1
|
Joue G, Navarro-Schröder T, Achtzehn J, Moffat S, Hennies N, Fuß J, Döller C, Wolbers T, Sommer T. Effects of estrogen on spatial navigation and memory. Psychopharmacology (Berl) 2024; 241:1037-1063. [PMID: 38407638 PMCID: PMC11031496 DOI: 10.1007/s00213-024-06539-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024]
Abstract
RATIONALE Animal studies suggest that the so-called "female" hormone estrogen enhances spatial navigation and memory. This contradicts the observation that males generally out-perform females in spatial navigation and tasks involving spatial memory. A closer look at the vast number of studies actually reveals that performance differences are not so clear. OBJECTIVES To help clarify the unclear performance differences between men and women and the role of estrogen, we attempted to isolate organizational from activational effects of estrogen on spatial navigation and memory. METHODS In a double-blind, placebo-controlled study, we tested the effects of orally administered estradiol valerate (E2V) in healthy, young women in their low-hormone menstrual cycle phase, compared to healthy, young men. Participants performed several first-person, environmentally rich, 3-D computer games inspired by spatial navigation and memory paradigms in animal research. RESULTS We found navigation behavior suggesting that sex effects dominated any E2 effects with men performing better with allocentric strategies and women with egocentric strategies. Increased E2 levels did not lead to general improvements in spatial ability in either sex but to behavioral changes reflecting navigation flexibility. CONCLUSION Estrogen-driven differences in spatial cognition might be better characterized on a spectrum of navigation flexibility rather than by categorical performance measures or skills.
Collapse
Affiliation(s)
- Gina Joue
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Tobias Navarro-Schröder
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030, Trondheim, Norway
| | - Johannes Achtzehn
- Department of Neurology with Experimental Neurology (CVK), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Scott Moffat
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA, 30332, USA
| | - Nora Hennies
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Johannes Fuß
- Institute of Forensic Psychiatry and Sex Research, University Duisburg-Essen, Hohlweg 26, 45147, Essen, Germany
| | - Christian Döller
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030, Trondheim, Norway
- Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103, Leipzig, Germany
| | - Thomas Wolbers
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Tobias Sommer
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
2
|
Calvo N, Einstein G. Steroid hormones: risk and resilience in women's Alzheimer disease. Front Aging Neurosci 2023; 15:1159435. [PMID: 37396653 PMCID: PMC10313425 DOI: 10.3389/fnagi.2023.1159435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
More women have Alzheimer disease (AD) than men, but the reasons for this phenomenon are still unknown. Including women in clinical research and studying their biology is key to understand not just their increased risk but also their resilience against the disease. In this sense, women are more affected by AD than men, but their reserve or resilience mechanisms might delay symptom onset. The aim of this review was to explore what is known about mechanisms underlying women's risk and resilience in AD and identify emerging themes in this area that merit further research. We conducted a review of studies analyzing molecular mechanisms that may induce neuroplasticity in women, as well as cognitive and brain reserve. We also analyzed how the loss of steroid hormones in aging may be linked to AD. We included empirical studies with human and animal models, literature reviews as well as meta-analyses. Our search identified the importance of 17-b-estradiol (E2) as a mechanism driving cognitive and brain reserve in women. More broadly, our analysis revealed the following emerging perspectives: (1) the importance of steroid hormones and their effects on both neurons and glia for the study of risk and resilience in AD, (2) E2's crucial role in women's brain reserve, (3) women's verbal memory advantage as a cognitive reserve factor, and (4) E2's potential role in linguistic experiences such as multilingualism and hearing loss. Future directions for research include analyzing the reserve mechanisms of steroid hormones on neuronal and glial plasticity, as well as identifying the links between steroid hormone loss in aging and risk for AD.
Collapse
Affiliation(s)
- Noelia Calvo
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Women’s College Research Institute, Toronto, ON, Canada
- Centre for Life Course and Aging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Koebele SV, Poisson ML, Palmer JM, Berns-Leone C, Northup-Smith SN, Peña VL, Strouse IM, Bulen HL, Patel S, Croft C, Bimonte-Nelson HA. Evaluating the Cognitive Impacts of Drospirenone, a Spironolactone-Derived Progestin, Independently and in Combination With Ethinyl Estradiol in Ovariectomized Adult Rats. Front Neurosci 2022; 16:885321. [PMID: 35692432 PMCID: PMC9177129 DOI: 10.3389/fnins.2022.885321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/14/2022] [Indexed: 11/15/2022] Open
Abstract
Oral contraceptives and hormone therapies require a progestogen component to prevent ovulation, curtail uterine hyperplasia, and reduce gynecological cancer risk. Diverse classes of synthetic progestogens, called progestins, are used as natural progesterone alternatives due to progesterone’s low oral bioavailability. Progesterone and several synthetic analogs can negatively impact cognition and reverse some neuroprotective estrogen effects. Here, we investigate drospirenone, a spironolactone-derived progestin, which has unique pharmacological properties compared to other clinically-available progestins and natural progesterone, for its impact on spatial memory, anxiety-like behavior, and brain regions crucial to these cognitive tasks. Experiment 1 assessed three drospirenone doses in young adult, ovariectomized rats, and found that a moderate drospirenone dose benefited spatial memory. Experiment 2 investigated this moderate drospirenone dose with and without concomitant ethinyl estradiol (EE) treatment, the most common synthetic estrogen in oral contraceptives. Results demonstrate that the addition of EE to drospirenone administration reversed the beneficial working memory effects of drospirenone. The hippocampus, entorhinal cortex, and perirhinal cortex were then probed for proteins known to elicit estrogen- and progestin- mediated effects on learning and memory, including glutamate decarboxylase (GAD)65, GAD67, and insulin-like growth factor receptor protein expression, using western blot. EE increased GAD expression in the perirhinal cortex. Taken together, results underscore the necessity to consider the distinct cognitive and neural impacts of clinically-available synthetic estrogen and progesterone analogs, and why they produce unique cognitive profiles when administered together compared to those observed when each hormone is administered separately.
Collapse
Affiliation(s)
- Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Mallori L. Poisson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Justin M. Palmer
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Claire Berns-Leone
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Steven N. Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Veronica L. Peña
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Isabel M. Strouse
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Haidyn L. Bulen
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Corissa Croft
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- *Correspondence: Heather A. Bimonte-Nelson,
| |
Collapse
|
4
|
Blaya MO, Raval AP, Bramlett HM. Traumatic brain injury in women across lifespan. Neurobiol Dis 2022; 164:105613. [PMID: 34995753 DOI: 10.1016/j.nbd.2022.105613] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 11/27/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability and a global public health challenge. Every year more than 50 million people suffer a TBI, and it is estimated that 50% of the global population will experience at least one TBI in their lifetime. TBI affects both men and women of all ages, however there is a male bias in TBI research as women have frequently been left out of the literature despite irrefutable evidence of male and female dimorphism in several posttraumatic measures. Women uniquely experience distinct life stages marked by levels of endogenous circulating sex hormones, as well as by physiological changes that are nonexistent in men. In addition to generalized sex-specific differences, a woman's susceptibility, neurological outcomes, and treatment success may vary considerably depending upon when in her lifespan she incurred a traumatic insult. How women impacted by TBI might differ from other women as a factor of age and physiology is not well understood. Furthermore, there is a gap in the knowledge of what happens when TBI occurs in the presence of certain sex-specific and sex-nonspecific variables, such as during pregnancy, with oral contraceptive use, in athletics, in cases of addiction and nicotine consumption, during perimenopause, postmenopause, in frailty, among others. Parsing out how hormone-dependent and hormone-independent lifespan variables may influence physiological, neurodegenerative, and functional outcomes will greatly contribute to future investigative studies and direct therapeutic strategies. The goal of this review is to aggregate the knowledge of prevalence, prognosis, comorbid risk, and response of women incurring TBI at differing phases of lifespan. We strive to illuminate commonalities and disparities among female populations, and to pose important questions to highlight gaps in the field in order to further the endeavor of targeted treatment interventions in a patient-specific manner.
Collapse
Affiliation(s)
- Meghan O Blaya
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA.
| |
Collapse
|
5
|
Sheppard PAS, Puri TA, Galea LAM. Sex Differences and Estradiol Effects in MAPK and Akt Cell Signaling across Subregions of the Hippocampus. Neuroendocrinology 2022; 112:621-635. [PMID: 34407537 DOI: 10.1159/000519072] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/16/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Rapid effects of estrogens within the hippocampus of rodents are dependent upon cell-signaling cascades, and activation of these cascades by estrogens varies by sex. Whether these pathways are rapidly activated within the dentate gyrus (DG) and CA1 by estrogens across sex and the anatomical longitudinal axis has been overlooked. METHODS Gonadally intact female and male rats were given either vehicle or physiological systemic low (1.1 µg/kg) or high (37.3 µg/kg) doses of 17β-estradiol 30 min prior to tissue collection. To control for the effects of circulating estrogens, an additional group of female rats was ovariectomized (OVX) and administered 17β-estradiol. Brains were extracted, and tissue punches of the CA1 and DG were taken along the longitudinal hippocampal axis (dorsal and ventral) and analyzed for key mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) cascade phosphoproteins. RESULTS Intact females had higher Akt pathway phosphoproteins (pAkt, pGSK-3β, and pp70S6K) than males in the DG (dorsal and ventral) and lower pERK1/2 in the dorsal DG. Most effects of 17β-estradiol on cell signaling occurred in OVX animals. In OVX animals, 17β-estradiol increased cell signaling of MAPK and Akt phosphoproteins (pERK1/2, pJNK, pAkt, and pGSK-3β) in the CA1 and pERK1/2 and pJNK DG. DISCUSSION/CONCLUSIONS Systemic 17β-estradiol treatment rapidly alters phosphoprotein levels in the hippocampus, dependent on reproductive status, and intact females have greater expression of Akt phosphoproteins than that in intact males in the DG. These findings shed light on underlying mechanisms of sex differences in hippocampal function and response to interventions that affect MAPK or Akt signaling.
