1
|
Zhvania MG, Japaridze N, Tizabi Y, Pochkhidze N, Lobzhanidze G. Effects of high-intensity chronic noise on spatial memory in male versus female rats. Eur J Neurosci 2024; 60:5581-5590. [PMID: 39180282 DOI: 10.1111/ejn.16514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
The detrimental effects of high-intensity noise on the auditory system and emotional status, including the induction of anxiety, are well documented. Preclinical as well as epidemiological and clinical studies have solidly established differential responses between males and females to various stressful stimuli, including high-intensity white noise (HIWN). However, whether chronic exposure to noise affects cognitive functions and whether this effect is sex dependent has not been adequately addressed. In this study, we used two cognitive test paradigms, such as the Morris water maze (MWM) and the multi-branch maze (MBM), to test the effect of chronic HIWN on indices of spatial learning and memory in both male and female Wistar rats. Our findings indicate that daily (1 h) exposure to 100 dB of noise for 30 consecutive days induces different task-dependent responses in male versus female rats. For example, in the acquisition phase of MWM, female rats exposed to noise outperformed their male counterparts at twice the speed. Similarly, in the MBM test, noise-exposed female rats outperformed the male rats in reaching the nest box. It is clear from these studies that noise impairs cognitive functions twice as negatively in male rats as in female rats. Thus, sex-related differences in spatial learning and memory in response to HIWN must be taken into consideration when investigating the neurobiological components and/or treatment modalities.
Collapse
Affiliation(s)
- Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Carl Zeiss Scientific and Education Center, New Vision University, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Nino Pochkhidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Giorgi Lobzhanidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
2
|
Mu E, Gurvich C, Kulkarni J. Estrogen and psychosis - a review and future directions. Arch Womens Ment Health 2024:10.1007/s00737-023-01409-x. [PMID: 38221595 DOI: 10.1007/s00737-023-01409-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/02/2023] [Indexed: 01/16/2024]
Abstract
The link between sex hormones and schizophrenia has been suspected for over a century; however, scientific evidence supporting the pharmacotherapeutic effects of exogenous estrogen has only started to emerge during the past three decades. Accumulating evidence from epidemiological and basic research suggests that estrogen has a protective effect in women vulnerable to schizophrenia. Such evidence has led multiple researchers to investigate the role of estrogen in schizophrenia and its use in treatment. This narrative review provides an overview of the effects of estrogen as well as summarizes the recent work regarding estrogen as a treatment for schizophrenia, particularly the use of new-generation selective estrogen receptor modulators.
Collapse
Affiliation(s)
- Eveline Mu
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Caroline Gurvich
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jayashri Kulkarni
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Fehr T, Janssen WG, Park J, Baxter MG. Neonatal exposures to sevoflurane in rhesus monkeys alter synaptic ultrastructure in later life. iScience 2022; 25:105685. [PMID: 36567715 PMCID: PMC9772858 DOI: 10.1016/j.isci.2022.105685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/02/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Repeated or prolonged early life exposure to anesthesia is neurotoxic in animals and associated with neurocognitive impairment in later life in humans. We used electron microscopy with unbiased stereological sampling to assess synaptic ultrastructure in dorsolateral prefrontal cortex (dlPFC) and hippocampal CA1 of female and male rhesus monkeys, four years after three 4-h exposures to sevoflurane during the first five postnatal weeks. This allowed us to ascertain long-term consequences of anesthesia exposure without confounding effects of surgery or illness. Synapse areas were reduced in the largest synapses in CA1 and dlPFC, predominantly in perforated spinous synapses in CA1 and nonperforated spinous synapses in dlPFC. Mitochondrial morphology and localization changed subtly in both areas. Synapse areas in CA1 correlated with response to a mild social stressor. Thus, exposure to anesthesia in infancy can cause long-term ultrastructural changes in primates, which may be substrates for long-term alterations in synaptic transmission and behavioral deficits.
Collapse
Affiliation(s)
- Tristan Fehr
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - William G.M. Janssen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Janis Park
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark G. Baxter
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA,Corresponding author
| |
Collapse
|
4
|
Lee J, Kim HJ. Normal Aging Induces Changes in the Brain and Neurodegeneration Progress: Review of the Structural, Biochemical, Metabolic, Cellular, and Molecular Changes. Front Aging Neurosci 2022; 14:931536. [PMID: 35847660 PMCID: PMC9281621 DOI: 10.3389/fnagi.2022.931536] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
Aging is accompanied by many changes in brain and contributes to progressive cognitive decline. In contrast to pathological changes in brain, normal aging brain changes have relatively mild but important changes in structural, biochemical and molecular level. Representatively, aging associated brain changes include atrophy of tissues, alteration in neurotransmitters and damage accumulation in cellular environment. These effects have causative link with age associated changes which ultimately results in cognitive decline. Although several evidences were found in normal aging changes of brain, it is not clearly integrated. Figuring out aging related changes in brain is important as aging is the process that everyone goes through, and comprehensive understanding may help to progress further studies. This review clarifies normal aging brain changes in an asymptotic and comprehensive manner, from a gross level to a microscopic and molecular level, and discusses potential approaches to seek the changes with cognitive decline.
Collapse
Affiliation(s)
- Jiseon Lee
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Hee-Jin Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| |
Collapse
|
5
|
Oliva CA, Rivera DS, Mariqueo TA, Bozinovic F, Inestrosa NC. Differential Role of Sex and Age in the Synaptic Transmission of Degus (Octodon degus). Front Integr Neurosci 2022; 16:799147. [PMID: 35295186 PMCID: PMC8918727 DOI: 10.3389/fnint.2022.799147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Octodon degus are a diurnal long-lived social animal widely used to perform longitudinal studies and complex cognitive tasks to test for physiological conditions with similitude in human behavior. They show a complex social organization feasible to be studied under different conditions and ages. Several aspects in degus physiology demonstrated that these animals are susceptible to environmental conditions, such as stress, fear, feeding quality, and isolation. However, the relevance of these factors in life of this animal depends on sex and age. Despite its significance, there are few studies with the intent to characterize neurological parameters that include these two parameters. To determine the basal neurophysiological status, we analyzed basic electrophysiological parameters generated during basal activity or synaptic plasticity in the brain slices of young and aged female and male degus. We studied the hippocampal circuit of animals kept in social ambient in captivity under controlled conditions. The study of basal synaptic activity in young animals (12–24 months old) was similar between sexes, but female degus showed more efficient synaptic transmission than male degus. We found the opposite in aged animals (60–84 months old), where male degus had a more efficient basal transmission and facilitation index than female degus. Furthermore, female and male degus develop significant but not different long-term synaptic plasticity (LTP). However, aged female degus need to recruit twice as many axons to evoke the same postsynaptic activity as male degus and four times more when compared to young female degus. These data suggest that, unlike male degus, the neural status of aged female degus change, showing less number or functional axons available at advanced ages. Our data represent the first approach to incorporate the effect of sex along with age progression in basal neural status.
Collapse
Affiliation(s)
- Carolina A. Oliva
- Center of Aging and Regeneration UC, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Carolina A. Oliva,
| | - Daniela S. Rivera
- GEMA Center for Genomics, Ecology & Environment, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago, Chile
| | - Trinidad A. Mariqueo
- Centro de Investigaciones Médicas, Laboratorio de Neurofarmacología, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Francisco Bozinovic
- Center of Applied Ecology and Sustainability, Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Center of Aging and Regeneration UC, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
- Nibaldo C. Inestrosa,
| |
Collapse
|
6
|
Tirkkonen A, Kekäläinen T, Aukee P, Kujala UM, Laakkonen EK, Kokko K, Sipilä S. Bidirectional associations between cognitive functions and walking performance among middle-aged women. Menopause 2021; 29:200-209. [PMID: 34930867 PMCID: PMC8785773 DOI: 10.1097/gme.0000000000001896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE This study investigated whether (1) cognitive functions change after the transition from the perimenopausal to the postmenopausal stage, (2) cognitive functions and walking are associated in middle-aged women, and (3) cognitive functions assessed in perimenopause are associated with walking after reaching the postmenopause or vice versa. METHODS In total, 342 women, categorized as early (n = 158) or late perimenopausal (n = 184), were included in the study and followed up until postmenopausal. Psychomotor speed, executive functions related to set-shifting and updating, working memory, and visual memory were assessed. Walking was assessed with walking speed, walking distance, and dual-task cost in walking speed. Data was analyzed using the paired-samples t test, Wilcoxon signed rank test, multiple linear regression analysis, and structural equation modeling. RESULTS We found small but significant improvements in psychomotor speed (P = 0.01) and working memory (P < 0.001) among early perimenopausal and in psychomotor speed (P = 0.001), set-shifting (P = 0.02), visual memory (P = 0.002), and working memory (P < 0.001) among late perimenopausal women after the transition from peri- to postmenopause. Walking speed (β = 0.264, P = 0.001) and dual-task cost (β = 0.160, P = 0.03) were associated with updating, and walking distance was associated with updating and set-shifting (β = 0.198, P = 0.02, β=-0.178 P = 0.04 respectively) among the late perimenopausal women. We found no longitudinal associations between cognitive functions and walking. CONCLUSION Cognitive performance remained unchanged or improved after reaching postmenopause. Cognitive functions and walking were associated during the late perimenopause, but the association depended on the cognitive process and nature of the physical task. Cognitive performance was not associated with walking after reaching postmenopause or vice versa.
