors and aromatase immunoreactivity. Both treated and control neurites continue to grow and branch with increasing time in culture. Treatment with testosterone (10 nM) for 3 days significantly (P < 0.05) increased both total neurite outgrowth (35%) and soma size (8%) in the HPOA and outgrowth (21%) and number of branch points (33%) in the CTX. These findings indicate that testosterone-induced somal enlargement and neurite outgrowth in fetal lamb neurons may contribute to the development of a fully masculine sheep brain.
Collapse
Affiliation(s)
- Radhika C. Reddy
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Rebecka Amodei
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, United States of America
| | - Fred Stormshak
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Mary Meaker
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Charles E. Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
13
|
Kinch CD, Ibhazehiebo K, Jeong JH, Habibi HR, Kurrasch DM. Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish. Proc Natl Acad Sci U S A 2015; 112:1475-80. [PMID: 25583509 PMCID: PMC4321238 DOI: 10.1073/pnas.1417731112] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Bisphenol A (BPA), a ubiquitous endocrine disruptor that is present in many household products, has been linked to obesity, cancer, and, most relevant here, childhood neurological disorders such as anxiety and hyperactivity. However, how BPA exposure translates into these neurodevelopmental disorders remains poorly understood. Here, we used zebrafish to link BPA mechanistically to disease etiology. Strikingly, treatment of embryonic zebrafish with very low-dose BPA (0.0068 μM, 1,000-fold lower than the accepted human daily exposure) and bisphenol S (BPS), a common analog used in BPA-free products, resulted in 180% and 240% increases, respectively, in neuronal birth (neurogenesis) within the hypothalamus, a highly conserved brain region involved in hyperactivity. Furthermore, restricted BPA/BPS exposure specifically during the neurogenic window caused later hyperactive behaviors in zebrafish larvae. Unexpectedly, we show that BPA-mediated precocious neurogenesis and the concomitant behavioral phenotype were not dependent on predicted estrogen receptors but relied on androgen receptor-mediated up-regulation of aromatase. Although human epidemiological results are still emerging, an association between high maternal urinary BPA during gestation and hyperactivity and other behavioral disturbances in the child has been suggested. Our studies here provide mechanistic support that the neurogenic period indeed may be a window of vulnerability and uncovers previously unexplored avenues of research into how endocrine disruptors might perturb early brain development. Furthermore, our results show that BPA-free products are not necessarily safer and support the removal of all bisphenols from consumer merchandise.
Collapse
Affiliation(s)
- Cassandra D Kinch
- Departments of Biological Sciences and Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Kingsley Ibhazehiebo
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Joo-Hyun Jeong
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| | | | - Deborah M Kurrasch
- Medical Genetics and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada T2N 4N1
| |
Collapse
|
14
|
Gürer B, Kertmen H, Kasim E, Yilmaz ER, Kanat BH, Sargon MF, Arikok AT, Ergüder BI, Sekerci Z. Neuroprotective effects of testosterone on ischemia/reperfusion injury of the rabbit spinal cord. Injury 2015; 46:240-8. [PMID: 25467821 DOI: 10.1016/j.injury.2014.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/25/2014] [Accepted: 11/03/2014] [Indexed: 02/02/2023]
Abstract
AIM Previous studies demonstrated the neuroprotective effects of testosterone, but no previous study has examined the neuroprotective effects of testosterone on spinal cord ischemia/reperfusion injury. The purpose of this study was to evaluate whether testosterone could protect the spinal cord from ischemia/reperfusion injury. METHODS Rabbits were randomised into four groups of eight animals as follows: group 1 (control), group 2 (ischemia), group 3 (methylprednisolone) and group 4 (testosterone). In the control group only a laparotomy was performed. In all other groups, the spinal cord ischemia model was created by the occlusion of the aorta just caudal to the renal artery. Levels of malondialdehyde and catalase were analysed, as were the activities of caspase-3, myeloperoxidase, and xanthine oxidase. Histopathological and ultrastructural evaluations were performed. Neurological evaluation was performed with the Tarlov scoring system. RESULTS After ischemia-reperfusion injury, increases were found in caspase-3 activity, myeloperoxidase activity, malondialdehyde levels, and xanthine oxidase activity. In contrast, decreases in catalase levels were observed. After the administration of testosterone, decreases were observed in caspase-3 activity, myeloperoxidase activity, malondialdehyde levels, and xanthine oxidase activity, whereas catalase levels increased. Furthermore, testosterone treatment showed improved results concerning histopathological scores, ultrastructural score and Tarlov scores. CONCLUSIONS Our results revealed for the first time that testosterone exhibits meaningful neuroprotective activity following ischemia-reperfusion injury of the spinal cord.
Collapse
Affiliation(s)
- Bora Gürer
- Ministry of Health, Fatih Sultan Mehmet Education and Research Hospital, Neurosurgey Clinic, Istanbul, Turkey.
| | - Hayri Kertmen
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Emin Kasim
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Erdal Resit Yilmaz
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| | - Burhan Hakan Kanat
- Ministry of Health, Elazig Education and Research Hospital, General Surgery Clinic, Elazig, Turkey
| | - Mustafa Fevzi Sargon
- Hacettepe University, Faculty of Medicine, Department of Anatomy, Ankara, Turkey
| | - Ata Türker Arikok
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Department of Pathology, Ankara, Turkey
| | - Berrin Imge Ergüder
- Ankara University, Faculty of Medicine, Department of Biochemistry, Ankara, Turkey
| | - Zeki Sekerci
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Neurosurgey Clinic, Ankara, Turkey
| |
Collapse
|
15
|
Pallarés ME, Antonelli MC. Hormonal modulation of catecholaminergic neurotransmission in a prenatal stress model. ADVANCES IN NEUROBIOLOGY 2015; 10:45-59. [PMID: 25287535 DOI: 10.1007/978-1-4939-1372-5_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Our laboratory has a long-standing interest in the effects of prenatal stress (PS) on various neurotransmitter pathways and the morphology of the developing brain as well as in behavioural aspects of the offspring. Employing a commonly used PS paradigm in which the dams were subjected to restraint stress during the last week of gestation, we observed that several of these pathways were altered in the offspring brain. In this chapter, we will summarize and discuss the results obtained with the main catecholaminergic pathways, namely dopamine (DA) and norepinephrine (NE). In our hands, PS produces an increase in dopamine D2-type receptors in limbic areas, a decreased DA release after amphetamine stimulation in prefrontal cortex (PFC) and an increase in NE release in the same area of the adult offspring brain. In addition, DA uptake is altered at prepubertal stages that persist through adulthood. However, the expression of the step-limiting enzyme of the DA synthesis, tyrosine hydroxylase (TH), is only impaired at early stages of development after PS in the neuronal bodies. At the nuclear regulation level, dopaminergic transcription factors Nurr1 and Ptx3 showed a high vulnerability to PS showing changes along the lifespan. It was striking to observe that many impairments observed in most of these pathways differed depending on whether they were tested before or after puberty indicating a particular sensitivity of the systems to variations in gonadal hormones peaks. In fact, we observed that PS induced long-term effects on the male offspring reproductive system and spermatogenesis development, particularly by inducing a long-term imbalance of circulating sexual hormone levels. Our findings suggest that PS exerts long-term effects on various neurotransmitter pathways altering the normal connectivity between brain areas. Since the developing forebrain was shown to be influenced by androgen exposure, and PS was shown to disrupt prenatal testosterone surges, our results suggest that prenatal insults might be affecting the organizational role of androgens during brain development and differentially modulating their activational role during pubertal brain maturation.
