1
|
Kim GW, Park K, Kim YH, Jeong GW. Altered brain morphology and functional connectivity in postmenopausal women: automatic segmentation of whole-brain and thalamic subnuclei and resting-state fMRI. Aging (Albany NY) 2024; 16:4965-4979. [PMID: 38526330 PMCID: PMC11006480 DOI: 10.18632/aging.205662] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/07/2024] [Indexed: 03/26/2024]
Abstract
The transition to menopause is associated with various physiological changes, including alterations in brain structure and function. However, menopause-related structural and functional changes are poorly understood. The purpose of this study was not only to compare the brain volume changes between premenopausal and postmenopausal women, but also to evaluate the functional connectivity between the targeted brain regions associated with structural atrophy in postmenopausal women. Each 21 premenopausal and postmenopausal women underwent magnetic resonance imaging (MRI). T1-weighted MRI and resting-state functional MRI data were used to compare the brain volume and seed-based functional connectivity, respectively. In statistical analysis, multivariate analysis of variance, with age and whole brain volume as covariates, was used to evaluate surface areas and subcortical volumes between the two groups. Postmenopausal women showed significantly smaller cortical surface, especially in the left medial orbitofrontal cortex (mOFC), right superior temporal cortex, and right lateral orbitofrontal cortex, compared to premenopausal women (p < 0.05, Bonferroni-corrected) as well as significantly decreased functional connectivity between the left mOFC and the right thalamus was observed (p < 0.005, Monte-Carlo corrected). Although postmenopausal women did not show volume atrophy in the right thalamus, the volume of the right pulvinar anterior, which is one of the distinguished thalamic subnuclei, was significantly decreased (p < 0.05, Bonferroni-corrected). Taken together, our findings suggest that diminished brain volume and functional connectivity may be linked to menopause-related symptoms caused by the lower sex hormone levels.
Collapse
Affiliation(s)
- Gwang-Won Kim
- Advanced Institute of Aging Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kwangsung Park
- Advanced Institute of Aging Science, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Yun-Hyeon Kim
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Gwang-Woo Jeong
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
2
|
Oughli HA, Nguyen SA, Siddarth P, Fox M, Milillo M, Ercoli L, Lavretsky H. The Effect of Cumulative Lifetime Estrogen Exposure on Cognition in Depressed Versus Non-Depressed Older Women. J Geriatr Psychiatry Neurol 2022; 35:832-839. [PMID: 35410535 PMCID: PMC9574476 DOI: 10.1177/08919887221090216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Two-thirds of individuals living with Alzheimer's disease are women. Declining estrogen levels influence mood and cognition. Cumulative lifetime estrogen exposure (CLEE) correlates with cognition later in life. We examined the relationship of CLEE to depression and cognition in older women with major depression compared to non-depressed women. DESIGN Older women (age ≥60 years) with depression were compared to non-depressed women using a lifetime estrogen exposure questionnaire. CLEE was defined as combined durations of reproductive span (age of menopause minus age of menarche) and any post-menopausal hormone replacement therapy use. Higher vs lower CLEE groups were based on a median of 474 months of estrogen exposure. SETTING University hospital outpatient research program. PARTICIPANTS 135 women ≥60 years; 64 depressed and 71 non-depressed. MEASURMENTS Participants completed a comprehensive cognitive test battery. General linear models were used to examine the association between cognitive domain scores and CLEE in depressed and non-depressed women, controlling for age, education, and ethnicity. RESULTS Depressed and non-depressed groups had significantly different levels of CLEE, measured in months: mean 495.7 (SD 108.6) vs 456.4 (SD 66.0) months, F(1,130) = 5.01, p = .03. Within the non-depressed participants, higher CLEE was associated with improved delayed recall (F(1,59) = 5.94, p = .02, effect size = .61), while no such relationship was observed in the depressed group. CONCLUSION Higher CLEE was associated with improvement in delayed recall among non-depressed, but not among depressed participants. This suggests a protective role of estrogen on memory in non-depressed older postmenopausal women. Further research should examine the role of the CLEE in antidepressant response and cognitive decline.
Collapse
Affiliation(s)
- Hanadi Ajam Oughli
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| | - Sarah A. Nguyen
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| | - Prabha Siddarth
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| | - Molly Fox
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
- Department of Anthropology, University of California Los Angeles, Los Angeles, CA, USA
| | - Michaela Milillo
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| | - Linda Ercoli
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| | - Helen Lavretsky
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Noachtar IA, Hidalgo-Lopez E, Pletzer B. Duration of oral contraceptive use relates to cognitive performance and brain activation in current and past users. Front Endocrinol (Lausanne) 2022; 13:885617. [PMID: 36204097 PMCID: PMC9530450 DOI: 10.3389/fendo.2022.885617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies indicate effects of oral contraceptive (OC) use on spatial and verbal cognition. However, a better understanding of the OC effects is still needed, including the differential effects of androgenic or anti-androgenic OC use and whether the possible impact persists beyond the OC use. We aim to investigate the associations of OC use duration with spatial and verbal cognition, differentiating between androgenic and anti-androgenic OC. Using functional magnetic resonance imaging (MRI), we scanned a group of 94 past and current OC-users in a single session. We grouped current OC users (N=53) and past OC users with a natural cycle (N=41) into androgenic and anti-androgenic user. Effects of OC use duration were observed for current use and after discontinuation. Duration of OC use was reflected only in verbal fluency performance but not navigation: The longer the current OC use, the less words were produced in the verbal fluency task. During navigation, deactivation in the caudate and postcentral gyrus was duration-dependent in current androgenic OC users. Only during the verbal fluency task, duration of previous OC use affects several brain parameters, including activation of the left putamen and connectivity between right-hemispheric language areas (i.e., right inferior frontal gyrus and right angular gyrus). The results regarding performance and brain activation point towards stronger organizational effects of OCs on verbal rather than spatial processing. Irrespective of the task, a duration-dependent connectivity between the hippocampus and various occipital areas was observed. This could suggest a shift in strategy or processing style with long-term contraceptive use during navigation/verbal fluency. The current findings suggest a key role of the progestogenic component of OCs in both tasks. The influence of OC use on verbal fluency remains even after discontinuation which further points out the importance of future studies on OC effects and their reversibility.
Collapse
|
4
|
Ambrase A, Lewis CA, Barth C, Derntl B. Influence of ovarian hormones on value-based decision-making systems: Contribution to sexual dimorphisms in mental disorders. Front Neuroendocrinol 2021; 60:100873. [PMID: 32987043 DOI: 10.1016/j.yfrne.2020.100873] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/28/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Women and men exhibit differences in behavior when making value-based decisions. Various hypotheses have been proposed to explain these findings, stressing differences in functional lateralization of the brain, functional activation, neurotransmitter involvement and more recently, sex hormones. While a significant interaction of neurotransmitter systems and sex hormones has been shown for both sexes, decision-making in women might be particularly affected by variations of ovarian hormones. In this review we have gathered information from animal and human studies on how ovarian hormones affect decision-making processes in females by interacting with neurotransmitter systems at functionally relevant brain locations and thus modify the computation of decision aspects. We also review previous findings on impaired decision-making in animals and clinical populations with substance use disorder and depression, emphasizing how little we know about the role of ovarian hormones in aberrant decision-making.
Collapse
Affiliation(s)
- Aiste Ambrase
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany
| | - Carolin A Lewis
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; Emotion Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; International Max Planck Research School on Neuroscience of Communication: Function, Structure, and Plasticity, Leipzig, Germany
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany; TübingenNeuroCampus, University of Tübingen, Tübingen, Germany; LEAD Research School and Graduate Network, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Ishunina TA. Alternative splicing in aging and Alzheimer's disease: Highlighting the role of tau and estrogen receptor α isoforms in the hypothalamus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:177-189. [PMID: 34266591 DOI: 10.1016/b978-0-12-819973-2.00012-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human genes show the highest efficacy of alternative splicing (AS) in the brain as compared to other tissues. Within the brain, a remarkably rich diversity of AS events was identified in the hypothalamus. The AS frequency is increased in the aging brain. Such AS events, as intron retention and accumulation of circular RNAs, were acknowledged as some of the main hallmarks of the aging brain. In Alzheimer's disease (AD) pivotal (tau gene, in particular), risk, candidate and other genes show significant alterations in AS. Therefore AD has been suggested to be a disease of dysregulated AS. One of the reported risk factors for AD is estrogen deficiency that may interfere with the extension of neurobrillary tangles. Mounting evidence suggests that estrogens may decrease hyperphosphorylated tau deposition in the brain. Furthermore, AS of estrogen receptor α (ERα) mRNA is decreased in AD brain areas with the highest tau load. These potential interactions among tau, estrogens, and ERα AS may be important for the development of therapeutic and preventive strategies for AD. The intriguing point is that the amount of splice variants of ERα in the hypothalamus and the hippocampus is increased in aging and decreased in AD, while ERα is one of the regulators of AS and is subject to AS itself.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Department of Histology, Embryology and Cytology, Kursk State Medical University, Kursk, Russia.
| |
Collapse
|
6
|
Wang D, Lu R, Rempala G, Sadee W. Ligand-Free Estrogen Receptor α (ESR1) as Master Regulator for the Expression of CYP3A4 and Other Cytochrome P450 Enzymes in the Human Liver. Mol Pharmacol 2019; 96:430-440. [PMID: 31399483 PMCID: PMC6724575 DOI: 10.1124/mol.119.116897] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/01/2019] [Indexed: 12/25/2022] Open
Abstract
Cytochrome P450 3A4 isoform (CYP3A4) transcription is controlled by hepatic transcription factors (TFs), but how TFs dynamically interact remains uncertain. We hypothesize that several TFs form a regulatory network with nonlinear, dynamic, and hierarchical interactions. To resolve complex interactions, we have applied a computational approach for estimating Sobol's sensitivity indices (SSI) under generalized linear models to existing liver RNA expression microarray data (GSE9588) and RNA-seq data from genotype-tissue expression (GTEx), generating robust importance ranking of TF effects and interactions. The SSI-based analysis identified TFs and interacting TF pairs, triplets, and quadruplets involved in CYP3A4 expression. In addition to known CYP3A4 TFs, estrogen receptor α (ESR1) emerges as key TF with the strongest main effect and as the most frequently included TF interacting partner. Model predictions were validated using small interfering RNA (siRNA)/short hairpin RNA (shRNA) gene knockdown and clustered regularly interspaced short palindromic repeats (CRISPR)-mediated transcriptional activation of ESR1 in biliary epithelial Huh7 cells and human hepatocytes in the absence of estrogen. Moreover, ESR1 and known CYP3A4 TFs mutually regulate each other. Detectable in both male and female hepatocytes without added estrogen, the results demonstrate a role for unliganded ESR1 in CYP3A4 expression consistent with unliganded ESR1 signaling reported in other cell types. Added estrogen further enhances ESR1 effects. We propose a hierarchical regulatory network for CYP3A4 expression directed by ESR1 through self-regulation, cross regulation, and TF-TF interactions. We also demonstrate that ESR1 regulates the expression of other P450 enzymes, suggesting broad influence of ESR1 on xenobiotics metabolism in human liver. Further studies are required to understand the mechanisms underlying role of ESR1 in P450 regulation. SIGNIFICANCE STATEMENT: This study focuses on identifying key transcription factors and regulatory networks for CYP3A4, the main drug metabolizing enzymes in liver. We applied a new computational approach (Sobol's sensitivity analysis) to existing hepatic gene expression data to determine the role of transcription factors in regulating CYP3A4 expression, and used molecular genetics methods (siRNA/shRNA gene knockdown and CRISPR-mediated transcriptional activation) to test these interactions in life cells. This approach reveals a robust network of TFs, including their putative interactions and the relative impact of each interaction. We find that ESR1 serves as a key transcription factor function in regulating CYP3A4, and it appears to be acting at least in part in a ligand-free fashion.