Collapse
Affiliation(s)
- Anna Tirkkonen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Tiia Kekäläinen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Pauliina Aukee
- Department of Obstetrics and Gynecology, Central Finland Health Care District, Jyväskylä, Finland
| | - Urho M. Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Eija K. Laakkonen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Katja Kokko
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sarianna Sipilä
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
7
|
Leithead AB, Tasker JG, Harony‐Nicolas H. The interplay between glutamatergic circuits and oxytocin neurons in the hypothalamus and its relevance to neurodevelopmental disorders. J Neuroendocrinol 2021; 33:e13061. [PMID: 34786775 PMCID: PMC8951898 DOI: 10.1111/jne.13061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/14/2021] [Accepted: 10/30/2021] [Indexed: 11/27/2022]
Abstract
Oxytocin (OXT) neurons of the hypothalamus are at the center of several physiological functions, including milk ejection, uterus contraction, and maternal and social behavior. In lactating females, OXT neurons show a pattern of burst firing and inter-neuron synchronization during suckling that leads to pulsatile release of surges of OXT into the bloodstream to stimulate milk ejection. This pattern of firing and population synchronization may be facilitated in part by hypothalamic glutamatergic circuits, as has been observed in vitro using brain slices obtained from male rats and neonates. However, it remains unknown how hypothalamic glutamatergic circuits influence OXT cell activity outside the context of lactation. In this review, we summarize the in vivo and in vitro studies that describe the synchronized burst firing pattern of OXT neurons and the implication of hypothalamic glutamate in this pattern of firing. We also make note of the few studies that have traced glutamatergic afferents to the hypothalamic paraventricular and supraoptic nuclei. Finally, we discuss the genetic findings implicating several glutamatergic genes in neurodevelopmental disorders, including autism spectrum disorder, thus underscoring the need for future studies to investigate the impact of these mutations on hypothalamic glutamatergic circuits and the OXT system.
Collapse
Affiliation(s)
- Amanda B. Leithead
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Jeffrey G. Tasker
- Neurobiology DivisionDepartment of Cell and Molecular BiologyTulane UniversityNew OrleansLAUSA
| | - Hala Harony‐Nicolas
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mindich Child Health and Development Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
8
|
Sinha P, Rani A, Kumar A, Riva A, Brant JO, Foster TC. Examination of CA1 Hippocampal DNA Methylation as a Mechanism for Closing of Estrogen's Critical Window. Front Aging Neurosci 2021; 13:717032. [PMID: 34421577 PMCID: PMC8371553 DOI: 10.3389/fnagi.2021.717032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/15/2021] [Indexed: 02/01/2023] Open
Abstract
There is a critical window for estrogen replacement therapy, beyond which estradiol (E2) fails to enhance cognition and N-methyl-D-aspartate (NMDA) receptor function, and E2-responsive transcription decreases. Much less attention has been given to the mechanism for closing of the critical window, which is thought to involve the decline in estrogen signaling cascades, possibly involving epigenetic mechanisms, including DNA methylation. This study investigated changes in DNA methylation in region CA1 of the hippocampus of ovariectomized female rats over the course of brain aging and in response to E2-treatment, using whole genome bisulfite sequencing. Differential methylation of CpG and non-CpG (CHG and CHH) sites and associated genes were characterized in aged controls (AC), middle-age controls (MC), and young controls (YC) and differential methylation in response to E2-treatment (T) was examined in each age group (AT-AC, MT-MC, and YT-YC). Possible candidate genes for the closing of the critical window were defined as those that were hypomethylated by E2-treatment in younger animals, but were unresponsive in aged animals. Gene ontology categories for possible critical window genes were linked to response to hormones (Adcyap1, Agtr2, Apob, Ahr, Andpro, Calm2, Cyp4a2, Htr1b, Nr3c2, Pitx2, Pth, Pdk4, Slc2a2, Tnc, and Wnt5a), including G-protein receptor signaling (Gpr22 and Rgs4). Other possible critical window genes were linked to glutamate synapses (Nedd4, Grm1, Grm7, and Grin3a). These results suggest that decreased E2 signaling with advanced age, and/or prolonged E2 deprivation, results in methylation of E2-responsive genes, including those involved in rapid E2 signaling, which may limit subsequent transcription.
Collapse
Affiliation(s)
- Puja Sinha
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, United States
| | - Jason Orr Brant
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Genetics and Genomics Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
9
|
Bilash OM, Actor-Engel HS, Sherpa AD, Chen YW, Aoki C. Suppression of food restriction-evoked hyperactivity in activity-based anorexia animal model through glutamate transporters GLT-1 at excitatory synapses in the hippocampus. Synapse 2021; 75:e22197. [PMID: 33619810 DOI: 10.1002/syn.22197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 12/22/2022]
Abstract
Severe voluntary food restriction is the defining symptom of anorexia nervosa (AN), but anxiety and excessive exercise are maladaptive symptoms that contribute significantly to the severity of AN and which individuals with AN have difficulty suppressing. We hypothesized that the excitability of hippocampal pyramidal neurons, known to contribute to anxiety, leads to the maladaptive behavior of excessive exercise. Conversely, since glutamate transporter GLT-1 dampens the excitability of hippocampal pyramidal neurons through the uptake of ambient glutamate and suppression of the GluN2B-subunit containing NMDA receptors (GluN2B-NMDARs), GLT-1 may contribute toward dampening excessive exercise. This hypothesis was tested using the mouse model of AN, called activity-based anorexia (ABA), whereby food restriction evokes the maladaptive behavior of excessive wheel running (food restriction-evoked running, FRER). We tested whether individual differences in ABA vulnerability of mice, quantified based on FRER, correlated with individual differences in the levels of GLT-1 at excitatory synapses of the hippocampus. Electron microscopic immunocytochemistry (EM-ICC) was used to quantify GLT-1 levels at the excitatory synapses of the hippocampus. The FRER seen in individual mice varied more than 10-fold, and Pearson correlation analyses revealed a strong negative correlation (p = .02) between FRER and GLT-1 levels at the axon terminals of excitatory synapses and at the surrounding astrocytic plasma membranes. Moreover, synaptic levels of GluN2B-NMDARs correlated strongly with GLT-1 levels at perisynaptic astrocytic plasma membranes. There is at present no accepted pharmacotherapy for AN, and little is known about the etiology of this deadly illness. Current findings suggest that drugs increasing GLT-1 expression may reduce AN severity through the reduction of GluN2B-NMDAR activity.
Collapse
Affiliation(s)
- Olesia M Bilash
- The Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Center for Neural Science, New York University, New York, NY, USA
| | | | - Ang D Sherpa
- Center for Neural Science, New York University, New York, NY, USA
| | - Yi-Wen Chen
- Center for Neural Science, New York University, New York, NY, USA
| | - Chiye Aoki
- The Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Center for Neural Science, New York University, New York, NY, USA
| |
Collapse
|
10
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
11
|
17α Estradiol promotes plasticity of spared inputs in the adult amblyopic visual cortex. Sci Rep 2019; 9:19040. [PMID: 31836739 PMCID: PMC6910995 DOI: 10.1038/s41598-019-55158-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
The promotion of structural and functional plasticity by estrogens is a promising approach to enhance central nervous system function in the aged. However, how the sensitivity to estrogens is regulated across brain regions, age and experience is poorly understood. To ask if estradiol treatment impacts structural and functional plasticity in sensory cortices, we examined the acute effect of 17α-Estradiol in adult Long Evans rats following chronic monocular deprivation, a manipulation that reduces the strength and selectivity of deprived eye vision. Chronic monocular deprivation decreased thalamic input from the deprived eye to the binocular visual cortex and accelerated short-term depression of the deprived eye pathway, but did not change the density of excitatory synapses in primary visual cortex. Importantly, we found that the classical estrogen receptors ERα and ERβ were robustly expressed in the adult visual cortex, and that a single dose of 17α-Estradiol reduced the expression of the calcium-binding protein parvalbumin, decreased the integrity of the extracellular matrix and increased the size of excitatory postsynaptic densities. Furthermore, 17α-Estradiol enhanced experience-dependent plasticity in the amblyopic visual cortex, by promoting response potentiation of the pathway served by the non-deprived eye. The promotion of plasticity at synapses serving the non-deprived eye may reflect selectivity for synapses with an initially low probability of neurotransmitter release, and may inform strategies to remap spared inputs around a scotoma or a cortical infarct.
Collapse
|
12
|
Youlten SE, Baldock PA. Using mouse genetics to understand human skeletal disease. Bone 2019; 126:27-36. [PMID: 30776501 DOI: 10.1016/j.bone.2019.02.015] [Citation(s) in RCA: 4] [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: 12/03/2018] [Revised: 01/25/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
Abstract
Technological advances have enabled the study of the human genome in incredible detail with relative ease. However, our ability to interpret the functional significance of the millions of genetic variants present within each individual is limited. As a result, the confident assignment of disease-causing variant calls remains a significant challenge. Here we explore how mouse genetics can help address this deficit in functional genomic understanding. Underpinned by marked genetic correspondence, skeletal biology shows inter-species similarities which provide important opportunities to use data from mouse models to direct research into the genetic basis of skeletal pathophysiology. In this article we outline critical resources that may be used to establish genotype/phenotype relationships in skeletal tissue, identify genes with established skeletal effects and define the transcriptome of critical skeletal cell types. Finally, we outline how these mouse resources might be utilized to progress from a list of human sequence variants toward plausible gene candidates that contribute to skeletal disease.