Collapse
Affiliation(s)
- María Eugenia Pallarés
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
16
|
Reddy R, Estill C, Meaker M, Stormshak F, Roselli CE. Sex differences in expression of oestrogen receptor α but not androgen receptor mRNAs in the foetal lamb brain. J Neuroendocrinol 2014; 26:321-8. [PMID: 24730418 PMCID: PMC4071168 DOI: 10.1111/jne.12152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 03/12/2014] [Accepted: 03/16/2014] [Indexed: 11/26/2022]
Abstract
Gonadal steroid hormones play important roles during critical periods of development to organise brain structures that control sexually dimorphic neuroendocrine responses and behaviours. Specific receptors for androgens and oestrogens must be expressed at appropriate times during development to mediate these processes. The present study was performed to test for sex differences in the relative expression of oestrogen receptor (ER)α and androgen receptor (AR) mRNA during the window of time in gestation that is critical for behavioural masculinisation and differentiation of the ovine sexually dimorphic nucleus (oSDN) in the sheep. In addition, we examined whether ERα and AR mRNA expression is localised within the nascent oSDN and could be involved in its development. Using the quantitative real-time polymerase chain reaction, we found that females expressed more ERα mRNA than males in medial preoptic area and medial basal hypothalamus during the mid-gestational critical period for brain sexual differentiation. No sex differences were found for AR mRNA in any tissue examined or for ERα in amygdala and frontal cortex. Using radioactive in situ hybridisation, we found that the distributions of ERα and AR mRNA overlapped with aromatase mRNA, which delineates the boundaries of the developing oSDN and identifies this nucleus as a target for both androgens and oestrogens. These data demonstrate that the transcriptional machinery for synthesising gonadal steroid receptors is functional in the foetal lamb brain during the critical period for sexual differentiation and suggest that possible mechanisms for establishing dimorphisms controlled by gonadal steroids may exist at the level of steroid hormone receptor expression.
Collapse
Affiliation(s)
- Radhika Reddy
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239-3098
| | - Charles Estill
- College of Veterinary Medicine Oregon State University, Corvallis, OR 97331-4501
| | - Mary Meaker
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
| | - Fred Stormshak
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
| | - Charles E. Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239-3098
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
| |
Collapse
|
17
|
Pan M, Zhang C. Stimulatory effect of gonadal hormones on fetal rat hippocampal neural proliferation requires neurotrophin receptor activation in vitro. Neurosci Lett 2013; 546:1-5. [DOI: 10.1016/j.neulet.2013.04.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/26/2013] [Accepted: 04/12/2013] [Indexed: 11/15/2022]
|
18
|
Regional distribution of 5α-reductase type 2 in the adult rat brain: an immunohistochemical analysis. Psychoneuroendocrinology 2013; 38:281-93. [PMID: 22776423 PMCID: PMC3762250 DOI: 10.1016/j.psyneuen.2012.06.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/18/2012] [Accepted: 06/18/2012] [Indexed: 11/20/2022]
Abstract
The enzyme 5α-reductase (5αR) catalyzes the conversion of testosterone and other Δ(4)-3-ketosteroids into their 5α-reduced metabolites. Of the five members of the 5αR family, the type 2 enzyme (5αR2) plays a key role in androgen metabolism, and is abundantly distributed in the urogenital system. Although 5αR2 has been reported to be highly expressed in the brain during early developmental stages, little is currently known on its anatomical and cellular distribution in the adult brain. Thus, the present study was designed to determine the detailed localization of 5αR2 in the adult rat brain, using a highly specific polyclonal antibody against this isoform. Parasagittal and coronal sections revealed 5αR2 immunoreactivity throughout most brain regions, with strong immunolabeling in the layers III and VI of the prefrontal and somatosensory cortex, olfactory bulb, thalamic nuclei, CA3 field of hippocampus, basolateral amygdala and Purkinje cell layer of cerebellum. Lower 5αR2 levels were detected in the hypothalamus and midbrain. Moreover, double labeling fluorescence with confocal laser scanning microscopy (CLSM) revealed that 5αR2 is localized in neurons, but not in glial cells. Specifically, the enzyme was documented in the pyramidal neurons of the cortex by CLSM analysis of simultaneous Golgi-Cox and immunofluorescent staining. Finally, low levels of 5αR2 expression were identified in GABAergic cells across the cortex, hippocampus and striatum. These findings show that, in the adult brain, 5αR2 is distributed in critical regions for behavioral regulation, suggesting that the functional role of this isoform is present throughout the entire lifespan of the individual.
Collapse
|
19
|
Burkhardt MS, Foster JK, Martins RN. The Significance of Age-Related Androgen Depletion in Cognitive Impairment: A Review. BRAIN IMPAIR 2012. [DOI: 10.1375/brim.5.2.166.58254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractThe potential role of supplementing sex steroids for the prevention and delay of age-related cognitive decline has received a great deal of recent interest. Although the biological plausibility of hormone treatment has received considerable support, clinical studies of cognitive functioning after hormonal treatment in postmenopausal women with and without dementia have produced mixed results. Much less attention has been given to the corresponding role of androgens in men. In order to establish the relevance of hormonal supplementation for men in delaying or preventing cognitive decline, it is of importance to evaluate both adrenal and gonadal contributions to androgen status. Additionally, consideration must also be given to the potential interactions of androgens with risk and protective factors (e.g., apolipoprotein E genotype and education). Here we review experimental and epidemiological studies of the significance of androgens for cognitive function.
Collapse
|
20
|
Aloisi AM, Ceccarelli I, Fiorenzani P, Maddalena M, Rossi A, Tomei V, Sorda G, Danielli B, Rovini M, Cappelli A, Anzini M, Giordano A. Aromatase and 5-alpha reductase gene expression: modulation by pain and morphine treatment in male rats. Mol Pain 2010; 6:69. [PMID: 20977699 PMCID: PMC2978140 DOI: 10.1186/1744-8069-6-69] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/26/2010] [Indexed: 01/18/2023] Open
Abstract
Background The steroid hormone testosterone has been found to be greatly reduced by opioids in different experimental and clinical conditions. The purpose of this study on male rats was to determine the effects of a single injection of morphine (5 mg/Kg) on persistent pain (formalin test) and the single or combined effects on p450-aromatase and 5-alpha reductase type 1 mRNA expression in the brain, liver and testis. Testosterone was determined in the plasma and in the brain, morphine was assayed in the plasma. Results In the morphine-treated rats, there were increases of 5-alpha reductase mRNA expression in the liver and aromatase mRNA expression in the brain and gonads. Morphine was detected in the blood of all morphine-treated rats even though there were no clear analgesic affects in the formalin-treated animals three hours after treatment. Testosterone was greatly reduced in the plasma and brain in morphine-treated subjects. Conclusions It appears that morphine administration can induce long-lasting genomic effects in different body areas which contribute to the strong central and peripheral testosterone levels. These changes were not always accompanied by behavioral modifications.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- Department of Physiology, Neuroscience and Applied Physiology Unit, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Swamydas M, Bessert D, Skoff R. Sexual dimorphism of oligodendrocytes is mediated by differential regulation of signaling pathways. J Neurosci Res 2010; 87:3306-19. [PMID: 19084904 DOI: 10.1002/jnr.21943] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Sexual dimorphism of white matter has not been considered important, the assumption being that sex hormones are not essential for glial development. We recently showed exogenous hormones in vivo differentially regulate in male and female rodents the life span of oligodendrocytes (Olgs) and amount of myelin (Cerghet et al. [2006] J. Neurosci. 26:1439-1447). To determine which hormones regulate male and female Olg development, we prepared enriched Olg cultures grown in serum-free medium with estrogen (E2), progesterone (P2), and dihydrotestosterone (DHT) or their combinations. P2 significantly increased the number of Olgs in both sexes, but more so in females; E2 had minor effects on Olg numbers; and DHT reduced Olgs numbers in both sexes, but more so in females. Combinations of hormones affected Olg numbers differently from single hormones. The change in Olg numbers was due to changes not in proliferation but rather in survival. P2 increased pAKT by many-fold, but MAPK levels were unchanged, indicating that activation of the Akt pathway by P2 is sufficient to regulate Olg differentiation. DHT reduced pAkt in both sexes but differentially increased pMAPK in males and decreased it in females. Stressing Olgs reveals that both sexes are protected by P2, but females are slightly better protected than males. Females always showed greater differences than males regarding changes in Olg numbers and in signaling molecules. Given the greater fluctuation of neurosteroids in women than in men and the higher incidence of multiple sclerosis (MS) in women, these sexually dimorphic differences may contribute to differences in male and female MS lesions.