Collapse
Affiliation(s)
- Danxin Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Clinical Sciences, Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas (R.L.); and Mathematical Bioscience Institute, (G.R.) and Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine (W.S.), Ohio State University, Columbus, Ohio
| | - Rong Lu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Clinical Sciences, Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas (R.L.); and Mathematical Bioscience Institute, (G.R.) and Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine (W.S.), Ohio State University, Columbus, Ohio
| | - Grzegorz Rempala
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Clinical Sciences, Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas (R.L.); and Mathematical Bioscience Institute, (G.R.) and Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine (W.S.), Ohio State University, Columbus, Ohio
| | - Wolfgang Sadee
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Clinical Sciences, Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas (R.L.); and Mathematical Bioscience Institute, (G.R.) and Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine (W.S.), Ohio State University, Columbus, Ohio
| |
Collapse
|
7
|
Anamthathmakula P, Kyathanahalli C, Ingles J, Hassan SS, Condon JC, Jeyasuria P. Estrogen receptor alpha isoform ERdelta7 in myometrium modulates uterine quiescence during pregnancy. EBioMedicine 2019; 39:520-530. [PMID: 30502052 PMCID: PMC6355643 DOI: 10.1016/j.ebiom.2018.11.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/04/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Circulating estrogen (E2) levels are high throughout pregnancy and increase towards term, however its local tissue specific actions vary across gestation. For example, myometrial E2 regulated uterotonic action is disabled until term, whereas it's proliferative function is maintained in the breast. We have identified gestationally regulated splicing events, mediated by hnRNPG and modulated by E2 that generate alternatively spliced estrogen receptor alpha (ERα) variants (ERΔ7 and ERα46) in the myometrium. These variants allow for differential, gestationally regulated, modulation of the uterotonic action of E2. METHODS Human myometrium isolated from preterm and term non-laboring and laboring pregnant women were analyzed for ERα isoforms and splice factor levels. Lentiviral mediated shRNA knockdown of hnRNPG and overexpression of ERΔ7 were performed in human myometrial (hTERT-HM) cells. Functional 3D collagen contraction assays were executed. FINDINGS ERΔ7 acts as a dominant negative repressor of the uterotonic action of ERα66 and ERα46 isoforms through the regulation of the myometrial gap junction protein GJA1. Elimination of hnRNPG inhibits the generation of ERΔ7 while overexpression of ERΔ7 inhibited GJA1 expression. Moreover in vivo human myometrial hnRNPG levels decline at term in an E2 dependent manner resulting in a withdrawal of ERΔ7 levels and its tocolytic action at term. INTERPRETATION Our findings implicate the unique role of ERΔ7 as a modulator of myometrial quiescence and define the mechanism of ERΔ7 generation, through hormonally regulated splicing events. FUND: This study was supported by NIH OPRU U01 supplement (HD047905), University of Pittsburgh and Wayne State University Perinatal Research Initiative (USA).
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Judith Ingles
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Perinatology Research Branch, NICHD, Bethesda, MD 20892, USA
| | - Jennifer C Condon
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Pancharatnam Jeyasuria
- Department of Obstetrics and Gynecology, Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
8
|
Shin NY, Bak Y, Nah Y, Han S, Kim DJ, Kim SJ, Lee JE, Lee SG, Lee SK. Disturbed retrieval network and prospective memory decline in postpartum women. Sci Rep 2018; 8:5476. [PMID: 29615788 PMCID: PMC5882973 DOI: 10.1038/s41598-018-23875-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 03/12/2018] [Indexed: 11/09/2022] Open
Abstract
Prospective memory (PM) refers to the ability to remember to execute an intended action in the future. For successful PM performance, both top-down strategic monitoring and bottom-up spontaneous retrieval processes need to be appropriately recruited. We assessed PM performance and used fMRI to discover relevant neural correlates and possible predictors for PM performance in 25 postpartum and 26 nulliparous age- and education-matched women. Postpartum women showed decreased PM performance, a higher number of nocturnal awakenings, and lower estradiol level. The postpartum women had decreased functional connectivity (FC) in the right hippocampus and ventral frontoparietal networks (FPN) during retrieval-dominant PM trials relative to maintenance-dominant ongoing trials in the PM block. On multivariate analyses, decreased FC between the right hippocampus and ventral FPN and a higher number of nocturnal awakenings were independent predictors for poor PM performance after adjusting for age, education, estradiol level, and depressive symptoms. On mediation analyses, the estradiol level was found to have an indirect effect on PM accuracy via altered FC as a mediator. This suggests that decreased FC within the spontaneous retrieval-related regions including the right hippocampus and ventral FPN, disrupted sleep rhythms, and decreased estradiol level may contribute to poor PM performance in postpartum women.
Collapse
Affiliation(s)
- Na-Young Shin
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yunjin Bak
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoonjin Nah
- Department of Psychology, Yonsei University, Seoul, Korea
| | - Sanghoon Han
- Department of Psychology, Yonsei University, Seoul, Korea
| | - Dong Joon Kim
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Se Joo Kim
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Guk Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Owens SJ, Murphy CE, Purves-Tyson TD, Weickert TW, Shannon Weickert C. Considering the role of adolescent sex steroids in schizophrenia. J Neuroendocrinol 2018; 30. [PMID: 28941299 DOI: 10.1111/jne.12538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/06/2017] [Accepted: 09/20/2017] [Indexed: 12/29/2022]
Abstract
Schizophrenia is a disabling illness that is typically first diagnosed during late adolescence to early adulthood. It has an unremitting course and is often treatment-resistant. Many clinical aspects of the illness suggest that sex steroid-nervous system interactions may contribute to the onset, course of symptoms and the cognitive impairment displayed by men and women with schizophrenia. Here, we discuss the actions of oestrogen and testosterone on the brain during adolescent development and in schizophrenia from the perspective of experimental studies in animals, human post-mortem studies, magnetic resonance imaging studies in living humans and clinical trials of sex steroid-based treatments. We present evidence of potential beneficial, as well as detrimental, effects of both testosterone and oestrogen. We provide a rationale for the necessity to further elucidate sex steroid mechanisms of action at different ages, sexes and brain regions to more fully understand the role of testosterone and oestrogen in the pathophysiology of schizophrenia. The weight of the evidence suggests that sex steroid hormones influence mammalian brain function, including both cognition and emotion, and that pharmaceutical agents aimed at sex steroid receptors appear to provide a novel treatment avenue to reduce symptoms and improve cognition in men and women with schizophrenia.
Collapse
Affiliation(s)
- S J Owens
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - C E Murphy
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - T D Purves-Tyson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - T W Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - C Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
10
|
Orhan FÖ, Kurutaş EB, Doğaner A, Türker E, Özcü SŞT, Güngör M, Çakmak S. Serum levels of GPER-1 in euthymic bipolar patients. Neuropsychiatr Dis Treat 2018; 14:855-862. [PMID: 29618927 PMCID: PMC5875407 DOI: 10.2147/ndt.s158822] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Estrogen and its receptors have been suggested as playing a role in the pathogenesis of bipolar disorder (BD). Estrogen functions through the estrogen receptors alpha and beta and the recently discovered G-protein-coupled estrogen receptor-1 (GPER-1). The aim of this study was to evaluate serum GPER-1 levels in euthymic BD patients. PATIENTS AND METHODS The study population consisted of 38 euthymic outpatients meeting the criteria for BD in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition and 35 age- and gender-matched healthy controls. Medical histories were obtained and physical examinations and laboratory tests conducted. RESULTS Serum GPER-1 levels were measured in both patients and controls and found to be significantly higher in the BD patients than in controls. These results were not influenced by the medications in use. CONCLUSION The results of this study demonstrated that GPER-1 may play a role in BD pathophysiology.
Collapse
Affiliation(s)
- Fatma Özlem Orhan
- Department of Psychiatry, Faculty of Medicine, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| | - Ergül Belge Kurutaş
- Department of Biochemistry, Faculty of Medicine, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| | - Adem Doğaner
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| | - Ebru Türker
- Department of Psychiatry, Faculty of Medicine, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| | - Safiye Şeyma Taner Özcü
- Department of Bioengineering, Institute of Science, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| | - Meltem Güngör
- Vocational High School, Toros University, Mersin, Turkey
| | - Seyfettin Çakmak
- Department of Psychiatry, Faculty of Medicine, Kahramanmaraş Sütçüimam University, Kahramanmaras, Turkey
| |
Collapse
|
11
|
Maney DL. Polymorphisms in sex steroid receptors: From gene sequence to behavior. Front Neuroendocrinol 2017; 47:47-65. [PMID: 28705582 PMCID: PMC6312198 DOI: 10.1016/j.yfrne.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/05/2017] [Accepted: 07/08/2017] [Indexed: 01/09/2023]
Abstract
Sex steroid receptors have received much interest as potential mediators of human behaviors and mental disorders. Candidate gene association studies have identified about 50 genetic variants of androgen and estrogen receptors that correlate with human behavioral phenotypes. Because most of these polymorphisms lie outside coding regions, discerning their effect on receptor function is not straightforward. Thus, although discoveries of associations improve our ability to predict risk, they have not greatly advanced our understanding of underlying mechanisms. This article is intended to serve as a starting point for psychologists and other behavioral biologists to consider potential mechanisms. Here, I review associations between polymorphisms in sex steroid receptors and human behavioral phenotypes. I then consider ways in which genetic variation can affect processes such as mRNA transcription, splicing, and stability. Finally, I suggest ways that hypotheses about mechanism can be tested, for example using in vitro assays and/or animal models.
Collapse
Affiliation(s)
- Donna L Maney
- Department of Psychology, 36 Eagle Row, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
12
|
Ishii H, Hattori Y, Munetomo A, Watanabe H, Sakuma Y, Ozawa H. Characterization of rodent constitutively active estrogen receptor α variants and their constitutive transactivation mechanisms. Gen Comp Endocrinol 2017; 248:16-26. [PMID: 28412386 DOI: 10.1016/j.ygcen.2017.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/24/2017] [Accepted: 04/12/2017] [Indexed: 12/17/2022]
Abstract
Estrogen receptor α (ERα) mRNAs exhibit remarkable heterogeneity owing to complicated alternative splicing. Some encode C-terminally-truncated ERα proteins, which display ligand-independent transactivation or dominant-negative activity. We previously characterized C-terminally-truncated ERα mRNA variants with cryptic sequences in humans and mice, and demonstrated that helices in the ligand-binding domains (LBDs) of ERα variants contribute to ligand-independent transcriptional activity. However, existence of non-conventional coding exons and generation of constitutively active ERα variants have remained to be examined in rats. To comparatively analyze modular organization and splicing profiles of the ERα genes, the range of C-terminally-truncated ERα variants was explored in rats and mice using rapid amplification of cDNA ends and RT-PCR cloning. Furthermore, their functions were determined in transiently transfected cells using expression constructs and luciferase reporter assays. Multiple cryptic exons and C-terminally-truncated ERα variant mRNAs were identified in rats and mice. Naturally occurring and artificially truncated variants/constructs lacking helix 5 potentially exhibited gain-of-function in transfected cells. Although cryptic exons and splicing profiles were poorly conserved among humans, mice, and rats, constitutively active variants were generated from the ERα genes. Moreover, the primary mechanism of ligand-independent activation by C-terminally-truncated ERα variants is C-terminus to helix 5 deletion in the LBD. This comparative study documented the complexity of ERα genes, mRNAs, and proteins, and further determined the underlying structural basis of ligand-independent activation by C-terminally-truncated ERα variants.
