1
|
Ciallella HL, Russo DP, Aleksunes LM, Grimm FA, Zhu H. Revealing Adverse Outcome Pathways from Public High-Throughput Screening Data to Evaluate New Toxicants by a Knowledge-Based Deep Neural Network Approach. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:10875-10887. [PMID: 34304572 PMCID: PMC8713073 DOI: 10.1021/acs.est.1c02656] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Traditional experimental testing to identify endocrine disruptors that enhance estrogenic signaling relies on expensive and labor-intensive experiments. We sought to design a knowledge-based deep neural network (k-DNN) approach to reveal and organize public high-throughput screening data for compounds with nuclear estrogen receptor α and β (ERα and ERβ) binding potentials. The target activity was rodent uterotrophic bioactivity driven by ERα/ERβ activations. After training, the resultant network successfully inferred critical relationships among ERα/ERβ target bioassays, shown as weights of 6521 edges between 1071 neurons. The resultant network uses an adverse outcome pathway (AOP) framework to mimic the signaling pathway initiated by ERα and identify compounds that mimic endogenous estrogens (i.e., estrogen mimetics). The k-DNN can predict estrogen mimetics by activating neurons representing several events in the ERα/ERβ signaling pathway. Therefore, this virtual pathway model, starting from a compound's chemistry initiating ERα activation and ending with rodent uterotrophic bioactivity, can efficiently and accurately prioritize new estrogen mimetics (AUC = 0.864-0.927). This k-DNN method is a potential universal computational toxicology strategy to utilize public high-throughput screening data to characterize hazards and prioritize potentially toxic compounds.
Collapse
Affiliation(s)
- Heather L Ciallella
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
| | - Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
- Department of Chemistry, Rutgers University Camden, Camden, New Jersey 08102, United States
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Fabian A Grimm
- ExxonMobil Biomedical Sciences, Inc., Annandale, New Jersey 08801, United States
| | - Hao Zhu
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
- Department of Chemistry, Rutgers University Camden, Camden, New Jersey 08102, United States
| |
Collapse
|
2
|
Ciallella HL, Russo DP, Aleksunes LM, Grimm FA, Zhu H. Predictive modeling of estrogen receptor agonism, antagonism, and binding activities using machine- and deep-learning approaches. J Transl Med 2021; 101:490-502. [PMID: 32778734 PMCID: PMC7873171 DOI: 10.1038/s41374-020-00477-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 11/23/2022] Open
Abstract
As defined by the World Health Organization, an endocrine disruptor is an exogenous substance or mixture that alters function(s) of the endocrine system and consequently causes adverse health effects in an intact organism, its progeny, or (sub)populations. Traditional experimental testing regimens to identify toxicants that induce endocrine disruption can be expensive and time-consuming. Computational modeling has emerged as a promising and cost-effective alternative method for screening and prioritizing potentially endocrine-active compounds. The efficient identification of suitable chemical descriptors and machine-learning algorithms, including deep learning, is a considerable challenge for computational toxicology studies. Here, we sought to apply classic machine-learning algorithms and deep-learning approaches to a panel of over 7500 compounds tested against 18 Toxicity Forecaster assays related to nuclear estrogen receptor (ERα and ERβ) activity. Three binary fingerprints (Extended Connectivity FingerPrints, Functional Connectivity FingerPrints, and Molecular ACCess System) were used as chemical descriptors in this study. Each descriptor was combined with four machine-learning and two deep- learning (normal and multitask neural networks) approaches to construct models for all 18 ER assays. The resulting model performance was evaluated using the area under the receiver- operating curve (AUC) values obtained from a fivefold cross-validation procedure. The results showed that individual models have AUC values that range from 0.56 to 0.86. External validation was conducted using two additional sets of compounds (n = 592 and n = 966) with established interactions with nuclear ER demonstrated through experimentation. An agonist, antagonist, or binding score was determined for each compound by averaging its predicted probabilities in relevant assay models as an external validation, yielding AUC values ranging from 0.63 to 0.91. The results suggest that multitask neural networks offer advantages when modeling mechanistically related endpoints. Consensus predictions based on the average values of individual models remain the best modeling strategy for computational toxicity evaluations.
Collapse
Affiliation(s)
- Heather L Ciallella
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Fabian A Grimm
- ExxonMobil Biomedical Sciences, Inc., Annandale, NJ, USA
| | - Hao Zhu
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA.
- Department of Chemistry, Rutgers University, Camden, NJ, USA.
| |
Collapse
|
3
|
Premachandran H, Zhao M, Arruda-Carvalho M. Sex Differences in the Development of the Rodent Corticolimbic System. Front Neurosci 2020; 14:583477. [PMID: 33100964 PMCID: PMC7554619 DOI: 10.3389/fnins.2020.583477] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, a growing body of research has shown sex differences in the prevalence and symptomatology of psychopathologies, such as depression, anxiety, and fear-related disorders, all of which show high incidence rates in early life. This has highlighted the importance of including female subjects in animal studies, as well as delineating sex differences in neural processing across development. Of particular interest is the corticolimbic system, comprising the hippocampus, amygdala, and medial prefrontal cortex. In rodents, these corticolimbic regions undergo dynamic changes in early life, and disruption to their normative development is believed to underlie the age and sex-dependent effects of stress on affective processing. In this review, we consolidate research on sex differences in the hippocampus, amygdala, and medial prefrontal cortex across early development. First, we briefly introduce current principles on sexual differentiation of the rodent brain. We then showcase corticolimbic regional sex differences in volume, morphology, synaptic organization, cell proliferation, microglia, and GABAergic signaling, and explain how these differences are influenced by perinatal and pubertal gonadal hormones. In compiling this research, we outline evidence of what and when sex differences emerge in the developing corticolimbic system, and illustrate how temporal dynamics of its maturational trajectory may differ in male and female rodents. This will help provide insight into potential neural mechanisms underlying sex-specific critical windows for stress susceptibility and behavioral emergence.
Collapse
Affiliation(s)
| | - Mudi Zhao
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
4
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
5
|
Meitzen J, Britson KA, Tuomela K, Mermelstein PG. The expression of select genes necessary for membrane-associated estrogen receptor signaling differ by sex in adult rat hippocampus. Steroids 2019; 142:21-27. [PMID: 28962849 PMCID: PMC5874170 DOI: 10.1016/j.steroids.2017.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/14/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
Abstract
17β-estradiol can rapidly modulate neuron function via membrane estrogen receptors (ERs) in a sex-specific manner. For example, female rat hippocampal neurons express palmitoylated versions of ERα and ERβ that associate with the plasma membrane. These membrane-associated ERs are organized by caveolin proteins into functional signaling microdomains with metabotropic glutamate receptors (mGluRs). ER/mGluR signaling mediates several sex-specific estradiol actions on hippocampal neuron function. An important unanswered question regards the mechanism by which sex-specific membrane-associated ER signaling is generated, especially since it has been previously demonstrated that mGluR action is not sex-specific. One possibility is that the genes necessary for the ER membrane complex are differentially expressed between males and females, including genes that encode ERα and β, caveolin 1 and 3, and/or the palmitoylacyltransferases DHHC-7 and -21. Thus we used qPCR to test the hypothesis that these genes show sex differences in expression in neonatal and adult rat hippocampus. As an additional control we tested the expression of the 20 other DHHC palmitoylacyltransferases with no known connections to ER. In neonatal hippocampus, no sex differences were detected in gene expression. In adult hippocampus, the genes that encode caveolin 1 and DHHC-7 showed decreased expression in females compared to males. Thus, select genes differ by sex at specific developmental stages, arguing for a more nuanced model than simple widespread perinatal emergence of sex differences in all genes enabling sex-specific estradiol action. These findings enable the generation of new hypotheses regarding the mechanisms by which sex differences in membrane-associated ER signaling are programmed.
Collapse
Affiliation(s)
- John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, United States; W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States.
| | - Kyla A Britson
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Krista Tuomela
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul G Mermelstein
- Dept. of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Kight KE, McCarthy MM. Sex differences and estrogen regulation of BDNF gene expression, but not propeptide content, in the developing hippocampus. J Neurosci Res 2017; 95:345-354. [PMID: 27870444 DOI: 10.1002/jnr.23920] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/29/2023]
Abstract
Sex differences in adult brain function are frequently determined developmentally through the actions of steroid hormones during sensitive periods of prenatal and early postnatal life. In rodents, various cellular end points of the developing brain are affected by estradiol that is derived from the aromatization of circulating testosterone and/or synthesized within the brain. We have previously described a sex difference in neurogenesis in the hippocampus of neonatal rats that is modulated by estradiol. In this report, we examined a potential downstream regulator of the effects of estradiol on hippocampal cell proliferation by measuring gene expression of brain-derived neurotrophin (BDNF) in male and female neonatal rats in response to estradiol. Males had higher baseline BDNF gene expression in dentate gyrus and CA1 regions of the hippocampus compared with females. Neonatal administration of exogenous estradiol resulted in opposite effects on BDNF expression in these areas of the neonatal hippocampus, such that BDNF transcripts increased in CA1 but decreased in dentate. Blocking endogenous estradiol signaling by antagonizing estrogen receptors decreased BDNF expression in the dentate of males, but not females, and had no effect in CA1. Interestingly, this sex difference and response to estradiol was not mirrored by translational output, as no differences in BDNF precursor peptide were observed. The sex- and region-specific effects of estradiol on BDNF expression in the neonatal hippocampus suggest a complex functional relationship between these pleiotropic factors in regulating developmental neurogenesis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katherine E Kight
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Margaret M McCarthy
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Thaung Zaw JJ, Howe PRC, Wong RHX. Does phytoestrogen supplementation improve cognition in humans? A systematic review. Ann N Y Acad Sci 2017; 1403:150-163. [DOI: 10.1111/nyas.13459] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Jay Jay Thaung Zaw
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan New South Wales Australia
| | - Peter Ranald Charles Howe
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan New South Wales Australia
- Division of Research and Innovation University of Southern Queensland Raceview Queensland Australia
| | - Rachel Heloise Xiwen Wong
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan New South Wales Australia
| |
Collapse
|
9
|
Good E, Perschke S, Lopez R, Chang S, Kinsler A, Snowman A, Lacombe J, Fedock M, Zeppetello R, Zysk JR. Profiling Established Cell Lines as a Means to Screening Diversity. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/108705719800300310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cell lines provide a readily available source of target material for functional and molecular binding screens in drug discovery. The Cell PROFILE® program at NovaScreen® represents an effort to identify receptors and enzymes expressed in established cell lines that are relevant to important drug screening endeavors. In this report, we present data on a selected number of receptors and enzymes for four cell lines studied in this survey. The objective of this survey was not to compare one cell line with another, but to illustrate the diversity of pharmacologic targets and the untapped potential of databases for readily obtainable cell lines. The following cell lines, which are all derived from human tumors, were included in this study (with some relevant pharmacologic/pathologic targets): HT-29, derived from an adenocarcinoma of the colon (colorectal cancer); SK-N-MC, derived from a neuroepithelioma (NPY receptors, apoptosis, HIV type I infection); H-4, derived from a neuroglioma (Alzheimer's disease); and LNCaP, derived from a prostate carcinoma (androgen receptor, prostate cancer). Specific to this survey were receptor-binding assays for androgens, corticotropin-releasing factor, endothelin, GABA, NMDA, somatostatin, and alpha and beta adrenergic ligands, as well as binding sites for ion channels. A comparison of specific binding of these various sites between target tissues routinely used in our assays and established cell lines reveals a diversity of receptors heretofore not reported for the latter and represents a potential database for screening and pharmacologic research.