Collapse
Affiliation(s)
- Muthulekha Swamydas
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | |
Collapse
|
22
|
Vu TT, Hirst JJ, Stark M, Wright IMR, Palliser HK, Hodyl N, Clifton VL. Changes in human placental 5alpha-reductase isoenzyme expression with advancing gestation: effects of fetal sex and glucocorticoid exposure. Reprod Fertil Dev 2009; 21:599-607. [PMID: 19383266 DOI: 10.1071/rd08224] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 02/16/2009] [Indexed: 01/17/2023] Open
Abstract
5alpha-reduced steroids, including allopregnanolone, suppress neuronal activity and can have neuroprotective effects in the fetus. 5alpha-reductases in the placenta may contribute precursors to brain allopregnanolone synthesis. Preterm birth and glucocorticoids, administered for fetal lung maturation or for maternal asthma, may influence reductase expression. The aims of the present study were to evaluate placental 5alpha-reductase isoform expression during late gestation and to examine fetal sex differences and the effects of glucocorticoid therapies on the expression of these enzymes. Expression of the two 5alpha-reductase isoenzymes was measured in placental samples, whereas cortisol concentrations were measured in cord blood, from two cohorts. The first cohort consisted of women who delivered preterm and received betamethasone treatment (n=41); the second cohort consisted of women who delivered at term and were either healthy controls (n=30) or asthmatics who had used glucocorticoids (n=24). Placental expression of both isoenzymes increased with advancing gestation and there were marked sex differences in levels of 5alpha-reductase I (P<0.05), but not of 5alpha-reductase II. The expression of both enzymes was positively correlated with cortisol levels (P<0.05), but there was no effect of recent glucocorticoid exposure. These findings suggest that the preterm neonate may have lower developmental exposure to 5alpha-reduced steroids and may lack steroid-mediated neuroprotection depending on fetal sex.
Collapse
Affiliation(s)
- Thi T Vu
- Mothers and Babies Research Centre and School of Biomedical Sciences, John Hunter Hospital Campus, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | | | | | |
Collapse
|
23
|
Negri-Cesi P, Colciago A, Pravettoni A, Casati L, Conti L, Celotti F. Sexual differentiation of the rodent hypothalamus: hormonal and environmental influences. J Steroid Biochem Mol Biol 2008; 109:294-9. [PMID: 18403198 DOI: 10.1016/j.jsbmb.2008.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Brain sexual differentiation is a complex developmental phenomenon influenced by the genetic background, sex hormone secretions and environmental inputs, including pollution. The main hormonal drive to masculinize and defeminize the rodent brain is testosterone secreted by the testis. The hormone does not influence sex brain differentiation only in its native configuration, but it mostly needs local conversion into active metabolites (estradiol and DHT) through the action of specific enzymatic systems: the aromatase and 5alpha-reductase (5alpha-R), respectively. This allows the hormone to control target cell gene expression either through the estrogen (ER) or the androgen (AR) receptors. The developmental profile of testosterone metabolizing enzymes, different in the two sexes, is therefore of the utmost importance in affecting the bioavailability of the steroids active in brain differentiation. Widely diffused pollutants, like polychlorinated biphenyls (PCBs) are able to affect the production and/or action of testosterone metabolites, exerting detrimental influences on reproduction and sex behavior. The main studies performed in our and other laboratories concerning the pattern of expression and the control of the enzymatic systems involved in brain androgen action and metabolism are shortly reviewed. Some recent data on the influence exerted by PCBs on these metabolic systems are also reported.
Collapse
Affiliation(s)
- Paola Negri-Cesi
- Department of Endocrinology, University of Milano, via Balzaretti 9, 20133 Milano, Italy.
| | | | | | | | | | | |
Collapse
|
24
|
Bodo C. A role for the androgen receptor in the sexual differentiation of the olfactory system in mice. BRAIN RESEARCH REVIEWS 2008; 57:321-31. [PMID: 17915335 PMCID: PMC2348186 DOI: 10.1016/j.brainresrev.2007.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 07/10/2007] [Accepted: 08/11/2007] [Indexed: 10/22/2022]
Abstract
Olfactory signals play a central role in the identification of a mating partner in rodents, and the behavioral response to these cues varies markedly between the sexes. As several other sexually dimorphic traits, this response is thought to differentiate as a result of exposure of the developing individual to gonadal steroids, but both the identity of the specific steroid signal and the neural structures targeted for differentiation on this particular case are largely unknown. The present review summarizes results obtained in our lab using genetic males affected by the testicular feminization syndrome (Tfm) as experimental model, and that led to the identification of a role for non-aromatized gonadal steroids acting through the androgen receptor (AR) in the differentiation of olfactory cues processing in mice. The existing literature about AR-mediated sexual differentiation of the CNS in animal models is discussed, along with potential targets for the action of non-aromatized gonadal steroids in either one of the subsystems that detect and process olfactory information in rodents.
Collapse
Affiliation(s)
- Cristian Bodo
- Graduate Program in Neuroscience, 1300 Jefferson Park Avenue, Room 1229, Jordan Hall, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
25
|
Rosario ER, Pike CJ. Androgen regulation of beta-amyloid protein and the risk of Alzheimer's disease. ACTA ACUST UNITED AC 2007; 57:444-53. [PMID: 17658612 PMCID: PMC2390933 DOI: 10.1016/j.brainresrev.2007.04.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/25/2007] [Accepted: 04/27/2007] [Indexed: 01/08/2023]
Abstract
Advancing age is the most significant risk factor for the development of Alzheimer's disease (AD), however the age-related changes that underlie this effect remain unclear. In men, one normal consequence of aging is a robust decline in circulating and brain levels of the sex steroid hormone testosterone. Testosterone depletion leads to functional impairments and increased risk of disease in androgen-responsive tissues throughout the body, including brain. In this review we discuss the relationship between age-related testosterone depletion and the development of AD. Specifically, we focus on androgen regulation of beta-amyloid protein (Abeta), the accumulation of which is a key initiating factor in AD pathogenesis. Emerging data suggest that the regulatory actions of androgens on both Abeta and the development of AD support consideration of androgen therapy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Emily R Rosario
- Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | | |
Collapse
|
26
|
Nguyen TVV, Yao M, Pike CJ. Flutamide and cyproterone acetate exert agonist effects: induction of androgen receptor-dependent neuroprotection. Endocrinology 2007; 148:2936-43. [PMID: 17347309 DOI: 10.1210/en.2006-1469] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Androgens can exert profound effects on the organization, development, and function of the nervous system through activation of androgen receptors (ARs). Nonsteroidal and steroidal antiandrogens antagonize AR-mediated, classic genomic actions of androgens. However, emerging studies in nonneuronal cells indicate that antiandrogens can act as partial agonists for the AR. Here we investigated the effects of the antiandrogens flutamide and cyproterone acetate on neuroprotection induced by dihydrotestosterone (DHT). We observed that, although flutamide and cyproterone acetate blocked androgen-induced gene expression, they failed to inhibit DHT protection against apoptotic insults in cultured hippocampal neurons. Interestingly, flutamide and cyproterone acetate alone, like DHT, significantly reduced apoptosis. Furthermore, the protective actions of flutamide and cyproterone acetate were observed specifically in AR-expressing cell lines, suggesting a role for AR in the agonist effects of antiandrogens. Our results indicate that, in contrast to the classic antiandrogen properties of flutamide and cyproterone acetate, these AR modulators display agonist activities at the level of neuroprotection. These findings provide new insight into the agonist vs. antagonist properties of antiandrogens, information that will be crucial to understanding the neural implications of clinically used AR-modulating drugs.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- University of Southern California, Davis School of Gerontology, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | | | | |
Collapse
|
27
|
Ciofi P, Lapirot OC, Tramu G. An androgen-dependent sexual dimorphism visible at puberty in the rat hypothalamus. Neuroscience 2007; 146:630-42. [PMID: 17395386 DOI: 10.1016/j.neuroscience.2007.02.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Revised: 01/24/2007] [Accepted: 02/06/2007] [Indexed: 11/18/2022]
Abstract
Morphological studies in rodents have well documented the masculinization of the perinatal brain by estradiol derived from aromatized testosterone, and the resulting irreversible quantitative sex-differences generated in cell numbers or expression of chemical phenotypes. Here, using immunohistochemistry, we explored how this applies to the postnatal development and masculinization of the neurokinin B (NKB)-containing system of the arcuate nucleus/median eminence complex (ARC/ME). In adult rats, NKB-immunoreactive neurons exhibit an unusual, qualitative sexual dimorphism of their ventral axonal projections: to the neuropil in females, to capillary vessels in males. In adults, there was no sex-difference in the numbers of NKB-immunoreactive perikarya or capillary vessels in the ARC/ME, suggesting that this sexual dimorphism cannot be explained by the existence of supernumerary structures. At birth (day 0) the NKB system was immature in both sexes, and while its adult features emerged progressively until puberty in females, they did not develop before puberty (day 40) in males, revealing a sexual dimorphism only late postnatally. When males were orchidectomized at day 30, the masculine distribution of NKB-immunoreactive axons expected at day 40 was not seen, while it was apparent after chronic treatment with testosterone or dihydrotestosterone, suggesting a testicular masculinizing action via androgen receptors at puberty. Moreover in these prepubertal-orchidectomized males, the distribution of NKB-immunoreactive axons was surprisingly feminized by chronic estradiol alone, suggesting that NKB neurons are not irreversibly programmed before puberty. Last, in adult females, the distribution of NKB-immunoreactive axons was feminine 30 days after ovariectomy, and it was masculinized after concurrent chronic dihydrotestosterone, suggesting that NKB neurons remain responsive to androgens late in reproductive life. Thus, the sexual differentiation of the hypothalamus proceeds well beyond the perinatal period and includes the epigenetic action of non-aromatizable androgens upon subsets of neurons that have retained bipotent features.