Collapse
Affiliation(s)
- Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Yujiro Hattori
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Neurosurgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Arisa Munetomo
- Laboratory of Behavior Neuroscience, Faculty of Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo 112-8681, Japan.
| | - Hiroshi Watanabe
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Yasuo Sakuma
- University of Tokyo Health Sciences, 4-11 Ochiai, Tama, Tokyo 206-0033, Japan.
| | - Hitoshi Ozawa
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| |
Collapse
|
13
|
Lai YJ, Yu D, Zhang JH, Chen GJ. Cooperation of Genomic and Rapid Nongenomic Actions of Estrogens in Synaptic Plasticity. Mol Neurobiol 2016; 54:4113-4126. [PMID: 27324789 PMCID: PMC5509832 DOI: 10.1007/s12035-016-9979-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022]
Abstract
Neuroplasticity refers to the changes in the molecular and cellular processes of neural circuits that occur in response to environmental experiences. Clinical and experimental studies have increasingly shown that estrogens participate in the neuroplasticity involved in cognition, behavior, and memory. It is generally accepted that estrogens exert their effects through genomic actions that occur over a period of hours to days. However, emerging evidence indicates that estrogens also rapidly influence the neural circuitry through nongenomic actions. In this review, we provide an overview of the genomic and nongenomic actions of estrogens and discuss how these actions may cooperate in synaptic plasticity. We then summarize the role of epigenetic modifications, synaptic protein synthesis, and posttranslational modifications, and the splice variants of estrogen receptors in the complicated network of estrogens. The combination of genomic and nongenomic mechanisms endows estrogens with considerable diversity in modulating neural functions including synaptic plasticity.
Collapse
Affiliation(s)
- Yu-Jie Lai
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College of Central South University, Haikou Municipal Hospital, Haikou, Hainan, 570208, China
| | - Dan Yu
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College of Central South University, Haikou Municipal Hospital, Haikou, Hainan, 570208, China
| | - John H Zhang
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China.
| |
Collapse
|
14
|
Adjunctive selective estrogen receptor modulator increases neural activity in the hippocampus and inferior frontal gyrus during emotional face recognition in schizophrenia. Transl Psychiatry 2016; 6:e795. [PMID: 27138794 PMCID: PMC5070055 DOI: 10.1038/tp.2016.59] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/29/2016] [Accepted: 03/05/2016] [Indexed: 12/18/2022] Open
Abstract
Estrogen has been implicated in the development and course of schizophrenia with most evidence suggesting a neuroprotective effect. Treatment with raloxifene, a selective estrogen receptor modulator, can reduce symptom severity, improve cognition and normalize brain activity during learning in schizophrenia. People with schizophrenia are especially impaired in the identification of negative facial emotions. The present study was designed to determine the extent to which adjunctive raloxifene treatment would alter abnormal neural activity during angry facial emotion recognition in schizophrenia. Twenty people with schizophrenia (12 men, 8 women) participated in a 13-week, randomized, double-blind, placebo-controlled, crossover trial of adjunctive raloxifene treatment (120 mg per day orally) and performed a facial emotion recognition task during functional magnetic resonance imaging after each treatment phase. Two-sample t-tests in regions of interest selected a priori were performed to assess activation differences between raloxifene and placebo conditions during the recognition of angry faces. Adjunctive raloxifene significantly increased activation in the right hippocampus and left inferior frontal gyrus compared with the placebo condition (family-wise error, P<0.05). There was no significant difference in performance accuracy or reaction time between active and placebo conditions. To the best of our knowledge, this study provides the first evidence suggesting that adjunctive raloxifene treatment changes neural activity in brain regions associated with facial emotion recognition in schizophrenia. These findings support the hypothesis that estrogen plays a modifying role in schizophrenia and shows that adjunctive raloxifene treatment may reverse abnormal neural activity during facial emotion recognition, which is relevant to impaired social functioning in men and women with schizophrenia.
Collapse
|
15
|
Hattori Y, Ishii H, Munetomo A, Watanabe H, Morita A, Sakuma Y, Ozawa H. Human C-terminally truncated ERα variants resulting from the use of alternative exons in the ligand-binding domain. Mol Cell Endocrinol 2016; 425:111-22. [PMID: 26835991 DOI: 10.1016/j.mce.2016.01.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 11/29/2022]
Abstract
The nuclear receptor genes contain alternative internal and terminal exons, with alternative exon incorporation yielding mRNA variants that encode various receptor types, including some with C-terminal truncation that exhibit constitutive activation or dominant-negative transcriptional transactivation. However, C-terminally truncated estrogen receptor α (ERα) variants with alternative sequences have rarely been reported in humans. Therefore, we assessed human ERα genomic organization and alternative splicing profiles, and identified both alternative exons and C-terminally truncated ERα variants. These naturally occurring C-terminally truncated ERα proteins were localized in the nuclei of transfected cells. In addition, ERαi45c and ERαΔ5 variants exhibited constitutive transactivation of an estrogen responsive element-driven promoter in transfected cells. We manufactured expression vectors encoding artificially truncated ERα constructs and evaluated their transactivation abilities to establish mechanisms determining the constitutive activity and dominant-negative properties of truncated variants. Lack of the region encoded in exon 8 eliminated basal and ligand-induced transcriptional transactivation. The C-terminally truncated ERα variants/constructs containing the helices 5 in their ligand-binding domains did not exhibit constitutive transactivation. Furthermore, we demonstrated that truncation from C-termini to helices 5 in the variant ligand-binding domains was required for constitutive activation and found that the remnant regions of the ligand-binding domains and variant-specific sequences influenced transcriptional transactivation efficiency. In conclusion, we elucidated the structural and functional features of novel C-terminally truncated ERα variants and revealed the mechanisms underlying constitutive transactivation by C-terminally truncated nuclear receptor variants.
Collapse
Affiliation(s)
- Yujiro Hattori
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Neurosurgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Arisa Munetomo
- Laboratory of Behavior Neuroscience, Faculty of Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo 112-8681, Japan
| | - Hiroshi Watanabe
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Akio Morita
- Department of Neurosurgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yasuo Sakuma
- University of Tokyo Health Sciences, 4-11 Ochiai, Tama, Tokyo 206-0033, Japan
| | - Hitoshi Ozawa
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| |
Collapse
|
16
|
Hampson E, Phillips SD, Duff-Canning SJ, Evans KL, Merrill M, Pinsonneault JK, Sadée W, Soares CN, Steiner M. Working memory in pregnant women: Relation to estrogen and antepartum depression. Horm Behav 2015; 74:218-27. [PMID: 26187710 PMCID: PMC8693635 DOI: 10.1016/j.yhbeh.2015.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 10/23/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Subjective changes in concentration and memory are commonly reported by women during the second or third trimesters of pregnancy, but the nature of the problem is poorly understood. We hypothesized that these self-reports might reflect difficulties in working memory (WM). It was further hypothesized that antepartum depression (depression arising during pregnancy) may play an etiological role, either on its own or due to secondary changes in endocrine function or sleep. Using WM tasks that emphasized executive control processes mediated by the prefrontal cortex (PFC) we compared pregnant women tested at 34-36 weeks of gestation (n = 28) with age- and education-matched non-pregnant controls (n = 26). All pregnant women were screened for depression. Evidence of a WM disturbance was found, and was evident only among pregnant women showing depressive symptoms. In contrast, pregnant women who were not depressed showed WM performance that equalled, or even significantly exceeded, non-pregnant controls. No significant differences were observed on control tests of other cognitive functions. Multiple regression revealed that serum estradiol concentrations, along with severity of depressive affect but not sleep disruption, significantly predicted variation in the WM scores. In agreement with studies of estradiol and WM in other contexts, higher estradiol was associated with better WM, while higher levels of depressive symptoms predicted poorer WM. We conclude that memory disturbance during gestation might not be as widespread as commonly believed, but can be seen among women experiencing antepartum depression. The high level of WM performance found in healthy, non-depressed, pregnant women is discussed from an adaptationist perspective.
Collapse
Affiliation(s)
- Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, ON, Canada; Graduate Program in Neuroscience, University of Western Ontario, London, ON, Canada.
| | - Shauna-Dae Phillips
- Women's Health Concerns Clinic, St. Joseph's Healthcare and Medical Sciences Program, McMaster University, Hamilton, ON, Canada
| | | | - Kelly L Evans
- Department of Psychology, University of Western Ontario, London, ON, Canada
| | - Mia Merrill
- Graduate Program in Neuroscience, University of Western Ontario, London, ON, Canada
| | - Julia K Pinsonneault
- Department of Pharmacology and Program in Pharmacogenomics, Ohio State University, Columbus, OH, USA
| | - Wolfgang Sadée
- Department of Pharmacology and Program in Pharmacogenomics, Ohio State University, Columbus, OH, USA
| | - Claudio N Soares
- Women's Health Concerns Clinic, St. Joseph's Healthcare, Department of Psychiatry and Behavioural Neurosciences and Obstetrics & Gynecology, McMaster University, Hamilton, ON, Canada
| | - Meir Steiner
- Women's Health Concerns Clinic, St. Joseph's Healthcare, Department of Psychiatry and Behavioural Neurosciences and Obstetrics & Gynecology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
17
|
Weickert TW, Weinberg D, Lenroot R, Catts SV, Wells R, Vercammen A, O'Donnell M, Galletly C, Liu D, Balzan R, Short B, Pellen D, Curtis J, Carr VJ, Kulkarni J, Schofield PR, Weickert CS. Adjunctive raloxifene treatment improves attention and memory in men and women with schizophrenia. Mol Psychiatry 2015; 20:685-94. [PMID: 25980345 PMCID: PMC4444978 DOI: 10.1038/mp.2015.11] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/03/2014] [Accepted: 12/19/2014] [Indexed: 11/09/2022]
Abstract
There is increasing clinical and molecular evidence for the role of hormones and specifically estrogen and its receptor in schizophrenia. A selective estrogen receptor modulator, raloxifene, stimulates estrogen-like activity in brain and can improve cognition in older adults. The present study tested the extent to which adjunctive raloxifene treatment improved cognition and reduced symptoms in young to middle-age men and women with schizophrenia. Ninety-eight patients with a diagnosis of schizophrenia or schizoaffective disorder were recruited into a dual-site, thirteen-week, randomized, double-blind, placebo-controlled, crossover trial of adjunctive raloxifene treatment in addition to their usual antipsychotic medications. Symptom severity and cognition in the domains of working memory, attention/processing speed, language and verbal memory were assessed at baseline, 6 and 13 weeks. Analyses of the initial 6-week phase of the study using a parallel groups design (with 39 patients receiving placebo and 40 receiving raloxifene) revealed that participants receiving adjunctive raloxifene treatment showed significant improvement relative to placebo in memory and attention/processing speed. There was no reduction in symptom severity with treatment compared with placebo. There were significant carryover effects, suggesting some cognitive benefits are sustained even after raloxifene withdrawal. Analysis of the 13-week crossover data revealed significant improvement with raloxifene only in attention/processing speed. This is the first study to show that daily, oral adjunctive raloxifene treatment at 120 mg per day has beneficial effects on attention/processing speed and memory for both men and women with schizophrenia. Thus, raloxifene may be useful as an adjunctive treatment for cognitive deficits associated with schizophrenia.