Collapse
Affiliation(s)
- Evelyn Good
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Scott Perschke
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Rani Lopez
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Sonia Chang
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - April Kinsler
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Adele Snowman
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Jason Lacombe
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Michael Fedock
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - Rene Zeppetello
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| | - John R. Zysk
- NovaScreen®, a Division of Oceanix Biosciences Corp., 7170 Standard Drive, Hanover, MD 21076
| |
Collapse
|
10
|
Barbie-Shoshani Y, Shoham S, Bejar C, Weinstock M. Sex-Specific Effects of Prenatal Stress on Memory and Markers of Neuronal Activity in Juvenile Rats. Dev Neurosci 2016; 38:206-219. [DOI: 10.1159/000446981] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 05/20/2016] [Indexed: 11/19/2022] Open
Abstract
Stress during pregnancy can increase the incidence of emotional problems, learning and language difficulties in human infants and pre-adolescents. Most preclinical studies in rats that attempted to find experimental support for these observations were performed in adult male offspring, but the results are inconsistent. The aim of the current study was to examine the effect of prenatal stress on novel object recognition (NOR) and spatial learning and memory in the Morris water maze (MWM) of juvenile rats of both sexes. By the use of fluorescence immunohistochemistry and protein measurements by Western blot, we measured the expression of markers of neurogenesis (doublecortin, DCX) and neuronal activity that are important for synaptic plasticity and learning (c-fos, GluR1, nNOS). Since neuronal activity in the developing and adult brain can be regulated by astrocytes, we also measured the number of astrocytes and the expression of two astroglial proteins (GFAP and S100B) in the stress-responsive hippocampal dentate gyrus (DG). Experiments were performed on littermates of rats in which its effects on behavior were measured. We found for the first time that juvenile females performed better than males in the NOR and MWM tests. They also had higher densities of DCX and c-fos in the DG, together with the expression of nNOS and GluR1 in the subgranular zone (SGZ) of the DG. There were no sex differences in the expression of GFAP and S100B in astrocytes. Prenatal stress did not affect NOR in females, but improved it in males, together with an increase in DCX+ and c-fos, the number of GFAP-expressing astrocytes and the intensity of GFAP and S100B immunofluorescence in the DG. Staining intensity of GluR1 and nNOS in the hilus and SGZ of the DG, and protein expression in the whole DG, was unchanged in prenatally stressed males. Thus, prenatal stress changed the behavior and expression of key proteins in the DG to resemble that in females. A reduction in plasma testosterone, which although not attaining statistical significance was associated with that in anogenital distance, may contribute to the effect of prenatal stress in males. In females, prenatal stress had no effect on c-fos, DCX or the number of astrocytes but reduced the staining intensity of GluR1 and nNOS. Protein expression of nNOS was also significantly lower than that in prenatally stressed males. The differential effects of prenatal stress on hippocampal neuronal and glial markers may help to explain the sex-dependent effect on spatial learning in prepubertal rats.
Collapse
|
11
|
Chen F, Zhou L, Bai Y, Zhou R, Chen L. Hypothalamic-pituitary-adrenal axis hyperactivity accounts for anxiety- and depression-like behaviors in rats perinatally exposed to bisphenol A. J Biomed Res 2014; 29:250-8. [PMID: 26060449 PMCID: PMC4449493 DOI: 10.7555/jbr.29.20140058] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/10/2014] [Accepted: 10/13/2014] [Indexed: 11/04/2022] Open
Abstract
Accumulating studies have proved that perinatal exposure to environmental dose causes long-term potentiation in anxiety/depression-related behaviors in rats. Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis is one of the most consistent biological findings in anxiety- and depression-related disorders. The HPA axis is reported to be susceptible to developmental reprogramming. The present study focused on HPA reactivity in postnatal day (PND) 80 male rats exposed perinatally to environmental-dose BPA. When female breeders were orally administered 2 μg/(kg.day) BPA from gestation day 10 to lactation day 7, their offspring (PND 80 BPA-exposed rats) showed obvious anxiety/depression-like behaviors. Notably, significant increase in serum corticosterone and adrenocorticotropin, and corticotropin-releasing hormone mRNA were detected in BPA-exposed rats before or after the mild stressor. Additionally, the level of glucocorticoid receptor mRNA in the hippocampus, but not the hypothalamus, was decreased in BPA-exposed rats. The levels of hippocampal mineralocorticoid receptor mRNA, neuronal nitric oxide synthase and phosphorylated cAMP response element binding protein were increased in BPA-exposed rats. In addition, the testosterone level was in BPA-exposed rats. The results indicate that reprogramming-induced hyperactivity of the HPA axis is an important link between perinatal BPA exposure and persistent potentiation in anxiety and depression.
Collapse
Affiliation(s)
- Fang Chen
- State Key Lab of Reproductive Medicine
| | - Libin Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | - Rong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
12
|
Zuloaga DG, Zuloaga KL, Hinds LR, Carbone DL, Handa RJ. Estrogen receptor β expression in the mouse forebrain: age and sex differences. J Comp Neurol 2014; 522:358-71. [PMID: 23818057 DOI: 10.1002/cne.23400] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/15/2012] [Accepted: 06/19/2012] [Indexed: 12/11/2022]
Abstract
Estrogen receptors regulate multiple brain functions, including stress, sexual, and memory-associated behaviors as well as controlling neuroendocrine and autonomic function. During development, estrogen signaling is involved in programming adult sex differences in physiology and behavior. Expression of estrogen receptor α changes across development in a region-specific fashion. By contrast, estrogen receptor β (ERβ) is expressed in many brain regions, yet few studies have explored sex and developmental differences in its expression, largely because of the absence of selective reagents for anatomical localization of the protein. This study utilized bacterial artificial chromosome transgenic mice expressing ERβ identified by enhanced green fluorescent protein (EGFP) to compare expression levels and distribution of ERβ in the male and female mouse forebrain on the day of birth (P0), on postnatal day 4 (P4), and on P21. By using qualitative analysis, we mapped the distribution of ERβ-EGFP and found developmental alterations in ERβ expression within the cortex, hippocampus, and hypothalamic regions including the arcuate, ventromedial, and paraventricular nuclei. We also report a sex difference in ERβ in the bed nucleus of the stria terminalis, with males showing greater expression at P4 and P21. Another sex difference was found in the anteroventral periventricular nucleus of P21, but not P0 or P4, mice, in which ERβ-EGFP-immunoreactive cells were densely clustered near the third ventricle in females but not males. These developmental changes and sex differences in ERβ indicate a mechanism through which estrogens might differentially affect brain functions or program adult physiology at select times during development.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, 85004-2157
| | | | | | | | | |
Collapse
|
13
|
Waddell J, Bowers JM, Edwards NS, Jordan CL, McCarthy MM. Dysregulation of neonatal hippocampal cell genesis in the androgen insensitive Tfm rat. Horm Behav 2013; 64:144-52. [PMID: 23747829 PMCID: PMC3753588 DOI: 10.1016/j.yhbeh.2013.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 05/23/2013] [Accepted: 05/29/2013] [Indexed: 12/20/2022]
Abstract
The first two weeks of life are a critical period for hippocampal development. At this time gonadal steroid exposure organizes sex differences in hippocampal sensitivity to activational effects of steroids, hippocampal cell morphology and hippocampus dependent behaviors. Our laboratory has characterized a robust sex difference in neonatal neurogenesis in the hippocampus that is mediated by estradiol. Here, we extend our knowledge of this sex difference by comparing the male and female hippocampus to the androgen insensitive testicular feminized mutant (Tfm) rat. In the neonatal Tfm rat hippocampus, fewer newly generated cells survive compared to males or females. This deficit in cell genesis is partially recovered with the potent androgen DHT, but is more completely recovered following estradiol administration. Tfm rats do not differ from males or females in the level of endogenous estradiol in the neonatal hippocampus, suggesting other mechanisms mediate a differential sensitivity to estradiol in male, female and Tfm hippocampus. We also demonstrate disrupted performance on a hippocampal-dependent contextual fear discrimination task. Tfm rats generalize fear across contexts, and do not exhibit significant loss of fear during extinction exposure. These results extend prior reports of exaggerated response to stress in Tfm rats, and following gonadectomy in normal male rats.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Pediatrics, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
14
|
Brain-derived neurotrophic factor-estrogen interactions in the hippocampal mossy fiber pathway: implications for normal brain function and disease. Neuroscience 2012; 239:46-66. [PMID: 23276673 DOI: 10.1016/j.neuroscience.2012.12.029] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/13/2012] [Indexed: 12/17/2022]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) and the steroid hormone estrogen exhibit potent effects on hippocampal neurons during development and in adulthood. BDNF and estrogen have also been implicated in the etiology of diverse types of neurological disorders or psychiatric illnesses, or have been discussed as potentially important in treatment. Although both are typically studied independently, it has been suggested that BDNF mediates several of the effects of estrogen in the hippocampus, and that these interactions play a role in the normal brain as well as disease. Here we focus on the mossy fiber (MF) pathway of the hippocampus, a critical pathway in normal hippocampal function, and a prime example of a location where numerous studies support an interaction between BDNF and estrogen in the rodent brain. We first review the temporal and spatially regulated expression of BDNF and estrogen in the MFs, as well as their receptors. Then we consider the results of studies that suggest that 17β-estradiol alters hippocampal function by its influence on BDNF expression in the MF pathway. We also address the hypothesis that estrogen influences the hippocampus by mechanisms related not only to the mature form of BDNF, acting at trkB receptors, but also by regulating the precursor, proBDNF, acting at p75NTR. We suggest that the interactions between BDNF and 17β-estradiol in the MFs are potentially important in the normal function of the hippocampus, and have implications for sex differences in functions that depend on the MFs and in diseases where MF plasticity has been suggested to play an important role, Alzheimer's disease, epilepsy and addiction.