Collapse
Affiliation(s)
- Scott E Youlten
- Division of Bone Biology, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, NSW, 2010, Australia.
| | - Paul A Baldock
- Division of Bone Biology, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Sydney, NSW, 2010, Australia; University of Notre Dame Australia, Sydney, NSW, 2010, Australia
| |
Collapse
|
13
|
Kandasamy M, Radhakrishnan RK, Poornimai Abirami GP, Roshan SA, Yesudhas A, Balamuthu K, Prahalathan C, Shanmugaapriya S, Moorthy A, Essa MM, Anusuyadevi M. Possible Existence of the Hypothalamic-Pituitary-Hippocampal (HPH) Axis: A Reciprocal Relationship Between Hippocampal Specific Neuroestradiol Synthesis and Neuroblastosis in Ageing Brains with Special Reference to Menopause and Neurocognitive Disorders. Neurochem Res 2019; 44:1781-1795. [PMID: 31254250 DOI: 10.1007/s11064-019-02833-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022]
|
14
|
Zárate SC, Traetta ME, Codagnone MG, Seilicovich A, Reinés AG. Humanin, a Mitochondrial-Derived Peptide Released by Astrocytes, Prevents Synapse Loss in Hippocampal Neurons. Front Aging Neurosci 2019; 11:123. [PMID: 31214013 PMCID: PMC6555273 DOI: 10.3389/fnagi.2019.00123] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/09/2019] [Indexed: 01/13/2023] Open
Abstract
Astroglial cells are crucial for central nervous system (CNS) homeostasis. They undergo complex morpho-functional changes during aging and in response to hormonal milieu. Ovarian hormones positively affect different astroglia parameters, including regulation of cell morphology and release of neurotrophic and neuroprotective factors. Thus, ovarian hormone loss during menopause has profound impact in astroglial pathophysilogy and has been widely associated to the process of brain aging. Humanin (HN) is a secreted mitochondrial-encoded peptide with neuroprotective effects. It is localized in several tissues with high metabolic rate and its expression decreases with age. In the brain, humanin has been found in glial cells in physiological conditions. We previously reported that surgical menopause induces hippocampal mitochondrial dysfunction that mimics an aging phenotype. However, the effect of ovarian hormone deprivation on humanin expression in this area has not been studied. Also, whether astrocytes express and release humanin and the regulation of such processes by ovarian hormones remain elusive. Although humanin has also proven to be beneficial in ameliorating cognitive impairment induced by different insults, its putative actions on structural synaptic plasticity have not been fully addressed. In a model of surgical menopause in rats, we studied hippocampal humanin expression and localization by real-time quantitative polymerase chain reaction (RT-qPCR) and double immunohistochemistry, respectively. Humanin production and release and ovarian hormone regulation of such processes were studied in cultured astrocytes by flow cytometry and ELISA, respectively. Humanin effects on glutamate-induced structural synaptic alterations were determined in primary cultures of hippocampal neurons by immunocytochemistry. Humanin expression was lower in the hippocampus of ovariectomized rats and its immunoreactivity colocalized with astroglial markers. Chronic ovariectomy also promoted the presence of less complex astrocytes in this area. Ovarian hormones increased humanin intracellular content and release by cultured astrocytes. Humanin prevented glutamate-induced dendritic atrophy and reduction in puncta number and total puncta area for pre-synaptic marker synaptophysin in cultured hippocampal neurons. In conclusion, astroglial functional and morphological alterations induced by chronic ovariectomy resemble an aging phenotype and could affect astroglial support to neuronal function by altering synaptic connectivity and functionality. Reduced astroglial-derived humanin may represent an underlying mechanism for synaptic dysfunction and cognitive decline after menopause.
Collapse
Affiliation(s)
- Sandra Cristina Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Gabriel Codagnone
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Gabriela Reinés
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Waters EM, Mazid S, Dodos M, Puri R, Janssen WG, Morrison JH, McEwen BS, Milner TA. Effects of estrogen and aging on synaptic morphology and distribution of phosphorylated Tyr1472 NR2B in the female rat hippocampus. Neurobiol Aging 2019; 73:200-210. [PMID: 30384123 DOI: 10.1016/j.neurobiolaging.2018.09.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/10/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022]
Abstract
Age and estrogens may impact the mobility of N-methyl-D-aspartate receptors (NMDARs) in hippocampal synapses. Here, we used serial section immunogold electron microscopy to examine whether phosphorylated tyrosine 1472 NR2B (pY1472), which is involved in the surface expression of NMDARs, is altered in the dorsal hippocampus of young (3-4 months old) and aged (∼24 months old) ovariectomized rats treated with 17β-estradiol or vehicle for 2 days. The number of gold particles labeling pY1472 was higher in presynaptic and postsynaptic compartments of aged rats with low estradiol (vehicle-treated) compared to other groups. In terminals, pY1472 levels were elevated in aged rats but reduced by estradiol treatment to levels seen in young rats. Conversely, the mitochondria number was lower in aged females but was restored to young levels by estradiol. In the postsynaptic density and dendritic spines, estradiol reduced pY1472 in young and aged rats. As phosphorylation at Y1472 blocks NR2B endocytosis, reduction of pY1472 by estradiol suggests another mechanism through which estrogen enhances synaptic plasticity by altering localization of NMDAR subunits within synapses.
Collapse
Affiliation(s)
- Elizabeth M Waters
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Sanoara Mazid
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Mariana Dodos
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rishi Puri
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John H Morrison
- Department of Neurology, Center for Neuroscience, The California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Teresa A Milner
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2018. [PMID: 29311911 DOI: 10.3389/fnagi.2017.00430/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
17
|
Wang S, Zhu J, Xu T. 17β-estradiol (E2) promotes growth and stability of new dendritic spines via estrogen receptor β pathway in intact mouse cortex. Brain Res Bull 2017; 137:241-248. [PMID: 29288734 DOI: 10.1016/j.brainresbull.2017.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/01/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022]
Abstract
The steroid hormone 17β-estradiol (E2) remodels neural circuits at the synaptic level in the mammalian hippocampus and cortex. However, the underlying mechanism of synapse dynamics remains unclear. To elucidate the mechanism, we traced individual dendritic spines on layer V pyramidal neurons of the primary sensory cortex in adult female mice under E2 intervention using two-photon in vivo imaging microscopy. We confirmed the increase of the spine density upon E2 treatment in the intact mouse cortex. Furthermore, we found that this increase is due to the promotion of spine formation and the stability of newly formed spines. E2 treatment doesn't alter the elimination rate of pre-existing spines. Our results also indicate that the activation of the estrogen receptor β (ERβ) mimics the effects of E2 administration on spine dynamics. Taken together, our findings suggest that estrogen promotes growth and stability of new dendritic spines via the ERβ pathway in the intact cortex of female mice.
Collapse
Affiliation(s)
- Shaofang Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jun Zhu
- Chengdu Military General Hospital, Chengdu, China
| | - Tonghui Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
18
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2017; 9:430. [PMID: 29311911 PMCID: PMC5743731 DOI: 10.3389/fnagi.2017.00430] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
19
|
Pike CJ. Sex and the development of Alzheimer's disease. J Neurosci Res 2017; 95:671-680. [PMID: 27870425 DOI: 10.1002/jnr.23827] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022]
Abstract
Men and women exhibit differences in the development and progression of Alzheimer's disease (AD). The factors underlying the sex differences in AD are not well understood. This Review emphasizes the contributions of sex steroid hormones to the relationship between sex and AD. In women, events that decrease lifetime exposure to estrogens are generally associated with increased AD risk, whereas estrogen-based hormone therapy administered near the time of menopause may reduce AD risk. In men, estrogens do not exhibit age-related reduction and are not significantly associated with AD risk. Rather, normal age-related depletions of testosterone in plasma and brain predict enhanced vulnerability to AD. Both estrogens and androgens exert numerous protective actions in the adult brain that increase neural functioning and resilience as well as specifically attenuating multiple aspects of AD-related neuropathology. Aging diminishes the activational effects of sex hormones in sex-specific manners, which is hypothesized to contribute to the relationship between aging and AD. Sex steroid hormones may also drive sex differences in AD through their organizational effects during developmental sexual differentiation of the brain. Specifically, sex hormone actions during early development may confer inherent vulnerability of the female brain to development of AD in advanced age. The combined effects of organizational and activational effects of sex steroids yield distinct sex differences in AD pathogenesis, a significant variable that must be more rigorously considered in future research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|
20
|
Karoglu ET, Halim DO, Erkaya B, Altaytas F, Arslan-Ergul A, Konu O, Adams MM. Aging alters the molecular dynamics of synapses in a sexually dimorphic pattern in zebrafish ( Danio rerio ). Neurobiol Aging 2017; 54:10-21. [DOI: 10.1016/j.neurobiolaging.2017.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 10/20/2022]
|
21
|
Crimins JL, Wang ACJ, Yuk F, Puri R, Janssen WGM, Hara Y, Rapp PR, Morrison JH. Diverse Synaptic Distributions of G Protein-coupled Estrogen Receptor 1 in Monkey Prefrontal Cortex with Aging and Menopause. Cereb Cortex 2017; 27:2022-2033. [PMID: 26941383 PMCID: PMC5909633 DOI: 10.1093/cercor/bhw050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Age- and menopause-related impairment in working memory mediated by the dorsolateral prefrontal cortex (dlPFC) occurs in humans and nonhuman primates. Long-term cyclic 17β-estradiol treatment rescues cognitive deficits in aged ovariectomized rhesus monkeys while restoring highly plastic synapses. Here we tested whether distributions of G protein-coupled estrogen receptor 1 (GPER1) within monkey layer III dlPFC synapses are sensitive to age and estradiol, and coupled to cognitive function. Ovariectomized young and aged monkeys administered vehicle or estradiol were first tested on a delayed response test of working memory. Then, quantitative serial section immunoelectron microscopy was used to determine the distributions of synaptic GPER1. GPER1-containing nonperforated axospinous synapse density was reduced with age, and partially restored with estrogen treatment. The majority of synapses expressed GPER1, which was predominately localized to presynaptic cytoplasm and mitochondria. GPER1 was also abundant at plasmalemmas, and within cytoplasmic and postsynaptic density (PSD) domains of dendritic spines. GPER1 levels did not differ with age or treatment, and none of the variables examined were tightly associated with cognitive function. However, greater representation of GPER1 subjacent to the PSD accompanied higher synapse density. These data suggest that GPER1 is positioned to support diverse functions key to synaptic plasticity in monkey dlPFC.