Collapse
Affiliation(s)
- T W Weickert
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia,School of Psychiatry University of New South Wales Neuroscience Research Australia Barker Street, Randwick 2031, New South Wales Australia. E-mail:
| | - D Weinberg
- Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - R Lenroot
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia
| | - S V Catts
- Neuroscience Research Australia, Randwick, New South Wales, Australia,School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - R Wells
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - A Vercammen
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia,School of Psychology, Australian Catholic University, Strathfield, New South Wales, Australia
| | - M O'Donnell
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - C Galletly
- Discipline of Psychiatry, School of Medicine, the University of Adelaide, Adelaide, South Australia, Australia,Northern Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - D Liu
- Discipline of Psychiatry, School of Medicine, the University of Adelaide, Adelaide, South Australia, Australia,Northern Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - R Balzan
- Discipline of Psychiatry, School of Medicine, the University of Adelaide, Adelaide, South Australia, Australia,School of Psychology, Flinders University, Adelaide, South Australia, Australia
| | - B Short
- Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - D Pellen
- Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - J Curtis
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia
| | - V J Carr
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia
| | - J Kulkarni
- Alfred Psychiatric Research Centre, Melbourne, Victoria, Australia
| | - P R Schofield
- Neuroscience Research Australia, Randwick, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia,School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - C S Weickert
- School of Psychiatry, University of New South Wales, Kensington, New South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales, Australia,Schizophrenia Research Institute, Darlinghurst, New South Wales, Australia
| |
Collapse
|
18
|
Ji E, Weickert CS, Lenroot R, Catts SV, Vercammen A, White C, Gur RE, Weickert TW. Endogenous testosterone levels are associated with neural activity in men with schizophrenia during facial emotion processing. Behav Brain Res 2015; 286:338-46. [DOI: 10.1016/j.bbr.2015.03.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/07/2015] [Accepted: 03/11/2015] [Indexed: 12/17/2022]
|
19
|
Petersen N, Touroutoglou A, Andreano JM, Cahill L. Oral contraceptive pill use is associated with localized decreases in cortical thickness. Hum Brain Mapp 2015; 36:2644-54. [PMID: 25832993 DOI: 10.1002/hbm.22797] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 01/10/2023] Open
Abstract
Oral contraceptive pills (OCs), which are used to prevent pregnancy by the majority of women in the United States, contain steroid hormones that may affect the brain's structure and function. In this investigation, we tested the hypothesis that OC use is associated with differences in brain structure using a hypothesis-driven, surface-based approach. In 90 women, (44 OC users, 46 naturally-cycling women), we compared the cortical thickness of brain regions that participate in the salience network and the default mode network, as well as the volume of subcortical regions in these networks. We found that OC use was associated with significantly lower cortical thickness measurements in the lateral orbitofrontal cortex and the posterior cingulate cortex. These regions are believed to be important for responding to rewards and evaluating internal states/incoming stimuli, respectively. Further investigations are needed to determine if cortical thinning in these regions are associated with behavioral changes, and also to identify whether OC use is causally or only indirectly related to these changes in brain morphology.
Collapse
Affiliation(s)
- Nicole Petersen
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, California.,Department of Neurobiology and Behavior, Bonney Research Laboratory, University of California, Irvine, California
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Massachusetts.,Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Massachusetts.,Psychiatric Neuroimaging Division, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Massachusetts
| | - Joseph M Andreano
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Massachusetts.,Psychiatric Neuroimaging Division, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Massachusetts
| | - Larry Cahill
- Department of Neurobiology and Behavior, Bonney Research Laboratory, University of California, Irvine, California
| |
Collapse
|
20
|
Barth C, Villringer A, Sacher J. Sex hormones affect neurotransmitters and shape the adult female brain during hormonal transition periods. Front Neurosci 2015; 9:37. [PMID: 25750611 PMCID: PMC4335177 DOI: 10.3389/fnins.2015.00037] [Citation(s) in RCA: 425] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/26/2015] [Indexed: 12/21/2022] Open
Abstract
Sex hormones have been implicated in neurite outgrowth, synaptogenesis, dendritic branching, myelination and other important mechanisms of neural plasticity. Here we review the evidence from animal experiments and human studies reporting interactions between sex hormones and the dominant neurotransmitters, such as serotonin, dopamine, GABA and glutamate. We provide an overview of accumulating data during physiological and pathological conditions and discuss currently conceptualized theories on how sex hormones potentially trigger neuroplasticity changes through these four neurochemical systems. Many brain regions have been demonstrated to express high densities for estrogen- and progesterone receptors, such as the amygdala, the hypothalamus, and the hippocampus. As the hippocampus is of particular relevance in the context of mediating structural plasticity in the adult brain, we put particular emphasis on what evidence could be gathered thus far that links differences in behavior, neurochemical patterns and hippocampal structure to a changing hormonal environment. Finally, we discuss how physiologically occurring hormonal transition periods in humans can be used to model how changes in sex hormones influence functional connectivity, neurotransmission and brain structure in vivo.
Collapse
Affiliation(s)
- Claudia Barth
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences Leipzig, Germany ; Clinic of Cognitive Neurology, University of Leipzig Leipzig, Germany ; Leipzig Research Center for Civilization Diseases, University of Leipzig Leipzig, Germany ; Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig Leipzig, Germany ; Berlin School of Mind and Brain, Mind and Brain Institute Berlin, Germany
| | - Julia Sacher
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences Leipzig, Germany ; Clinic of Cognitive Neurology, University of Leipzig Leipzig, Germany
| |
Collapse
|
21
|
Hampson E, Levy-Cooperman N, Korman JM. Estradiol and mental rotation: relation to dimensionality, difficulty, or angular disparity? Horm Behav 2014; 65:238-48. [PMID: 24394702 DOI: 10.1016/j.yhbeh.2013.12.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 12/23/2013] [Accepted: 12/26/2013] [Indexed: 01/28/2023]
Abstract
Several studies have reported that performance on spatial rotation tests is better at menses than at high estradiol phases of the menstrual cycle in women. These effects are debated because nearly all reports of menstrual cycle variability have relied on a single test, the Mental Rotations Test (MRT, Vandenberg and Kuse, 1978). In the present study, we investigated key features of the MRT that might be responsible for its association with estradiol levels. We hypothesized that associations could be demonstrated for other tasks that share the same characteristics. Forty-four women ages 20-38 years, matched on education and general ability, were assessed at low (n=24) or high (n=20) estradiol stages of the menstrual cycle on a set of spatial tests that varied in dimensionality, plane of rotation, angular disparity, and effortfulness. Saliva was used to quantify estradiol and progesterone. Low estradiol was found to be associated with significantly better accuracy on the MRT and also on a mental rotation task that required large angles of rotation but employed only two-dimensional object representations and rotations limited to the picture plane. In contrast, a task using identical stimuli that required only small angles of rotation did not show an estradiol effect. A group difference also was seen on a test of perceptual closure. The results confirm that the estradiol effect is not limited to the MRT, and identify the rotational element, but also aspects of figural perception, as possible processes that may be responsive to estrogens. These findings advance our understanding by showing an association between estradiol and discrete spatial processes. Implications for understanding the origins of the robust sex difference commonly observed on the MRT are discussed.
Collapse
Affiliation(s)
- Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, Ontario, Canada; Graduate Program in Neuroscience, University of Western Ontario, London, Ontario, Canada.
| | | | - Jennifer M Korman
- Department of Psychology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
22
|
Moore L, Kyaw M, Vercammen A, Lenroot R, Kulkarni J, Curtis J, O'Donnell M, Carr VJ, Shannon Weickert C, Weickert TW. Serum testosterone levels are related to cognitive function in men with schizophrenia. Psychoneuroendocrinology 2013; 38:1717-28. [PMID: 23490072 DOI: 10.1016/j.psyneuen.2013.02.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 01/16/2013] [Accepted: 02/04/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sex steroids such as oestrogen and testosterone are potent neurodevelopmental hormones that also play a role in neuromodulation and neuroprotection of the mature brain. Sex steroid hormones may also be involved in the pathophysiology of schizophrenia as reduced circulating sex steroid levels and changes in brain sex steroid receptors are found in people with schizophrenia compared to controls. In men with schizophrenia, recent studies have documented an inverse correlation between serum testosterone and negative symptoms. Our study sought to confirm whether men with schizophrenia had lower levels of testosterone relative to controls and to determine whether lower testosterone levels were related to higher symptom severity and impaired cognition. METHOD Circulating serum hormone levels (testosterone, oestrogen, and prolactin), cognitive function and symptoms were assessed in 29 chronically ill men with schizophrenia or schizoaffective disorder. Twenty healthy men were recruited as a comparison group. A series of regression analyses were performed to determine the extent to which circulating sex steroid hormone levels predict cognition and symptoms in men with schizophrenia. RESULTS We did not find a significant difference in serum testosterone levels between groups. However, circulating testosterone levels significantly predicted performance on verbal memory, processing speed, and working memory in men with schizophrenia. With the exception of an effect of oestrogen on verbal memory, circulating sex steroid levels did not predict cognitive function in healthy men. Testosterone levels were not related to positive or negative symptom severity, but testosterone influenced excitement/hostility levels in our schizophrenia sample. CONCLUSIONS The results suggest that circulating sex steroids may modulate cognitive deficits associated with schizophrenia.
Collapse
Affiliation(s)
- L Moore
- Schizophrenia Research Institute, Darlinghurst, New South Wales (NSW), Australia; Neuroscience Research Australia, Randwick, NSW, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Acosta JI, Hiroi R, Camp BW, Talboom JS, Bimonte-Nelson HA. An update on the cognitive impact of clinically-used hormone therapies in the female rat: models, mazes, and mechanisms. Brain Res 2013; 1514:18-39. [PMID: 23333453 PMCID: PMC3739440 DOI: 10.1016/j.brainres.2013.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/09/2013] [Indexed: 01/05/2023]
Abstract
In women, ovarian hormone loss associated with menopause has been related to cognitive decline. Hormone therapy (HT) may ameliorate some of these changes. Understanding the cognitive impact of female steroids, including estrogens, progestogens, and androgens, is key to discovering treatments that promote brain health in women. The preclinical literature has presented elegant and methodical experiments allowing a better understanding of parameters driving the cognitive consequences of ovarian hormone loss and HT. Animal models have been a valuable tool in this regard, and will be vital to future discoveries. Here, we provide an update on the literature evaluating the impact of female steroid hormones on cognition, and the putative mechanisms mediating these effects. We focus on preclinical work that was done with an eye toward clinical realities. Parameters that govern the cognitive efficacy of HT, from what we know thus far, include but are not limited to: type, dose, duration, and route of HT, age at HT initiation, timing of HT relative to ovarian hormone loss, memory type examined, menopause history, and hormone receptor status. Researchers have identified intricate relationships between some of these factors by studying their individual effects on cognition. As of late, there is increased focus on studying interactions between these variables as well as multiple hormone types when administered concomitantly. This is key to translating preclinical data to the clinic, wherein women typically have concurrent exposure to endogenous ovarian hormones as well as exogenous combination HTs, which include both estrogens and progestins. Gains in understanding the parameters of HT effects on cognition provide exciting novel avenues that can inform clinical treatments, eventually expanding the window of opportunity to optimally enhance cognition and brain health in aging women. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- J I Acosta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | |
Collapse
|
24
|
Dominguez R, Dewing P, Kuo J, Micevych P. Membrane-initiated estradiol signaling in immortalized hypothalamic N-38 neurons. Steroids 2013; 78:607-13. [PMID: 23296142 PMCID: PMC3636190 DOI: 10.1016/j.steroids.2012.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 11/18/2022]
Abstract
Regulation of sexual reproduction by estradiol involves the activation of estrogen receptors (ERs) in the hypothalamus. Of the two classical ERs involved in reproduction, ERα appears to be the critical isoform. The role of ERα in reproduction has been found to involve a nuclear ERα that induces a genomic mechanism of action. More recently, a plasma membrane ERα has been shown to trigger signaling pathways involved in reproduction. Mechanisms underlying membrane-initiated estradiol signaling are emerging, including evidence that activation of plasma membrane ERα involves receptor trafficking. The present study examined the insertion of ERα into the plasma membrane of N-38 neurons, an immortalized murine hypothalamic cell line. We identified, using western blotting and PCR that N-38 neurons express full-length 66kDa ERα and a 52kDa ERα spliced variant missing the fourth exon - ERαΔ4. Using surface biotinylation, we observed that treatment of N-38 neurons with estradiol or with a membrane impermeant estradiol elevated plasma membrane ERα protein levels, indicating that membrane signaling increased receptor insertion into the cell membrane. Insertion of ERα was blocked by the ER antagonist ICI 182,780 or with the protein kinase C (PKC) pathway inhibitor bisindolylmaleimide (BIS). Downstream membrane-initiated signaling was confirmed by estradiol activation of PKC-theta (PKCθ) and the release of intracellular calcium. These results indicate that membrane ERα levels in N-38 neurons are dynamically autoregulated by estradiol.