Collapse
|
15
|
Sex and stress hormone influences on the expression and activity of brain-derived neurotrophic factor. Neuroscience 2012; 239:295-303. [PMID: 23211562 DOI: 10.1016/j.neuroscience.2012.10.073] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 01/02/2023]
Abstract
The neurotrophin, brain-derived neurotrophic factor (BDNF), is recognized as a key component in the regulation of CNS ontogeny, homeostasis and adult neuroplasticity. The importance of BDNF in CNS development and function is well documented by numerous reports from animal studies linking abnormal BDNF signaling to metabolic disturbances and anxiety or depressive-like behavior. Despite the diverse roles for BDNF in nearly all aspects of CNS physiology, the regulation of BDNF expression, as well as our understanding of the signaling mechanisms associated with this neurotrophin, remains incomplete. However, links between sex hormones such as estradiol and testosterone, as well as endogenous and synthetic glucocorticoids (GCs), have emerged as important mediators of BDNF expression and function. Examples of such regulation include brain region-specific induction of Bdnf mRNA in response to estradiol. Additional studies have also documented regulation of the expression of the high-affinity BDNF receptor Tropomyosin-Related Kinase B by estradiol, thus implicating sex steroids not only in the regulation of BDNF expression, but also in mechanisms of signaling associated with it. In addition to gonadal steroids, further evidence also suggests functional interaction between BDNF and GCs, such as in the regulation of corticotrophin-releasing hormone and other important neuropeptides. In this review, we provide an overview of the roles played by selected sex or stress hormones in the regulation of BDNF expression and signaling in the CNS.
Collapse
|
16
|
Zuloaga DG, Carbone DL, Quihuis A, Hiroi R, Chong DL, Handa RJ. Perinatal dexamethasone-induced alterations in apoptosis within the hippocampus and paraventricular nucleus of the hypothalamus are influenced by age and sex. J Neurosci Res 2012; 90:1403-12. [PMID: 22388926 DOI: 10.1002/jnr.23026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 12/06/2011] [Accepted: 12/08/2011] [Indexed: 01/18/2023]
Abstract
Exposure to high levels of glucocorticoids (GCs) during development leads to long-term changes in hypothalamic-pituitary-adrenal (HPA) axis regulation, although little is known about the neural mechanisms that underlie these alterations. In this study, we investigated the effects of late gestational (days 18-22) or postnatal (days 4-6) administration of the GC receptor agonist dexamethasone (DEX) on an apoptosis marker in two brain regions critical to HPA axis regulation, the hippocampus and the hypothalamic paraventricular nucleus (PVN). One day after the final DEX injection, male and female rats were sacrificed, and brains were processed for immunohistochemical detection of cleaved caspase-3, an apoptotic cell death indicator. DEX increased cleaved caspase-3 immunoreactivity in the CA1 hippocampal region of both sexes following prenatal but not postnatal treatment. Prenatal DEX also increased caspase-3 immunoreactivity in the CA3 region, an elevation that tended to be greater in females. In contrast, postnatal DEX resulted in a much smaller, albeit significant, induction in CA3 caspase-3 compared with prenatal treatment. Quantitative real-time PCR analysis revealed that prenatal but not postnatal DEX-induced hippocampal cleaved caspase-3 correlated with elevated mRNA of the proapoptotic gene Bad. Few caspase-3-ir cells were identified within the PVN regardless of treatment age, although postnatal but not prenatal DEX increased this number. However, the region immediately surrounding the PVN (peri-PVN) showed significant increases in caspase-3-ir cells following pre- and postnatal DEX. Together these findings indicate that developmental GC exposure increases apoptosis in HPAaxis-associated brain regions in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Poimenova A, Markaki E, Rahiotis C, Kitraki E. Corticosterone-regulated actions in the rat brain are affected by perinatal exposure to low dose of bisphenol A. Neuroscience 2010; 167:741-9. [DOI: 10.1016/j.neuroscience.2010.02.051] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 02/18/2010] [Accepted: 02/19/2010] [Indexed: 10/19/2022]
|
18
|
Larkin JW, Binks SL, Li Y, Selvage D. The role of oestradiol in sexually dimorphic hypothalamic-pituitary-adrena axis responses to intracerebroventricular ethanol administration in the rat. J Neuroendocrinol 2010; 22:24-32. [PMID: 19912475 PMCID: PMC2818645 DOI: 10.1111/j.1365-2826.2009.01934.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Systemic ethanol (EtOH) administration activates the hypothalamic-pituitary-adrenal (HPA) axis of rats in a sexually dimorphic manner. The present studies tested the role played by the central nervous system (CNS) in this phenomenon. To localise the effects of the drug to the brain, we utilised an experimental paradigm whereby a small, nontoxic amount of the drug was delivered via intracerebroventricular (i.c.v.) injection. EtoH administered i.c.v. rapidly diffuses throughout the cerebrospinal fluid and brain, and does not cause neuronal damage or have any long-term physiological or behavioural effects. Experimental groups included intact males, intact cycling females, and ovariectomised (OVX) animals with or without replacement oestradiol (E(2)). Intracerebroventricular EtOH-induced HPA hormonal activation was determined by measuring plasma adrenocorticotrophin (ACTH) levels. Activation of brain areas that both regulate HPA function and are responsive to gonadal hormones was determined using expression of the transcription factor c-fos (Fos) as a marker of neuronal activity. We observed sex- and oestrous cycle- dependent differences in HPA activation by EtOH as measured by both these parameters. ACTH secretion was highest in females in pro-oestrus or oestrus, just prior to or after the endogenous peak of E(2), as was Fos expression in the paraventricular nucleus of the hypothalamus (PVN) and the locus coreuleus (LC) of the brainstem. In OVX animals, E(2) replacement caused an increase in PVN and LC Fos expression in response to i.c.v. EtOH compared to OVX controls, but a decrease in ACTH secretion. Taken together, these results indicate that at the level of the CNS, EtOH stimulates HPA activity more robustly at times when the effects of E(2) are high, but that E(2) alone is not responsible for this effect. The data further suggest that the LC plays an important role in the circuitry, which appears to be different from that activated following the systemic administration of EtOH.
Collapse
Affiliation(s)
- J W Larkin
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID, USA
| | | | | | | |
Collapse
|
19
|
Thorp AA, Sinn N, Buckley JD, Coates AM, Howe PRC. Soya isoflavone supplementation enhances spatial working memory in men. Br J Nutr 2009; 102:1348-54. [PMID: 19480732 DOI: 10.1017/s0007114509990201] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Females perform better in certain memory-related tasks than males. Sex differences in cognitive performance may be attributable to differences in circulating oestrogen acting on oestrogen beta receptors (ERbeta) which are prevalent in brain regions such as the hippocampus, frontal lobe and cortex that mediate cognitive functions. Since soya isoflavones are known to activate ERbeta, chronic isoflavone supplementation in males may improve cognitive performance in memory-related tasks. A 12-week double-blind, placebo-controlled cross-over trial was conducted in thirty-four healthy men to investigate the effect of isoflavone supplementation on cognitive function. Volunteers were randomised to take four capsules/d containing soya isoflavones (116 mg isoflavone equivalents/d: 68 mg daidzein, 12 mg genistein, 36 mg glycitin) or placebo for 6 weeks, and the alternate treatment during the following 6 weeks. Assessments of memory (verbal episodic, auditory and working), executive function (planning, attention, mental flexibility) and visual-spatial processing were performed at baseline and after each treatment period. Isoflavone supplementation significantly improved spatial working memory (P = 0.01), a test in which females consistently perform better than males. Compared with placebo supplementation, there were 18 % fewer attempts (P = 0.01), 23 % fewer errors (P = 0.02) and 17 % less time (P = 0.03) required to correctly identify the requisite information. Isoflavones did not affect auditory and episodic memory (Paired Associate Learning, Rey's Auditory Verbal Learning Task, Backward Digit Span and Letter-Number Sequencing), executive function (Trail Making and Initial Letter Fluency Task) or visual-spatial processing (Mental Rotation Task). Isoflavone supplementation in healthy males may enhance cognitive processes which appear dependent on oestrogen activation.