Collapse
Affiliation(s)
- Paul A S Sheppard
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tanvi A Puri
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
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 PMCID: PMC11548941 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
|
7
|
Santos VR, Pun RYK, Arafa SR, LaSarge CL, Rowley S, Khademi S, Bouley T, Holland KD, Garcia-Cairasco N, Danzer SC. PTEN deletion increases hippocampal granule cell excitability in male and female mice. Neurobiol Dis 2017; 108:339-351. [PMID: 28855130 DOI: 10.1016/j.nbd.2017.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/10/2017] [Accepted: 08/26/2017] [Indexed: 02/06/2023] Open
Abstract
Deletion of the mTOR pathway inhibitor PTEN from postnatally-generated hippocampal dentate granule cells causes epilepsy. Here, we conducted field potential, whole cell recording and single cell morphology studies to begin to elucidate the mechanisms by which granule cell-specific PTEN-loss produces disease. Cells from both male and female mice were recorded to identify sex-specific effects. PTEN knockout granule cells showed altered intrinsic excitability, evident as a tendency to fire in bursts. PTEN knockout granule cells also exhibited increased frequency of spontaneous excitatory synaptic currents (sEPSCs) and decreased frequency of inhibitory currents (sIPSCs), further indicative of a shift towards hyperexcitability. Morphological studies of PTEN knockout granule cells revealed larger dendritic trees, more dendritic branches and an impairment of dendrite self-avoidance. Finally, cells from both female control and female knockout mice received more sEPSCs and more sIPSCs than corresponding male cells. Despite the difference, the net effect produced statistically equivalent EPSC/IPSC ratios. Consistent with this latter observation, extracellularly evoked responses in hippocampal slices were similar between male and female knockouts. Both groups of knockouts were abnormal relative to controls. Together, these studies reveal a host of physiological and morphological changes among PTEN knockout cells likely to underlie epileptogenic activity. SIGNIFICANCE STATEMENT Hyperactivation of the mTOR pathway is associated with numerous neurological diseases, including autism and epilepsy. Here, we demonstrate that deletion of the mTOR negative regulator, PTEN, from a subset of hippocampal dentate granule impairs dendritic patterning, increases excitatory input and decreases inhibitory input. We further demonstrate that while granule cells from female mice receive more excitatory and inhibitory input than males, PTEN deletion produces mostly similar changes in both sexes. Together, these studies provide new insights into how the relatively small number (≈200,000) of PTEN knockout granule cells instigates the development of the profound epilepsy syndrome evident in both male and female animals in this model.
Collapse
Affiliation(s)
- Victor R Santos
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Raymund Y K Pun
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Salwa R Arafa
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; University of Cincinnati, College of Pharmacy, Cincinnati, OH 45267, United States
| | - Candi L LaSarge
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Shane Rowley
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Shadi Khademi
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Tom Bouley
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Katherine D Holland
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Norberto Garcia-Cairasco
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, United States.
| |
Collapse
|
8
|
Koebele SV, Bimonte-Nelson HA. The endocrine-brain-aging triad where many paths meet: female reproductive hormone changes at midlife and their influence on circuits important for learning and memory. Exp Gerontol 2016; 94:14-23. [PMID: 27979770 DOI: 10.1016/j.exger.2016.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 01/15/2023]
Abstract
Female mammals undergo natural fluctuations in sex steroid hormone levels throughout life. These fluctuations span from early development, to cyclic changes associated with the menstrual or estrous cycle and pregnancy, to marked hormone flux during perimenopause, and a final decline at reproductive senescence. While the transition to reproductive senescence is not yet fully understood, the vast majority of mammals experience this spontaneous, natural phenomenon with age, which has broad implications for long-lived species. Indeed, this post-reproductive life stage, and its transition, involves significant and enduring physiological changes, including considerably altered sex steroid hormone and gonadotropin profiles that impact multiple body systems, including the brain. The endocrine-brain-aging triad is especially noteworthy, as many paths meet and interact. Many of the brain regions affected by aging are also sensitive to changes in ovarian hormone levels, and aging and reproductive senescence are both associated with changes in memory performance. This review explores how menopause is related to cognitive aging, and discusses some of the key neural systems and molecular factors altered with age and reproductive hormone level changes, with an emphasis on brain regions important for learning and memory.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
9
|
Hamson DK, Roes MM, Galea LAM. Sex Hormones and Cognition: Neuroendocrine Influences on Memory and Learning. Compr Physiol 2016; 6:1295-337. [DOI: 10.1002/cphy.c150031] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
10
|
Koebele SV, Bimonte-Nelson HA. Modeling menopause: The utility of rodents in translational behavioral endocrinology research. Maturitas 2016; 87:5-17. [PMID: 27013283 DOI: 10.1016/j.maturitas.2016.01.015] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 01/25/2016] [Indexed: 01/31/2023]
Abstract
The human menopause transition and aging are each associated with an increase in a variety of health risk factors including, but not limited to, cardiovascular disease, osteoporosis, cancer, diabetes, stroke, sexual dysfunction, affective disorders, sleep disturbances, and cognitive decline. It is challenging to systematically evaluate the biological underpinnings associated with the menopause transition in the human population. For this reason, rodent models have been invaluable tools for studying the impact of gonadal hormone fluctuations and eventual decline on a variety of body systems. While it is essential to keep in mind that some of the mechanisms associated with aging and the transition into a reproductively senescent state can differ when translating from one species to another, animal models provide researchers with opportunities to gain a fundamental understanding of the key elements underlying reproduction and aging processes, paving the way to explore novel pathways for intervention associated with known health risks. Here, we discuss the utility of several rodent models used in the laboratory for translational menopause research, examining the benefits and drawbacks in helping us to better understand aging and the menopause transition in women. The rodent models discussed are ovary-intact, ovariectomy, and 4-vinylcylohexene diepoxide for the menopause transition. We then describe how these models may be implemented in the laboratory, particularly in the context of cognition. Ultimately, we aim to use these animal models to elucidate novel perspectives and interventions for maintaining a high quality of life in women, and to potentially prevent or postpone the onset of negative health consequences associated with these significant life changes during aging.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
11
|
Au A, Feher A, McPhee L, Jessa A, Oh S, Einstein G. Estrogens, inflammation and cognition. Front Neuroendocrinol 2016; 40:87-100. [PMID: 26774208 DOI: 10.1016/j.yfrne.2016.01.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/02/2016] [Accepted: 01/11/2016] [Indexed: 01/15/2023]
Abstract
The effects of estrogens are pleiotropic, affecting multiple bodily systems. Changes from the body's natural fluctuating levels of estrogens, through surgical removal of the ovaries, natural menopause, or the administration of exogenous estrogens to menopausal women have been independently linked to an altered immune profile, and changes to cognitive processes. Here, we propose that inflammation may mediate the relationship between low levels of estrogens and cognitive decline. In order to determine what is known about this connection, we review the literature on the cognitive effects of decreased estrogens due to oophorectomy or natural menopause, decreased estrogens' role on inflammation--both peripherally and in the brain--and the relationship between inflammation and cognition. While this review demonstrates that much is unknown about the intersection between estrogens, cognition, inflammation, we propose that there is an important interaction between these literatures.