Collapse
Affiliation(s)
| | - Athena Ching-Jung Wang
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, CO 80045, USA
| | - Frank Yuk
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | - Rishi Puri
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | | | - Yuko Hara
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | - Peter R Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224, USA
| | - John H Morrison
- Fishberg Department of Neuroscience and Friedman Brain Institute
- Department of Geriatrics and Palliative Medicine
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- California National Primate Research Center, Davis, CA 95616, USA
- Department of Neurology, School of Medicine, University of California Davis, Davis 95616, USA
| |
Collapse
|
22
|
Abstract
UNLABELLED A decline in estradiol (E2)-mediated cognitive benefits denotes a critical window for the therapeutic effects of E2, but the mechanism for closing of the critical window is unknown. We hypothesized that upregulating the expression of estrogen receptor α (ERα) or estrogen receptor β (ERβ) in the hippocampus of aged animals would restore the therapeutic potential of E2 treatments and rejuvenate E2-induced hippocampal plasticity. Female rats (15 months) were ovariectomized, and, 14 weeks later, adeno-associated viral vectors were used to express ERα, ERβ, or green fluorescent protein (GFP) in the CA1 region of the dorsal hippocampus. Animals were subsequently treated for 5 weeks with cyclic injections of 17β-estradiol-3-benzoate (EB, 10 μg) or oil vehicle. Spatial memory was examined 48 h after EB/oil treatment. EB treatment in the GFP (GFP + EB) and ERβ (ERβ + EB) groups failed to improve episodic spatial memory relative to oil-treated animals, indicating closing of the critical window. Expression of ERβ failed to improve cognition and was associated with a modest learning impairment. Cognitive benefits were specific to animals expressing ERα that received EB treatment (ERα + EB), such that memory was improved relative to ERα + oil and GFP + EB. Similarly, ERα + EB animals exhibited enhanced NMDAR-mediated synaptic transmission compared with the ERα + oil and GFP + EB groups. This is the first demonstration that the window for E2-mediated benefits on cognition and hippocampal E2 responsiveness can be reinstated by increased expression of ERα. SIGNIFICANCE STATEMENT Estradiol is neuroprotective, promotes synaptic plasticity in the hippocampus, and protects against cognitive decline associated with aging and neurodegenerative diseases. However, animal models and clinical studies indicate a critical window for the therapeutic treatment such that the beneficial effects are lost with advanced age and/or with extended hormone deprivation. We used gene therapy to upregulate expression of the estrogen receptors ERα and ERβ and demonstrate that the window for estradiol's beneficial effects on memory and hippocampal synaptic function can be reinstated by enhancing the expression of ERα. Our findings suggest that the activity of ERα controls the therapeutic window by regulating synaptic plasticity mechanisms involved in memory.
Collapse
|
23
|
Naugle MM, Lozano SA, Guarraci FA, Lindsey LF, Kim JE, Morrison JH, Janssen WG, Yin W, Gore AC. Age and Long-Term Hormone Treatment Effects on the Ultrastructural Morphology of the Median Eminence of Female Rhesus Macaques. Neuroendocrinology 2016; 103:650-64. [PMID: 26536204 PMCID: PMC4860175 DOI: 10.1159/000442015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022]
Abstract
The median eminence (ME) of the hypothalamus comprises the hypothalamic nerve terminals, glia (especially tanycytes) and the portal capillary vasculature that transports hypothalamic neurohormones to the anterior pituitary gland. The ultrastructure of the ME is dynamically regulated by hormones and undergoes organizational changes during development and reproductive cycles in adult females, but relatively little is known about the ME during aging, especially in nonhuman primates. Therefore, we used a novel transmission scanning electron microscopy technique to examine the cytoarchitecture of the ME of young and aged female rhesus macaques in a preclinical monkey model of menopausal hormone treatments. Rhesus macaques were ovariectomized and treated for 2 years with vehicle, estradiol (E2), or estradiol + progesterone (E2 + P4). While the overall cytoarchitecture of the ME underwent relatively few changes with age and hormones, changes to some features of neural and glial components near the portal capillaries were observed. Specifically, large neuroterminal size was greater in aged compared to young adult animals, an effect that was mitigated or reversed by E2 alone but not by E2 + P4 treatment. Overall glial size and the density and tissue fraction of the largest subset of glia were greater in aged monkeys, and in some cases reversed by E2 treatment. Mitochondrial size was decreased by E2, but not E2 + P4, only in aged macaques. These results contrast substantially with work in rodents, suggesting that the ME of aging macaques is less vulnerable to age-related disorganization, and that the effects of E2 on monkeys' ME are age specific.
Collapse
Affiliation(s)
| | - Sateria A. Lozano
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - Fay A. Guarraci
- Department of Psychology, Southwestern University, Georgetown, TX
| | - Larry F. Lindsey
- Center for Learning and Memory, University of Texas at Austin, Austin, TX
| | - Ji E. Kim
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - John H. Morrison
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - William G.M. Janssen
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Weiling Yin
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - Andrea C. Gore
- Institute for Neuroscience, University of Texas at Austin, Austin, TX
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
- Institute for Cellular & Molecular Biology, University of Texas at Austin, Austin, TX
- Correspondence: Andrea C Gore, PhD, The University of Texas at Austin, 107 West Dean Keeton, C0875, Austin, TX, 78712, USA, ; Tel: +1-512-471-3669; Fax: +1-512-471-5002
| |
Collapse
|
24
|
Gogos A, Sbisa AM, Sun J, Gibbons A, Udawela M, Dean B. A Role for Estrogen in Schizophrenia: Clinical and Preclinical Findings. Int J Endocrinol 2015; 2015:615356. [PMID: 26491441 PMCID: PMC4600562 DOI: 10.1155/2015/615356] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/30/2022] Open
Abstract
Gender differences in schizophrenia have been extensively researched and it is being increasingly accepted that gonadal steroids are strongly attributed to this phenomenon. Of the various hormones implicated, the estrogen hypothesis has been the most widely researched one and it postulates that estrogen exerts a protective effect by buffering females against the development and severity of the illness. In this review, we comprehensively analyse studies that have investigated the effects of estrogen, in particular 17β-estradiol, in clinical, animal, and molecular research with relevance to schizophrenia. Specifically, we discuss the current evidence on estrogen dysfunction in schizophrenia patients and review the clinical findings on the use of estradiol as an adjunctive treatment in schizophrenia patients. Preclinical research that has used animal models and molecular probes to investigate estradiol's underlying protective mechanisms is also substantially discussed, with particular focus on estradiol's impact on the major neurotransmitter systems implicated in schizophrenia, namely, the dopamine, serotonin, and glutamate systems.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Alyssa M. Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jeehae Sun
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Andrew Gibbons
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Madhara Udawela
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
25
|
Arevalo MA, Azcoitia I, Gonzalez-Burgos I, Garcia-Segura LM. Signaling mechanisms mediating the regulation of synaptic plasticity and memory by estradiol. Horm Behav 2015; 74:19-27. [PMID: 25921586 DOI: 10.1016/j.yhbeh.2015.04.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/02/2015] [Accepted: 04/20/2015] [Indexed: 01/29/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". Estradiol participates in the regulation of the function and plasticity of synaptic circuits in key cognitive brain regions, such as the prefrontal cortex and the hippocampus. The mechanisms elicited by estradiol are mediated by the regulation of transcriptional activity by nuclear estrogen receptors and by intracellular signaling cascades activated by estrogen receptors associated with the plasma membrane. In addition, the mechanisms include the interaction of estradiol with the signaling of other factors involved in the regulation of cognition, such as brain derived neurotrophic factor, insulin-like growth factor-1 and Wnt. Modifications in these signaling pathways by aging or by a long-lasting ovarian hormone deprivation after menopause may impair the enhancing effects of estradiol on synaptic plasticity and cognition.