Collapse
Affiliation(s)
- Alicia A Thorp
- Nutritional Physiology Research Centre and ATN Centre for Metabolic Fitness, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | | | | | | | | |
Collapse
|
20
|
McEwen BS, Gould E, Orchinik M, Weiland NG, Woolley CS. Oestrogens and the structural and functional plasticity of neurons: implications for memory, ageing and neurodegenerative processes. CIBA FOUNDATION SYMPOSIUM 2007; 191:52-66; discussion 66-73. [PMID: 8582205 DOI: 10.1002/9780470514757.ch4] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Oestrogens have numerous effects on the brain, beginning during gestation and continuing on into adulthood. Many of these actions involve areas of the brain that are not primarily involved in reproduction, such as the basal forebrain, hippocampus, caudate putamen, midbrain raphe and brainstem locus coeruleus. This paper describes three actions of oestrogens that are especially relevant to brain mechanisms involved in memory processes and their alterations during ageing and neurodegenerative diseases: (1) the regulation of cholinergic neurons by oestradiol in the rat basal forebrain, involving induction of choline acetyltransferase and acetylcholinesterase according to a sexually dimorphic pattern; (2) the regulation of synaptogenesis in the CA1 region of the hippocampus by oestrogens and progestins during the four- to five-day oestrus cycle of the female rat. Formation of new excitatory synapses is induced by oestradiol and involves N-methyl-D-aspartate receptors; removal of these synapses involves intracellular progestin receptors; (3) sex differences in hippocampal structure, which may help to explain differences in the strategies that male and female rats use to solve spatial navigation problems. During the period of development when testosterone is elevated in the male, aromatase and oestrogen receptors are also elevated, making it likely that this pathway is involved in the masculinization of hippocampal structure.
Collapse
Affiliation(s)
- B S McEwen
- Laboratory of Neuroendocrinology, Rockefeller University, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
21
|
Jelks KB, Wylie R, Floyd CL, McAllister AK, Wise P. Estradiol targets synaptic proteins to induce glutamatergic synapse formation in cultured hippocampal neurons: critical role of estrogen receptor-alpha. J Neurosci 2007; 27:6903-13. [PMID: 17596438 PMCID: PMC6672227 DOI: 10.1523/jneurosci.0909-07.2007] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Estradiol mediates structural changes at synapses of the hippocampus, an area in the brain important for learning and memory. This study was designed to test the hypothesis that estradiol mediates subcellular changes of synaptic proteins to induce new synapses via an estrogen receptor (ER)-mediated process. To elucidate the mechanisms involved in glutamatergic synapse formation, we investigated effects of estradiol on synaptic proteins in cultured hippocampal neurons using immunocytochemistry and confocal microscopy. Synaptic protein distribution and size were identified with antibodies to the presynaptic vesicular glutamate transporter protein (vGlut1) and postsynaptic NMDA receptor (NR1 subunit). We observed an increase in synapse density, as detected by NR1 and vGlut1 colocalization, along dendrites of neurons cultured in steroid-stripped media and exposed to estradiol (10 nM) for 48 h. Additionally, the NR1 subunit was enriched at synaptic clusters. Immunocytochemistry and confocal imaging revealed punctate staining of extranuclear ERs along dendrites of hippocampal neurons expressing NR1. Estradiol increased the density of both ER-alpha and ER-beta protein clusters along dendrites. To test whether ERs play an important functional role in the estradiol-induced synaptogenesis, we used the ER antagonist [7alpha,17beta-[9[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol (ICI 182,780)] and the ER-alpha- and ER-beta-specific agonists [1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT) and 2,3-bis(4-hydroxyphenyl) propionitrile (DPN), respectively]. ICI 182,780 blocked the increase in synapse density. Treatment with PPT, but not DPN, induced significant increases in synapse density that mimicked treatment with estradiol. Together, our results demonstrate that estradiol stimulates glutamatergic synapse formation in the developing hippocampus through an ER-alpha-dependent mechanism. These findings carry profound implications regarding the potential of estrogen to influence learning, memory, and possibly hormone-modulated neurodegeneration.
Collapse
Affiliation(s)
| | - Rebecca Wylie
- Department of Neurobiology, Physiology, and Behavior and
| | - Candace L. Floyd
- Department of Physical Medicine and Rehabilitation, University of Alabama–Birmingham, Birmingham, Alabama 35249-7330, and
| | | | - Phyllis Wise
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195-1237
| |
Collapse
|
22
|
Corrieri L, Della Seta D, Canoine V, Fusani L. Developmental exposure to xenoestrogen enhances spatial learning in male rats. Horm Behav 2007; 51:620-5. [PMID: 17428485 DOI: 10.1016/j.yhbeh.2007.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 02/28/2007] [Accepted: 03/01/2007] [Indexed: 11/20/2022]
Abstract
Steroid hormones have profound effects on the development and function of the nervous system. Environmental estrogens or xenoestrogens are manmade or are natural compounds, which mimics the action of estrogen hormones. The experimental evidence for impairment of cognitive functions in humans and mammals following exposure to xenoestrogens has been fiercely debated. The strongest arguments against such studies have been that the route, time course, and intensity of exposure did not simulate environmental exposure, and that the chemicals tested have additional, non-estrogenic toxic effects, hindering a generalization of actual "xenoestrogenic" effects. Here we show that an environmental-like exposure to the pure estrogen, 17alpha-ethynylestradiol (EE2) during development enhances spatial learning abilities in adult male Sprague-Dawley rats. To simulate an environmental exposure, we used a very low dose (4 ng/kg/day) of EE2 equivalent to concentrations measured in European and US streams which was given orally with a non-invasive method, and we extended the treatment for the entire course of development, from conception to puberty. The animals were tested in a Morris water maze protocol at 6 months of age. Male rats treated with EE2 during development showed a faster learning during the training phase, and remembered better the position of the hidden platform in the short term. Our study demonstrates that actual levels of exposure to xenoestrogens can permanently alter cognitive abilities of a mammalian species.
Collapse
Affiliation(s)
- Luca Corrieri
- Dipartimento di Fisiologia, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | | | | | | |
Collapse
|
23
|
Miles C, Green R, Hines M. Estrogen treatment effects on cognition, memory and mood in male-to-female transsexuals. Horm Behav 2006; 50:708-17. [PMID: 16884726 DOI: 10.1016/j.yhbeh.2006.06.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Revised: 06/14/2006] [Accepted: 06/14/2006] [Indexed: 11/29/2022]
Abstract
Gonadal hormones, particularly estrogens, have been suggested to influence memory and cognitive tasks that show sex differences. Previously, we reported that male-to-female (M-F) transsexuals undergoing estrogen treatment for sex re-assignment scored higher on verbal Paired Associate Learning (PAL) than a transsexual control group awaiting estrogen treatment. The present study used a more robust design to examine further associations between estrogen and cognition. We assessed additional aspects of memory, including visual, spatial, object and location memory, other cognitive abilities that show reliable sex differences, including verbal and visual-spatial abilities, and mood variables that could mediate associations between estrogen and cognition. In addition to comparing groups of individuals on and off estrogen, we used two repeated measures designs (AB and BA). The AB group was tested prior to hormone treatment and then again after treatment had begun; the BA group was tested while on estrogen treatment and then again when hormones had been withdrawn prior to surgery. Few changes in memory or cognition were observed, and changes that were observed were not consistent across study designs. The lack of significant effects did not relate to mood changes or to the sexual orientation of participants. These findings suggest that estrogen treatment associated with sex change for M-F transsexuals has little or no influence on sex-typed aspects of cognition or memory.
Collapse
Affiliation(s)
- Clare Miles
- Department of Psychology, City University, London, UK
| | | | | |
Collapse
|
24
|
Sandau US, Handa RJ. Localization and developmental ontogeny of the pro-apoptotic Bnip3 mRNA in the postnatal rat cortex and hippocampus. Brain Res 2006; 1100:55-63. [PMID: 16765336 DOI: 10.1016/j.brainres.2006.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Revised: 05/01/2006] [Accepted: 05/02/2006] [Indexed: 11/30/2022]
Abstract
Naturally occurring cell death occurs during the first two postnatal weeks in the rat cortex and hippocampus. During this process, apoptosis is initiated by activating or altering expression of pro-apoptotic members of the Bcl-2 family. Bnip3 is a pro-apoptotic member of the Bcl-2 family that induces cell death by opening the mitochondrial permeability transition pore. To date, Bnip3 expression in the central nervous system has only been examined during hypoxia-mediated apoptosis in the adult rat brain. In this study, we investigated the localization and ontogeny of Bnip3 mRNA expression in the postnatal male and female rat brain. Bnip3 mRNA was localized by in situ hybridization in the neonatal cortex, hippocampus, habenula and thalamus. Using quantitative real-time RT-PCR, Bnip3 mRNA levels were found to be greatest at postnatal day 6.5 in the female anterior and posterior cingulate cortices and hippocampus. Bnip3 mRNA expression also increased in the male anterior cingulate cortex at postnatal day 6.5. However, a developmental change in Bnip3 levels did not occur in the male posterior cingulate cortex and hippocampus. In the anterior cingulate cortex on postnatal day 6.0 and adulthood, female rats had significantly greater levels of Bnip3 mRNA compared to that of males. Altering levels of testosterone in the neonatal rat did not alter the sex differences in Bnip3 mRNA levels. The transient increase in Bnip3 mRNA expression correlates with naturally occurring cell death in the neonatal rat cortex and hippocampus. Thus, Bnip3 may be a mediator of developmental apoptosis in the postnatal rat brain.
Collapse
Affiliation(s)
- Ursula S Sandau
- Department of Biomedical Sciences, Colorado State University, W103 Anatomy, 1617 Campus Delivery, Fort Collins, CO 80523-1617, USA
| | | |
Collapse
|
25
|
Lund TD, Hinds LR, Handa RJ. The androgen 5alpha-dihydrotestosterone and its metabolite 5alpha-androstan-3beta, 17beta-diol inhibit the hypothalamo-pituitary-adrenal response to stress by acting through estrogen receptor beta-expressing neurons in the hypothalamus. J Neurosci 2006; 26:1448-56. [PMID: 16452668 PMCID: PMC6675494 DOI: 10.1523/jneurosci.3777-05.2006] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Estrogen receptor beta (ERbeta) and androgen receptor (AR) are found in high levels within populations of neurons in the hypothalamus. To determine whether AR or ERbeta plays a role in regulating hypothalamo-pituitary-adrenal (HPA) axis function by direct action on these neurons, we examined the effects of central implants of 17beta-estradiol (E2), 5alpha-dihydrotestosterone (DHT), the DHT metabolite 5alpha-androstan-3beta, 17beta-diol (3beta-diol), and several ER subtype-selective agonists on the corticosterone and adrenocorticotropin (ACTH) response to immobilization stress. In addition, activation of neurons in the paraventricular nucleus (PVN) was monitored by examining c-fos mRNA expression. Pellets containing these compounds were stereotaxically implanted near the PVN of gonadectomized male rats. Seven days later, animals were killed directly from their home cage (nonstressed) or were restrained for 30 min (stressed) before they were killed. Compared with controls, E2 and the ERalpha-selective agonists moxestrol and propyl-pyrazole-triol significantly increased the stress induced release of corticosterone and ACTH. In contrast, central administration of DHT, 3beta-diol, and the ERbeta-selective compound diarylpropionitrile significantly decreased the corticosterone and ACTH response to immobilization. Cotreatment with the ER antagonist tamoxifen completely blocked the effects of 3beta-diol and partially blocked the effect of DHT, whereas the AR antagonist flutamide had no effect. Moreover, DHT, 3beta-diol, and diarylpropionitrile treatment significantly decreased restraint-induced c-fos mRNA expression in the PVN. Together, these studies indicate that the inhibitory effects of DHT on HPA axis activity may be in part mediated via its conversion to 3beta-diol and subsequent binding to ERbeta.