Collapse
Affiliation(s)
- April Au
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Anita Feher
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Lucy McPhee
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Ailya Jessa
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Soojin Oh
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| | - Gillian Einstein
- University of Toronto, 100 St. George Street, 4F Sidney Smith Hall, Dept. of Psychology, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
12
|
Yagi S, Chow C, Lieblich SE, Galea LAM. Sex and strategy use matters for pattern separation, adult neurogenesis, and immediate early gene expression in the hippocampus. Hippocampus 2015; 26:87-101. [PMID: 26179150 DOI: 10.1002/hipo.22493] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2015] [Indexed: 12/12/2022]
Abstract
Adult neurogenesis in the dentate gyrus (DG) plays a crucial role for pattern separation, and there are sex differences in the regulation of neurogenesis. Although sex differences, favoring males, in spatial navigation have been reported, it is not known whether there are sex differences in pattern separation. The current study was designed to determine whether there are sex differences in the ability for separating similar or distinct patterns, learning strategy choice, adult neurogenesis, and immediate early gene (IEG) expression in the DG in response to pattern separation training. Male and female Sprague-Dawley rats received a single injection of the DNA synthesis marker, bromodeoxyuridine (BrdU), and were tested for the ability of separating spatial patterns in a spatial pattern separation version of delayed nonmatching to place task using the eight-arm radial arm maze. Twenty-seven days following BrdU injection, rats received a probe trial to determine whether they were idiothetic or spatial strategy users. We found that male spatial strategy users outperformed female spatial strategy users only when separating similar, but not distinct, patterns. Furthermore, male spatial strategy users had greater neurogenesis in response to pattern separation training than all other groups. Interestingly, neurogenesis was positively correlated with performance on similar pattern trials during pattern separation in female spatial strategy users but negatively correlated with performance in male idiothetic strategy users. These results suggest that the survival of new neurons may play an important positive role for pattern separation of similar patterns in females. Furthermore, we found sex and strategy differences in IEG expression in the CA1 and CA3 regions in response to pattern separation. These findings emphasize the importance of studying biological sex on hippocampal function and neural plasticity.
Collapse
Affiliation(s)
- Shunya Yagi
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carmen Chow
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephanie E Lieblich
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
13
|
Koebele SV, Bimonte-Nelson HA. Trajectories and phenotypes with estrogen exposures across the lifespan: What does Goldilocks have to do with it? Horm Behav 2015; 74:86-104. [PMID: 26122297 PMCID: PMC4829405 DOI: 10.1016/j.yhbeh.2015.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/14/2015] [Accepted: 06/04/2015] [Indexed: 01/04/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Estrogens impact the organization and activation of the mammalian brain in both sexes, with sex-specific critical windows. Throughout the female lifespan estrogens activate brain substrates previously organized by estrogens, and estrogens can induce non-transient brain and behavior changes into adulthood. Therefore, from early life through the transition to reproductive senescence and beyond, estrogens are potent modulators of the brain and behavior. Organizational, reorganizational, and activational hormone events likely impact the trajectory of brain profiles during aging. A "brain profile," or quantitative brain measurement for research purposes, is typically a snapshot in time, but in life a brain profile is anything but static--it is in flux, variable, and dynamic. Akin to this, the only thing continuous and consistent about hormone exposures across a female's lifespan is that they are noncontinuous and inconsistent, building and rebuilding on past exposures to create a present brain and behavioral landscape. Thus, hormone variation is especially rich in females, and is likely the destiny for maximal responsiveness in the female brain. The magnitude and direction of estrogenic effects on the brain and its functions depend on a myriad of factors; a "Goldilocks" phenomenon exists for estrogens, whereby if the timing, dose, and regimen for an individual are just right, markedly efficacious effects present. Data indicate that exogenously-administered estrogens can bestow beneficial cognitive effects in some circumstances, especially when initiated in a window of opportunity such as the menopause transition. Could it be that the age-related reduction in efficacy of estrogens reflects the closure of a late-in-life critical window occurring around the menopause transition? Information from classic and contemporary works studying organizational/activational estrogen actions, in combination with acknowledging the tendency for maximal responsiveness to cyclicity, will elucidate ways to extend sensitivity and efficacy into post-menopause.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA.
| |
Collapse
|
14
|
Hojo Y, Munetomo A, Mukai H, Ikeda M, Sato R, Hatanaka Y, Murakami G, Komatsuzaki Y, Kimoto T, Kawato S. Estradiol rapidly modulates spinogenesis in hippocampal dentate gyrus: Involvement of kinase networks. Horm Behav 2015; 74:149-56. [PMID: 26122288 DOI: 10.1016/j.yhbeh.2015.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 01/15/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Estradiol (E2) is locally synthesized within the hippocampus and the gonads. Rapid modulation of hippocampal synaptic plasticity by E2 is essential for synaptic regulation. The molecular mechanisms of modulation through the synaptic estrogen receptor (ER) and its downstream signaling, however, are largely unknown in the dentate gyrus (DG). We investigated the E2-induced modulation of dendritic spines in male adult rat hippocampal slices by imaging Lucifer Yellow-injected DG granule cells. Treatments with 1 nM E2 increased the density of spines by approximately 1.4-fold within 2h. Spine head diameter analysis showed that the density of middle-head spines (0.4-0.5 μm) was significantly increased. The E2-induced spine density increase was suppressed by blocking Erk MAPK, PKA, PKC and LIMK. These suppressive effects by kinase inhibitors are not non-specific ones because the GSK-3β antagonist did not inhibit E2-induced spine increase. The ER antagonist ICI 182,780 also blocked the E2-induced spine increase. Taken together, these results suggest that E2 rapidly increases the density of spines through kinase networks that are driven by synaptic ER.
Collapse
Affiliation(s)
- Yasushi Hojo
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan; Bioinformatics Project of Japan Science and Technology Agency, University of Tokyo, Japan
| | - Arisa Munetomo
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Hideo Mukai
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan; Bioinformatics Project of Japan Science and Technology Agency, University of Tokyo, Japan
| | - Muneki Ikeda
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Rei Sato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Yusuke Hatanaka
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Gen Murakami
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan; Bioinformatics Project of Japan Science and Technology Agency, University of Tokyo, Japan
| | - Yoshimasa Komatsuzaki
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Tetsuya Kimoto
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153-8902, Japan; Bioinformatics Project of Japan Science and Technology Agency, University of Tokyo, Japan; Department of Urology, Juntendo University, Graduate School of Medicine, Tokyo 113-8431, Japan.
| |
Collapse
|
15
|
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
|
16
|
Lipatova O, Byrd D, Green JT, Toufexis DJ. Effects of continuous vs. cycling estrogen replacement on the acquisition, retention and expression of place- and response-learning in the open-field tower maze. Neurobiol Learn Mem 2014; 114:81-9. [PMID: 24837787 DOI: 10.1016/j.nlm.2014.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/23/2014] [Accepted: 05/05/2014] [Indexed: 01/28/2023]
Abstract
Estrogen has been shown to either enhance or impair learning and memory in female rats. The use of different experimental paradigms or estrogen treatment regimens may contribute to these disparate findings. In order to assess the effect of different estradiol (E2) treatments on several aspects of cognition, we trained ovariectomized female rats with either continuous, cycling, or vehicle E2 replacement, in an open-field tower maze task (OFTM) designed to test reference memory in a low-stress environment. In addition, in order to compare two distinct learning and memory systems, rats were trained to use either a dorsolateral striatum-based response type learning or a hippocampal-based place type learning to solve the maze. Results showed that cyclic, but not continuous, E2 replacement facilitated the acquisition of spatial memory in place-learners. Neither E2 regimen affected acquisition in response-learners. Additionally, when all experimental groups were performing at asymptote, rats were evaluated for performance stability by changing the location of their start position in the OFTM. Both regimens of E2 disrupted the expression of spatial memory in place-learners following the novel start position. However, E2 replacement protected ovariectomized female rats from the disruption of memory expression following a start position change in response-learners. Additionally all experimental groups performed equally well when tested following a 21-day period during which rats were absent from the maze. These results suggest that E2 fluctuation is particularly important in the acquisition of hippocampal-mediated spatial learning, and that hippocampal-based memory may be subject to disruption following environmental change, while striatum-based memory is subject to protection.
Collapse
Affiliation(s)
- Olga Lipatova
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States.
| | - Dennis Byrd
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| | - John T Green
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| | - Donna J Toufexis
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| |
Collapse
|
17
|
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
|
18
|
Galea LAM, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol 2013; 25:1039-61. [PMID: 23822747 DOI: 10.1111/jne.12070] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/23/2013] [Accepted: 06/29/2013] [Indexed: 12/12/2022]
Abstract
The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex- and experience-dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.
Collapse
Affiliation(s)
- L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
19
|
Baxter MG, Roberts MT, Gee NA, Lasley BL, Morrison JH, Rapp PR. Multiple clinically relevant hormone therapy regimens fail to improve cognitive function in aged ovariectomized rhesus monkeys. Neurobiol Aging 2013; 34:1882-90. [PMID: 23369546 PMCID: PMC3622837 DOI: 10.1016/j.neurobiolaging.2012.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 12/10/2012] [Accepted: 12/22/2012] [Indexed: 10/27/2022]
Abstract
Preclinical studies in aged, surgically-menopausal rhesus monkeys have revealed powerful benefits of intermittent estrogen injections on prefrontal cortex-dependent working memory, together with corresponding effects on dendritic spine morphology in the prefrontal cortex. This contrasts with the inconsistent effects of hormone therapy (HT) reported in clinical studies in women. Factors contributing to this discrepancy could include differences in the formulation and sequence of HT regimens, resulting in different neurobiological outcomes. The current study evaluated, in aging surgically menopausal rhesus monkeys, the cognitive effects of 4 HT regimens modeled directly on human clinical practice, including continuous estrogen treatment opposed by progesterone. None of the regimens tested produced any cognitive effect, despite yielding physiologically relevant serum hormone levels, as intended. These findings have implications for the design of regimens that might optimize the benefits of hormone treatment for healthy aging, and suggest that common HT protocols used by women may fail to result in substantial cognitive benefit, at least via direct effects on the prefrontal cortex.