Collapse
Affiliation(s)
- P Ciofi
- Inserm U862, F-33077 Bordeaux, France; Université Victor Ségalen Bordeaux 2, F-33077 Bordeaux, France.
| | | | | |
Collapse
|
28
|
Pike CJ, Rosario ER, Nguyen TVV. Androgens, aging, and Alzheimer's disease. Endocrine 2006; 29:233-41. [PMID: 16785599 DOI: 10.1385/endo:29:2:233] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Revised: 10/24/2005] [Accepted: 10/24/2005] [Indexed: 01/03/2023]
Abstract
Testosterone depletion is a normal consequence of aging in men that is associated with senescent effects in androgen- responsive tissues. We discuss new evidence that one consequence of testosterone depletion in men is an increased risk for the development of Alzheimer's disease (AD). Furthermore, we discuss two candidate mechanisms by which testosterone may affect AD pathogenesis. First, testosterone has been identified as an endogenous regulator of beta-amyloid, a protein that abnormally accumulates in AD brain and is implicated as a causal factor in the disease. Second, findings from several different paradigms indicate that testosterone has both neurotrophic and neuroprotective functions. These new findings support the clinical evaluation of androgen-based therapies for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Christian J Pike
- Andrus Gerontology Center, University of Southern California, Los Angeles, CA 90089-0191, USA.
| | | | | |
Collapse
|
29
|
Zhao H, Tian ZZ, Chen BY. Electroacupuncture stimulates hypothalamic aromatization. Brain Res 2005; 1037:164-70. [PMID: 15777765 DOI: 10.1016/j.brainres.2005.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2004] [Revised: 12/22/2004] [Accepted: 01/01/2005] [Indexed: 11/18/2022]
Abstract
We have previously reported that the repeated electroacupuncture (EA) stimulation significantly increased the concentrations of circulating estradiol and restored the depressed function of the hypothalamus-pituitary-ovary axis (HPOA) in ovariectomized (OVX) rats. We hypothesize that extragonadal aromatization in specific brain areas might be responsible for these changes. Thus, various assays, including radiometric assay, Western blot, and reverse transcriptase polymerase chain reaction (RT-PCR), were employed to determine the aromatization in the hypothalamus of rats that received both OVX and electroacupuncture (OVX + EA). The results showed that EA significantly increased the aromatase activity as well as the expressions of its mRNA and protein (P < 0.05) in the OVX rats. These results suggest that EA enhances brain aromatization, which might contribute to influence the function of gonadotropin-releasing hormone (GnRH) neurons and promote the hypofunction of the HPOA in the ovariectomized rats.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research, Shanghai Medical Center of Fudan University, PO Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P.R. China
| | | | | |
Collapse
|
30
|
Zhao H, Tian Z, Hao J, Chen B. Extragonadal aromatization increases with time after ovariectomy in rats. Reprod Biol Endocrinol 2005; 3:6. [PMID: 15661083 PMCID: PMC548297 DOI: 10.1186/1477-7827-3-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2004] [Accepted: 01/21/2005] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The circulating estrogen concentration elevated gradually along with time after ovariectomy in rats. To explore the source of the increased circulation estrogen, the extragonadal aromatization as well as the synthesis of androgen in the adrenal cortex of the ovariectomized rats was evaluated. METHODS Female rats were divided into twelve groups: 1 month after ovariectomy (OVX1M), OVX2M, OVX3M, OVX4M, OVX5M, OVX6M; intact 1 month (INT1M), INT2M, INT3M, INT4M, INT5M, INT6M. The blood concentration of testosterone (T) was measured by radioimmunoassay. The mRNA expressions of P450 aromatase in the liver and subcutaneous abdominal (SA) adipose as well as the adrenal cytochrome P450 17 alpha hydroxylase/lyase (P450c17) were semiquantified by RT-PCR. The P450 aromatase protein expressions in the liver and SA adipose were detected by Western blot. RESULTS The blood E2 concentrations increased gradually along with time after ovariectomy in the rats. The 58-kDa aromatase protein and mRNA expressions normalized to beta-actin in the OVX6M rats' SA adipose tissues showed higher levels than those from corresponding tissues in the INT6M (p < 0.05). And the ratios of aromatase mRNA and protein to beta-actin in the OVX6M rats' liver tissues increased significantly compared with those in the OVX1M rats (p < 0.05). The ratio of adrenal P450c17 to beta-actin in the OVX6M increased markedly, and was higher than OVX1M (p < 0.05), though the blood concentration of T decreased significantly in all the ovariectomized rats (p < 0.05). CONCLUSION Both the subcutaneous abdominal adipose tissues and the liver tissues contributed to the extragonadal aromatisation to promote the circulating E2 levels in the rats along with time after ovariectomy; the adrenal compensation might also be activated naturally.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Centre for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Zhanzhuang Tian
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Centre for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Junwei Hao
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Centre for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Boying Chen
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Centre for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| |
Collapse
|
31
|
Abizaid A, Mezei G, Sotonyi P, Horvath TL. Sex differences in adult suprachiasmatic nucleus neurons emerging late prenatally in rats. Eur J Neurosci 2004; 19:2488-96. [PMID: 15128402 DOI: 10.1111/j.0953-816x.2004.03359.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The suprachiasmatic nucleus (SCN) is implicated in the control of circadian rhythms of gonadal function. Although several structures surrounding the SCN are sensitive to the effects of gonadal steroids, similar effects in the SCN remain unclear. For example, there are conflicting data on whether the SCN is sexually differentiated. This study attempted to determine sex differences in the number of SCN cells generated during late gestation, and if testosterone mediates these differences. Pregnant female rats were treated with 5-bromo-2'-deoxyuridine (BrdU; 50 mg/kg) on gestational day 18 (E18), the day when aromatase activity peaks in the developing rat fetus. These animals were also given injections of oil or testosterone propionate (10 mg/0.1 mL peanut oil) from E15 until parturition. Litters were allowed to survive until adulthood and were killed on postnatal day 60 (PN60). Following fixation, brain sections containing the SCN from these rats were processed for BrdU immunocytochemistry. A second set of SCN sections was processed for immunocytochemistry detecting BrdU and some of the cell groups prevalent within the SCN. Data showed that female rats have a higher number of cells labeled with BrdU in the SCN, particularly in the medial and caudal SCN. This sex difference was abolished in animals treated with testosterone during late gestation. Double immunocytochemistry revealed that BrdU-labeled cells were neurons expressing calbindin-D28K, vasoactive intestinal peptide and, to a lesser degree, vasopressin. Our results unveiled a previously unknown effect of gonadal steroids on the developing SCN, which may contribute to the emergence of gender-specific circadian rhythms.