Collapse
Affiliation(s)
- Trent D Lund
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA.
| | | | | |
Collapse
|
26
|
Lemmens EMP, Lubbers T, Schijns OEMG, Beuls EAM, Hoogland G. Gender differences in febrile seizure-induced proliferation and survival in the rat dentate gyrus. Epilepsia 2005; 46:1603-12. [PMID: 16190931 DOI: 10.1111/j.1528-1167.2005.00252.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Febrile seizures are fever-associated early-life seizures that are thought play a role in the development of epilepsy. Seizure-induced proliferation of dentate granule cells has been demonstrated in several adult animal models and is thought to be an integral part of epileptogenesis. The aim of the present study was to investigate proliferation and survival of dentate gyrus (DG) cells born after early-life hyperthermia (HT)-induced seizures in male and female rats. METHODS At postnatal day (PN) 10, male and female rats were exposed to heated air to induce seizures. Littermates were used as normothermia controls. Convulsive behavior was observed by two researchers. From PN11 to PN16, rats were injected with bromodeoxyuridine (BrdU) to label dividing cells. The number of BrdU-immunoreactive cells in the DG was counted at PN17 and PN66. RESULTS At PN17, male as well as female HT rats had the same amount of BrdU-positive cells compared with controls. At PN66, significantly more BrdU-positive cells were left in HT females (53%) than in controls (44%, percentage of BrdU-positive cells at PN17), whereas no difference was found between HT males and male controls. The net result of proliferation and survival at PN66 was that female HT rats had the same number of BrdU-immunoreactive cells as controls, whereas male HT rats had 25% more BrdU-immunoreactive cells than did controls (p < 0.05). CONCLUSIONS Early-life seizures cause a sexually dimorphic cytogenic response that results in an increased population of newborn DG cells in young adult males, while leaving that of young adult females unaltered.
Collapse
Affiliation(s)
- Evi M P Lemmens
- Department of Neurosurgery, University Hospital Maastricht, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
27
|
Hamada T, Wada-Kiyama Y, Sakuma Y. Visualizing forebrain-specific usage of an estrogen receptor α promoter for receptor downregulation in the rat. ACTA ACUST UNITED AC 2005; 139:42-51. [PMID: 15953656 DOI: 10.1016/j.molbrainres.2005.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 04/29/2005] [Accepted: 05/09/2005] [Indexed: 11/20/2022]
Abstract
Transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of an estrogen receptor (ER) alpha promoter were generated to tag ERalpha-positive neurons in the brain. Two transgenes, one containing sequences for promoter A and DsRed and the other containing sequences for promoter 0/B and EGFP, were injected simultaneously into Wistar rat zygotes. Twenty-two founders with both transgenes were identified. Ten lines of these founders expressed the EGFP tag in the brains of their first filial generation, whereas none similarly expressed the DsRed tag. In two lines selected for the brightness of the EGFP fluorescence in their brains, tagged cells showed essentially the same patterns. Tagged cells were in the preoptic area (POA), bed nucleus of the stria terminalis (BNST), hypothalamic arcuate nucleus and medial amygdala. ERalpha-immunoreactive neurons were identified in all of these structures by immunohistochemistry. In ovariectomized females, approximately 75% of the EGFP-fluorescent cells in the POA-BNST were immunoreactive for ERalpha. In the POA-BNST, ovariectomy increased the number of EGFP-immunopositive cells and estrogen supplementation reversed this effect, indicating that the promoter 0/B is involved in estrogen-induced downregulation of ERalpha. EGFP was also present in cells in the cerebral cortex and hippocampus, which have not previously been associated with endocrine regulation. Conversely, only a few cells were tagged in the hypothalamic ventromedial nucleus, which contained many ERalpha-immunoreactive neurons. This discrepancy could have arisen as a result of differential promoter usage.
Collapse
Affiliation(s)
- Tomohiro Hamada
- Department of Physiology, Nippon Medical School, Sendagi 1, Bunkyo, Tokyo 113-8602, Japan.
| | | | | |
Collapse
|
28
|
Setchell KDR, Clerici C, Lephart ED, Cole SJ, Heenan C, Castellani D, Wolfe BE, Nechemias-Zimmer L, Brown NM, Lund TD, Handa RJ, Heubi JE. S-equol, a potent ligand for estrogen receptor beta, is the exclusive enantiomeric form of the soy isoflavone metabolite produced by human intestinal bacterial flora. Am J Clin Nutr 2005; 81:1072-9. [PMID: 15883431 DOI: 10.1093/ajcn/81.5.1072] [Citation(s) in RCA: 318] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The discovery of equol in human urine more than 2 decades ago and the finding that it is bacterially derived from daidzin, an isoflavone abundant in soy foods, led to the current nutritional interest in soy foods. Equol, unlike the soy isoflavones daidzein or genistein, has a chiral center and therefore can occur as 2 distinct diastereoisomers. OBJECTIVE Because it was unclear which enantiomer was present in humans, our objectives were to characterize the exact structure of equol, to examine whether the S- and R-equol enantiomers are bioavailable, and to ascertain whether the differences in their conformational structure translate to significant differences in affinity for estrogen receptors. DESIGN With the use of chiral-phase HPLC and mass spectrometry, equol was isolated from human urine and plasma, and its enantiomeric structure was defined. Human fecal flora were cultured in vitro and incubated with daidzein to ascertain the stereospecificity of the bacterial production of equol. The pharmacokinetics of S- and R- equol were determined in 3 healthy adults after single-bolus oral administration of both enantiomers, and the affinity of each equol enantiomer for estrogen receptors was measured. RESULTS Our studies definitively establish S-equol as the exclusive product of human intestinal bacterial synthesis from soy isoflavones and also show that both enantiomers are bioavailable. S-equol has a high affinity for estrogen receptor beta (K(i) = 0.73 nmol/L), whereas R-equol is relatively inactive. CONCLUSIONS Humans have acquired an ability to exclusively synthesize S-equol from the precursor soy isoflavone daidzein, and it is significant that, unlike R-equol, this enantiomer has a relatively high affinity for estrogen receptor beta.
Collapse
Affiliation(s)
- Kenneth D R Setchell
- Division of Pathology, Clinical Mass Spectrometry, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati 45229, OH, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Arabo A, Lefebvre M, Fermanel M, Caston J. Administration of 17alpha-ethinylestradiol during pregnancy elicits modifications of maternal behavior and emotional alteration of the offspring in the rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 156:93-103. [PMID: 15862632 DOI: 10.1016/j.devbrainres.2005.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 02/07/2005] [Accepted: 02/08/2005] [Indexed: 11/15/2022]
Abstract
Intraperitoneal administration of 17alpha-ethinylestradiol (15 microg.kg(-1)) in pregnant rats, every day from day 9 to day 14 of pregnancy, elicited a high percentage of abortions. Quantification of maternal behavior showed that treated dams took better care of their pups than control dams, injected with the vehicle only, did. Postnatal reflexes, which reflect maturational rate, were established more promptly in the offspring of treated dams than in the offspring of control dams. However, when adult, the rats born from treated dams developed anxiety- and depressive-like behaviors. All these results are explained by the effects of the exogenous estrogen on the developing brain of the fetuses.
Collapse
Affiliation(s)
- A Arabo
- Université de Rouen, Faculté des Sciences, UPRES EA 1780 76821 Mont-Saint-Aignan Cedex, France
| | | | | | | |
Collapse
|
30
|
Markham JA, McKian KP, Stroup TS, Juraska JM. Sexually dimorphic aging of dendritic morphology in CA1 of hippocampus. Hippocampus 2005; 15:97-103. [PMID: 15390161 DOI: 10.1002/hipo.20034] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During aging, rats of both sexes experience a decline in performance on hippocampal-dependent tasks. Investigations into the neuroanatomical correlates of this functional decline have been conducted almost exclusively in male subjects. In the present study, dendritic spine density in stratum radiatum and complexity of the entire apical dendritic tree were quantified using Golgi-Cox-stained tissue in young (3-5 months) and aged (19-22 months) rats of both sexes. Because both cognitive decline and hippocampal morphology may be influenced by ovarian hormonal state, young adult females were examined during either proestrus or estrus, and aged females were examined in one of two reproductively senescent states: persistent estrus or persistent diestrus. A sex difference in dendritic branching of CA1 pyramidal cells was found among young adults. However, this difference disappeared during aging, due to a reduction in branching with age for males but not for females. Spine density was not influenced by age or sex, nor did ovarian hormone status influence either measure. These results are consistent with our previous findings in the rat medial prefrontal cortex and primary motor cortex and with the human literature, which indicate that age-related atrophy of cognitive brain regions is more severe for males than females.