Collapse
Affiliation(s)
- Mark G Baxter
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
20
|
Velíšková J, Desantis KA. Sex and hormonal influences on seizures and epilepsy. Horm Behav 2013; 63:267-77. [PMID: 22504305 PMCID: PMC3424285 DOI: 10.1016/j.yhbeh.2012.03.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 11/20/2022]
Abstract
Epilepsy is the third most common chronic neurological disorder. Clinical and experimental evidence supports the role of sex and influence of sex hormones on seizures and epilepsy as well as alterations of the endocrine system and levels of sex hormones by epileptiform activity. Conversely, seizures are sensitive to changes in sex hormone levels, which in turn may affect the seizure-induced neuronal damage. The effects of reproductive hormones on neuronal excitability and seizure-induced damage are complex to contradictory and depend on different mechanisms, which have to be accounted for in data interpretation. Both estradiol and progesterone/allopregnanolone may have beneficial effects for patients with epilepsy. Individualized hormonal therapy should be considered as adjunctive treatment in patients with epilepsy to improve seizure control as well as quality of life.
Collapse
Affiliation(s)
- Jana Velíšková
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA.
| | | |
Collapse
|
21
|
Abstract
The nature and extent of the impact of gender and reproductive function on mood has been the subject of speculation and controversy for centuries. Over the past 50 years, however, it has become increasingly clear that not only is the brain a major target of reproductive steroid hormones, but additionally, the steroid hormones, as neuroregulators, create a context thai influences a broad range of brain activities; ie, neural actions and resultant behaviors are markedly different in the presence and absence of gonadal steroids. In turn, the actions of gonadal steroids are themselves context-dependent. Thus, even where it can be demonstrated thai gonadal steroids trigger mood disorders, the triggers are normal levels of gonadal steroids (to be contrasted with the mood disturbances accompanying endocrinopathies), and the mood disorders appear only in a subset of susceptible individuals. The context specificity and differential susceptibility to affective dysregulation seen in women with reproductive endocrine-related mood disorders are undoubtedly important underlying characteristics of a wide range of psychiatric disorders in which the triggers have not yet been identified. Consequently, reproductive endocrine-related mood disorders offer unparalleled promise for the identification of those contextual variables that permit biological stimuli to differentially translate into depression in individuals at risk.
Collapse
Affiliation(s)
- David R Rubinow
- Behavioral Endocrinology Branch, National Institute of Mental Health, Bethesda, Md, USA
| | | |
Collapse
|
22
|
Bayer J, Rune G, Kutsche K, Schwarze U, Kalisch R, Büchel C, Sommer T. Estrogen and the male hippocampus: Genetic variation in the aromatase gene predicting serum estrogen is associated with hippocampal gray matter volume in men. Hippocampus 2012; 23:117-21. [DOI: 10.1002/hipo.22059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2012] [Indexed: 01/11/2023]
|
23
|
Foster TC. Role of estrogen receptor alpha and beta expression and signaling on cognitive function during aging. Hippocampus 2012; 22:656-69. [PMID: 21538657 PMCID: PMC3704216 DOI: 10.1002/hipo.20935] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2011] [Indexed: 12/24/2022]
Abstract
This review presents evidence for the idea that the expression of estrogen receptor alpha and beta (ERα and ERβ) interacts with the level of estradiol (E2) to influence the etiology of age-related cognitive decline and responsiveness to E2 treatments. There is a nonmonotonic dose response curve for E2 influences on behavior and transcription. Evidence is mounting to indicate that the dose response curve is shifted according to the relative expression of ERα and ERβ. Recent work characterizing age-related changes in the expression of ERα and ERβ in the hippocampus, as well as studies using mutant mice, and viral mediated delivery of estrogen receptors indicate that an age-related shift in ERα/ERβ expression, combined with declining gonadal E2 can impact transcription, cell signaling, neuroprotection, and neuronal growth. Finally, the role of ERα/ERβ on rapid E2 signaling and synaptogenesis as it relates to hippocampal aging is discussed.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610-0244, USA.
| |
Collapse
|
24
|
Abstract
Oestrogen has important roles not only in the regulation of reproductive function, but also with respect to other functions, such as cognition, emotion and cardiovascular regulation. Oestrogen acts mainly via its oestrogen receptor (ER), namely, ERα and ERβ in target tissues, including the brain. During ageing, the actions of oestrogen are altered in both females and males, raising the possibility that the expression level of ER may be altered with age. Age-related changes in ER expression in female rat brain have been well demonstrated with regard to reproductive ageing, whereas very little is known about the effects of age on the expression of ERs, especially ERβ, in males. In the present study, which aimed to elucidate the effects of ageing on ERβ expression in the male brain at the transcriptional level, we performed in situ hybridisation using young (10weeks), middle-aged (12months) and old (24 months) gonadally-intact male rats. We revealed a wide distribution of ERβ mRNA-positive cells throughout the brain, and found that the number of ERβ mRNA-positive cells was reduced in several brain regions in males with ageing. ERβ mRNA-positive cells were decreased with age in layer 6 of the cerebral cortex, hippocampal CA1/CA3 regions, the dorsal endopiriform nucleus, the medial septal nucleus, various subregions of the amygdala (central, lateral, anterior cortical and posterolateral cortical subnuclei), the anteroventral periventricular nucleus, the substantia nigra pars compacta, the raphe magnus nucleus and the locus coeruleus. These results suggest that ERβ expression in male rat brain decreases with age at the transcriptional level and that these ageing effects are region-specific.
Collapse
Affiliation(s)
- N Yamaguchi
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Kohasu, Nankoku, Japan
| | | |
Collapse
|
25
|
Abstract
A promising strategy to delay and perhaps prevent Alzheimer's disease (AD) is to identify the age-related changes that put the brain at risk for the disease. A significant normal age change known to result in tissue-specific dysfunction is the depletion of sex hormones. In women, menopause results in a relatively rapid loss of estradiol and progesterone. In men, aging is associated with a comparatively gradual yet significant decrease in testosterone. We review a broad literature that indicates age-related losses of estrogens in women and testosterone in men are risk factors for AD. Both estrogens and androgens exert a wide range of protective actions that improve multiple aspects of neural health, suggesting that hormone therapies have the potential to combat AD pathogenesis. However, translation of experimental findings into effective therapies has proven challenging. One emerging treatment option is the development of novel hormone mimetics termed selective estrogen and androgen receptor modulators. Continued research of sex hormones and their roles in the aging brain is expected to yield valuable approaches to reducing the risk of AD.
Collapse
Affiliation(s)
- Anna M. Barron
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Japan
| | - Christian J. Pike
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
26
|
Wu Y, Moriya-Ito K, Iwakura T, Tsutiya A, Ichikawa M, Ohtani-Kaneko R. Sexually dimorphic effects of estrogen on spines in cultures of accessory olfactory bulb. Neurosci Lett 2011; 500:77-81. [DOI: 10.1016/j.neulet.2011.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 05/08/2011] [Accepted: 06/03/2011] [Indexed: 10/18/2022]
|
27
|
Aenlle KK, Foster TC. Aging alters the expression of genes for neuroprotection and synaptic function following acute estradiol treatment. Hippocampus 2011; 20:1047-60. [PMID: 19790252 DOI: 10.1002/hipo.20703] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study used microarray analysis to examine age-related changes in gene expression 6 and 12 h following a single estradiol injection in ovariectomized mice. Estradiol-responsive gene expression at the 6 h time point was reduced in aged (18 months) animals compared with young (4 months) and middle-aged (MA, 12 months) mice. Examination of gene clustering within biological and functional pathways indicated that young and MA mice exhibited increased expression of genes for cellular components of the synapse and decreased expression of genes related to oxidative phosphorylation and mitochondrial dysfunction. At the 12 h time point, estradiol-responsive gene expression increased in aged animals and decreased in young and MA mice compared with the 6 h time point. Gene clustering analysis indicated that aged mice exhibited increased expression of genes for signaling pathways that are rapidly influenced by estradiol. The age differences in gene expression for rapid signaling pathways may relate to disparity in basal pathway activity and estradiol mediated activation of rapid signaling cascades.