Collapse
Affiliation(s)
- Alfonso Abizaid
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
32
|
Zhao H, Tian Z, Cheng L, Chen B. Electroacupuncture enhances extragonadal aromatization in ovariectomized rats. Reprod Biol Endocrinol 2004; 2:18. [PMID: 15113414 PMCID: PMC411058 DOI: 10.1186/1477-7827-2-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Accepted: 04/27/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Repeated electroacupuncture (EA) stimulation is known to stimulate the activity of the hypothalamus-pituitary-adrenal axis, and to enhance the circulation level of estrogen in the ovariectomized rats. To explore the source of the increased circulation estrogen, the extragonadal aromatization was detected. METHODS Female rats were divided into five groups: 1) intact (INT), 2) intact with EA in specific points (INT+EA), 3) ovariectomized (OVX), 4) ovariectomized with EA in specific points (OVX+EA) and 5) ovariectomized with EA in non-specific points (OVX+C). Radiometric assay, Western blot and RT-PCR were adopted to determine the extragonadal aromatization in subcutaneous abdominal (SA) adipose and liver tissues of rats. The blood concentrations of estrogen, testosterone and corticosterone were measured by radioimmunoassay. RESULTS The aromatase activities of the SA adipose and liver tissues in the OVX+EA rats increased significantly (p < 0.01) compared with those in the INT, INT+EA and OVX rats. The 58-kDa aromatase protein and aromatase mRNA expressions normalized to beta-actin in the OVX+EA rats' SA adipose tissues showed higher levels than those from corresponding tissues in the INT and INT+EA rats (p < 0.05). And the ratios of aromatase mRNA and protein to beta-actin in the OVX+EA rats' liver tissues increased significantly compared with those in the OVX rats (p < 0.05). Furthermore, blood estrogen and corticosterone concentrations showed significant increase in the OVX+EA rats compared with the concentrations in the OVX and OVX+C rats (p < 0.05), but no statistical disparity occurred on the blood testosterone concentrations between the OVX+EA rats and the OVX ones. CONCLUSION Both the subcutaneous abdominal adipose and the liver tissues contributed to the effects of electroacupuncture on the extragonadal aromatization to promote the blood concentrations of estrogen in the ovariectomized rats.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Zhanzhuang Tian
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Lina Cheng
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| | - Boying Chen
- Department of Neurobiology and Integrative Medicine, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine, Research Department of Acupuncture), Shanghai Medical College of Fudan University (Formerly Shanghai Medical University), P.O. Box 291, 138 Yi-Xue-Yuan Road, 200032 Shanghai, P. R. China
| |
Collapse
|
33
|
Greenland KJ, Rekaris G, MacLean HE, Warne GL, Zajac JD. Application of differential display in the identification of androgen-regulated genes. Endocr Res 2004; 30:69-82. [PMID: 15098921 DOI: 10.1081/erc-120029886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Identification of androgen-regulated genes in neurons is an important step in understanding the mechanisms involved in androgen action. The aim of the current study was to identify androgen-responsive genes in the neural cells using the technique of differential display reverse transcription polymerase chain reaction (DDRT-PCR) on the human neuroblastoma cell line, SK-N-MC. Using this analysis, 18 putatively androgen-regulated cDNA species were identified, ranging in size from 280 to 800 bp. Of these, 14 were found to be negatively regulated and 4 positively regulated by androgens. Only 12 were successfully re-amplified, and of these, 8 were found to contain multiple species of cDNA fragments. When Northern analysis was conducted using the 21 different cDNA fragments as probes, only one was found to confirm the androgen regulation demonstrated via DDRT-PCR. While this putatively regulated gene remains to be fully characterized, future studies of may provide insights into the molecular mechanisms governing androgen action in neural cells.
Collapse
Affiliation(s)
- Karen J Greenland
- Centre for Hormone Research, Murdoch Children's Research Institute, University of Melbourne, Royal Children's Hospital, Victoria, Australia
| | | | | | | | | |
Collapse
|
34
|
Ammini AC, Gupta R, Kapoor A, Karak A, Kriplani A, Gupta DK, Kucheria K. Etiology, clinical profile, gender identity and long-term follow up of patients with ambiguous genitalia in India. J Pediatr Endocrinol Metab 2002; 15:423-30. [PMID: 12008689 DOI: 10.1515/jpem.2002.15.4.423] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There is little information on the profile of children with ambiguous genitalia in India. Presented here is an analysis of patients with ambiguous genitalia registered in a general endocrine clinic during the last 2 decades. Seventy-four patients (age 4 months to 36 years) were registered during this period. Fifty-two were more than 5 years old at the time of registration. Thirty-five were reared as females, 29 as males; nine children (4 months to 1 year old) were brought for sex assignment, and one (with epispadias) was brought for correction of urinary incontinence. Investigations revealed 28 patients with congenital adrenal hyperplasia, 14 dysgenetic male pseudohermaphroditism, ten true hermaphroditism, six partial androgen insensitivity, four castration and one epispadias. There were eight patients with perineal hypospadias with normal Leydig cell reserve (normal LH, FSH and testosterone response to LHRH). Sex of rearing and gender identity were concordant in all except the patients with perineal hypospadias with normal Leydig cell response. These observations support the theory that prenatal androgen exposure masculinizes the brain.
Collapse
Affiliation(s)
- A C Ammini
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi.
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Accumulating evidence suggests that testosterone has neurotrophic and perhaps neuroprotective actions. Thus, age-related depletion of testosterone may increase the brain's vulnerability to Alzheimer's disease and related disorders. To begin investigating this issue, cultured neurons were exposed to the Alzheimer-related insult beta-amyloid in the presence of testosterone. beta-Amyloid neurotoxicity was significantly reduced by testosterone via a rapid, estrogen-independent mechanism. These data may provide additional insight into the treatment of age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- C J Pike
- Andrus Gerontology Center, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA.
| |
Collapse
|
36
|
Roselli CE, Klosterman S, Resko JA. Anatomic relationships between aromatase and androgen receptor mRNA expression in the hypothalamus and amygdala of adult male cynomolgus monkeys. J Comp Neurol 2001; 439:208-23. [PMID: 11596049 DOI: 10.1002/cne.1343] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study mapped the regional locations of cells expressing cytochrome P450 aromatase (P450AROM) and androgen receptor (AR) mRNAs in the adult male macaque hypothalamus and amygdala by in situ hybridization histochemistry using monkey-specific cRNA probes. High densities of P450AROM and AR mRNA-containing neurons were observed in discrete hypothalamic areas involved in the regulation of gonadotropin secretion and reproductive behavior. P450AROM mRNA-containing neurons were most abundant in the medial preoptic nucleus, bed nucleus of the stria terminalis, and anterior hypothalamic area, whereas AR mRNA-containing neurons were most numerous in the ventromedial nucleus, arcuate nucleus, and tuberomamillary nucleus. Moderate to heavily labeled P450AROM mRNA-containing cells were present in the cortical and medial amygdaloid nuclei, which are known to have strong reciprocal inputs with the hypothalamus. Heavily labeled P450AROM mRNA-containing cells were found in the accessory basal amygdala nucleus, which projects to the cingulate cortex and hippocampus, areas that are important in the expression of emotional behaviors and memory processing. In contrast to P450AROM, the highest density of AR mRNA labeling in the temporal lobe was associated with the cortical amygdaloid nucleus and the pyramidal cells of the hippocampus. All areas that contained P450AROM mRNA-expressing cells also contained AR mRNA-expressing cells, but there were areas in which AR mRNA was expressed but not P450AROM mRNA. The apparent relative differences in the expression of P450AROM and AR mRNA-containing neurons within the monkey brain suggests that T acts through different signaling pathways in specific brain areas or within different cells from the same region.