Collapse
Affiliation(s)
- Reymundo Dominguez
- Laboratory of Neuroendocrinology of the Brain Research Institute, Departments of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1763, United States.
| | | | | | | |
Collapse
|
25
|
Micevych P, Christensen A. Membrane-initiated estradiol actions mediate structural plasticity and reproduction. Front Neuroendocrinol 2012; 33:331-41. [PMID: 22828999 PMCID: PMC3496015 DOI: 10.1016/j.yfrne.2012.07.003] [Citation(s) in RCA: 49] [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: 04/04/2012] [Revised: 07/11/2012] [Accepted: 07/13/2012] [Indexed: 12/17/2022]
Abstract
Over the years, our ideas about estrogen signaling have greatly expanded. In addition to estradiol having direct nuclear actions that mediate transcription and translation, more recent experiments have demonstrated membrane-initiated signaling. Both direct nuclear and estradiol membrane signaling can be mediated by the classical estrogen receptors, ERα and ERβ, which are two of the numerous putative membrane estrogen receptors. Thus far, however, only ERα has been shown to play a prominent role in regulating female reproduction and sexual behavior. Because ERα is a ligand-gated transcription factor and not a typical membrane receptor, trafficking to the cell membrane requires post-translational modifications. Two necessary modifications are palmitoylation and association with caveolins, a family of scaffolding proteins. In addition to their role in trafficking, caveolin proteins also serve to determine ERα interactions with metabotropic glutamate receptors (mGluRs). It is through these complexes that ERα, which cannot by itself activate G proteins, is able to initiate intracellular signaling. Various combinations of ERα-mGluR interactions have been demonstrated throughout the nervous system from hippocampus to striatum to hypothalamus to dorsal root ganglion (DRG) in both neurons and astrocytes. These combinations of ER and mGluR allow estradiol to have both facilitative and inhibitory actions in neurons. In hypothalamic astrocytes, the estradiol-mediated release of intracellular calcium stores regulating neurosteroid synthesis requires ERα-mGluR1a interaction. In terms of estradiol regulation of female sexual receptivity, activation of ERα-mGluR1a signaling complex leads to the release of neurotransmitters and alteration of neuronal morphology. This review will examine estradiol membrane signaling (EMS) activating a limbic-hypothalamic lordosis regulating circuit, which involves ERα trafficking, internalization, and modifications of neuronal morphology in a circuit that underlies female sexual receptivity.
Collapse
Affiliation(s)
- Paul Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1763, United States.
| | | |
Collapse
|
26
|
Hesson J. Cumulative estrogen exposure and prospective memory in older women. Brain Cogn 2012; 80:89-95. [DOI: 10.1016/j.bandc.2012.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 04/30/2012] [Accepted: 05/02/2012] [Indexed: 11/27/2022]
|
27
|
Sinchak K, Wagner EJ. Estradiol signaling in the regulation of reproduction and energy balance. Front Neuroendocrinol 2012; 33:342-63. [PMID: 22981653 PMCID: PMC3496056 DOI: 10.1016/j.yfrne.2012.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Our knowledge of membrane estrogenic signaling mechanisms and their interactions that regulate physiology and behavior has grown rapidly over the past three decades. The discovery of novel membrane estrogen receptors and their signaling mechanisms has started to reveal the complex timing and interactions of these various signaling mechanisms with classical genomic steroid actions within the nervous system to regulate physiology and behavior. The activation of the various estrogenic signaling mechanisms is site specific and differs across the estrous cycle acting through both classical genomic mechanisms and rapid membrane-initiated signaling to coordinate reproductive behavior and physiology. This review focuses on our current understanding of estrogenic signaling mechanisms to promote: (1) sexual receptivity within the arcuate nucleus of the hypothalamus, (2) estrogen positive feedback that stimulates de novo neuroprogesterone synthesis to trigger the luteinizing hormone surge important for ovulation and estrous cyclicity, and (3) alterations in energy balance.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Blvd., Long Beach, CA 90840-9502, United States.
| | | |
Collapse
|
28
|
Abstract
The peak in incidence for schizophrenia is during late adolescence for both sexes, but within this time frame the peak is both earlier and steeper for males. Additionally, women have a second peak in incidence following menopause. Two meta-analyses have reported that men have an overall ∼40% greater chance of developing schizophrenia than do women (Aleman et al., 2003; McGrath et al., 2004). These and other findings have led to the suggestion that ovarian hormones may be protective against schizophrenia. Less explored is the potential role of testosterone in schizophrenia, although disruptions in steroid levels have also been reported in men with the illness. The relationship between increased gonadal hormone release per se and peri-adolescent vulnerability for psychiatric illness is difficult to tease apart from other potentially contributory factors in clinical studies, as adolescence is a turbulent period characterized by many social and biological changes. Despite the obvious opportunity provided by animal research, surprisingly little basic science effort has been devoted to this important issue. On the other hand, the animal work offers an understanding of the many ways in which gonadal steroids exert a powerful impact on the brain, both shaping its development and modifying its function during adulthood. Recently, investigators using preclinical models have described a greater male vulnerability to neurodevelopmental insults that are associated with schizophrenia; such studies may provide clinically relevant insights into the role of gonadal steroids in psychiatric illness.
Collapse
Affiliation(s)
- Julie A Markham
- Maryland Psychiatric Research Center, University of Maryland-Baltimore School of Medicine, P.O. Box 21247, Baltimore, MD 21228, USA.
| |
Collapse
|
29
|
Kuo J, Micevych P. Neurosteroids, trigger of the LH surge. J Steroid Biochem Mol Biol 2012; 131:57-65. [PMID: 22326732 PMCID: PMC3474707 DOI: 10.1016/j.jsbmb.2012.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 01/19/2012] [Accepted: 01/22/2012] [Indexed: 12/28/2022]
Abstract
Recent experiments from our laboratory are consistent with the idea that hypothalamic astrocytes are critical components of the central nervous system (CNS) mediated estrogen positive feedback mechanism. The "astrocrine hypothesis" maintains that ovarian estradiol rapidly increases free cytoplasmic calcium concentrations ([Ca(2+)](i)) that facilitate progesterone synthesis in astrocytes. This hypothalamic neuroprogesterone along with the elevated estrogen from the ovaries allows for the surge release of gonadotropin-releasing hormone (GnRH) that triggers the pituitary luteinizing hormone (LH) surge. A narrow range of estradiol stimulated progesterone production supports an "off-on-off" mechanism regulating the transition from estrogen negative feedback to estrogen positive feedback, and back again. The rapidity of the [Ca(2+)](i) response and progesterone synthesis support a non-genomic, membrane-initiated signaling mechanism. In hypothalamic astrocytes, membrane-associated estrogen receptors (mERs) signal through transactivation of the metabotropic glutamate receptor type 1a (mGluR1a), implying that astrocytic function is influenced by surrounding glutamatergic nerve terminals. Although other putative mERs, such as mERβ, STX-activated mER-Gα(q), and G protein-coupled receptor 30 (GPR30), are present and participate in membrane-mediated signaling, their influence in reproduction is still obscure since female reproduction be it estrogen positive feedback or lordosis behavior requires mERα. The astrocrine hypothesis is also consistent with the well-known sexual dimorphism of estrogen positive feedback. In rodents, only post-pubertal females exhibit this positive feedback. Hypothalamic astrocytes cultured from females, but not males, responded to estradiol by increasing progesterone synthesis. Estrogen autoregulates its own signaling by regulating levels of mERα in the plasma membrane of female astrocytes. In male astrocytes, the estradiol-induced increase in mERα was attenuated, suggesting that membrane-initiated estradiol signaling (MIES) would also be blunted. Indeed, estradiol induced [Ca(2+)](i) release in male astrocytes, but not to levels required to stimulate progesterone synthesis. Investigation of this sexual differentiation was performed using hypothalamic astrocytes from post-pubertal four core genotype (FCG) mice. In this model, genetic sex is uncoupled from gonadal sex. We demonstrated that animals that developed testes (XYM and XXM) lacked estrogen positive feedback, strongly suggesting that the sexual differentiation of progesterone synthesis is driven by the sex steroid environment during early development. This article is part of a Special Issue entitled 'Neurosteroids'.
Collapse
Affiliation(s)
- John Kuo
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Paul Micevych
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
- Corresponding author at: Department of Neurobiology, David Geffen School of Medicine at UCLA, 10833 LeConte Avenue, 73-078 CHS, Los Angeles, CA 90095-1763, United States. Tel.: +1 310 206 8265; fax: +1 310 825 2224. (P. Micevych)
| |
Collapse
|
30
|
Pinto PIS, Teodósio R, Socorro S, Power DM, Canário AVM. Structure, tissue distribution and estrogen regulation of splice variants of the sea bream estrogen receptor α gene. Gene 2012; 503:18-24. [PMID: 22579469 DOI: 10.1016/j.gene.2012.04.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 04/15/2012] [Accepted: 04/26/2012] [Indexed: 01/27/2023]
Abstract
Estrogen actions are mainly mediated by specific nuclear estrogen receptors (ERs), for which different genes and a diversity of transcript variants have been identified, mainly in mammals. In this study, we investigated the presence of ER splice variants in the teleost fish gilthead sea bream (Sparus auratus), by comparison with the genomic organization of the related species Takifugu rubripes. Two exon2-deleted ERα transcript variants were isolated from liver cDNA of estradiol-treated fish. The ΔE2 variant lacks ERα exon 2, generating a premature termination codon and a putative C-terminal truncated receptor, while the ΔE2,3* variant contains an in-frame deletion of exon 2 and part of exon 3 and codes for a putative ERα protein variant lacking most of the DNA-binding domain. Both variants were expressed at very low levels in several female and male sea bream tissues, and their expression was highly inducible in liver by estradiol-17β treatment with a strong positive correlation with the typical wild-type (wt) ERα response in this tissue. These findings identify novel estrogen responsive splice variants of fish ERα, and provide the basis for future studies to investigate possible modulation of wt-ER actions by splice variants.