Collapse
Affiliation(s)
- Kristina K Aenlle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
28
|
Waters EM, Yildirim M, Janssen WGM, Lou WYW, McEwen BS, Morrison JH, Milner TA. Estrogen and aging affect the synaptic distribution of estrogen receptor β-immunoreactivity in the CA1 region of female rat hippocampus. Brain Res 2011; 1379:86-97. [PMID: 20875808 PMCID: PMC3046233 DOI: 10.1016/j.brainres.2010.09.069] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 09/17/2010] [Accepted: 09/17/2010] [Indexed: 11/29/2022]
Abstract
Estradiol (E) mediates increased synaptogenesis in the hippocampal CA1 stratum radiatum (sr) and enhances memory in young and some aged female rats, depending on dose and age. Young female rats express more estrogen receptor α (ERα) immunolabeling in CA1sr spine synapse complexes than aged rats and ERα regulation is E sensitive in young but not aged rats. The current study examined whether estrogen receptor β (ERβ) expression in spine synapse complexes may be altered by age or E treatment. Young (3-4 months) and aged (22-23 months) female rats were ovariectomized 7 days prior to implantation of silastic capsules containing either vehicle (cholesterol) or E (10% in cholesterol) for 2 days. ERβ immunoreactivity (ir) in CA1sr was quantitatively analyzed using post-embedding electron microscopy. ERβ-ir was more prominent post-synaptically than pre-synaptically and both age and E treatment affected its synaptic distribution. While age decreased the spine synaptic complex localization of ERβ-ir (i.e., within 60 nm of the pre- and post-synaptic membranes), E treatment increased synaptic ERβ in both young and aged rats. In addition, the E treatment, but not age, increased dendritic shaft labeling. This data demonstrates that like ERα the levels of ERβ-ir decrease in CA1 axospinous synapses with age, however, unlike ERα the levels of ERβ-ir increase in these synapses in both young and aged rats in response to E. This suggests that synaptic ERβ may be a more responsive target to E, particularly in aged females.
Collapse
Affiliation(s)
- Elizabeth M Waters
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Barron AM, Hojo Y, Mukai H, Higo S, Ooishi Y, Hatanaka Y, Ogiue-Ikeda M, Murakami G, Kimoto T, Kawato S. Regulation of synaptic plasticity by hippocampus synthesized estradiol. Horm Mol Biol Clin Investig 2011; 7:361-75. [PMID: 25961274 DOI: 10.1515/hmbci.2011.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 07/21/2011] [Indexed: 01/29/2023]
Abstract
Estradiol is synthesized from cholesterol in hippocampal neurons of adult rats by cytochrome P450 and hydroxysteroid dehydrogenase enzymes. These enzymes are expressed in the glutamatergic neurons of the hippocampus. Surprisingly, the concentration of estradiol and androgen in the hippocampus is significantly higher than that in circulation. Locally synthesized estradiol rapidly and potently modulates synaptic plasticity within the hippocampus. E2 rapidly potentiates long-term depression and induces spinogenesis through synaptic estrogen receptors and kinases. The rapid effects of estradiol are followed by slow genomic effects mediated by both estrogen receptors located at the synapse and nucleus, modulating long-term potentiation and promoting the formation of new functional synaptic contacts. Age-related changes in hippocampally derived estradiol synthesis and distribution of estrogen receptors may alter synaptic plasticity, and could potentially contribute to age-related cognitive decline. Understanding factors which regulate hippocampal estradiol synthesis could lead to the identification of alternatives to conventional hormone therapy to protect against age-related cognitive decline.
Collapse
|
30
|
Zhang D, Guo Q, Bian C, Zhang J, Cai W, Su B. Expression of Steroid Receptor Coactivator-1 Was Regulated by Postnatal Development but Not Ovariectomy in the Hippocampus of Rats. Dev Neurosci 2011; 33:57-63. [DOI: 10.1159/000322978] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/20/2010] [Indexed: 11/19/2022] Open
|
31
|
Boulware MI, Kent BA, Frick KM. The impact of age-related ovarian hormone loss on cognitive and neural function. Curr Top Behav Neurosci 2011; 10:165-84. [PMID: 21533680 DOI: 10.1007/7854_2011_122] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
On average, women now live one-third of their lives after menopause. Because menopause has been associated with an elevated risk of dementia, an increasing body of research has studied the effects of reproductive senescence on cognitive function. Compelling evidence from humans, nonhuman primates, and rodents suggests that ovarian sex-steroid hormones can have rapid and profound effects on memory, attention, and executive function, and on regions of the brain that mediate these processes, such as the hippocampus and prefrontal cortex. This chapter will provide an overview of studies in humans, nonhuman primates, and rodents that examine the effects of ovarian hormone loss and hormone replacement on cognitive functions mediated by the hippocampus and prefrontal cortex. For humans and each animal model, we outline the effects of aging on reproductive function, describe how ovarian hormones (primarily estrogens) modulate hippocampal and prefrontal physiology, and discuss the effects of both reproductive aging and hormone treatment on cognitive function. Although this review will show that much has been learned about the effects of reproductive senescence on cognition, many critical questions remain for future investigation.
Collapse
Affiliation(s)
- Marissa I Boulware
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave, Milwaukee, WI 53211, USA
| | | | | |
Collapse
|
32
|
Influence of different estrogens on neuroplasticity and cognition in the hippocampus. Biochim Biophys Acta Gen Subj 2010; 1800:1056-67. [DOI: 10.1016/j.bbagen.2010.01.006] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 01/13/2010] [Accepted: 01/16/2010] [Indexed: 11/18/2022]
|
33
|
Daniel JM, Bohacek J. The critical period hypothesis of estrogen effects on cognition: Insights from basic research. Biochim Biophys Acta Gen Subj 2010; 1800:1068-76. [DOI: 10.1016/j.bbagen.2010.01.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 01/09/2010] [Accepted: 01/16/2010] [Indexed: 02/07/2023]
|
34
|
Williams TJ, Mitterling KL, Thompson LI, Torres-Reveron A, Waters EM, McEwen BS, Gore AC, Milner TA. Age- and hormone-regulation of opioid peptides and synaptic proteins in the rat dorsal hippocampal formation. Brain Res 2010; 1379:71-85. [PMID: 20828542 DOI: 10.1016/j.brainres.2010.08.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 08/13/2010] [Accepted: 08/31/2010] [Indexed: 02/05/2023]
Abstract
Circulating estrogen levels and hippocampal-dependent cognitive functions decline with aging. Moreover, the responses of hippocampal synaptic structure to estrogens differ between aged and young rats. We recently reported that estrogens increase levels of post-synaptic proteins, including PSD-95, and opioid peptides leu-enkephalin and dynorphin in the hippocampus of young animals. However, the influence of ovarian hormones on synaptic protein and opioid peptide levels in the aging hippocampus is understudied. Here, young (3- to 5-month-old), middle-aged (9- to 12-month-old), and aged (about 22-month-old) female rats were ovariectomized and then, 4 weeks later, subcutaneously implanted with a silastic capsule containing vehicle or 17β-estradiol. After 48 h, rats were subcutaneously injected with progesterone or vehicle and sacrificed 1 day later. Coronal sections through the dorsal hippocampus were processed for quantitative peroxidase immunohistochemistry of leu-enkephalin, dynorphin, synaptophysin, and PSD-95. With age, females showed opposing changes in leu-enkephalin and dynorphin levels in the mossy fiber pathway, particularly within the hilus, and regionally specific changes in synaptic protein levels. 17β-estradiol, with or without progesterone, altered leu-enkephalin levels in the dentate gyrus and synaptophysin levels in the CA1 of young but not middle-aged or aged females. Additionally, 17β-estradiol decreased synaptophysin levels in the CA3 of middle-aged females. Our results support and extend previous findings indicating 17β-estradiol modulation of hippocampal opioid peptides and synaptic proteins while demonstrating regional and age-specific effects. Moreover, they lend credence to the "window of opportunity" hypothesis during which hormone replacement can modulate hippocampal structure and circuitry to improve cognitive outcomes.
Collapse
Affiliation(s)
- Tanya J Williams
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Rodgers SP, Bohacek J, Daniel JM. Transient estradiol exposure during middle age in ovariectomized rats exerts lasting effects on cognitive function and the hippocampus. Endocrinology 2010; 151:1194-203. [PMID: 20068005 DOI: 10.1210/en.2009-1245] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We determined whether transient exposure to estradiol during middle age in ovariectomized rats would exert lasting effects on cognition and the brain beyond the period of exposure. Two experiments were conducted. Rats 10-11 months of age were ovariectomized and received vehicle control treatment throughout the experiment, continuous estradiol treatment throughout the experiment, or 40 d of transient exposure to estradiol that ended 3 d before behavioral training. In the first experiment, rats were trained on a radial-maze working memory task and killed 2 months after the termination of transient exposure to estradiol. The hippocampus was immunostained for choline acetyltransferase and estrogen receptors alpha (ER alpha) and beta (ER beta) by Western blotting. In a second experiment to determine the durability of treatment effects, rats were behaviorally tested every other month until brains were collected for Western blotting 8 months after the termination of transient exposure to estradiol. Maze testing included delay trials and scopolamine trials, in which dose-effect curves for the muscarinic receptor antagonist were determined. Transient exposure to estradiol enhanced working memory and attenuated amnestic effects of scopolamine as effectively as continuous estradiol exposure. Enhancements persisted for up to 7 months. Transient exposure to estradiol increased hippocampal levels of ER alpha and choline acetyltransferase 2 months and ER alpha 8 months after termination of the exposure. Neither estradiol treatment altered estrogen receptor beta levels. Results demonstrate that short-term treatment with estradiol during middle age enhances working memory well beyond the duration of treatment and suggest ER alpha as a potential mechanism for this effect.