Collapse
Affiliation(s)
- Maria-Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avenida Doctor Arce 37, E-28002 Madrid, Spain
| | - Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense, Calle José Antonio Novais 12, Ciudad Universitaria, E-28040 Madrid, Spain
| | - Ignacio Gonzalez-Burgos
- Laboratorio de Psicobiología, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jal. Mexico
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avenida Doctor Arce 37, E-28002 Madrid, Spain.
| |
Collapse
|
26
|
Winsauer PJ, Filipeanu CM, Weed PF, Sutton JL. Hormonal status and age differentially affect tolerance to the disruptive effects of delta-9-tetrahydrocannabinol (Δ(9)-THC) on learning in female rats. Front Pharmacol 2015; 6:133. [PMID: 26191005 PMCID: PMC4488627 DOI: 10.3389/fphar.2015.00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 06/15/2015] [Indexed: 12/28/2022] Open
Abstract
The effects of hormone status and age on the development of tolerance to Δ(9)-THC were assessed in sham-operated (intact) or ovariectomized (OVX) female rats that received either intraperitoneal saline or 5.6 mg/kg of Δ(9)-THC daily from postnatal day (PD) 75-180 (early adulthood onward) or PD 35-140 (adolescence onward). During this time, the four groups for each age (i.e., intact/saline, intact/THC, OVX/saline, and OVX/THC) were trained in a learning and performance procedure and dose-effect curves were established for Δ(9)-THC (0.56-56 mg/kg) and the cannabinoid type-1 receptor (CB1R) antagonist rimonabant (0.32-10 mg/kg). Despite the persistence of small rate-decreasing and error-increasing effects in intact and OVX females from both ages during chronic Δ(9)-THC, all of the Δ(9)-THC groups developed tolerance. However, the magnitude of tolerance, as well as the effect of hormone status, varied with the age at which chronic Δ(9)-THC was initiated. There was no evidence of dependence in any of the groups. Hippocampal protein expression of CB1R, AHA1 (a co-chaperone of CB1R) and HSP90β (a molecular chaperone modulated by AHA-1) was affected more by OVX than chronic Δ(9)-THC; striatal protein expression was not consistently affected by either manipulation. Hippocampal brain-derived neurotrophic factor expression varied with age, hormone status, and chronic treatment. Thus, hormonal status differentially affects the development of tolerance to the disruptive effects of delta-9-tetrahydrocannabinol (Δ(9)-THC) on learning and performance behavior in adolescent, but not adult, female rats. These factors and their interactions also differentially affect cannabinoid signaling proteins in the hippocampus and striatum, and ultimately, neural plasticity.
Collapse
Affiliation(s)
- Peter J. Winsauer
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center New OrleansNew Orleans, LA, USA
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center New OrleansNew Orleans, LA, USA
| | - Catalin M. Filipeanu
- Department of Pharmacology, Howard University College of MedicineWashington, DC, USA
| | - Peter F. Weed
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center New OrleansNew Orleans, LA, USA
| | - Jessie L. Sutton
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center New OrleansNew Orleans, LA, USA
| |
Collapse
|
27
|
Ultrastructural analyses in the hippocampus CA1 field in Shank3-deficient mice. Mol Autism 2015; 6:41. [PMID: 26137200 PMCID: PMC4486760 DOI: 10.1186/s13229-015-0036-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 06/22/2015] [Indexed: 11/10/2022] Open
Abstract
Background The genetics of autism spectrum disorder (hereafter referred to as “autism”) are rapidly unfolding, with a significant increase in the identification of genes implicated in the disorder. Many of these genes are part of a complex landscape of genetic variants that are thought to act together to cause the behavioral phenotype associated with autism. One of the few single-locus causes of autism involves a mutation in the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene. Previous electrophysiological studies in mice with Shank3 mutations demonstrated impairment in synaptic long-term potentiation, suggesting a potential disruption at the synapse. Methods To understand how variants in SHANK3 would lead to such impairments and manifest in the brain of patients with autism, we assessed the presence of synaptic pathology in Shank3-deficient mice at 5 weeks and 3 months of age, focusing on the stratum radiatum of the CA1 field. This study analyzed both Shank3 heterozygous and homozygous mice using an electron microscopy approach to determine whether there is a morphological correlate to the synaptic functional impairment. Results As both synaptic strength and plasticity are affected in Shank3-deficient mice, we hypothesized that there would be a reduction in synapse density, postsynaptic density length, and perforated synapse density. No differences were found in most parameters assessed. However, Shank3 heterozygotes had significantly higher numbers of perforated synapses at 5 weeks compared to 3 months of age and significantly higher numbers of perforated synapses compared to 5-week-old wildtype and Shank3 homozygous mice. Conclusions Although this finding represents preliminary evidence for ultrastructural alterations, it suggests that while major structural changes seem to be compensated for in Shank3-deficient mice, more subtle morphological alterations, affecting synaptic structure, may take place in an age-dependent manner.
Collapse
|
28
|
Mannella P, Simoncini T, Genazzani AR. Estrogens and progestins: molecular effects on brain cells. Horm Mol Biol Clin Investig 2015; 4:609-13. [PMID: 25961237 DOI: 10.1515/hmbci.2010.078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 12/06/2010] [Indexed: 11/15/2022]
Abstract
Sex steroids are known to regulate brain function and their role is so important that several diseases are strictly correlated with the onset of menopause when estrogen-progesterone deficiency makes neural cells much more vulnerable to toxic stimuli. Although in the past years several scientists have focused their studies on in vitro and in vivo effects of sex steroids on the brain, we are still far from complete knowledge. Indeed, contrasting results from large clinical trials have made the entire issue much more complicated. Currently we know that protective effects exerted by sex steroids depend on several factors among which the dose, the health of the cells and the type of molecule being used. In this review, we present an overview of the direct and indirect effects of estrogen and progesterone on the brain with specific focus on the molecular mechanisms by which these molecules act on neural cells.
Collapse
|
29
|
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: 387] [Impact Index Per Article: 43.0] [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
|
30
|
Immunohistochemical colocalization of estrogen receptor-α and GABA in adult female rat hippocampus. Ann Neurosci 2014; 19:112-5. [PMID: 25205981 PMCID: PMC4117079 DOI: 10.5214/ans.0972.7531.190305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 06/06/2012] [Accepted: 07/17/2012] [Indexed: 11/22/2022] Open
Abstract
Background Hippocampus is an important target for estrogen action. It is severely affected in patients of Alzheimer’s disease. Much of the current research related to estrogen and brain function is focused in two directions. Purpose By attempting to lacalize localizing ERs in GABAergic neurons of the hippocampus we tried to test the hypothesis that the action of estrogen in maintaining the neuronal plasticity and more specially the spine density of pyramidal neurons is through GABAergic neurons. Methods The present study was planned to demonstrate the detailed immunoreactive (IR) distribution pattern of estrogen receptors (ER) in GABAergic neurons of hippocampus. The study was conducted in adult female Wistar rats in estrous phase. 30 µm thick cryostat sections of hippocampal region were obtained from perfusion fixed (with 4% buffered Para formaldehyde) adult female rats (n = 15). The sections were processed free- floating for immunolocalization using the PAP protocol. First they were immunostained for ER using, mouse monoclonal anti-ER-α antibody with DAB as chromogen. Subsequently the same sections were immunostained for GABA using rabbit monoclonal anti-GABA antibody respectively with 9 amino 3 ethyl carbazole (AEC) as chromogen. Results The results showed ER were colocalised in GABAergic neurons in all the subfields of hippocampus with obvious variations. In Cornua ammonis (CA) maximum co-localised neurons were seen in CA3 region. Conclusions The view is strengthened by our results as it was established through previous studies that the immediate target neurons of estrogen in hippocampus is the GABAergic neurons.
Collapse
|
31
|
Humphreys GI, Ziegler YS, Nardulli AM. 17β-estradiol modulates gene expression in the female mouse cerebral cortex. PLoS One 2014; 9:e111975. [PMID: 25372139 PMCID: PMC4221195 DOI: 10.1371/journal.pone.0111975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/10/2014] [Indexed: 01/13/2023] Open
Abstract
17β-estradiol (E2) plays critical roles in a number of target tissues including the mammary gland, reproductive tract, bone, and brain. Although it is clear that E2 reduces inflammation and ischemia-induced damage in the cerebral cortex, the molecular mechanisms mediating the effects of E2 in this brain region are lacking. Thus, we examined the cortical transcriptome using a mouse model system. Female adult mice were ovariectomized and implanted with silastic tubing containing oil or E2. After 7 days, the cerebral cortices were dissected and RNA was isolated and analyzed using RNA-sequencing. Analysis of the transcriptomes of control and E2-treated animals revealed that E2 treatment significantly altered the transcript levels of 88 genes. These genes were associated with long term synaptic potentiation, myelination, phosphoprotein phosphatase activity, mitogen activated protein kinase, and phosphatidylinositol 3-kinase signaling. E2 also altered the expression of genes linked to lipid synthesis and metabolism, vasoconstriction and vasodilation, cell-cell communication, and histone modification. These results demonstrate the far-reaching and diverse effects of E2 in the cerebral cortex and provide valuable insight to begin to understand cortical processes that may fluctuate in a dynamic hormonal environment.
Collapse
Affiliation(s)
- Gwendolyn I. Humphreys
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yvonne S. Ziegler
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ann M. Nardulli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
Estradiol effects on memory depend on hormone levels and the interaction of different estrogen receptors within neural circuits. Estradiol induces gene transcription and rapid membrane signaling mediated by estrogen receptor-alpha (ERα), estrogen receptor-beta (ERβ), and a recently characterized G-protein coupled estrogen receptor, each with distinct distributions and ability to influence estradiol-dependent signaling. Investigations using receptor specific agonists suggest that all three receptors rapidly activate kinase-signaling and have complex dose-dependent influences on memory. Research employing receptor knockout mice demonstrate that ERα maintains transcription and memory as estradiol levels decline. This work indicates a regulatory role of ERβ in transcription and cognition, which depends on estradiol levels and the function of ERα. The regulatory role of ERβ is due in part to ERβ acting as a negative regulator of ERα-mediated transcription. Vector-mediated expression of estrogen receptors in the hippocampus provides an innovative research approach and suggests that memory depends on the relative expression of ERα and ERβ interacting with estradiol levels. Notably, the ability of estradiol to improve cognition declines with advanced age along with decreased expression of estrogen receptors. Thus, it will be important for future research to determine the mechanisms that regulate estrogen receptor expression during aging.