Collapse
Affiliation(s)
- P I S Pinto
- University of Algarve, Campus de Gambelas, Faro, Portugal.
| | | | | | | | | |
Collapse
|
31
|
Wend K, Wend P, Krum SA. Tissue-Specific Effects of Loss of Estrogen during Menopause and Aging. Front Endocrinol (Lausanne) 2012; 3:19. [PMID: 22654856 PMCID: PMC3356020 DOI: 10.3389/fendo.2012.00019] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/23/2012] [Indexed: 12/04/2022] Open
Abstract
The roles of estrogens have been best studied in the breast, breast cancers, and in the female reproductive tract. However, estrogens have important functions in almost every tissue in the body. Recent clinical trials such as the Women's Health Initiative have highlighted both the importance of estrogens and how little we know about the molecular mechanism of estrogens in these other tissues. In this review, we illustrate the diverse functions of estrogens in the bone, adipose tissue, skin, hair, brain, skeletal muscle and cardiovascular system, and how the loss of estrogens during aging affects these tissues. Early transcriptional targets of estrogen are reviewed in each tissue. We also describe the tissue-specific effects of selective estrogen receptor modulators (SERMs) used for the treatment of breast cancers and postmenopausal symptoms.
Collapse
Affiliation(s)
- Korinna Wend
- Orthopaedic Hospital Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California Los AngelesLos Angeles, CA, USA
| | - Peter Wend
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los AngelesLos Angeles, CA, USA
| | - Susan A. Krum
- Orthopaedic Hospital Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California Los AngelesLos Angeles, CA, USA
| |
Collapse
|
32
|
Ishunina TA, Swaab DF. Decreased alternative splicing of estrogen receptor-α mRNA in the Alzheimer's disease brain. Neurobiol Aging 2012; 33:286-296.e3. [DOI: 10.1016/j.neurobiolaging.2010.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 02/19/2010] [Accepted: 03/16/2010] [Indexed: 01/15/2023]
|
33
|
Al Sweidi S, Sánchez MG, Bourque M, Morissette M, Dluzen D, Di Paolo T. Oestrogen receptors and signalling pathways: implications for neuroprotective effects of sex steroids in Parkinson's disease. J Neuroendocrinol 2012; 24:48-61. [PMID: 21790809 DOI: 10.1111/j.1365-2826.2011.02193.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder with a higher incidence in the male population. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD, 17β-oestradiol but not androgens were shown to protect dopamine (DA) neurones. We report that oestrogen receptors (ER)α and β distinctly contribute to neuroprotection against MPTP toxicity, as revealed by examining the membrane DA transporter (DAT), the vesicular monoamine transporter 2 (VMAT2) and tyrosine hyroxylase in ER wild-type (WT) and knockout (ERKO) C57Bl/6 male mice. Intact ERKOβ mice had lower levels of striatal DAT and VMAT2, whereas ERKOα mice were the most sensitive to MPTP toxicity compared to WT and ERKOβ mice and had the highest levels of plasma androgens. In both ERKO mice groups, treatment with 17β-oestradiol did not provide neuroprotection against MPTP, despite elevated plasma 17β-oestradiol levels. Next, the recently described membrane G protein-coupled oestrogen receptor (GPER1) was examined in female Macaca fascicularis monkeys and mice. GPER1 levels were increased in the caudate nucleus and the putamen of MPTP-monkeys and in the male mouse striatum lesioned with methamphetamine or MPTP. Moreover, neuroprotective mechanisms in response to oestrogens transmit via Akt/glycogen synthase kinase-3 (GSK3) signalling. The intact and lesioned striata of 17β-oestradiol treated monkeys, similar to that of mice, had increased levels of pAkt (Ser 473)/βIII-tubulin, pGSK3 (Ser 9)/βIII-tubulin and Akt/βIII-tubulin. Hence, ERα, ERβ and GPER1 activation by oestrogens is imperative in the modulation of ER signalling and serves as a basis for evaluating nigrostriatal neuroprotection.
Collapse
Affiliation(s)
- S Al Sweidi
- Molecular Endocrinology and Genomic Research Center, CHUQ (CHUL), Quebec City, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Jacobs E, D'Esposito M. Estrogen shapes dopamine-dependent cognitive processes: implications for women's health. J Neurosci 2011; 31:5286-93. [PMID: 21471363 PMCID: PMC3089976 DOI: 10.1523/jneurosci.6394-10.2011] [Citation(s) in RCA: 266] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 02/09/2011] [Accepted: 02/11/2011] [Indexed: 01/02/2023] Open
Abstract
The prefrontal cortex (PFC) is exquisitely sensitive to its neurochemical environment. Minor fluctuations in cortical dopamine (DA) can profoundly alter working memory, a PFC-dependent cognitive function that supports an array of essential human behaviors. Dopamine's action in the PFC follows an inverted U-shaped curve, where an optimal DA level results in maximal function and insufficient or excessive DA impairs PFC function. In animals, 17β-estradiol (the major estrogen in most mammals, referred to henceforth as estradiol) has been shown to enhance DA activity, yet no human study has adequately addressed whether estradiol's impact on cognition occurs by way of modulating specific neurochemical systems. Here we examined the effects of endogenous fluctuations in estradiol on working memory in healthy young women as a function of baseline PFC DA [indexed by catechol-O-methyltransferase (COMT) Val(158)Met genotype and, at a finer scale, COMT enzyme activity]. The results demonstrate that estradiol status impacts working memory function and, crucially, the direction of the effect depends on indices of baseline DA. Moreover, consistent with a DA cortical efficiency hypothesis, functional MRI revealed that inferred optimal DA was associated with reduced PFC activity sustained across task blocks and selectively enhanced PFC activity on trials with the greatest demand for cognitive control. The magnitude of PFC activity during high control trials was predictive of an individual's performance. These findings show that although estrogen, considered in isolation, may have unpredictable effects on cognitive performance, its influence is clarified when considered within a larger neuromodulatory framework. Given the clinical prevalence of dopaminergic drugs, understanding the relationship between estrogen and DA is essential for advancing women's health.
Collapse
Affiliation(s)
- Emily Jacobs
- Henry H. Wheeler Jr. Brain Imaging Center, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720
| | - Mark D'Esposito
- Henry H. Wheeler Jr. Brain Imaging Center, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720
| |
Collapse
|
35
|
Micevych P, Sinchak K. The Neurosteroid Progesterone Underlies Estrogen Positive Feedback of the LH Surge. Front Endocrinol (Lausanne) 2011; 2:90. [PMID: 22654832 PMCID: PMC3356049 DOI: 10.3389/fendo.2011.00090] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 11/16/2011] [Indexed: 01/25/2023] Open
Abstract
Our understanding the steroid regulation of neural function has rapidly evolved in the past decades. Not long ago the prevailing thoughts were that peripheral steroid hormones carried information to the brain which passively responded to these steroids. These steroid actions were slow, taking hours to days to be realized because they regulated gene expression. Over the past three decades, discoveries of new steroid receptors, rapid membrane-initiated signaling mechanisms, and de novo neurosteroidogenesis have shed new light on the complexity of steroids actions within the nervous system. Sexual differentiation of the brain during development occurs predominately through timed steroid-mediated expression of proteins and long term epigenetic modifications. In contrast across the estrous cycle, estradiol release from developing ovarian follicles initially increases slowly and then at proestrus increases rapidly. This pattern of estradiol release acts through both classical genomic mechanisms and rapid membrane-initiated signaling in the brain to coordinate reproductive behavior and physiology. This review focuses on recently discovered estrogen receptor-α membrane signaling mechanisms that estradiol utilizes during estrogen positive feedback to stimulate de novo progesterone synthesis within the hypothalamus to trigger the luteinizing hormone (LH) surge important for ovulation and estrous cyclicity. The activation of these signaling pathways appears to be coordinated by the rising and waning of estradiol throughout the estrous cycle and integral to the negative and positive feedback mechanisms of estradiol. This differential responsiveness is part of the timing mechanism triggering the LH surge.
Collapse
Affiliation(s)
- Paul Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine, Brain Research Institute, University of CaliforniaLos Angeles, CA, USA
- *Correspondence: Paul Micevych, Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA. e-mail:
| | - Kevin Sinchak
- Department of Biological Sciences, California State UniversityLong Beach, CA, USA
| |
Collapse
|
36
|
Estradiol rapidly regulates membrane estrogen receptor alpha levels in hypothalamic neurons. J Neurosci 2010; 30:12589-96. [PMID: 20861365 DOI: 10.1523/jneurosci.1038-10.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Estrogen receptors (ERs) and estrogen-binding proteins have been localized intracellularly and on the cell surface. The membrane-associated proteins initiate signaling that activates a myriad of cellular responses including the modulation of ion channels and ultimately transcription. Although many of the downstream actions of membrane ERs, including ERα and ERβ, have been characterized, the mechanisms regulating membrane ER levels have remained elusive in the nervous system. In the present study, we used surface biotinylation to identify and study the estradiol regulation of membrane ERα in mixed-sex, cultured hypothalamic neurons from rat. Following surface biotinylation, Western blot analysis revealed full-length 66 kDa ERα and several ERα splice variants, most notably a biotinylated 52 kDa ERα-immunoreactive protein. Treatment of the neurons with estradiol caused a rapid and transient increase of the biotinylated 52 kDa and 66 kDa ERα proteins in the plasma membrane. Exposure of the neurons to estradiol also significantly increased internalization of 52 kDa and 66 kDa ERα membrane proteins, a measure of receptor activation. In the hypothalamus, membrane ERα signaling depends on transactivation of metabotropic glutamate receptor-1a (mGluR1a). Estradiol treatment increased the internalization of mGluR1a in parallel with ERα, a finding consistent with the hypothesis of an ERα-mGluR1a signaling unit. These results demonstrate that estradiol regulates the amount of ERα in the membrane, suggesting estradiol can regulate its own membrane signaling.
Collapse
|
37
|
Morris RW, Fung SJ, Rothmond DA, Richards B, Ward S, Noble PL, Woodward RA, Weickert CS, Winslow JT. The effect of gonadectomy on prepulse inhibition and fear-potentiated startle in adolescent rhesus macaques. Psychoneuroendocrinology 2010; 35:896-905. [PMID: 20042297 DOI: 10.1016/j.psyneuen.2009.12.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 11/22/2009] [Accepted: 12/01/2009] [Indexed: 10/20/2022]
Abstract
Sex steroids, such as testosterone, can regulate brain development, cognition and modify psychiatric conditions. However, the role of adolescent testosterone in the emergence of cognitive deficits relevant to psychiatric illness has not been directly studied in primates. We examined whether removing testosterone during adolescence in rhesus macaques would affect prepulse inhibition (PPI) and fear-potentiated startle (FPS), which are translational tests of cognition affected in psychiatric disorders. Prepubertal macaques (30 months old) were castrated (n=6) or sham operated (n=6), and PPI and (FPS) were tested before the onset of puberty (34 months old) and after the pubertal surge in sex hormones 16 months later (50 months old). As expected there were no differences between the gonadectomized and intact groups' level of startle amplitude, PPI or (FPS) before puberty. After puberty, the intact group displayed substantially less PPI than the gonadectomized group, consistent with evidence that PPI is attenuated by endogenous increases in sex hormones. At the end of the study, testosterone among the intact monkeys was also correlated with tyrosine hydroxylase levels in the putamen, suggesting the attenuation of PPI by gonadal sex hormones may be influenced by subcortical dopamine. Thus, puberty involves significant increases in sex hormones, which in turn may modulate subcortical dopamine synthesis and affect cognitive functions impaired in psychiatric illnesses such as schizophrenia.