Collapse
Affiliation(s)
- Shaefali P Rodgers
- Department of Psychology and Neuroscience Program, Tulane University, New Orleans, Louisiana 70118, USA
| | | | | |
Collapse
|
36
|
Barha CK, Dalton GL, Galea LAM. Low doses of 17alpha-estradiol and 17beta-estradiol facilitate, whereas higher doses of estrone and 17alpha- and 17beta-estradiol impair, contextual fear conditioning in adult female rats. Neuropsychopharmacology 2010; 35:547-59. [PMID: 19847162 PMCID: PMC3055382 DOI: 10.1038/npp.2009.161] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogens are known to exert significant structural and functional effects in the hippocampus of adult rodents. In particular, 17beta-estradiol can improve, impair, or have no effect on hippocampus-dependent learning and memory depending on dose and time of administration. The effects of other forms of estrogen, such as estrone and 17alpha-estradiol, on hippocampus-dependent learning have not been as thoroughly investigated. Therefore, the purpose of this study was to investigate the effects of 17beta-estradiol, estrone, and 17alpha-estradiol at three different doses on two different tasks: hippocampus-dependent contextual fear conditioning and hippocampus-independent cued fear conditioning. Adult ovariectomized female rats were injected with one of the estrogens at one of the three doses 30 mins before conditioning to assess the rapid effects of these estrogens on acquisition. Twenty-four hours later memory for the context was examined and 1 h later memory for the cue (tone) was assessed. Levels of synaptophysin were examined in the dorsal hippocampus of rats to identify a potential synaptic correlate of hormonal effects on contextual fear conditioning. Low 17beta-estradiol and 17alpha-estradiol enhanced, whereas high 17beta-estradiol and 17alpha-estradiol impaired, contextual fear conditioning. Only the middle dose of estrone severely impaired contextual fear conditioning. Estrogens did not alter performance in the hippocampus-independent cued task. Synaptophysin expression was increased by estrone (at a middle and high dose) and 17beta-estradiol (at a middle dose) in the CA3 region of the hippocampus and was not correlated with cognition. The results of this study indicate that estradiol can positively or negatively influence hippocampus-dependent learning and memory, whereas estrone impairs hippocampus-dependent learning and memory in a dose-dependent manner. These results have important therapeutic implications, as estrone, a main component of a widely used hormone replacement therapy, was shown to have either a negative effect or no effect on learning and memory. It may be possible to use 17alpha-estradiol and lower doses of estrogens as potential alternatives in hormone replacement therapies.
Collapse
Affiliation(s)
- Cindy K Barha
- Department of Psychology, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Gemma L Dalton
- Department of Psychology, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada,Department of Psychiatry, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Liisa AM Galea
- Department of Psychology, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada,Program in Neuroscience, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada,Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, Canada V6T 1Z4, Tel.: +1 604 822 6536, Fax: +1 604 822 3697, E-mail:
| |
Collapse
|
37
|
Opposite effects of early maternal deprivation on neurogenesis in male versus female rats. PLoS One 2009; 4:e3675. [PMID: 19180242 PMCID: PMC2629844 DOI: 10.1371/journal.pone.0003675] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 10/14/2008] [Indexed: 12/16/2022] Open
Abstract
Background Major depression is more prevalent in women than in men. The underlying neurobiological mechanisms are not well understood, but recent data shows that hippocampal volume reductions in depressed women occur only when depression is preceded by an early life stressor. This underlines the potential importance of early life stress, at least in women, for the vulnerability to develop depression. Perinatal stress exposure in rodents affects critical periods of brain development that persistently alter structural, emotional and neuroendocrine parameters in adult offspring. Moreover, stress inhibits adult hippocampal neurogenesis, a form of structural plasticity that has been implicated a.o. in antidepressant action and is highly abundant early postnatally. We here tested the hypothesis that early life stress differentially affects hippocampal structural plasticity in female versus male offspring. Principal Findings We show that 24 h of maternal deprivation (MD) at PND3 affects hippocampal structural plasticity at PND21 in a sex-dependent manner. Neurogenesis was significantly increased in male but decreased in female offspring after MD. Since no other structural changes were found in granule cell layer volume, newborn cell survival or proliferation rate, astrocyte number or gliogenesis, this indicates that MD elicits specific changes in subsets of differentiating cells and differentially affects immature neurons. The MD induced sex-specific effects on neurogenesis cannot be explained by differences in maternal care. Conclusions Our data shows that early environment has a critical influence on establishing sex differences in neural plasticity and supports the concept that the setpoint for neurogenesis may be determined during perinatal life. It is tempting to speculate that a reduced level of neurogenesis, secondary to early stress exposure, may contribute to maladaptation of the HPA axis and possibly to the increased vulnerability of women to stress-related disorders.
Collapse
|
38
|
Frick KM. Estrogens and age-related memory decline in rodents: what have we learned and where do we go from here? Horm Behav 2009; 55:2-23. [PMID: 18835561 PMCID: PMC2664384 DOI: 10.1016/j.yhbeh.2008.08.015] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 08/28/2008] [Accepted: 08/29/2008] [Indexed: 01/20/2023]
Abstract
The question of whether ovarian hormone therapy can prevent or reduce age-related memory decline in menopausal women has been the subject of much recent debate. Although numerous studies have demonstrated a beneficial effect of estrogen and/or progestin therapy for certain types of memory in menopausal women, recent clinical trials suggest that such therapy actually increases the risk of cognitive decline and dementia. Because rodent models have been frequently used to examine the effects of age and/or ovarian hormone deficiency on mnemonic function, rodent models of age-related hormone and memory decline may be useful in helping to resolve this issue. This review will focus on evidence suggesting that estradiol modulates memory, particularly hippocampal-dependent memory, in young and aging female rats and mice. Various factors affecting the mnemonic response to estradiol in aging females will be highlighted to illustrate the complications inherent to studies of estrogen therapy in aging females. Avenues for future development of estradiol-based therapies will also be discussed, and it is argued that an approach to drug development based on identifying the molecular mechanisms underlying estrogenic modulation of memory may lead to promising future treatments for reducing age-related mnemonic decline.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
39
|
Paris JJ, Frye CA. Estrous cycle, pregnancy, and parity enhance performance of rats in object recognition or object placement tasks. Reproduction 2008; 136:105-15. [PMID: 18390689 DOI: 10.1530/rep-07-0512] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ovarian hormone elevations are associated with enhanced learning/memory. During behavioral estrus or pregnancy, progestins, such as progesterone (P(4)) and its metabolite 5 alpha-pregnan-3 alpha-ol-20-one (3 alpha,5 alpha-THP), are elevated due, in part, to corpora luteal and placental secretion. During 'pseudopregnancy', the induction of corpora luteal functioning results in a hormonal milieu analogous to pregnancy, which ceases after about 12 days, due to the lack of placental formation. Multiparity is also associated with enhanced learning/memory, perhaps due to prior steroid exposure during pregnancy. Given evidence that progestins and/or parity may influence cognition, we investigated how natural alterations in the progestin milieu influence cognitive performance. In Experiment 1, virgin rats (nulliparous) or rats with two prior pregnancies (multiparous) were assessed on the object placement and recognition tasks, when in high-estrogen/P(4) (behavioral estrus) or low-estrogen/P(4) (diestrus) phases of the estrous cycle. In Experiment 2, primiparous or multiparous rats were tested in the object placement and recognition tasks when not pregnant, pseudopregnant, or pregnant (between gestational days (GDs) 6 and 12). In Experiment 3, pregnant primiparous or multiparous rats were assessed daily in the object placement or recognition tasks. Females in natural states associated with higher endogenous progestins (behavioral estrus, pregnancy, multiparity) outperformed rats in low progestin states (diestrus, non-pregnancy, nulliparity) on the object placement and recognition tasks. In earlier pregnancy, multiparous, compared with primiparous, rats had a lower corticosterone, but higher estrogen levels, concomitant with better object placement performance. From GD 13 until post partum, primiparous rats had higher 3 alpha,5 alpha-THP levels and improved object placement performance compared with multiparous rats.