Collapse
Affiliation(s)
- J A Markham
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61820, USA
| | | | | | | |
Collapse
|
31
|
Conejo NM, González-Pardo H, Cimadevilla JM, Argüelles JA, Díaz F, Vallejo-Seco G, Arias JL. Influence of gonadal steroids on the glial fibrillary acidic protein-immunoreactive astrocyte population in young rat hippocampus. J Neurosci Res 2004; 79:488-94. [PMID: 15619230 DOI: 10.1002/jnr.20372] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is known that expression of glial fibrillary acidic protein (GFAP) as an astrocyte-specific marker can be regulated by levels of circulating gonadal steroids during postnatal development. In addition, astrocytes play an important role in the physiology of the hippocampus, a brain region considered sexually dimorphic at the neuronal level in rodents. To evaluate the contribution of glial cells to gender-related differences in the hippocampus, we estimated the number of GFAP-immunoreactive (GFAP-IR) astrocytes in the hippocampus (CA1 and CA3 areas, dorsal and ventral regions) of male and female rats aged 30 days. Groups of 30-day-old masculinized females (TP-females; injected with testosterone propionate at birth) and feminized males (FLU-males, castrated and treated with flutamide, an androgen receptor antagonist) were included to assess the effects of gonadal hormones on these hippocampal astrocytes. Using the optical fractionator method, the total number of GFAP-IR cells found in CA1 and CA3 areas was significantly higher in males compared to that in age-matched females. This numerical pattern was reversed in TP-females and FLU-males in both hippocampal areas. In addition, more GFAP-IR cells were found in dorsal hippocampus than in the ventral region in the CA1 area from all experimental groups, whereas this result was found in the CA3 area from males and TP-females. Our results suggest an essential contribution of gonadal hormones to gender differences found in the astrocyte population of the rat hippocampus during development.
Collapse
Affiliation(s)
- N M Conejo
- Laboratory of Psychobiology, Faculty of Psychology, University of Oviedo, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Dalla C, Antoniou K, Papadopoulou-Daifoti Z, Balthazart J, Bakker J. Oestrogen-deficient female aromatase knockout (ArKO) mice exhibit depressive-like symptomatology. Eur J Neurosci 2004; 20:217-28. [PMID: 15245494 DOI: 10.1111/j.1460-9568.2004.03443.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We recently found that female aromatase knockout (ArKO) mice that are deficient in oestradiol due to a targeted mutation in the aromatase gene show deficits in sexual behaviour that cannot be corrected by adult treatment with oestrogens. We determined here whether these impairments are associated with changes in general levels of activity, anxiety or 'depressive-like' symptomatology due to chronic oestrogen deficiency. We also compared the neurochemical profile of ArKO and wild-type (WT) females, as oestrogens have been shown to modulate dopaminergic, serotonergic and noradrenergic brain activities. ArKO females did not differ from WT in spontaneous motor activity, exploration or anxiety. These findings are in line with the absence of major neurochemical alterations in hypothalamus, prefrontal cortex or striatum, which are involved in the expression of these behaviours. By contrast, ArKO females displayed decreased active behaviours, such as struggling and swimming, and increased passive behaviours, such as floating, in repeated sessions of the forced swim test, indicating that these females exhibit 'depressive-like' symptoms. Adult treatment with oestradiol did not reverse the behavioural deficits observed in the forced swim test, suggesting that they may be due to the absence of oestradiol during development. Accordingly, an increased serotonergic activity was observed in the hippocampus of ArKO females compared with WT, which was also not reversed by adult oestradiol treatment. The possible organizational role of oestradiol on the hippocampal serotonergic system and the 'depressive-like' profile of ArKO females provide new insights into the pathophysiology of depression and the increased vulnerability of women to depression.
Collapse
Affiliation(s)
- C Dalla
- Centre for Cellular and Molecular Neurobiology, University of Liëge, Liëge, Belgium
| | | | | | | | | |
Collapse
|
33
|
Lund TD, Munson DJ, Haldy ME, Handa RJ. Dihydrotestosterone may inhibit hypothalamo–pituitary–adrenal activity by acting through estrogen receptor in the male mouse. Neurosci Lett 2004; 365:43-7. [PMID: 15234470 DOI: 10.1016/j.neulet.2004.04.035] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Revised: 04/08/2004] [Accepted: 04/16/2004] [Indexed: 11/25/2022]
Abstract
The corticosterone (CORT) response to environmental perturbation has been shown to be enhanced by estrogen but inhibited by the androgen dihydrotestosterone (DHT). However, the mechanism of androgen's action has not been identified. This study examined the effects of estradiol benzoate (EB), the non-aromatizable androgen DHT, and the DHT metabolite 5alpha-androstan-3beta, 17beta-diol (3beta-diol) on the corticosterone response to stress. Adult male CBB6/F1 mice were gonadectomized and injected subcutaneously (once a day for 4 days) with the above compounds (controls received oil vehicle injections). Animals (within treatments) were randomly assigned to stress or non-stress conditions. The non-stress animals were taken directly from their home cages and killed. Animals were stressed by a 30 min restraint prior to being killed. Hormone levels were determined in plasma via radioimmunoassay. In agreement with previous studies, the CORT response to immobilization was enhanced by EB and inhibited by DHT. Surprisingly, 3beta-diol inhibited the CORT response similar to the effect of DHT. In a second study, concomitant injections of the androgen receptor antagonist flutamide only partially blocked DHT's, but had no effect on 3beta-diol's, inhibitory action. In contrast, injections with the estrogen receptor antagonist tamoxifen completely blocked the effects of 3beta-diol and partially blocked DHT's effect. Taken together these studies suggest that DHT's inhibitory effects may be, at least in part, via the estrogen receptor, through its conversion to 3beta-diol. These studies also suggest that the DHT metabolites may be functionally relevant when considering hormonal responses to stress.
Collapse
Affiliation(s)
- Trent D Lund
- Department of Biomedical Sciences, Colorado State University, Anatomy W103, 1617 Campus Delivery, Fort Collins, CO 80523-1670, USA.
| | | | | | | |
Collapse
|
34
|
Hilton GD, Ndubuizu AN, McCarthy MM. Neuroprotective effects of estradiol in newborn female rat hippocampus. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 150:191-8. [PMID: 15158082 DOI: 10.1016/j.devbrainres.2004.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/25/2004] [Indexed: 10/26/2022]
Abstract
Perinatal brain injury, consequent to hypoxic/ischemic events, is associated with the release of excess excitatory neurotransmitters, including glutamate. We have previously shown that administration of a glutamate receptor agonist, kainic acid (KA), to postnatal day 0 (PN0) and PN1 rats results in damage selective to the dentate gyrus of females. Pretreatment with the gonadal steroid estradiol prevents KA-induced damage to the female dentate gyrus. To begin to elucidate the cellular mechanism of the neuroprotective effects of estradiol in neonatal females, we have employed the estrogen receptor antagonists Tamoxifen and ICI 182,780 in vivo and in vitro, respectively. Peripheral administration of Tamoxifen, which crosses the blood-brain barrier, prevented estradiol-mediated neuroprotection against KA-induced damage in the dentate gyrus. The highly selective estrogen receptor antagonist ICI 182,780, which does not penetrate into the brain from the periphery, also prevented estradiol's protective effects on KA-induced cell death in cultured hippocampal neurons but only late in the time course of injury. The data suggest that the neuroprotection afforded by estradiol against KA-induced injury in the female is estrogen receptor mediated but may include an additional mechanism that is not antagonized at the receptor.
Collapse
Affiliation(s)
- Genell D Hilton
- Department of Physiology, University of Maryland, 655 W. Baltimore St., Room 5-014, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
35
|
Verdier-Sevrain S, Yaar M, Cantatore J, Traish A, Gilchrest BA. Estradiol induces proliferation of keratinocytes via a receptor mediated mechanism. FASEB J 2004; 18:1252-4. [PMID: 15208259 DOI: 10.1096/fj.03-1088fje] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In this study, we investigated the effects of estradiol on the proliferation of neonatal keratinocytes, the expression of estrogen receptor isoforms, and the signaling mechanisms by which estradiol mediates cell growth. We demonstrate that estradiol binds neonatal keratinocytes with high affinity (Kd=5.2nM) and limited capacity (Bmax of 14.2fmol/mg of protein), confirming the presence of estrogen binding sites. Using specific antibodies, we demonstrate that keratinocytes express both estrogen receptor (ER)-alpha and ER-beta. At physiological concentrations, estradiol up-regulates the level of ER-alpha receptors in keratinocytes and induces keratinocyte proliferation. The proliferative effect of estradiol requires the availability of functional estrogen receptors, as it is abrogated by anti-estrogen administration. Estradiol effect on keratinocyte proliferation is most likely mediated in part by activation of a nongenomic, membrane-associated, signaling pathway involving activation of the extracellular signal regulated kinases 1 and 2 and in part by the genomic signaling pathway through activation of nuclear receptors.
Collapse
Affiliation(s)
- S Verdier-Sevrain
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts 02118-2394, USA
| | | | | | | | | |
Collapse
|
36
|
Lund TD, Munson DJ, Haldy ME, Setchell KDR, Lephart ED, Handa RJ. Equol Is a Novel Anti-Androgen that Inhibits Prostate Growth and Hormone Feedback1. Biol Reprod 2004; 70:1188-95. [PMID: 14681200 DOI: 10.1095/biolreprod.103.023713] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Equol (7-hydroxy-3[4'hydroxyphenyl]-chroman) is the major metabolite of the phytoestrogen daidzein, one of the main isoflavones found abundantly in soybeans and soy foods. Equol may be an important biologically active molecule based on recent studies demonstrating that equol can modulate reproductive function. In this study, we examined the effects of equol on prostate growth and LH secretion and determined some of the mechanisms by which it might act. Administration of equol to intact male rats for 4-7 days reduced ventral prostate and epididymal weight and increased circulating LH levels. Using binding assays, we determined that equol specifically binds 5alpha-dihydrotestosterone (DHT), but not testosterone, dehydroepiandrosterone, or estrogen with high affinity. Equol does not bind the prostatic androgen receptor, and has a modest affinity for recombinant estrogen receptor (ER) beta, and no affinity for ERalpha. In castrated male rats treated with DHT, concomitant treatment with equol blocked DHT's trophic effects on the ventral prostate gland growth and inhibitory feedback effects on plasma LH levels without changes in circulating DHT. Therefore, equol can bind circulating DHT and sequester it from the androgen receptor, thus altering growth and physiological hormone responses that are regulated by androgens. These data suggest a novel model to explain equol's biological properties. The significance of equol's ability to specifically bind and sequester DHT from the androgen receptor have important ramifications in health and disease and may indicate a broad and important usage for equol in the treatment of androgen-mediated pathologies.