Collapse
|
38
|
Micevych P, Bondar G, Kuo J. Estrogen actions on neuroendocrine glia. Neuroendocrinology 2010; 91:211-22. [PMID: 20332598 PMCID: PMC2889254 DOI: 10.1159/000289568] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/18/2010] [Indexed: 12/25/2022]
Abstract
Astrocytes are the most abundant cells in the central nervous system (CNS). It appears that astrocytes are as diverse as neurons, having different phenotypes in various regions throughout the brain and participating in intercellular communication that involves signaling to neurons. It is not surprising then that astrocytes in the hypothalamus have an active role in the CNS regulation of reproduction. In addition to the traditional mechanism involving ensheathment of neurons and processes, astrocytes may have a critical role in regulating estrogen-positive feedback. Work in our laboratory has focused on the relationship between circulating estradiol and progesterone synthesized de novo in the brain. We have demonstrated that circulating estradiol stimulates the synthesis of progesterone in adult hypothalamic astrocytes, and this neuroprogesterone is critical for initiating the LH surge. Estradiol cell signaling is initiated at the cell membrane and involves the transactivation of metabotropic glutamate receptor type 1a (mGluR1a) leading to the release of intracellular stores of calcium. We used surface biotinylation to demonstrate that estrogen receptor-alpha (ERalpha) is present in the cell membrane and has an extracellular portion. Like other membrane receptors, ERalpha is inserted into the membrane and removed via internalization after agonist stimulation. This trafficking is directly regulated by estradiol, which rapidly and transiently increases the levels of membrane ERalpha, and upon activation, increases internalization that finally leads to ERalpha degradation. This autoregulation temporally limits membrane-initiated estradiol cell signaling. Thus, neuroprogesterone, the necessary signal for the LH surge, is released when circulating levels of estradiol peak on proestrus and activate progesterone receptors whose expression has been induced by the gradual rise of estradiol during follicular development.
Collapse
Affiliation(s)
- Paul Micevych
- Department of Neurobiology, Laboratory of Neuroendocrinology, UCLA Brain Research Institute, Los Angeles, Calif., USA
- *Paul Micevych, Department of Neurobiology, David Geffen School of Medicine at UCLA, 10833 LeConte Avenue, 73-078 CHS, Los Angeles, CA 90095-1763 (USA), Tel. +1 310 206 8265, Fax +1 310 825 2224, E-Mail
| | - Galyna Bondar
- Department of Neurobiology, Laboratory of Neuroendocrinology, UCLA Brain Research Institute, Los Angeles, Calif., USA
| | - John Kuo
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, Calif., USA
| |
Collapse
|
39
|
Abstract
Estradiol has rapid actions in the CNS that are mediated by membrane estrogen receptors (ERs) and activate cell signaling pathways through interaction with metabotropic glutamate receptors (mGluRs). Membrane-initiated estradiol signaling increases the free cytoplasmic calcium concentration ([Ca(2+)](i)) that stimulates the synthesis of neuroprogesterone in astrocytes. We used surface biotinylation to demonstrate that ERalpha has an extracellular portion. In addition to the full-length ERalpha [apparent molecular weight (MW), 66 kDa], surface biotinylation labeled an ERalpha-immunoreactive protein (MW, approximately 52 kDa) identified by both COOH- and NH(2)-directed antibodies. Estradiol treatment regulated membrane levels of both proteins in parallel: within 5 min, estradiol significantly increased membrane levels of the 66 and 52 kDa ERalpha. Internalization, a measure of membrane receptor activation, was also increased by estradiol with a similar time course. Continuous treatment with estradiol for 24-48 h reduced ERalpha levels, suggesting receptor downregulation. Estradiol also increased mGluR1a trafficking and internalization, consistent with the proposed ERalpha-mGluR1a interaction. Blocking ER with ICI 182,780 or mGluR1a with LY 367385 prevented ERalpha trafficking to and from the membrane. Estradiol-induced [Ca(2+)](i) flux was also significantly increased at the time of peak ERalpha activation/internalization. These results demonstrate that ERalpha is present in the membrane and has an extracellular portion. Furthermore, membrane levels and internalization of ERalpha are regulated by estradiol and mGluR1a ligands. The pattern of trafficking into and out of the membrane suggests that the changing concentration of estradiol during the estrous cycle regulates ERalpha to augment and then terminate membrane-initiated signaling.
Collapse
|
40
|
Taylor SE, Martin-Hirsch PL, Martin FL. Oestrogen receptor splice variants in the pathogenesis of disease. Cancer Lett 2009; 288:133-48. [PMID: 19608332 DOI: 10.1016/j.canlet.2009.06.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 06/12/2009] [Accepted: 06/16/2009] [Indexed: 02/06/2023]
Abstract
The full-length oestrogen receptor (ER) exists in most vertebrates as two separately encoded isoforms. ER splice variants represent truncated or otherwise modified versions of the full-length alpha or beta isoforms of the parent receptor. ERalpha is found on chromosome 6q and encodes a 595 amino acid protein, while ERbeta is found on chromosome 14q and encodes a 530 amino acid protein. These receptors possess differing ligand affinities, are differentially expressed in a tissue-specific fashion and may act antagonistically. Their altered expression has been implicated in the pathophysiology of a diverse range of conditions from cancer progression in hormone-responsive tissues to neurodegenerative disease. Variously co-expressed with full-length ERs, ER splice variants may have a positive or negative influence on transcription either by modifying the effect of the parent receptor or through their own intrinsic activity. To date, the vast majority of studies have used generic primers or antibodies against the full-length receptors and would not distinguish ER-mediated effects associated with various splice variants. Thus the evidence base of the influence of ER splice variants in normal developmental physiology and in the pathogenesis of disease is weak and greater understanding of their role will undoubtedly lead to new therapeutic strategies for disease intervention and treatment. This review aims to compile the current evidence for the presence of ER splice variants in humans, their physiological roles and clinical sequelae.
Collapse
|
41
|
Richards AB, Morris RW, Ward S, Schmitz S, Rothmond DA, Noble PL, Woodward RA, Winslow JT, Weickert CS. Gonadectomy negatively impacts social behavior of adolescent male primates. Horm Behav 2009; 56:140-8. [PMID: 19361511 PMCID: PMC2746978 DOI: 10.1016/j.yhbeh.2009.03.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 03/27/2009] [Accepted: 03/31/2009] [Indexed: 01/30/2023]
Abstract
Social behavior changes dramatically during primate adolescence. However, the extent to which testosterone and other gonadal hormones are necessary for adolescent social behavioral development is unknown. In this study, we determined that gonadectomy significantly impairs social dominance in naturalistic settings and changes reactions to social stimuli in experimental settings. Rhesus macaques were castrated (n= 6) or sham operated (n=6) at age 2.4 years, group-housed for 2 years, and ethograms were collected weekly. During adolescence the gonadally intact monkeys displayed a decrease in subordinate behaviors and an increase in dominant behaviors, which ultimately related to a rise in social status and rank in the dominance hierarchy. We measured monkey's reactions to emotional faces (fear, threat, neutral) of conspecifics of three ages (adult, peer, infant). Intact monkeys were faster to retrieve a treat in front of a threatening or infant face, while castrated monkeys did not show a differential response to different emotional faces or ages. No group difference in reaction to an innate fear-eliciting object (snake) was found. Approach and proximity responses to familiar vs unfamiliar conspecifics were tested, and intact monkeys spent more time proximal to a novel conspecific as compared to castrates who tended to spend more time with a familiar conspecific. No group differences in time spent with novel or familiar objects were found. Thus, gonadectomy resulted in the emergence of significantly different responses to social stimuli, but not non-social stimuli. Our work suggests that intact gonads, which are needed to produce adolescent increases in circulating testosterone, impact social behavior during adolescences in primates.
Collapse
|
42
|
Wong J, Weickert CS. Transcriptional interaction of an estrogen receptor splice variant and ErbB4 suggests convergence in gene susceptibility pathways in schizophrenia. J Biol Chem 2009; 284:18824-32. [PMID: 19439407 DOI: 10.1074/jbc.m109.013243] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Mounting evidence from clinical and basic research suggests that estrogen signaling may be altered in the brains of people with schizophrenia. Previously, we found that DNA sequence variation in the estrogen receptor (ER) alpha gene, lower ERalpha mRNA levels, and/or blunted ERalpha signaling is associated with schizophrenia. In this study, we asked whether the naturally occurring truncated ERalpha isoform, Delta7, which acts as a dominant negative, can attenuate gene expression induced by the wild-type (WT) receptor in an estrogen-dependent manner in neuronal (SHSY5Y) and non-neuronal (CHOK1 and HeLa) cells. In addition, we determined the extent to which ERalpha interacts with NRG1-ErbB4, a leading schizophrenia susceptibility pathway. Reductions in the transcriptionally active form of ErbB4 comprising the intracytoplasmic domain (ErbB4-ICD) have been found in schizophrenia, and we hypothesized that ERalpha and ErbB4 may converge to control gene expression. In the present study, we show that truncated Delta7-ERalpha attenuates WT-ERalpha-driven gene expression across a wide range of estrogen concentrations in cells that express functional ERalpha at base line or upon co-transfection of full-length ERalpha. Furthermore, we find that ErbB4-ICD can potentiate the transcriptional activity of WT-ERalpha at EREs in two cell lines and that this potentiation effect is abolished by the presence of Delta7-ERalpha. Immunofluorescence microscopy revealed nuclear co-localization of WT-ERalpha, Delta7-ERalpha, and ErbB4-ICD, whereas immunoprecipitation assays showed direct interaction. Our findings demonstrate convergence between ERalpha and ErbB4-ICD in the transcriptional control of ERalpha-target gene expression and suggest that this may represent a convergent pathway that may be disrupted in schizophrenia.
Collapse
Affiliation(s)
- Jenny Wong
- Schizophrenia Research Institute, Sydney, New South Wales 2052, Australia
| | | |
Collapse
|
43
|
Giegling I, Chiesa A, Calati R, Hartmann AM, Möller HJ, De Ronchi D, Rujescu D, Serretti A. Do the estrogen receptors 1 gene variants influence the temperament and character inventory scores in suicidal attempters and healthy subjects? Am J Med Genet B Neuropsychiatr Genet 2009; 150B:434-8. [PMID: 18680103 DOI: 10.1002/ajmg.b.30834] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Several studies investigated possible associations between single nucleotide polymorphisms and the temperament and character inventory (TCI) scores. The possible association between TCI scores and the estrogen receptors 1 (ESR1) polymorphisms, which are expressed in cerebral regions that are involved in the development of temperament and character, has not been investigated yet. The aim of the present study is to investigate possible associations between specific ESR1 polymorphisms and TCI scores. Two hundred eighty-nine healthy subjects and 111 suicide attempters were enrolled in the study. All subjects compiled TCI. Rs827421, rs1913474, rs1801132, rs722207, rs974276, and rs910416 in ESR1 were genotyped. Rs722207 was associated with Harm Avoidance (HA) in the healthy sample (P = 0.003). Further associations have been found for two HA subscales, HA2 and HA3, in the healthy sample. Additionally, the haplotype rs722207-rs974276 showed an association with HA (P = 0.0003) and HA2 (P = 0.0002) in the global sample and in healthy volunteers and HA3 showed an association in the global sample. Our study showed a moderate association between ESR1 variants and TCI scores. The main finding concerns the association between rs722207 and HA in the healthy sample. Further research is needed to replicate our findings and to investigate further ESR1 polymorphisms.