Collapse
Affiliation(s)
- Linda A Bean
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lara Ianov
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Zhou L, Fester L, Haghshenas S, de Vrese X, von Hacht R, Gloger S, Brandt N, Bader M, Vollmer G, Rune GM. Oestradiol-induced synapse formation in the female hippocampus: roles of oestrogen receptor subtypes. J Neuroendocrinol 2014; 26:439-47. [PMID: 24779550 DOI: 10.1111/jne.12162] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 04/22/2014] [Accepted: 04/26/2014] [Indexed: 01/09/2023]
Abstract
During the oestrus cycle, varying spine synapse density correlates positively with varying local synthesis of oestradiol in the hippocampus. In this context, the roles of the oestrogen receptor (ER) subtypes ERα and β are not fully understood. In the present study, we used neonatal hippocampal slice cultures from female rats because these cultures synthesise oestradiol and express both receptor subtypes, and inhibition of oestradiol synthesis in these cultures results in spine synapse loss. Using electron microscopy, we tested the effects on spine synapse density in response to agonists of both ERα and ERβ. Application of agonists to the cultures had no effect. After inhibition of oestradiol synthesis, however, agonists of ERα induced spine synapse formation, whereas ERβ agonists led to a reduction in spine synapse density in the CA1 region of these cultures. Consistently, up-regulation of ERβ in the hippocampus of adult female aromatase-deficient mice is paralleled by hippocampus-specific spine synapse loss in this mutant. Finally, we found an increase in spine synapses in the adult female ERβ knockout mouse, but no effect in the adult female ERα knockout mouse. Our data suggest antagonistic roles of ERβ and ERα in spine synapse formation in the female hippocampus, which may contribute to oestrus cyclicity of spine synapse density in the hippocampus.
Collapse
Affiliation(s)
- L Zhou
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
VAFAEE F, HOSSEINI M, SADEGHINIA HR, HADJZADEH MAR, SOUKHTANLOO M, RAHIMI M. The effects of soy extract on spatial learning and memory damage induced by global ischemia in ovariectomised rats. Malays J Med Sci 2014; 21:19-30. [PMID: 25246832 PMCID: PMC4163555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 04/05/2014] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND The effects of soy extract on memory as well as the oxidative damage to brain tissue induced by ischemia was investigated in ovariectomised (OVX) rats. METHODS THE RATS WERE DIVIDED INTO: 1) Sham; 2) OVX; 3) Sham‑Ischemia; 4) OVX‑Ischemia; 5) OVX-Ischemia-S 20; and 6) OVX-Ischemia-S 60. The common carotid artery was occluded (30 minutes), and it was then re-perfused. The OVX-Ischemia-S 20 and OVX-Ischemia-S 60 groups received 20 or 60 mg/kg of soy extract for eight weeks before the ischemia. RESULTS The Sham-Ischemia and OVX-Ischemia groups took a longer time to reach the platform while, spent a shorter time in the target quadrant (Q1) than the Sham and OVX. The escape latencies in the OVX-Ischemia-S 20 and OVX-Ischemia-S 60 groups were lower while, time spent in the Q1 was higher than that of the OVX-Ischemia. In the rotarod test, there were no significant differences between the groups. The hippocampal concentrations of malondialdehyde (MDA) in the Sham-Ischemia and OVX-Ischemia groups were higher than the Sham and OVX. Pre-treatment by 20 and 60 mg/kg of the extract reduced the MDA. CONCLUSION It is suggested that soy prevents memory impairment and brain tissue oxidative damage due to ischemia in OVX rats.
Collapse
Affiliation(s)
- Farzaneh VAFAEE
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| | - Mahmoud HOSSEINI
- Neurogenic Inflammation Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| | - Hamid Reza SADEGHINIA
- Pharmacological Research Center of Medicinal Plants and Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| | - Mosa Al-reza HADJZADEH
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| | - Mohammad SOUKHTANLOO
- Department of Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| | - Motaharah RAHIMI
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Postal Code 9177948564, Iran
| |
Collapse
|
35
|
Vedder LC, Bredemann TM, McMahon LL. Estradiol replacement extends the window of opportunity for hippocampal function. Neurobiol Aging 2014; 35:2183-92. [PMID: 24813636 DOI: 10.1016/j.neurobiolaging.2014.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 01/27/2023]
Abstract
We previously reported that treating aged female rats, ovariectomized (OVX) as young adults, with acute proestrous levels of 17β estradiol (E2) increases CA1 spine density, NMDAR to AMPAR ratio, GluN2B-mediated NMDAR current, and long-term potentiation at CA3-CA1 synapses if administered by 15, but not at 19-month post-OVX, defining the critical window of opportunity. Importantly, when rats are aged with ovaries intact until OVX at 20 months, hippocampal E2 responsiveness is maintained, indicating the deficit at 19-month post-OVX is a consequence of the duration of hormone deprivation and not chronological age. Here, we find the beneficial effect of E2 on novel object recognition in OVX rats was constrained by the same critical window. Furthermore, chronic low-level E2 replacement, commenced by 11-month post-OVX using subcutaneous capsules removed 2 weeks before acute proestrous E2 treatment, prevents the loss of hippocampal responsiveness at 19-month post-OVX. These data define the dynamic nature of the critical window showing that chronic replacement with physiological E2 levels within a certain period post-OVX can lengthen the window.
Collapse
Affiliation(s)
- Lindsey C Vedder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Teruko M Bredemann
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lori L McMahon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
Wang TJ, Chen JR, Wang WJ, Wang YJ, Tseng GF. Genistein partly eases aging and estropause-induced primary cortical neuronal changes in rats. PLoS One 2014; 9:e89819. [PMID: 24587060 PMCID: PMC3934964 DOI: 10.1371/journal.pone.0089819] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/26/2014] [Indexed: 01/11/2023] Open
Abstract
Gonadal hormones can modulate brain morphology and behavior. Recent studies have shown that hypogonadism could result in cortical function deficits. To this end, hormone therapy has been used to ease associated symptoms but the risk may outweigh the benefits. Here we explored whether genistein, a phytoestrogen, is effective in restoring the cognitive and central neuronal changes in late middle age and surgically estropause female rats. Both animal groups showed poorer spatial learning than young adults. The dendritic arbors and spines of the somatosensory cortical and CA1 hippocampal pyramidal neurons were revealed with intracellular dye injection and analyzed. The results showed that dendritic spines on these neurons were significantly decreased. Remarkably, genistein treatment rescued spatial learning deficits and restored the spine density on all neurons in the surgically estropause young females. In late middle age females, genistein was as effective as estradiol in restoring spines; however, the recovery was less thorough than on young OHE rats. Neither genistein nor estradiol rectified the shortened dendritic arbors of the aging cortical pyramidal neurons suggesting that dendritic arbors and spines are differently modulated. Thus, genistein could work at central level to restore excitatory connectivity and appears to be potent alternative to estradiol for easing aging and menopausal syndromes.
Collapse
Affiliation(s)
- Tsyr-Jiuan Wang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Wen-Jay Wang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Yueh-Jan Wang
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| |
Collapse
|
37
|
Bates K, Vink R, Martins R, Harvey A. Aging, cortical injury and Alzheimer's disease-like pathology in the guinea pig brain. Neurobiol Aging 2013; 35:1345-51. [PMID: 24360504 DOI: 10.1016/j.neurobiolaging.2013.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 10/26/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized histopathologically by the abnormal deposition of the proteins amyloid-beta (Aβ) and tau. A major issue for AD research is the lack of an animal model that accurately replicates the human disease, thus making it difficult to investigate potential risk factors for AD such as head injury. Furthermore, as age remains the strongest risk factor for most of the AD cases, transgenic models in which mutant human genes are expressed throughout the life span of the animal provide only limited insight into age-related factors in disease development. Guinea pigs (Cavia porcellus) are of interest in AD research because they have a similar Aβ sequence to humans and thus may present a useful non-transgenic animal model of AD. Brains from guinea pigs aged 3-48 months were examined to determine the presence of age-associated AD-like pathology. In addition, fluid percussion-induced brain injury was performed to characterize mechanisms underlying the association between AD risk and head injury. No statistically significant changes were detected in the overall response to aging, although we did observe some region-specific changes. Diffuse deposits of Aβ were found in the hippocampal region of the oldest animals and alterations in amyloid precursor protein processing and tau immunoreactivity were observed with age. Brain injury resulted in a strong and sustained increase in amyloid precursor protein and tau immunoreactivity without Aβ deposition, over 7 days. Guinea pigs may therefore provide a useful model for investigating the influence of environmental and non-genetic risk factors on the pathogenesis of AD.