Collapse
Affiliation(s)
- C E Roselli
- Department of Physiology & Pharmacology, Oregon Health Science University, Portland, Oregon 97201-3098, USA.
| | | | | |
Collapse
|
37
|
Peterson RS, Saldanha CJ, Schlinger BA. Rapid upregulation of aromatase mRNA and protein following neural injury in the zebra finch (Taeniopygia guttata). J Neuroendocrinol 2001; 13:317-23. [PMID: 11264718 DOI: 10.1046/j.1365-2826.2001.00647.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The expression of aromatase (oestrogen synthase) within the vertebrate central nervous system (CNS) is key in the provision of local oestrogens to neural circuits. Aromatase expression appears to be exclusively neuronal under normal conditions. However, some in vitro studies suggest the presence of astrocytic aromatase in songbirds and mammals. Recently, aromatase in reactive astrocytes has been demonstrated in response to neural injury in the mammalian CNS. Since the glial aromatase expression first documented in cultures of the songbird telencephalon may reflect processes similar to those in response to mammalian neural injury, we investigated whether injury alters the pattern of aromatase-expression in the zebra finch, a species with very high levels of forebrain aromatase expression. Adult males received a penetrating neural injury to the right hemisphere and were killed either 24 or 72 h later. Controls were anaesthetized and otherwise unmanipulated. We determined the expression of aromatase mRNA and protein using in situ hybridization and immunocytochemistry, respectively. Both the transcription and translation of aromatase is dramatically upregulated around the lesion site in response to neural injury in the zebra finch forebrain. This effect is robust and rapid, occurring within 24 h of the injury itself. Cells that upregulate aromatase appear to be reactive astrocytes based upon morphology. The hemisphere contralateral to the injury and both hemispheres in control birds showed the normal, exclusively neuronal pattern of aromatase expression. The upregulation of aromatase in astrocytes may provide high levels of oestrogen available to modulate processes such as CNS repair. Injury-induced upregulation of astrocytic aromatase may be a general characteristic of the injured vertebrate brain.
Collapse
Affiliation(s)
- R S Peterson
- Department of Physiological Science and Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, 621 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
38
|
Roselli CE, Stormshak F, Resko JA. Distribution of aromatase mRNA in the ram hypothalamus: an in situ hybridization study. J Neuroendocrinol 2000; 12:656-64. [PMID: 10849210 DOI: 10.1046/j.1365-2826.2000.00496.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The regional distribution of neurones expressing aromatase mRNA in the ram hypothalamus was examined by in situ hybridization using 33P-labelled cRNA probes. The highest amounts of hybridization signal were observed in the central part of the medial preoptic nucleus and posterior medial part of the bed nucleus of the stria terminalis. Moderate amounts of hybridization signal were observed in the anteroventral periventricular preoptic nucleus, medial preoptic nucleus and a broad band extending between the medial preoptic nucleus and bed nucleus of the stria terminalis. Low levels of hybridization signal were observed in the organum vasculosum of the lamina terminalis, anterior part of the medial preoptic nucleus, and central part of the ventromedial nucleus of the hypothalamus. The presence of aromatase mRNA within neurones of the steroid-sensitive hypothalamic circuit supports a role for aromatization in the mechanism of testosterone action on reproductive function in male sheep. The distribution of aromatase mRNA in the ovine hypothalamus was similar to that described for other vertebrate species, suggesting a high degree of functional conservation across species.
Collapse
Affiliation(s)
- C E Roselli
- Department of Physiology and Pharmacology, School of Medicine, Oregon Health Sciences University, Portland, Oregon 97201-3098, USA.
| | | | | |
Collapse
|
39
|
Kruijver FP, Zhou JN, Pool CW, Hofman MA, Gooren LJ, Swaab DF. Male-to-female transsexuals have female neuron numbers in a limbic nucleus. J Clin Endocrinol Metab 2000; 85:2034-41. [PMID: 10843193 DOI: 10.1210/jcem.85.5.6564] [Citation(s) in RCA: 157] [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
Transsexuals experience themselves as being of the opposite sex, despite having the biological characteristics of one sex. A crucial question resulting from a previous brain study in male-to-female transsexuals was whether the reported difference according to gender identity in the central part of the bed nucleus of the stria terminalis (BSTc) was based on a neuronal difference in the BSTc itself or just a reflection of a difference in vasoactive intestinal polypeptide innervation from the amygdala, which was used as a marker. Therefore, we determined in 42 subjects the number of somatostatin-expressing neurons in the BSTc in relation to sex, sexual orientation, gender identity, and past or present hormonal status. Regardless of sexual orientation, men had almost twice as many somatostatin neurons as women (P < 0.006). The number of neurons in the BSTc of male-to-female transsexuals was similar to that of the females (P = 0.83). In contrast, the neuron number of a female-to-male transsexual was found to be in the male range. Hormone treatment or sex hormone level variations in adulthood did not seem to have influenced BSTc neuron numbers. The present findings of somatostatin neuronal sex differences in the BSTc and its sex reversal in the transsexual brain clearly support the paradigm that in transsexuals sexual differentiation of the brain and genitals may go into opposite directions and point to a neurobiological basis of gender identity disorder.
Collapse
Affiliation(s)
- F P Kruijver
- Graduate School Neurosciences Amsterdam, The Netherlands Institute for Brain Research.
| | | | | | | | | | | |
Collapse
|
40
|
Ryzhavskii BY, Rudman YY. Effect of deoxycorticosterone acetate administration to pregnant rats on the brain of their offspring. Bull Exp Biol Med 2000; 129:330-1. [PMID: 10977908 DOI: 10.1007/bf02439258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/1999] [Indexed: 10/24/2022]
Abstract
Effects of deoxycorticosterone acetate injected to rats on gestation days 18 and 20 on morphometric indexes of the brain and RNA concentration in neurons of neocortical layer V in 1-day-old offspring were studied. The brain in 1-day-old prenatally treated rats was more developed than in the offspring of intact and control females.
Collapse
Affiliation(s)
- B Y Ryzhavskii
- Department of Histology, Far-Eastern State Medical University, Khabarovsk
| | | |
Collapse
|
41
|
Abstract
Androgens exert profound effects on the organization and function of the central nervous system. These effects are mediated by the androgen receptor (AR), a ligand-dependent transcription factor. The mechanisms of AR regulation in neural tissue, however, remain to be fully elucidated. Characterizing this process can provide important information regarding receptor function and AR gene regulation in the brain. Previously, it was shown that testosterone (T) up-regulated neural AR in a dose-dependent manner in both male and female mice. In the present study, whether AR was differentially regulated by the natural agonists T and dihydrotestosterone (DHT) or the nonsteroidal antagonist flutamide (FLU) was assessed. Males were gonadectomized and AR levels were allowed to decline to baseline 3 days after surgery. Changes in AR protein content produced by the various treatments were measured by semiquantitative Western blot of limbic system extracts. Treatment with T or DHT significantly augmented AR 3 and 7 h after hormone administration, but only DHT sustained this increase for 21 h. This difference also was observed when males were given T plus finasteride (FIN, a 5alpha reductase inhibitor). The findings demonstrate that the two endogenous ligands have differential time course effects on neural AR. The antiandrogen FLU failed to up-regulate AR at doses up to 100 times higher than T or DHT. When administered concomitantly with T or DHT, it effectively inhibited the augmentation of AR normally seen 3 h after androgen treatment. While immunohistochemical studies showed that FLU was able to promote nuclear translocation of AR, Western analysis revealed that FLU, in contrast to T and DHT, failed to maintain the integrity of AR. The results demonstrate that (a) the endogenous androgens T and DHT regulate AR differently, suggesting a potential cellular mechanism that may contribute to the difference in neural target gene sensitivity to these androgens; (b) up-regulation of AR occurs only in the presence of agonists; (c) the mechanism of action of FLU in the brain involves inhibition of AR protein up-regulation normally seen in response to androgen; and (d) FLU promotes AR nuclear translocation but not augmentation of cellular AR populations. These findings demonstrate that in vivo AR regulation in the brain basically parallels mechanisms proposed from results obtained with transfected cells and cell lines.