Collapse
Affiliation(s)
- Jason J Paris
- The University at Albany, SUNY, 1400 Washington Avenue, Albany, New York 12222, USA
| | | |
Collapse
|
40
|
Walf AA, Koonce CJ, Frye CA. Estradiol or diarylpropionitrile administration to wild type, but not estrogen receptor beta knockout, mice enhances performance in the object recognition and object placement tasks. Neurobiol Learn Mem 2008; 89:513-21. [PMID: 18313947 DOI: 10.1016/j.nlm.2008.01.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 01/17/2008] [Accepted: 01/17/2008] [Indexed: 11/18/2022]
Abstract
Cognitive processes mediated by the hippocampus and cortex are influenced by estradiol (E(2)); however, the mechanisms by which E(2) has these effects are not entirely clear. As such, studies were conducted to begin to address the role of actions at the beta form of the intracellular estrogen receptor (ERbeta) for E(2)'s cognitive effects in adult female mice. We investigated whether E(2) improved performance of wild type (WT) and ERbeta knockout (betaERKO) mice in tasks considered to be mediated by the cortex and hippocampus, the object recognition and object placement tasks. WT and betaERKO mice were ovariectomized (ovx) and E(2) (0.1 mg/kg), an ERbeta selective ER modulator (SERM), diarylpropionitrile (DPN; 0.1 mg/kg), or oil vehicle was administered to mice following training in these tasks. We hypothesized that if E(2) has mnemonic effects, in part, due to its actions at ERbeta, then WT mice administered E(2) or DPN would have improved performance compared to vehicle WT controls, which would not be different from betaERKO mice administered vehicle, E(2) or DPN. Alternatively, activation of ERalpha (with E(2), which is a ligand for both ERalpha and ERbeta) may produce opposing effects on cognition and/or the activation of ERalpha and ERbeta vs. either receptor isoform alone may produce a different pattern of effects. Results obtained supported the hypothesis that ERbeta activation is important for mnemonic effects. Ovx WT, but not betaERKO, mice administered E(2) or DPN had a greater percentage of time exploring a novel object in the object recognition task and a displaced object in the object placement task. Thus, actions at ERbeta may be important for E(2) or SERMs to enhance cognitive performance of female mice in the object recognition and placement tasks.
Collapse
Affiliation(s)
- Alicia A Walf
- Department of Psychology, The University at Albany, State University of New York, Albany, NY 12222, USA
| | | | | |
Collapse
|
41
|
Aenlle KK, Kumar A, Cui L, Jackson TC, Foster TC. Estrogen effects on cognition and hippocampal transcription in middle-aged mice. Neurobiol Aging 2007; 30:932-45. [PMID: 17950954 DOI: 10.1016/j.neurobiolaging.2007.09.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/06/2007] [Accepted: 09/12/2007] [Indexed: 10/22/2022]
Abstract
Young and middle-aged female mice were ovariectomized and given cyclic injections of either estradiol or vehicle treatments. During the fifth week after surgery the Morris water maze was used to assess cognitive function. Age and treatment effects emerged over the course of spatial training such that middle-aged vehicle treated mice exhibited deficits in acquiring a spatial search strategy compared to younger vehicle treated mice and middle-age estradiol treated mice. Following behavioral characterization, mice were maintained on their injection schedule until week seven and hippocampi were collected 24h after the last injection. Hippocampal RNA was extracted and genes responsive to age and estrogen were identified using cDNA microarrays. Estradiol treatment in middle-aged mice altered the expression of genes related to transcriptional regulation, biosynthesis, growth, neuroprotection, and elements of cell signaling pathways. Expression profiles for representative genes were confirmed in a separate set of animals using oligonucleotide arrays and RT-PCR. Our results indicate that estrogen treatment in middle-aged animals may promote hippocampal health during the aging process.
Collapse
Affiliation(s)
- Kristina K Aenlle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, P.O. Box 100244, Gainesville, FL 32610-0244, USA
| | | | | | | | | |
Collapse
|
42
|
Schumacher M, Guennoun R, Ghoumari A, Massaad C, Robert F, El-Etr M, Akwa Y, Rajkowski K, Baulieu EE. Novel perspectives for progesterone in hormone replacement therapy, with special reference to the nervous system. Endocr Rev 2007; 28:387-439. [PMID: 17431228 DOI: 10.1210/er.2006-0050] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The utility and safety of postmenopausal hormone replacement therapy has recently been put into question by large clinical trials. Their outcome has been extensively commented upon, but discussions have mainly been limited to the effects of estrogens. In fact, progestagens are generally only considered with respect to their usefulness in preventing estrogen stimulation of uterine hyperplasia and malignancy. In addition, various risks have been attributed to progestagens and their omission from hormone replacement therapy has been considered, but this may underestimate their potential benefits and therapeutic promises. A major reason for the controversial reputation of progestagens is that they are generally considered as a single class. Moreover, the term progesterone is often used as a generic one for the different types of both natural and synthetic progestagens. This is not appropriate because natural progesterone has properties very distinct from the synthetic progestins. Within the nervous system, the neuroprotective and promyelinating effects of progesterone are promising, not only for preventing but also for reversing age-dependent changes and dysfunctions. There is indeed strong evidence that the aging nervous system remains at least to some extent sensitive to these beneficial effects of progesterone. The actions of progesterone in peripheral target tissues including breast, blood vessels, and bones are less well understood, but there is evidence for the beneficial effects of progesterone. The variety of signaling mechanisms of progesterone offers exciting possibilities for the development of more selective, efficient, and safe progestagens. The recognition that progesterone is synthesized by neurons and glial cells requires a reevaluation of hormonal aging.
Collapse
Affiliation(s)
- Michael Schumacher
- INSERM UMR 788, 80, rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gresack JE, Kerr KM, Frick KM. Short-term environmental enrichment decreases the mnemonic response to estrogen in young, but not aged, female mice. Brain Res 2007; 1160:91-101. [PMID: 17572392 DOI: 10.1016/j.brainres.2007.05.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 05/18/2007] [Accepted: 05/19/2007] [Indexed: 12/29/2022]
Abstract
The present study was designed to examine if 4 weeks of exposure to an enriched housing environment affects the ability of estrogen to facilitate object recognition in young and aged female mice. Object recognition was tested using a novel object recognition task. Ovariectomized young and aged female mice were maintained in standard or enriched housing for 4 weeks prior to and then throughout object recognition testing. Immediately after training, mice were injected intraperitoneally with vehicle or 0.2 mg/kg 17 beta-estradiol and then were re-tested 24 and 48 h later. Among young females, estradiol alone improved object recognition at both delays relative to chance, an effect not present in enriched females treated with estradiol. Enrichment alone had no significant effect on object recognition in young females at either delay. In contrast, enrichment alone in aged females significantly enhanced both 24- and 48-h object recognition relative to chance, an effect not present in mice treated with both enrichment and estradiol. Estradiol alone had no effect on object recognition in aged females at either delay. Together, these data indicate that estradiol and enrichment alone differentially affect object recognition in young and aged females. However, the fact that the combination of estradiol and enrichment treatments did not affect object recognition at either age suggests that co-administration of both treatments is less effective than the most effective single treatment at each age.
Collapse
Affiliation(s)
- Jodi E Gresack
- Department of Psychology, Yale University, New Haven, CT 06520, USA
| | | | | |
Collapse
|
44
|
Chiba S, Suzuki M, Yamanouchi K, Nishihara M. Involvement of Granulin in Estrogen-Induced Neurogenesis in the Adult Rat Hippocampus. J Reprod Dev 2007; 53:297-307. [PMID: 17179653 DOI: 10.1262/jrd.18108] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies have demonstrated the presence of neurogenesis in the adult mammalian hippocampus, and it has been suggested that estrogen and various growth factors influence the processes of adult neurogenesis. The present study assessed cell proliferation in the dentate gyrus and the mRNA expression levels of granulin, insulin-like growth factor-I (IGF-I), and brain-derived neurotrophic factor (BDNF) in the hippocampus 4 h after treatment with estradiol benzoate (EB) in 3- and 12-month old ovariectomized rats. At 3 months of age, mRNA expression of granulin precursor and cell proliferation were increased by EB treatment, although the mRNA expressions of IGF-I and BDNF remained unchanged. At 12 months of age, however, neither mRNA expression of the three genes nor cell proliferation in the dentate gyrus were affected by EB treatment. In addition, 17beta-estradiol enhanced the proliferation of neural progenitor cells derived from hippocampal tissue of 3-month-old female rats in vitro; this was inhibited by neutralization of granulin with specific antibody. These results suggest that estrogen induces granulin gene expression in the hippocampus and that the product of this gene is involved in the mitogenic effects of estrogen in the dentate gyrus, although the responses to estrogen decline with age.
Collapse
Affiliation(s)
- Shuichi Chiba
- Department of Veterinary Physiology, Veterinary Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
45
|
Abstract
In the late 1980s, the finding that the dentate gyrus contains more granule cells in the male than in the female of certain mouse strains provided the first indication that the dentate gyrus is a significant target for the effects of sex steroids during development. Gonadal hormones also play a crucial role in shaping the function and morphology of the adult brain. Besides reproduction-related processes, sex steroids participate in higher brain operations such as cognition and mood, in which the hippocampus is a critical mediator. Being part of the hippocampal formation, the dentate gyrus is naturally involved in these mechanisms and as such, this structure is also a critical target for the activational effects of sex steroids. These activational effects are the results of three major types of steroid-mediated actions. Sex steroids modulate the function of dentate neurons under normal conditions. In addition, recent research suggests that hormone-induced cellular plasticity may play a larger role than previously thought, particularly in the dentate gyrus. Specifically, the regulation of dentate gyrus neurogenesis and synaptic remodeling by sex steroids received increasing attention lately. Finally, the dentate gyrus is influenced by gonadal hormones in the context of cellular injury, and the work in this area demonstrates that gonadal hormones have neuroprotective potential. The expression of estrogen, progestin, and androgen receptors in the dentate gyrus suggests that sex steroids, which could be of gonadal origin and/or synthesized locally in the dentate gyrus, may act directly on dentate cells. In addition, gonadal hormones could also influence the dentate gyrus indirectly, by subcortical hormone-sensitive structures such as the cholinergic septohippocampal system. Importantly, these three sex steroid-related themes, functional effects in the normal dentate gyrus, mechanisms involving neurogenesis and synaptic remodeling, as well as neuroprotection, have substantial implications for understanding normal cognitive function, with clinical importance for epilepsy, Alzheimer's disease and mental disorders.