Collapse
Affiliation(s)
- Ina Giegling
- Department of Psychiatry, Ludwig Maximilians University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Estrogen receptor gene 1 variants are not associated with suicidal behavior. Psychiatry Res 2008; 160:1-7. [PMID: 18511131 DOI: 10.1016/j.psychres.2007.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 01/12/2007] [Accepted: 05/09/2007] [Indexed: 11/24/2022]
Abstract
Estrogen is thought to be involved in the pathophysiology of depression and suicidal behaviors. We studied gene variants of estrogen receptor alpha (rs827421, rs1913474, rs1801132, rs722207, rs974276 and rs910416) in 167 German suicide attempters (affective spectrum n=107, schizophrenia spectrum n=35, borderline personality disorder n=25), 92 German individuals who committed suicide and 312 German healthy subjects. Single markers and haplotype analysis in relation to suicidal behaviors (suicide attempters/completers) did not reveal any significant association. These were also not associated with related features, such as violence or impulsivity of suicide attempt, State-Trait Anger Expression Inventory (STAXI) and Questionnaire for Measuring Factors of Aggression (FAF) scores. In conclusion, our study does not support the hypothesis that estrogen receptor alpha gene variants are major contributors to suicide or to anger- or aggression-related behaviors.
Collapse
|
45
|
Montague D, Weickert CS, Tomaskovic-Crook E, Rothmond DA, Kleinman JE, Rubinow DR. Oestrogen receptor alpha localisation in the prefrontal cortex of three mammalian species. J Neuroendocrinol 2008; 20:893-903. [PMID: 18445128 PMCID: PMC2719673 DOI: 10.1111/j.1365-2826.2008.01743.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oestrogen modulates cognitive function and affective behaviours subserved by the prefrontal cortex (PFC). Identifying and localising oestrogen receptor (ER)alpha, in human PFC will contribute to our understanding of the molecular mechanism of oestrogen action in this region. Inferences about the site of action of oestrogen in human brain are derived largely from studies performed in nonhuman mammalian species; however, the congruence of findings across species has not been demonstrated. Furthermore, the laminar, cellular, and subcellular localisation of ERalpha in the cortex is debated. Therefore, we compared the distribution of ERalpha in human dorsolateral prefrontal cortex (DLPFC) with that of monkey DLPFC and rat medial PFC. Immunohistochemistry performed on frontal cortex from the three species demonstrated ERalpha positive cells throughout all layers of the PFC, in pyramidal and nonpyramidal neurones, with both nuclear and cytoplasmic immunoreactivity. Western blot analyses and preabsorption studies confirmed that the antibody used recognised ERalpha and not ERbeta. A strong ERalpha immunoreactive band corresponding to the full-length ERalpha protein (65-67 kDa) in the frontal cortex of all three species matched the size of the predominant immunoreactive band detected in breast cancer cell lines known to express ERalpha. Additionally, other ERalpha immunoreactive proteins of varying molecular weight in breast cancer cells, rat ovary and mammalian brain were detected, suggesting that ERalpha may exist in more than one form in the mammalian frontal cortex. The present study provides evidence that ERalpha protein exists in neurones in mammalian PFC and that ERalpha is anatomically well-positioned to directly mediate oestrogen action in these neurones.
Collapse
Affiliation(s)
- D Montague
- Behavioural Endocrinology Branch, National Institute of Mental Health, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
46
|
Schirmer A, Escoffier N, Li QY, Li H, Strafford-Wilson J, Li WI. What grabs his attention but not hers? Estrogen correlates with neurophysiological measures of vocal change detection. Psychoneuroendocrinology 2008; 33:718-27. [PMID: 18395352 DOI: 10.1016/j.psyneuen.2008.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 02/17/2008] [Accepted: 02/20/2008] [Indexed: 11/17/2022]
Abstract
Prior research revealed sex differences in the processing of unattended changes in speaker prosody. The present study aimed at investigating the role of estrogen in mediating these effects. To this end, the electroencephalogram (EEG) was recorded while participants watched a silent movie with subtitles and passively listened to a syllable sequence that contained occasional changes in speaker prosody. In one block, these changes were neutral, whereas in another block they were emotional. Estrogen values were obtained for each participant and correlated with the mismatch negativity (MMN) amplitude elicited in the EEG. As predicted, female listeners had higher estrogen values than male listeners and showed reduced MMN amplitudes to neutral as compared to emotional change in speaker prosody. Moreover, in both, male and female listeners, MMN amplitudes were negatively correlated with estrogen when the change in speaker prosody was neutral, but not when it was emotional. This suggests that estrogen is associated with reduced distractibility by neutral, but not emotional, events. Emotional events are spared from this reduction in distractibility and more likely to penetrate voluntary attention directed elsewhere. Taken together, the present findings provide evidence for a role of estrogen in human cognition and emotion.
Collapse
Affiliation(s)
- Annett Schirmer
- Faculty of Arts and Social Sciences, Department of Psychology, National University of Singapore, 9 Arts Link, Block AS4, Level 2, Singapore 117570, Singapore.
| | | | | | | | | | | |
Collapse
|
47
|
Weickert CS, Miranda-Angulo AL, Wong J, Perlman WR, Ward SE, Radhakrishna V, Straub RE, Weinberger DR, Kleinman JE. Variants in the estrogen receptor alpha gene and its mRNA contribute to risk for schizophrenia. Hum Mol Genet 2008; 17:2293-309. [PMID: 18424448 PMCID: PMC2465798 DOI: 10.1093/hmg/ddn130] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Estrogen modifies human emotion and cognition and impacts symptoms of schizophrenia. We hypothesized that the variation in the estrogen receptor alpha (ESR1) gene and cortical ESR1 mRNA is associated with schizophrenia. In a small case–control genetic association analysis of postmortem brain tissue, genotype CC (rs2234693) and haplotypes containing the C allele of a single-nucleotide polymorphism (SNP) in intron1 (PvuII) were more frequent in African American schizophrenics (P = 0.01–0.001). In a follow-up family-based association analysis, we found overtransmission of PvuII allele C and a PvuII C-containing haplotype (P = 0.01–0.03) to African American and Caucasian patients with schizophrenia. Schizophrenics with the ‘at risk’ PvuII genotype had lower ESR1 mRNA levels in the frontal cortex. Eighteen ESR1 splice variants and decreased frequencies of the wild-type ESR1 mRNA were detected in schizophrenia. In one patient, a unique ESR1 transcript with a genomic insert encoding a premature stop codon and a truncated ESR1 protein lacking most of the estrogen binding domain was the only transcript detected. Using a luciferase assay, we found that mRNA encoding a truncated ESR1 significantly attenuates gene expression at estrogen-response elements demonstrating a dominant negative function. An intron 6 SNP [rs2273207(G)] was associated with an ESR1 splice variant missing exon seven. The T allele of another intron 6 SNP was part of a 3′ haplotype less common in schizophrenia [rs2273206(T), rs2273207(G), rs2228480(G)]. Thus, the variation in the ESR1 gene is associated with schizophrenia and the mechanism of this association may involve alternative gene regulation and transcript processing.
Collapse
|
48
|
Abstract
In the present review we discuss recent findings showing that, in addition to the canonical estrogen receptor-alpha (ERalpha), the level of various ERalpha splice variants is changed in the human brain in aging and Alzheimer's disease (AD) at both the mRNA and protein level and that they should be considered for the understanding of estrogen effects on the brain and estrogen therapy pitfalls. Indeed, the expression pattern of certain splice forms is brain area-specific. Thus, the major isoform found in the mamillary body (MB) appeared to be del.7 (deletion of exon 7), while in the hippocampus del.4 (exon 4 omitted) was expressed at the highest level. Furthermore, while transcripts missing exons 7 and 2 declined with aging in the MB of patients >60 years old, no age-related alterations were determined for a number of splice variants in the hippocampus. A novel MB1 isoform with a 168-bp deletion within the transactivation function 1 of ERalpha turned out to accumulate in the histaminergic tuberomamillary nucleus of postmenopausal women. Finally, the level of alternatively spliced ERalpha may also change in AD in a brain area-specific manner and so affect the sensitivity to estrogen therapy.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Department of Histology and Embryology, Kursk State Medical University, Kursk, Russia.
| | | |
Collapse
|
49
|
Aizawa S, Yamamuro Y. Estradiol regulates alternative splicing of estrogen receptor-alpha mRNA in differentiated NG108-15 neuronal cells. Life Sci 2008; 82:692-8. [PMID: 18258268 DOI: 10.1016/j.lfs.2008.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 12/10/2007] [Accepted: 01/02/2008] [Indexed: 10/22/2022]
Abstract
The biological actions of estrogen are mostly conveyed through interaction with two different types of estrogen receptor (ER), ER-alpha and ER-beta. With regard to ER-alpha, an alternatively spliced form and its translated product, truncated estrogen receptor product-1 (TERP-1), have been identified in the rat pituitary. TERP-1 has the ability to inhibit the ER binding to DNA response element by forming hetero-dimers with the wild-type ER. Furthermore, TERP-1 expression increased concurrently with serum estrogen levels. Although estrogen also plays important roles in the central nervous system, the existence and regulatory mechanism of alternatively spliced ER-alpha mRNA expression has remained unclear. The present study evaluated the expression of the alternatively spliced form of the ER-alpha gene, and examined the influence of a representative ER ligand, 17beta-estradiol (E2), on the expression in differentiated NG108-15 neuronal cells. A real-time RT-PCR analysis using primer sets designed to amplify from exons 3 to 4, exons 4 to 5, exons 5 to 6, exons 6 to 7, and exons 7 to 8 of the mouse ER-alpha gene revealed the existence of alternatively spliced ER-alpha mRNA and its putative transcription initiation site, located between exon 4 and exon 5. Although E2 had no apparent effect on the overall expression of ER-alpha mRNA, it reduced the incidence of the alternatively spliced form of ER-alpha. The down-regulation by E2 predominantly arose via binding to nuclear ERs. The present study demonstrated that alternatively spliced ER-alpha mRNA is expressed in differentiated NG108-15 neuronal cells, and provides evidence for the functional up-regulation of ER-alpha via the ligand-binding activation of ERs.
Collapse
Affiliation(s)
- Shu Aizawa
- Department of Animal Science, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-8510, Japan
| | | |
Collapse
|
50
|
Ishunina TA, Fischer DF, Swaab DF. Estrogen receptor alpha and its splice variants in the hippocampus in aging and Alzheimer's disease. Neurobiol Aging 2006; 28:1670-81. [PMID: 17010478 DOI: 10.1016/j.neurobiolaging.2006.07.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 07/04/2006] [Accepted: 07/21/2006] [Indexed: 11/28/2022]
Abstract
Clinical and experimental studies show that estrogens can have beneficial effects on hippocampus-dependent cognitive functions that may be mediated by estrogen receptor (ER)alpha. We investigated whether menopause and Alzheimer's disease (AD) cause changes in this ER subtype. Immunocytochemical staining of ERalpha, aromatase and Golgi complex (GC) was performed on paraffin embedded hippocampal tissue from women of the pre-, peri- and postmenopausal age. Canonical ERalpha mRNA amplicons, ERalpha splice variants (del.2, del.4, del.7, MB1) and aromatase transcripts were measured by Q-PCR in frozen hippocampal samples of AD and matched control cases. Nuclear ERalpha, aromatase and the GC enhanced during aging in women indicating availability of locally synthesized estrogens that may up-regulate ERalpha by which neuronal metabolism can be augmented in the hippocampus after the menopause. In AD cases canonical and alternatively spliced ERalpha mRNA, and aromatase gene expression were down-regulated suggesting a deficit in local estrogen levels and diminished signalling through ERalpha. The major ERalpha splice variants in the hippocampus were found to be MB1 and del.4. A novel ERalpha isoform TADDI was isolated and sequenced from two female patients. It lacks 31 bp at the junction between exons 3 and 4 with an insertion of 13 nucleotides from the middle of the exon 2.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | | | | |
Collapse
|