Collapse
Affiliation(s)
- S Lu
- Department of Biological Sciences, 111 Research Drive, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | | | | | | |
Collapse
|
42
|
Garcia-Segura LM, Wozniak A, Azcoitia I, Rodriguez JR, Hutchison RE, Hutchison JB. Aromatase expression by astrocytes after brain injury: implications for local estrogen formation in brain repair. Neuroscience 1999; 89:567-78. [PMID: 10077336 DOI: 10.1016/s0306-4522(98)00340-6] [Citation(s) in RCA: 298] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recent evidence indicates that 17beta-estradiol may have neuroprotective and neuroregenerative properties. Estradiol is formed locally in neural tissue from precursor androgens. The expression of aromatase, the enzyme that catalyses the conversion of androgens to estrogens, is restricted, under normal circumstances, to specific neuronal populations. These neurons are located in brain areas in which local estrogen formation may be involved in neuroendocrine control and in the modulation of reproductive or sex dimorphic behaviours. In this study the distribution of aromatase immunoreactivity has been assessed in the brain of mice and rats after a neurotoxic lesion induced by the systemic administration of kainic acid. This treatment resulted in the induction of aromatase expression by reactive glia in the hippocampus and in other brain areas that are affected by kainic acid. The reactive glia were identified as astrocytes by co-localization of aromatase with glial fibrillary acidic protein and by ultrastructural analysis. No immunoreactive astrocytes were detected in control animals. The same result, the de novo induction of aromatase expression in reactive astrocytes on the hippocampus, was observed after a penetrating brain injury. Furthermore, using a 3H2O assay, aromatase activity was found to increase significantly in the injured hippocampus. These findings indicate that although astrocytes do not normally express aromatase, the enzyme expression is induced in these glial cells by different forms of brain injury. The results suggest a role for local astroglial estrogen formation in brain repair.
Collapse
Affiliation(s)
- L M Garcia-Segura
- MRC Neuroendocrine Development and Behaviour Group, The Barbraham Institute, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
43
|
Küppers E, Beyer C. Expression of aromatase in the embryonic and postnatal mouse striatum. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 63:184-8. [PMID: 9838103 DOI: 10.1016/s0169-328x(98)00279-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Estrogen influences striatal activity and the development of the nigrostriatal system. This study is concerned with the ontogenetic and postnatal expression of aromatase in the mouse striatum. Aromatase activity and mRNA expression were detectable in the embryonic striatum and increased postnatally with no differences between sexes. Aromatase-positive cells were uniformly distributed within the striatum. These data demonstrate that estrogen formation is an intrinsic property of striatal cells and suggest that estrogen may be important for striatal development and function.
Collapse
Affiliation(s)
- E Küppers
- Abteilung Anatomie und Zellbiologie, Universität Ulm, D-89069, Ulm, Germany
| | | |
Collapse
|
44
|
Roselli CE, Klosterman SA. Sexual differentiation of aromatase activity in the rat brain: effects of perinatal steroid exposure. Endocrinology 1998; 139:3193-201. [PMID: 9645693 DOI: 10.1210/endo.139.7.6101] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Androgens regulate aromatase activity in the medial preoptic area and other components of the brain circuit that mediates male sexual behavior. The levels of aromatase activity within these brain regions are greater in males than in females. As the activation of copulation requires aromatization of testosterone to estradiol, this quantitative enzymatic difference between sexes could contribute to the greater behavioral response displayed by males. The present study was designed to test the hypothesis that gender differences in brain aromatase activity of adult rats are dependent on the sexual differentiation of the brain that occurs during perinatal exposure to gonadal hormones. Aromatase activity was measured in vitro in microdissected brain samples using a sensitive radiometric assay. We examined the effect of pre- and postnatal treatment with testosterone propionate or diethylstilbestrol on basal levels and androgen responsiveness of aromatase in adults. In addition, we examined what effect prepubertal gonadectomy exerts on enzyme regulation. Our results demonstrate that perinatal treatments with gonadal hormones that are known to differentiate sexual behavior can completely masculinize the capacity for aromatization in the adult female. The process that differentiates aromatase expression appears to depend on androgen exposure and, in part, local estrogen synthesis, as diethylstilbestrol was able to substitute for testosterone propionate. We also observed that prepubertal gonadectomy reduced the levels of aromatase activity measured in adult brain, suggesting that gonadal hormones that are secreted during puberty may enhance the expression of aromatase activity in adulthood. From this study, we conclude that testosterone and/or its estrogenic metabolites act on the developing brain to determine the gender-specific capacity for aromatization and to regulate androgen responsiveness within components of the neural circuitry that mediates male sexual behavior.
Collapse
Affiliation(s)
- C E Roselli
- Department of Physiology and Pharmacology, Oregon Health Sciences University, Portland 97201-3098, USA.
| | | |
Collapse
|
45
|
Poletti A, Coscarella A, Negri-Cesi P, Colciago A, Celotti F, Martini L. 5 alpha-reductase isozymes in the central nervous system. Steroids 1998; 63:246-51. [PMID: 9618779 DOI: 10.1016/s0039-128x(98)00018-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The enzyme 5 alpha-reductase (5 alpha-R) activates several delta 4-3keto steroids to more potent derivatives which may also acquire new biological actions. Testosterone gives rise to the most potent natural androgen dihydrotestosterone (DHT), and progesterone to dihydroprogesterone (DHP), a precursor of the endogenous anxiolytic/anesthetic steroid tetrahydroprogesterone (THP). Two isoforms of 5 alpha-R, with a limited degree of homology, different biochemical properties and distinct tissue distribution have been cloned: 5 alpha-R type 1 and type 2. In androgen-dependent structures DHT is almost exclusively formed by 5 alpha-R type 2; 5 alpha-R type 1 is widely distributed in the body, with the highest levels in the liver, and may be involved in steroid catabolism. In the brain, the roles of the two isozymes are still largely unknown. This brief review will summarize recent experimental data from our laboratory which try to assign possible functional roles to the process of 5 alpha-reduction, and to the two 5 alpha-R isoforms in the CNS.
Collapse
Affiliation(s)
- A Poletti
- Istituto di Endocrinologia, Università di Milano, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Karolczak M, Küppers E, Beyer C. Developmental expression and regulation of aromatase- and 5alpha-reductase type I mRNA in the male and female mouse hypothalamus. J Neuroendocrinol 1998; 10:267-74. [PMID: 9630396 DOI: 10.1046/j.1365-2826.1998.00200.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Androgen metabolites synthesized by neural aromatase and 5alpha-reductase are implicated in many aspects of mammalian brain development and, in particular, in the masculinization of distinct central nervous system structures and brain functions. The present study was designed to determine (1) the developmental profile of aromatase- and 5alpha-reductase type I mRNA expression in the mouse hypothalamus and (2) to relate ontogenetic sex differences in aromatase activity which have been described in the past to sex-specific aromatase gene expression. In addition, we analysed the effect of androgens on the perinatal regulation of hypothalamic aromatase and 5alpha-reductase type I mRNA expression. By applying semiquantitative reverse transcription-polymerase chain reaction analysis, we found hypothalamic aromatase mRNA expression to be developmentally regulated and to display sex differences at birth and on postnatal day 15 with higher mRNA levels in males. Newborn males and females, which were treated in utero with the androgen receptor antagonist cyproterone actetate, exhibited significantly reduced aromatase mRNA levels compared with untreated controls. In contrast to aromatase, expression levels of hypothalamic 5alpha-reductase mRNA did not reveal a clear-cut developmental profile or sex differences, and no regulatory role for androgens in controlling 5alpha-reductase mRNA expression was found. In conclusion, these results demonstrate perinatal sex differences in hypothalamic aromatase- but not 5alpha-reductase gene expression and suggest that sex differences in perinatal aromatase activity are reflected by corresponding differences in mRNA levels. Androgens are found to control brain estrogen formation pretranslationally at the level of aromatase gene expression. Our findings imply that sex differences in androgen availability and responsiveness are important regulatory factors for aromatase expression in the developing male hypothalamus.