Collapse
Affiliation(s)
- Trent D Lund
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80524, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Nuñez JL, Huppenbauer CB, McAbee MD, Juraska JM, DonCarlos LL. Androgen receptor expression in the developing male and female rat visual and prefrontal cortex. JOURNAL OF NEUROBIOLOGY 2003; 56:293-302. [PMID: 12884268 DOI: 10.1002/neu.10236] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gonadal steroid hormones are known to influence the development of the cerebral cortex of mammals. Steroid hormone action involves hormone binding to cytoplasmic or nuclear receptors, followed by DNA binding and gene transcription. The goals of the present study were twofold: to determine whether androgen receptors are present during development in two known androgen sensitive regions of the rat cerebral cortex, the primary visual cortex (Oc1) and the anterior cingulate/frontal cortex (Cg1/Fr2); and to determine whether androgen receptor (AR) expression in these regions differs between developing males and females. We used immunocytochemistry to detect AR protein on postnatal days 0, 4, and 10, and in situ hybridization to detect AR mRNA on postnatal day 10 in male and female rats. The level of AR expression was specific to the cortical region, with higher AR immunoreactive cell density and more AR mRNA in Oc1 than in Cg1/Fr2. AR immunoreactive cell density increased with age in both regions. Finally, on postnatal day 10, males had a higher AR immunoreactive cell density and more AR mRNA in Oc1 than did females. Thus, the presence of ARs may allow androgens to directly influence the development the cerebral cortex.
Collapse
Affiliation(s)
- J L Nuñez
- Neuroscience Program and Department of Psychology, University of Illinois, Champaign, Illinois 61820, USA
| | | | | | | | | |
Collapse
|
38
|
Mihalick SM. Perinatal exposure to diethylstilbestrol improves olfactory discrimination learning in male and female Swiss-Webster mice. Neurobiol Learn Mem 2003; 80:55-62. [PMID: 12737934 DOI: 10.1016/s1074-7427(03)00020-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
During late prenatal and early postnatal brain development, estrogen induces structural sex differences that correspond to behavioral differences in certain domains such as learning and memory. The typically superior performance of males is attributed to the action of elevated concentrations of estrogen, derived inside neurons from the aromatization of testosterone. In contrast, female performance appears dependent on minimal estrogenic activity. Rat models of the relationship between hormones and cognitive behavior predominate the field, but the advent of genetically modified mice as research tools necessitates development of analogous mouse models. This study examined how early postnatal exposure to the synthetic estrogen diethylstilbestrol (DES) affected the ability of male and female Swiss-Webster mice to learn a two-choice olfactory discrimination and three repeated reversals. Mice treated with subcutaneous injections of DES from postnatal days 1-10 learned reversals more readily than oil-treated controls, a difference that became evident after repeated testing. DES-exposed males and females learned reversals at a comparable rate, suggesting that early postnatal estrogen exposure does not influence this mode of learning through a sexually differentiated mechanism in mice. An analysis of response patterns during qualitatively different phases of reversal learning revealed that DES-induced improvements probably were not due to greater inhibitory control. Instead, DES appeared to enhance associative ability. Early postnatal estrogen exposure may have the potential to preserve certain cognitive skills in adulthood.
Collapse
Affiliation(s)
- Sheila M Mihalick
- Eunice Kennedy Shriver Center for Mental Retardation, University of Massachusetts Medical School, USA.
| |
Collapse
|
39
|
Abstract
We have developed a model for prenatal hypoxia-ischemia in which muscimol, a selective gamma-aminobutyric acid A (GABA(A)) receptor agonist, administered to newborn rats, induces hippocampal damage. In the neonatal rat brain, activation of GABA(A) receptors leads to membrane depolarization and neuronal excitation. Because of our previous detection of sex differences in this model and the considerable interest in the neuroprotective effects of estradiol in the adult brain, we now investigate the effect of pretreatment with high physiological levels of estradiol in our model of prenatal hypoxia-ischemia. We used unbiased stereology to assess neuron number in the hippocampal formation of control, muscimol-treated, and estradiol- plus muscimol-treated animals. Muscimol decreased neuron number in the hippocampus, with damage exacerbated by pretreatment with estradiol. A hippocampal culture paradigm was developed to mirror the in vivo investigation. We observed elevated cytotoxicity (using the lactate dehydrogenase assay) by 48 h after treatment with estradiol plus muscimol, but decreased cytotoxicity between 2 and 24 h after treatment. To determine whether the actions of estradiol on muscimol-induced damage were via the estrogen receptor, hippocampal cultures were pretreated with ICI 182,780, a selective estrogen receptor antagonist. Treatment with ICI 182,780 blocked the potentiating effect of estradiol on the late period of cytotoxicity, but had no effect on the protective actions of estradiol during the early period of cytotoxicity. There appears to be a biphasic action of estradiol in our model of neonatal brain injury that involves early nongenomic, nonreceptor-mediated protection, followed by late deleterious receptor-mediated effects.
Collapse
Affiliation(s)
- Joseph L Nuñez
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | |
Collapse
|
40
|
Hilton GD, Nuñez JL, McCarthy MM. Sex differences in response to kainic acid and estradiol in the hippocampus of newborn rats. Neuroscience 2003; 116:383-91. [PMID: 12559094 DOI: 10.1016/s0306-4522(02)00716-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Premature and full-term human infants are at considerable risk of excitotoxic-mediated brain damage due to hypoxia-ischemia, infection or other trauma. Glutamate receptor activation is a major source of excitoxicity in the adult and developing brain, and the hippocampus is particularly vulnerable to damage. The seven-day-old rat is a widely used model of pediatric brain damage, in large part due to the relative insensitivity of the brain to exogenous glutamate treatment prior to this age. We have reexamined the possible role of glutamate in pediatric brain damage in the newborn rat using kainic acid treatment and attending to the sex of the animal as well as the effects of pretreatment with the gonadal steroid estradiol. Consistent with previous studies, we found no evidence of damage 7 days posttreatment in the CA1 region of the hippocampus in males or females. There was also little to no damage in the CA2/3 or dentate gyrus of males. In females, however, kainic-acid treatment induced substantial damage in the dentate gyrus and moderate damage in CA2/3, as assessed by neuron number and regional volume. Pretreatment with estradiol was protective against kainic acid-induced damage in females but was permissive for damage in the dentate gyrus of males. Estradiol treatment in the absence of kainic acid treatment was also neuroprotective in females in that it increased neuron number and volume throughout the hippocampal formation, suggesting that the basis of the sex difference observed in hippocampal volume was hormonally mediated. There was no effect of exogenous estradiol given to males in the absence of kainic acid. We conclude that the newborn female rat brain, but not the male, is sensitive to glutamate-mediated toxicity and that gonadal steroids play a complex role in both naturally occurring sex differences in hippocampal volume and response to injury.
Collapse
Affiliation(s)
- G D Hilton
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
41
|
Nuñez JL, Sodhi J, Juraska JM. Ovarian hormones after postnatal day 20 reduce neuron number in the rat primary visual cortex. JOURNAL OF NEUROBIOLOGY 2002; 52:312-21. [PMID: 12210098 DOI: 10.1002/neu.10092] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previous work from our lab has documented a sex difference in neuron number in the binocular region of the adult rat primary visual cortex (Oc1B), with males having 19% more neurons than females. In the present study, the role of developmental steroid hormones in the formation of this difference was explored. Male and female rats underwent neonatal hormone manipulation (female + testosterone or dihydrotestosterone; male + flutamide) followed by gonadectomy on postnatal day 20. Animals that did not undergo hormone manipulation were either gonadectomized or sham operated at day 20. Neuron number was quantified in the monocular (Oc1M) and binocular (Oc1B) subfields of the adult rat primary visual cortex using the optical disector technique. As adults, day 20 gonadectomized females, as well as females + testosterone and females + dihydrotestosterone, had significantly more neurons than intact females. There was no difference in neuron number between postnatal day 20 gonadectomized males, males + flutamide, and intact males. Also, intact males had significantly more neurons than intact females in both in Oc1M and Oc1B. It appears that ovarian steroids after day 20 are the primary cause of the lower number of neurons in the primary visual cortex of the female rat.
Collapse
Affiliation(s)
- Joseph L Nuñez
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | |
Collapse
|
42
|
Abstract
Oestrogens induce the development of female reproductive tissues. Endogenous human oestrogens include oestradiol, oestrone and oestriol. Oestrogen signalling in target tissues is dependent on the tissue concentration of oestrogen and the interaction of oestrogen receptors with an array of cell-specific co-regulator proteins. The diverse mechanisms of oestrogen signalling are complex and incompletely understood. In puberty, oestrogen is derived from both gonadal and peripheral sources. Originally, oestrogen was only thought to drive feminization in females; now, oestrogen is known to be important for pubertal development of males as well. Oestrogen is required for normal maturation of the neuroendocrine-gonadal axis and bone in both sexes, and a variety of other tissues are also responsive to oestrogen. Abnormal puberty can be associated with either excessive or inadequate oestrogen production. Girls deficient in oestrogen should receive replacement in physiological doses. Aromatase inhibitors and anti-oestrogens may prove to be useful therapeutic tools in some types of abnormal puberty.