Collapse
Affiliation(s)
- Kristyn Bates
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia; School of Psychiatry and Clinical Neuroscience, The University of Western Australia, Crawley, Western Australia, Australia; The McCusker Foundation for Alzheimer's Disease Research Inc, Nedlands, Western Australia, Australia.
| | - Robert Vink
- School of Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Ralph Martins
- School of Psychiatry and Clinical Neuroscience, The University of Western Australia, Crawley, Western Australia, Australia; The McCusker Foundation for Alzheimer's Disease Research Inc, Nedlands, Western Australia, Australia; School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Alan Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
38
|
Phosphorylation of cofilin regulates extinction of conditioned aversive memory via AMPAR trafficking. J Neurosci 2013; 33:6423-33. [PMID: 23575840 DOI: 10.1523/jneurosci.5107-12.2013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Actin dynamics provide an important mechanism for the modification of synaptic plasticity, which is regulated by the actin depolymerizing factor (ADF)/cofilin. However, the role of cofilin regulated actin dynamics in memory extinction process is still unclear. Here, we observed that extinction of conditioned taste aversive (CTA) memory led to temporally enhanced ADF/cofilin activity in the infralimbic cortex (IrL) of the rats. Moreover, temporally elevating ADF/cofilin activity in the IrL could accelerate CTA memory extinction by facilitating AMPAR synaptic surface recruitment, whereas inhibition of ADF/cofilin activity abolished AMPAR synaptic surface trafficking and impaired memory extinction. Finally, we observed that ADF/cofilin-regulated synaptic plasticity was not directly coupled to morphological changes of postsynaptic spines. These findings may help us understand the role of ADF/cofilin-regulated actin dynamics in memory extinction and suggest that appropriate manipulating ADF/cofilin activity might be a suitable way for therapeutic treatment of memory disorders.
Collapse
|
39
|
Arimoto JM, Wong A, Rozovsky I, Lin SW, Morgan TE, Finch CE. Age increase of estrogen receptor-α (ERα) in cortical astrocytes impairs neurotrophic support in male and female rats. Endocrinology 2013; 154:2101-13. [PMID: 23515288 PMCID: PMC3740484 DOI: 10.1210/en.2012-2046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rodent models show decreased neuronal responses to estradiol (E2) during aging (E2-desensitization) in association with reduced neuronal estrogen receptor (ER)-α, but little is known about age changes of E2-dependent astrocytic neurotrophic support. Because elevated expression of astrocyte glial fibrillary acidic protein (GFAP) is associated with impaired neurotrophic activity and because the GFAP promoter responds to ERα, we investigated the role of astrocytic ERα and ERβ in impaired astrocyte neurotrophic activity during aging. In vivo and in vitro, ERα was increased greater than 50% with age in astrocytes from the cerebral cortex of male rats (24 vs 3 months), whereas ERβ did not change. In astrocytes from 3-month-old males, experimentally increasing the ERα to ERβ ratio induced the aging phenotype of elevated GFAP and impaired E2-dependent neurite outgrowth. In 24-month-old male astrocytes, lowering ERα reversed the age elevation of GFAP and partially restored E2-dependent neurite outgrowth. Mixed glia (astrocytes to microglia, 3:1) of both sexes also showed these age changes. In a model of perimenopause, mixed glia from 9- to 15-month rats showed E2 desensitization: 9-month regular cyclers retained young-like ERα to ERβ ratios and neurotrophic activity, whereas 9-month noncyclers had elevated ERα and GFAP but low E2-dependent neurotrophic activity. In vivo, ERα levels in cortical astrocytes were also elevated. The persisting effects of ovarian acyclicity in vitro are hypothesized to arise from steroidal perturbations during ovarian senescence. These findings suggest that increased astrocyte ERα expression during aging contributes to the E2 desensitization of the neuronal responses in both sexes.
Collapse
Affiliation(s)
- Jason M Arimoto
- Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, California 90089-0191, USA
| | | | | | | | | | | |
Collapse
|
40
|
Chisholm NC, Juraska JM. Factors influencing the cognitive and neural effects of hormone treatment during aging in a rodent model. Brain Res 2013; 1514:40-9. [PMID: 23419893 DOI: 10.1016/j.brainres.2013.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 02/06/2013] [Accepted: 02/11/2013] [Indexed: 12/18/2022]
Abstract
Whether hormone treatment alters brain structure or has beneficial effects on cognition during aging has recently become a topic of debate. Although previous research has indicated that hormone treatment benefits memory in menopausal women, several newer studies have shown no effect or detrimental effects. These inconsistencies emphasize the need to evaluate the role of hormones in protecting against age-related cognitive decline in an animal model. Importantly, many studies investigating the effects of estrogen and progesterone on cognition and related brain regions have used young adult animals, which respond differently than aged animals. However, when only the studies that have examined the effects of hormone treatment in an aging model are reviewed, there are still varied behavioral and neural outcomes. This article reviews some of the important factors that can influence the behavioral and neural outcomes of hormone treatment including the type of estrogen administered, whether or not estrogen is combined with progesterone and if so, the type of progesterone used, as well as the route, mode, and length of treatment. How these factors influence cognitive outcomes highlights the importance of study design and avoiding generalizations from a small number of studies. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Psychology, University of Illinois at Urbana - Champaign, Champaign, IL 61820, USA.
| | | |
Collapse
|
41
|
Daniel JM. Estrogens, estrogen receptors, and female cognitive aging: the impact of timing. Horm Behav 2013; 63:231-7. [PMID: 22587940 DOI: 10.1016/j.yhbeh.2012.05.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 10/28/2022]
Abstract
Estrogens have been shown to be protective agents against neurodegeneration and associated cognitive decline in aging females. However, clinical data have been equivocal as to the benefits to the brain and cognition of estrogen therapy in postmenopausal women. One factor that is proposed to be critical in determining the efficacy of hormone therapy is the timing of its initiation. The critical period or window of opportunity hypothesis proposes that following long-term ovarian hormone deprivation, the brain and cognition become insensitive to exogenously administered estrogens. In contrast, if estrogens are administered during a critical period near the time of cessation of ovarian function, they will exert beneficial effects. The focus of the current review is the examination of evidence from rodent models investigating the critical period hypothesis. A growing body of experimental data indicates that beneficial effects of 17β-estradiol (estradiol) on cognition and on cholinergic function and hippocampal plasticity, both of which have been linked to the ability of estradiol to exert beneficial effects on cognition, are attenuated if estradiol is administered following a period of long-term ovarian hormone deprivation. Further, emerging data implicate loss of estrogen receptor alpha (ERα) in the brain resulting from long-term hormone deprivation as a basis for the existence of the critical period. A unifying model is proposed by which the presence or absence of estrogens during a critical period following the cessation of ovarian function permanently alters the system resulting in decreased or increased risk, respectively, of neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Program in Neuroscience, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
42
|
Buwalda B, Schagen SB. Is basic research providing answers if adjuvant anti-estrogen treatment of breast cancer can induce cognitive impairment? Life Sci 2013; 93:581-8. [PMID: 23353876 DOI: 10.1016/j.lfs.2012.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/27/2012] [Accepted: 12/21/2012] [Indexed: 12/23/2022]
Abstract
Adjuvant treatment of cancer by chemotherapy is associated with cognitive impairment in some cancer survivors. Breast cancer patients are frequently also receiving endocrine therapy with selective estrogen receptor modulators (SERMs) and/or aromatase inhibitors (AIs) to suppress the growth of estradiol sensitive breast tumors. Estrogens are well-known, however, to target brain areas involved in the regulation of cognitive behavior. In this review clinical and basic preclinical research is reviewed on the actions of estradiol, SERMs and AIs on brain and cognitive functioning to see if endocrine therapy potentially induces cognitive impairment and in that respect may contribute to the detrimental effects of chemotherapy on cognitive performance in breast cancer patients. Although many clinical studies may be underpowered to detect changes in cognitive function, current basic and clinical reports suggest that there is little evidence that AIs may have a lasting detrimental effect on cognitive performance in breast cancer patients. The clinical data on SERMs are not conclusive, but some studies do suggest that tamoxifen administration may form a risk for cognitive functioning particularly in older women. An explanation may come from basic preclinical research which indicates that tamoxifen often acts agonistic in the absence of estradiol but antagonistic in the presence of endogenous estradiol. It could be hypothesized that the negative effects of tamoxifen in older women is related to the so-called window of opportunity for estrogen. Administration of SERMs beyond this so-called window of opportunity may not be effective or might even have detrimental effects similar to estradiol.
Collapse
Affiliation(s)
- Bauke Buwalda
- Behavioral Physiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| | | |
Collapse
|
43
|
Cui J, Shen Y, Li R. Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med 2013; 19:197-209. [PMID: 23348042 DOI: 10.1016/j.molmed.2012.12.007] [Citation(s) in RCA: 461] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/18/2012] [Accepted: 12/27/2012] [Indexed: 01/05/2023]
Abstract
Estrogens are the primary female sex hormones and play important roles in both reproductive and non-reproductive systems. Estrogens can be synthesized in non-reproductive tissues such as liver, heart, muscle, bone and brain, and tissue-specific estrogen synthesis is consistent with a diversity of estrogen actions. In this article we review tissue and cell-specific estrogen synthesis and estrogen receptor signaling in three parts: (i) synthesis and metabolism, (ii) the distribution of estrogen receptors and signaling, and (iii) estrogen functions and related disorders, including cardiovascular diseases, osteoporosis, Alzheimer's disease (AD), and Parkinson disease (PD). This comprehensive review provides new insights into estrogens by giving a better understanding of the tissue-specific estrogen effects and their roles in various diseases.