Collapse
Affiliation(s)
- M Karolczak
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany
| | | | | |
Collapse
|
47
|
Melcangi RC, Poletti A, Cavarretta I, Celotti F, Colciago A, Magnaghi V, Motta M, Negri-Cesi P, Martini L. The 5alpha-reductase in the central nervous system: expression and modes of control. J Steroid Biochem Mol Biol 1998; 65:295-9. [PMID: 9699883 DOI: 10.1016/s0960-0760(98)00030-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The present paper will summarize two important aspects of the interactions between steroids and the brain, which have recently been studied in the authors' laboratory. In particular the paper will consider data on: (1) the significance of the two isoforms of the 5alpha-R during brain ontogenesis and development, and (2) the cross-talk between glial and neuronal elements, particularly in relation to the metabolism of sex hormones. (1) The data obtained have shown that the 5alpha-R type 1 enzyme is constitutively expressed in the rat CNS at all stages of brain development. Moreover, the expression of the 5alpha-R type 1 is similar in males and in females, and does not appear to be controlled by androgens. The gene expression of the 5alpha-R type 2 is totally different. This isoform appears to be expressed in the rat brain almost exclusively in the late fetal/early post-natal life and is controlled by testosterone. (2) The present data show that two cell lines derived respectively from a rat glioma (C6 cell line) and from a human astrocytoma (1321N1 cell line) are able to convert testosterone and progesterone into their corresponding 5alpha-reduced metabolites dihydrotestosterone and dihydroprogesterone. The possibility that secretory products of normal and tumoral brain cells might be able to influence steroid metabolism occurring in the two glial cell lines previously mentioned has been considered.
Collapse
Affiliation(s)
- R C Melcangi
- Department of Endocrinology, University of Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Beyer C, Raab H. Nongenomic effects of oestrogen: embryonic mouse midbrain neurones respond with a rapid release of calcium from intracellular stores. Eur J Neurosci 1998; 10:255-62. [PMID: 9753134 DOI: 10.1046/j.1460-9568.1998.00045.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Evidence is emerging that oestrogen, besides acting via classical nuclear receptors, can rapidly influence the physiology of nerve cells through other mechanisms. Oestrogens have been shown to modulate the differentiation and function of embryonic midbrain dopaminergic neurones by stimulating neurite outgrowth, expression of tyrosine hydroxylase mRNA, dopamine uptake and release in spite of the fact that dopaminergic cells in the prenatal midbrain do not express the classical oestrogen receptor. This study therefore intended to unravel possible signal transduction pathways activated by oestrogen which might be associated with the above oestrogen effects. As a physiological second-messenger mechanism, we studied the influence of oestrogen on fluctuations of intracellular Ca2+ levels [Ca2+]i by microspectrofluorimetry of the Ca2+-sensitive indicator Fura-2, in primary cultures from embryonic mouse midbrains. 17Beta-estradiol (10 nM-1 pM) but not 17alpha-estradiol increased [Ca2+]i within 1-3 s in a dose-dependent way. Removal of extracellular Ca2+ abrogated K+-stimulated Ca2+ rise but did not affect 17beta-estradiol stimulation. Pretreatment of cells with thapsigargin (1 microM, 10 min), an inhibitor of Ca2+-pumping ATPases in the endoplasmic reticulum, abolished the 17beta-estradiol effect but not the K+-stimulated [Ca2+]i rise. Oestrogen effects on [Ca2+]i were completely mimicked by using a membrane-impermeant oestrogen-BSA construct. In order to identify oestrogen-sensitive cells, some cultures were subsequently immunostained for microtubule-associated protein II, tyrosine hydroxylase, or GABA. All oestrogen-sensitive cells were immunocytochemically characterized as neurones, and about half of these responsive neurones was found to be dopaminergic or GABAergic. These results demonstrate that 17beta-estradiol is capable of rapidly modulating physiological parameters of developing midbrain neurones by directly interacting with specific membrane binding sites coupled to a signal transduction mechanism that causes a calcium release from intracellular Ca2+ stores. It is suggested that oestrogen effects on differentiation and function of midbrain dopaminergic neurones are mediated by intracellular Ca2+ signalling.
Collapse
Affiliation(s)
- C Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany.
| | | |
Collapse
|
49
|
MacLusky NJ, Bowlby DA, Brown TJ, Peterson RE, Hochberg RB. Sex and the developing brain: suppression of neuronal estrogen sensitivity by developmental androgen exposure. Neurochem Res 1997; 22:1395-414. [PMID: 9355112 DOI: 10.1023/a:1022027408234] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The developmental effects of androgen play a central role in sexual differentiation of the mammalian central nervous system. The cellular mechanisms responsible for mediating these effects remain incompletely understood. A considerable amount of evidence has accumulated indicating that one of the earliest detectable events in the mechanism of sexual differentiation is a selective and permanent reduction in estrogen receptor concentrations in specific regions of the brain. Using quantitative autoradiographic methods, it has been possible to precisely map the regional distribution of estrogen receptors in the brains of male and female rats, as well as to study the development of sexual dimorphisms in receptor distribution. Despite previous data suggesting that the left and right sides of the brain may be differentially responsive to early androgen exposure, there is no significant right-left asymmetry in estrogen receptor distribution, in either sex. Significant sex differences in receptor density are, however, observed in several regions of the preoptic area, the bed nucleus of the stria terminalis and the ventromedial nucleus of the hypothalamus, particularly in its most rostral and caudal aspects. In the periventricular preoptic area of the female, highest estrogen receptor density occurs in the anteroventral periventricular region: binding in this region is reduced by approximately 50% in the male, as compared to the female. These data are consistent with the hypothesis that androgen-induced defeminization of feminine behavioral and neuroendocrine responses to estrogen may involve selective reductions in the estrogen sensitivity of critical components of the neural circuitry regulating these responses, mediated in part through a reduction in estrogen receptor biosynthesis.
Collapse
Affiliation(s)
- N J MacLusky
- Division of Reproductive Science, Toronto Hospital Research Institute, Ontario, Canada
| | | | | | | | | |
Collapse
|
50
|
Hutchison JB, Beyer C, Hutchison RE, Wozniak A. Sex differences in the regulation of embryonic brain aromatase. J Steroid Biochem Mol Biol 1997; 61:315-22. [PMID: 9365207 DOI: 10.1016/s0960-0760(97)80029-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oestrogen formed from androgen by aromatization plays a critical role in the sexual differentiation of the male brain and behaviour. A question which has still to be answered is what regulates the gender-specific changes in aromatase activity forming oestrogen during sensitive periods of brain growth. Using a primary cell culture technique and sexed embryos, we have shown that in the fetal mouse brain, oestrogen formation in the male is neuronal rather than glial and aromatase activity is regionally localized, being higher in the hypothalamus than in the cortex. The aromatase activity measured from cells in culture has the same enzyme binding affinity (apparent Km approximately 40 nM) as intact brain samples. Neurones developing in the embryonic male brain (embryonic day (ED) 15) contain higher aromatase activity (Vmax, 895 fmol/h/mg protein) than the female (Vmax, 604). Although a sex difference exists at early stages of embryonic development (ED 13), the embryonic aromatase system is regulated by steroids later in fetal development. The developing aromatase-containing neuroblasts probably form processes which connect to other aromatase neurones. Immunoreactive staining with an aromatase polyclonal antibody identifies an increase in numbers of aromatase-immunoreactive hypothalamic neuronal cell bodies following testosterone treatment. Testosterone treatment also causes both stimulation of neurite growth and branching as well as functional maturation of aromatase neurones. In particular, there is an increase in aromatase activity per neurone as well as a dramatic increase in the number of neurones expressing the enzyme. Both the functional and morphological changes depend on androgen receptor stimulation for several days in vitro. This conclusion is supported by colocalization studies which reveal a high number of fetal hypothalamic aromatase neurones co-expressing androgen receptor. We conclude that testosterone influences the growth of male hypothalamic neurones containing aromatase at a sensitive period of brain development. Endogenous steroid inhibitors of aromatase, probably formed within the neuroglia, also play a role in the control of oestrogen production. An endogenous 5alpha-reduced metabolite of testosterone, 5alpha-androstanedione, is almost as potent in inhibiting neuronal hypothalamic aromatase activity (Ki = 23 nM) as the synthetic non-steroidal inhibitors such as the imidazole, fadrozole, and the triazoles, arimidex and letrozole. It is clear that the oestrogen-forming capacity of the male hypothalamus has the special characteristics and plasticity of regulation which could affect brain differentiation at specific steroid-sensitive stages in ontogeny.
Collapse
Affiliation(s)
- J B Hutchison
- MRC Neuroendocrine Development and Behaviour Group, The Babraham Institute, Cambridge, U.K
| | | | | | | |
Collapse
|