Collapse
Affiliation(s)
- Laura C Alonso
- Department of Medicine, Section of Endocrinology, The University of Chicago Hospitals, 5841 S. Maryland Avenue, MC 1027, Chicago, IL 60637-1470, USA
| | | |
Collapse
|
43
|
McEwen BS. Invited review: Estrogens effects on the brain: multiple sites and molecular mechanisms. J Appl Physiol (1985) 2001; 91:2785-801. [PMID: 11717247 DOI: 10.1152/jappl.2001.91.6.2785] [Citation(s) in RCA: 474] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Besides their well-established actions on reproductive functions, estrogens exert a variety of actions on many regions of the nervous system that influence higher cognitive function, pain mechanisms, fine motor skills, mood, and susceptibility to seizures; they also appear to have neuroprotective actions in relation to stroke damage and Alzheimer's disease. Estrogen actions are now recognized to occur via two different intracellular estrogen receptors, ER-alpha and ER-beta, that reside in the cell nuclei of some nerve cells, as well as by some less well-characterized mechanisms. In the hippocampus, such nerve cells are sparse in number and yet appear to exert a powerful influence on synapse formation by neurons that do not have high levels of nuclear estrogen receptors. However, we also find nonnuclear estrogen receptors outside of the cell nuclei in dendrites, presynaptic terminals, and glial cells, where estrogen receptors may couple to second messenger systems to regulate a variety of cellular events and signal to the nuclear via transcriptional regulators such as CREB. Sex differences exist in many of the actions of estrogens in the brain, and the process of sexual differentiation appears to affect many brain regions outside of the traditional brain areas involved in reproductive functions. Finally, the aging brain is responsive to actions of estrogens, which have neuroprotective effects both in vivo and in vitro. However, in an animal model, the actions of estrogens on the hippocampus appear to be somewhat attenuated with age. In the future, estrogen actions over puberty and in pregnancy and lactation should be further explored and should be studied in both the hypothalamus and the extrahypothalamic regions.
Collapse
Affiliation(s)
- B S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, 1230 York Ave., New York, NY 10021, USA.
| |
Collapse
|
44
|
Dugard ML, Tremblay-Leveau H, Mellier D, Caston J. Prenatal exposure to ethinylestradiol elicits behavioral abnormalities in the rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2001; 129:189-99. [PMID: 11506863 DOI: 10.1016/s0165-3806(01)00205-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pregnant rats were i.p. injected with a solution of 17alpha-ethinylestradiol (15 microg kg(-1)) every day between day 9 and day 14 of pregnancy and the behavior of the offspring was compared to that of rats born from dams injected with the vehicle only during the same gestational period. The percentage of neonatal death was dramatically high in the prenatally treated group. Growth of the surviving animals was even better than that of controls, but when adult, they exhibited a number of behavioral abnormalities: increased spontaneous motor activity, decreased exploratory behavior, impaired cognitive processing, qualitatively different exploratory drive, and/or persevering behavior, increased anxiety-like behavior and social neophobia. These behavioral alterations, which resemble a number of psychiatric syndromes, suggest that ethinylestradiol altered the ontogenesis of different parts of the central nervous system involved in cognitive and emotional processes. However, it cannot be excluded that the changes in behavior of ethinylestradiol exposed offspring were due to the abnormal maternal behavior of the estradiol treated dams.
Collapse
Affiliation(s)
- M L Dugard
- Laboratoire PSY.CO, U.F.R. de Psychologie, Université de Rouen, 76821 Mont-Saint-Aignan Cedex, France
| | | | | | | |
Collapse
|
45
|
Savaskan E, Olivieri G, Meier F, Ravid R, Müller-Spahn F. Hippocampal estrogen beta-receptor immunoreactivity is increased in Alzheimer's disease. Brain Res 2001; 908:113-9. [PMID: 11454321 DOI: 10.1016/s0006-8993(01)02610-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Post-menopausal estrogen use reduces the risk and severity of Alzheimer's disease (AD). The present study investigates the distribution of both estrogen receptors ER alpha and ER beta in the human hippocampus in aged controls and in AD cases with immunohistochemistry. No ER alpha immunoreactivity was observed both in controls and in AD cases. On the other hand, ER beta was observed in some neuronal cells in the hippocampal subfields CA1--4, in astrocytes and in extracellular deposits both in controls and AD cases. The ER beta immunoreactivity was distinctly increased in all AD cases in cellular and extracellular localizations indicating a role for ER beta-mediated estrogen effects in AD-related neuropathology. This study provides the first demonstration of ER beta in human hippocampus in aged controls compared to AD cases.
Collapse
Affiliation(s)
- E Savaskan
- Department of Psychiatry, University of Basel, Wilhelm Klein-Str.27, CH-4025 Basel, Switzerland.
| | | | | | | | | |
Collapse
|
46
|
Solum DT, Handa RJ. Localization of estrogen receptor alpha (ER alpha) in pyramidal neurons of the developing rat hippocampus. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2001; 128:165-75. [PMID: 11412902 DOI: 10.1016/s0165-3806(01)00171-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
During development, estrogen has a variety of effects on morphological, biochemical and electrophysiological properties of hippocampal neurons. Correspondingly, estrogen receptor (ER) binding and mRNA increase transiently in the developing hippocampus. In this study, we used immunocytochemistry to determine the localization of the ER alpha subtype in the developing rat hippocampus. Nuclear staining was present in pyramidal cells and some interneurons of the CA1 and CA3 regions of the developing rat hippocampus. Little or no immunoreactivity was observed in postnatal day (P)0 animals (day of birth=P0), however, beginning on P4, ER alpha-immunoreactivity (ER alpha-ir) was visible and reached maximal levels by P10. These levels subsequently declined to low levels so that by P15, levels approximated those of adult females. Western blot analysis confirmed that this antibody recognized a 67 kDa protein, characteristic of the full-length ER alpha protein, in the hippocampus and pituitary. Furthermore, most of the ER alpha-immunopositive cells in the hippocampus were located in the pyramidal cell layer, and did not co-localize appreciably with gamma-aminobutyric acid (GABA) at any age examined. We conclude, based on the immunocytochemical localization of ER alpha, that the effects of estrogen on biochemistry and morphology of the developing hippocampus may be direct through the ER alpha subtype in hippocampal pyramidal cells.
Collapse
Affiliation(s)
- D T Solum
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University Chicago, Maywood, IL 60301, USA
| | | |
Collapse
|
47
|
Lee SJ, McEwen BS. Neurotrophic and neuroprotective actions of estrogens and their therapeutic implications. Annu Rev Pharmacol Toxicol 2001; 41:569-91. [PMID: 11264469 DOI: 10.1146/annurev.pharmtox.41.1.569] [Citation(s) in RCA: 400] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Originally known for its regulation of reproductive functions, estradiol, a lipophilic hormone that can easily cross plasma membranes as well as the blood-brain barrier, maintains brain systems subserving arousal, attention, mood, and cognition. In addition, both synthetic and natural estrogens exert neurotrophic and neuroprotective effects. There is increasing evidence that estrogen actions are mediated by nongenomic as well as direct and indirect genomic pathways. Although in vitro models have provided the most extensive evidence for neurotrophic and neuroprotective actions to date, there are also in vivo studies that support these actions.
Collapse
Affiliation(s)
- S J Lee
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10021, USA.
| | | |
Collapse
|
48
|
Price RH, Butler CA, Webb P, Uht R, Kushner P, Handa RJ. A splice variant of estrogen receptor beta missing exon 3 displays altered subnuclear localization and capacity for transcriptional activation. Endocrinology 2001; 142:2039-49. [PMID: 11316771 DOI: 10.1210/endo.142.5.8130] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
There are two separate estrogen receptors (ERs), ERalpha and ERbeta. The ERbeta gene is variably spliced, and in some cases variant expression is high. Besides the full-length ERbeta (equivalent to ERbeta1), splice variants can encode proteins bearing an insert within the ligand-binding domain (beta2), a deletion of exon 3 (ERbeta1delta3) disrupting the DNA-binding domain, or both (ERbeta2delta3). Here we examine the intracellular localization and transcriptional properties of each of the ERbeta splice variants heterologously expressed in cultured cells. In accordance with ERalpha, ERbeta1 and ERbeta2 are both distributed in a reticular pattern within the nucleus after exposure to ligand. In contrast, ERbeta1delta3 and ERbeta2delta3 localize to discrete spots within the nucleus in the presence of ER agonists. In the presence of ER antagonists, the delta3 variants are distributed diffusely within the nucleus. We also show that the spots are stable nuclear structures to which the delta3 variants localize in a ligand-dependent manner. Coactivator proteins of ER colocalize with delta3 variants in the spots in the presence of agonists. The delta3 variants of ERbeta can activate luciferase reporter constructs containing an activator protein complex-1 site, but not an estrogen response element (ERE). These data suggest that without an intact DNA-binding domain, ERbeta is functionally altered, allowing localization to discrete nuclear spots and activation from activator protein-1-containing reporter genes.
Collapse
Affiliation(s)
- R H Price
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, Maywood, Illinois 60153, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Estrogen modulates the morphology and physiology of the rat hippocampus and enhances cognitive function. While estrogen receptor (alpha and beta) messenger RNAs have been detected in the hippocampus, the presence of functional protein remains uncertain. The present study used a new radiolabeled estrogen, [125I]estrogen, and in vivo autoradiography to address this question. Nuclear uptake and retention of [125I]estrogen was detected in the pyramidal cells of CA1-CA3, with the majority of cells in the ventral horn of CA2 and CA3 being labeled. Additional labeled cells were scattered throughout the strata oriens and radiatum and the hilus of the dentate gyrus. Since the number and distribution of labeled cells in the hippocampus was more than expected, in situ hybridization was used to assess the localization of estrogen receptor (alpha and beta) messenger RNAs in this brain region. The results revealed that both estrogen receptors are expressed in regions where [125I]estrogen binding was seen, although the intensity of estrogen receptor-alpha hybridization signal appears to be stronger when compared with estrogen receptor-beta.The results of these studies have demonstrated the presence of estrogen receptors in rat hippocampus and shown that the distribution of binding sites was much greater than expected, particularly in the pyramidal cells of the ventral hippocampus. These observations challenge our current thinking about steroid hormones and their mechanism(s) of action in a region associated with learning and memory and affected by the neurodegenerative conditions of aging.
Collapse
Affiliation(s)
- P J Shughrue
- Women's Health Research Institute, Wyeth-Ayerst Research, 145 King of Prussia Road, Radnor, PA 19087, USA.
| | | |
Collapse
|
50
|
|