Collapse
Affiliation(s)
- Jie Cui
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, USA
| | | | | |
Collapse
|
44
|
Effects of long-term treatment with estrogen and medroxyprogesterone acetate on synapse number in the medial prefrontal cortex of aged female rats. Menopause 2012; 19:804-11. [PMID: 22617337 DOI: 10.1097/gme.0b013e31824d1fc4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study investigated the effects of long-term hormone treatment, including the most commonly prescribed progestin, medroxyprogesterone acetate, during aging on synaptophysin-labeled boutons, a marker of synapses, in the medial prefrontal cortex (mPFC) of rats. METHODS Female Long Evans hooded rats were ovariectomized at middle age (12-13 mo) and were placed in one of four groups: no replacement (n = 5), 17β-estradiol alone (n = 6), estradiol and progesterone (n = 7), or estradiol and medroxyprogesterone acetate (n = 4). Estradiol was administered in the drinking water and progestogens were administered via subcutaneous pellets that were replaced every 90 days. After 7 months of hormone replacement, the animals were euthanized, and the brains were stained for synaptophysin, a membrane component of synaptic vesicles. The density of synaptophysin-labeled boutons was quantified in the mPFC using unbiased stereology and multiplied by the volume of the mPFC to obtain the total number. RESULTS Animals receiving estradiol and medroxyprogesterone acetate had significantly more synaptophysin-labeled boutons in the mPFC than did animals not receiving replacement (P < 0.03) and those receiving estradiol and progesterone (P < 0.02). In addition, there was a nonsignificant trend for animals receiving estradiol alone to have more synapses than those receiving estradiol and progesterone. CONCLUSIONS This study is the first to examine the effects of estradiol and medroxyprogesterone acetate during rat aging on cortical synaptic number. Estradiol with medroxyprogesterone acetate, but not progesterone, resulted in a greater number of synapses in the mPFC during aging than did no replacement.
Collapse
|
45
|
Dickstein DL, Weaver CM, Luebke JI, Hof PR. Dendritic spine changes associated with normal aging. Neuroscience 2012; 251:21-32. [PMID: 23069756 DOI: 10.1016/j.neuroscience.2012.09.077] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 12/14/2022]
Abstract
Given the rapid rate of population aging and the increased incidence of cognitive decline and neurodegenerative diseases with advanced age, it is important to ascertain the determinants that result in cognitive impairment. It is also important to note that much of the aged population exhibit 'successful' cognitive aging, in which cognitive impairment is minimal. One main goal of normal aging studies is to distinguish the neural changes that occur in unsuccessful (functionally impaired) subjects from those of successful (functionally unimpaired) subjects. In this review, we present some of the structural adaptations that neurons and spines undergo throughout normal aging and discuss their likely contributions to electrophysiological properties and cognition. Structural changes of neurons and dendritic spines during aging, and the functional consequences of such changes, remain poorly understood. Elucidating the structural and functional synaptic age-related changes that lead to cognitive impairment may lead to the development of drug treatments that can restore or protect neural circuits and mediate cognition and successful aging.
Collapse
Affiliation(s)
- D L Dickstein
- Fishberg Department of Neuroscience, Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; Computational Neurobiology and Imaging Center, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
46
|
Abstract
Women are relatively protected against schizophrenia. The illness has a similar rate in women and men, but it starts later in women and is less severe. It is tempting to attribute this to the neuroprotective effect of estrogen, but the story is not straightforward and contains many unknowns. Women begin their schizophrenia trajectory later in development compared with men and this probably accounts for their relatively superior prognosis. Estrogen agonists are potential therapeutic agents but need to be proven safe, and the timing of administration may be crucial. This article examines what is known about estrogen and the development of schizophrenia.
Collapse
Affiliation(s)
- Mary V Seeman
- Centre for Addiction and Mental Health, University of Toronto 250 College St. Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Mortillo S, Elste A, Ge Y, Patil SB, Hsiao K, Huntley GW, Davis RL, Benson DL. Compensatory redistribution of neuroligins and N-cadherin following deletion of synaptic β1-integrin. J Comp Neurol 2012; 520:2041-52. [PMID: 22488504 DOI: 10.1002/cne.23027] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
β1-containing integrins are required for persistent synaptic potentiation in hippocampus and regulate hippocampal-dependent learning. Based largely on indirect evidence, there is a prevailing assumption that β1-integrins are localized at synapses, where they contribute to synapse adhesion and signaling, but this has not been examined directly. Here we investigate the fine localization of β1-integrin in adult mouse hippocampus using high-resolution immunogold labeling, with a particular emphasis on synaptic labeling patterns. We find that β1-integrins localize to synapses in CA1 and are concentrated postsynaptically. At the postsynaptic membrane, β1-integrins are found more commonly clustered near active zone centers rather than at the peripheral edges. In mice harboring a conditional deletion of β1-integrins, labeling for N-cadherin and neuroligins increases. Western blots show increased levels of N-cadherin in total lysates and neuroligins increase selectively in synaptosomes. These data suggest there is a dynamic, compensatory adjustment of synaptic adhesion. Such adjustment is specific only for certain cell adhesion molecules (CAMs), because labeling for SynCAM is unchanged. Together, our findings demonstrate unequivocally that β1-integrin is an integral synaptic adhesion protein, and suggest that adhesive function at the synapse reflects a cooperative and dynamic network of multiple CAM families.
Collapse
Affiliation(s)
- Steven Mortillo
- Department of Neuroscience, Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Morphological and molecular changes in aging rat prelimbic prefrontal cortical synapses. Neurobiol Aging 2012; 34:200-10. [PMID: 22727942 DOI: 10.1016/j.neurobiolaging.2012.05.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 05/23/2012] [Accepted: 05/24/2012] [Indexed: 01/07/2023]
Abstract
Age-related impairments of executive functions appear to be related to reductions of the number and plasticity of dendritic spine synapses in the prefrontal cortex (PFC). Experimental evidence suggests that synaptic plasticity is mediated by the spine actin cytoskeleton, and a major pathway regulating actin-based plasticity is controlled by phosphorylated LIM kinase (pLIMK). We asked whether aging resulted in altered synaptic density, morphology, and pLIMK expression in the rat prelimbic region of the PFC. Using unbiased electron microscopy, we found an approximate 50% decrease in the density of small synapses with aging, while the density of large synapses remained unchanged. Postembedding immunogold revealed that pLIMK localized predominantly to the postsynaptic density where it was increased in aging synapses by approximately 50%. Furthermore, the age-related increase in pLIMK occurred selectively within the largest subset of prelimbic PFC synapses. Because pLIMK is known to inhibit actin filament plasticity, these data support the hypothesis that age-related increases in pLIMK may explain the stability of large synapses at the expense of their plasticity.
Collapse
|
49
|
Inagaki T, Kaneko N, Zukin RS, Castillo PE, Etgen AM. Estradiol attenuates ischemia-induced death of hippocampal neurons and enhances synaptic transmission in aged, long-term hormone-deprived female rats. PLoS One 2012; 7:e38018. [PMID: 22675505 PMCID: PMC3366987 DOI: 10.1371/journal.pone.0038018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 05/02/2012] [Indexed: 11/18/2022] Open
Abstract
Background Transient global forebrain ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons, and the ovarian hormone 17β-estradiol (E2) reduces neuronal loss in young and middle-aged females. The neuroprotective efficacy of E2 after a prolonged period of hormone deprivation is controversial, and few studies examine this issue in aged animals given E2 treatment after induction of ischemia. Methodology/Principal Findings The present study investigated the neuroprotective effects of E2 administered immediately after global ischemia in aged female rats (15–18 months) after 6 months of hormone deprivation. We also used electrophysiological methods to assess whether CA1 synapses in the aging hippocampus remain responsive to E2 after prolonged hormone withdrawal. Animals were ovariohysterectomized and underwent 10 min global ischemia 6 months later. A single dose of E2 (2.25 µg) infused intraventricularly after reperfusion significantly increased cell survival, with 45% of CA1 neurons surviving vs 15% in controls. Ischemia also induced moderate loss of CA3/CA4 pyramidal cells. Bath application of 1 nM E2 onto brain slices derived from non-ischemic aged females after 6 months of hormone withdrawal significantly enhanced excitatory transmission at CA1 synapses evoked by Schaffer collateral stimulation, and normal long-term potentiation (LTP) was induced. The magnitude of LTP and of E2 enhancement of field excitatory postsynaptic potentials was indistinguishable from that recorded in slices from young rats. Conclusions/Significance The data demonstrate that 1) acute post-ischemic infusion of E2 into the brain ventricles is neuroprotective in aged rats after 6 months of hormone deprivation; and 2) E2 enhances synaptic transmission in CA1 pyramidal neurons of aged long-term hormone deprived females. These findings provide evidence that the aging hippocampus remains responsive to E2 administered either in vivo or in vitro even after prolonged periods of hormone withdrawal.
Collapse
Affiliation(s)
- Tomoko Inagaki
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Naoki Kaneko
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Pablo E. Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Anne M. Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
50
|
Frick KM. Building a better hormone therapy? How understanding the rapid effects of sex steroid hormones could lead to new therapeutics for age-related memory decline. Behav Neurosci 2012; 126:29-53. [PMID: 22289043 DOI: 10.1037/a0026660] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A wealth of data collected in recent decades has demonstrated that ovarian sex-steroid hormones, particularly 17β-estradiol (E2), are important trophic factors that regulate the function of cognitive regions of the brain such as the hippocampus. The loss of hormone cycling at menopause is associated with cognitive decline and dementia in women, and the onset of memory decline in animal models. However, hormone therapy is not currently recommended to prevent or treat cognitive decline, in part because of its detrimental side effects. In this article, it is proposed that investigations of the rapid effects of E2 on hippocampal function be used to further the design of new drugs that mimic the beneficial effects of E2 on memory without the side effects of current therapies. A conceptual model is presented for elucidating the molecular and biochemical mechanisms through which sex-steroid hormones modulate memory, and a specific hypothesis is proposed to account for the rapid memory-enhancing effects of E2. Empirical support for this hypothesis is discussed as a means of stimulating the consideration of new directions for the development of hormone-based therapies to preserve memory function in menopausal women.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 East Hartford Avenue, Milwaukee, WI 53211, USA.
| |
Collapse
|