Collapse
Affiliation(s)
- Tibor Hajszan
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Biophysics, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - Teresa A Milner
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Medical College of Cornell University, New York, NY, USA
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Csaba Leranth
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
46
|
Farr TD, Carswell HVO, Gallagher L, Condon B, Fagan AJ, Mullin J, Macrae IM. 17β-Estradiol treatment following permanent focal ischemia does not influence recovery of sensorimotor function. Neurobiol Dis 2006; 23:552-62. [PMID: 16759876 DOI: 10.1016/j.nbd.2006.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 04/07/2006] [Accepted: 04/24/2006] [Indexed: 11/23/2022] Open
Abstract
The development of therapy to aid poststroke recovery is essential. The female hormone 17beta-estradiol has been shown to promote synaptogenesis; the purpose of this study was to attempt to harness these mechanisms to promote repair and recovery in the peri-infarct zone. Rats were ovariectomized, tested for sensorimotor function, and the middle cerebral artery permanently occluded (MCAO). Infarct volumes were calculated using MRI, and damage was equivalent in all animals prior to implantation of either 17beta-estradiol or placebo pellets. Animals were tested for functional recovery for 28 days and tissue processed for synaptic marker syntaxin immunohistochemistry. The stroke induced a significant behavioral deficit, which persisted out to 28 days, and was not significantly different between 17beta-estradiol and placebo treatment groups. There was no difference in syntaxin immunostaining between groups in either the peri-infarct cortex or in the dendritic CA1 reference region. In conclusion, 17beta-estradiol treatment, delivered poststroke, did not influence recovery of function or synaptogenesis.
Collapse
Affiliation(s)
- Tracy D Farr
- 7TMRI Facility and Wellcome Surgical Institute, Division of Clinical Neuroscience, University of Glasgow, Garscube Estate, Bearsden Road, Glasgow, Scotland G61 1QH, UK.
| | | | | | | | | | | | | |
Collapse
|
47
|
Chaovipoch P, Jelks KAB, Gerhold LM, West EJ, Chongthammakun S, Floyd CL. 17beta-estradiol is protective in spinal cord injury in post- and pre-menopausal rats. J Neurotrauma 2006; 23:830-52. [PMID: 16774470 DOI: 10.1089/neu.2006.23.830] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The neuroprotective effects of 17 beta -estradiol have been shown in models of central nervous system injury, including ischemia, brain injury, and more recently, spinal cord injury (SCI). Recent epidemiological trends suggest that SCIs in elderly women are increasing; however, the effects of menopause on estrogen-mediated neuroprotection are poorly understood. The objective of this study was to evaluate the effects of 17beta-estradiol and reproductive aging on motor function, neuronal death, and white matter sparing after SCI of post- and pre-menopausal rats. Two-month-old or 1- year-old female rats were ovariectomized and implanted with a silastic capsule containing 180 microg/mL of 17beta-estradiol or vehicle. Complete crush SCI at T8-9 was performed 1 week later. Additional animals of each age group were left ovary-intact but were spinal cord injured. The Basso, Beattie, Bresnahan (BBB) locomotor test was performed. Spinal cords were collected on post-SCI days 1, 7, and 21, and processed for histological markers. Administration of 17beta-estradiol to ovariectomized rats improved recovery of hind-limb locomotion, increased white matter sparing, and decreased apoptosis in both the post- and pre-menopausal rats. Also, ovary-intact 1-year-old rats did worse than ovary-intact 2-month-old rats, suggesting that endogenous estrogen confers neuroprotection in young rats, which is lost in older animals. Taken together, these data suggest that estrogen is neuroprotective in SCI and that the loss of endogenous estrogen-mediated neuroprotective seen in older rats can be attenuated with exogenous administration of 17beta-estradiol.
Collapse
Affiliation(s)
- Pimonporn Chaovipoch
- Department of Neurological Surgery, University of California, Davis, California 95616, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Gresack JE, Frick KM. Effects of continuous and intermittent estrogen treatments on memory in aging female mice. Brain Res 2006; 1115:135-47. [PMID: 16920082 DOI: 10.1016/j.brainres.2006.07.067] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 07/20/2006] [Accepted: 07/21/2006] [Indexed: 10/24/2022]
Abstract
The manner in which hormone therapy is given to postmenopausal women may significantly influence its ability to reduce age-associated memory loss. To test the hypothesis that a regimen that approximates the timing of estrogen surges in the natural cycle is more beneficial for memory than a regimen that provides continuous levels of estrogen, we examined the effects of continuous and intermittent estrogen regimens on spatial and object memory in aging female mice. Mice (18 months) were treated with 0.2 mg/kg 17beta-estradiol (E(2)) or vehicle (VEH) for 3 months following ovariectomy. A fast-acting water-soluble cyclodextrin-encapsulated E(2) was used to ensure metabolism within 24 h. Vehicle-treated mice received daily injections of 2-hydroxypropyl-beta-cyclodextrin vehicle. The continuous estradiol group (Contin E(2)) was injected daily with estradiol. The intermittent group (Twice/wk E(2)) received estradiol every 4 days and vehicle on all other days. Mice (21 months) were tested in water-escape motivated 8-arm radial arm maze (WRAM) and object recognition tasks. During WRAM acquisition, the Twice/wk E(2) group committed significantly more reference memory errors than VEH and Contin E(2) groups, and tended to make more working memory errors than the VEH group. The Contin E(2) group did not differ from VEH on either WRAM measure. Additionally, the Twice/wk E(2) group tended to exhibit impaired object recognition. Thus, neither treatment improved spatial or object memory. Indeed, intermittent estradiol was detrimental to both types of memory. These results suggest that the timing of administration may play an important role in the mnemonic response of aging females to estrogen.
Collapse
Affiliation(s)
- Jodi E Gresack
- Department of Psychology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
49
|
Harburger LL, Bennett JC, Frick KM. Effects of estrogen and progesterone on spatial memory consolidation in aged females. Neurobiol Aging 2006; 28:602-10. [PMID: 16621169 DOI: 10.1016/j.neurobiolaging.2006.02.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 02/03/2006] [Accepted: 02/27/2006] [Indexed: 11/21/2022]
Abstract
Interpretation of data illustrating that estrogen, with or without progestin, is detrimental to memory in post-menopausal women is complicated by the fact that little is known about the effects of progestins on memory. The present study examined if estrogen, alone or with progesterone, affects spatial memory consolidation in ovariectomized aged female mice. Mice received eight training trials in a spatial Morris water maze followed immediately by injection of water-soluble 17beta-estradiol (E(2); 0.2 mg/kg) or vehicle. Mice were re-tested 24 h later. All mice learned to find the platform on Day 1. On Day 2, the performance of control, but not E(2) mice, deteriorated, suggesting that E(2) enhanced memory for the platform location. In a second experiment, mice were injected with E(2) and 10 or 20 mg/kg water-soluble progesterone. The 10 mg/kg dose of progesterone did not affect estrogen's ability to enhance spatial memory consolidation, but 20 mg/kg blocked this effect. These data indicate that estrogen can improve spatial memory consolidation in aged females and that this effect can be attenuated by progesterone.
Collapse
|
50
|
Gulinello M, Lebesgue D, Jover-Mengual T, Zukin RS, Etgen AM. Acute and chronic estradiol treatments reduce memory deficits induced by transient global ischemia in female rats. Horm Behav 2006; 49:246-60. [PMID: 16125703 PMCID: PMC4169120 DOI: 10.1016/j.yhbeh.2005.07.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 07/01/2005] [Accepted: 07/15/2005] [Indexed: 11/28/2022]
Abstract
Transient global ischemia induces selective, delayed neuronal death in the hippocampal CA1 and delayed cognitive deficits. Estrogen treatment ameliorates hippocampal injury associated with global ischemia. Although much is known about the impact of estrogen on neuronal survival, relatively little is known about its impact on functional outcome assessed behaviorally. We investigated whether long-term estradiol (21-day pellets implanted 14 days prior to ischemia) or acute estradiol (50 microg infused into the lateral ventricles immediately after ischemia) attenuates ischemia-induced cell loss and improves visual and spatial working memory in ovariectomized female rats. Global ischemia significantly impaired visual and spatial memory, assessed by object recognition and object placement tests at 6-9 days. Global ischemia did not affect locomotion, exploration, or anxiety-related behaviors, assessed by an open-field test at 6 days. Long-term estradiol prevented the ischemia-induced deficit in visual working memory, maintaining normal performance in tests with retention intervals of up to 1 h. Long-term estradiol also prevented ischemia-induced deficits in spatial memory tests with short (1 and 7 min), but not longer (15 min), retention intervals. Acute estradiol significantly improved visual memory assessed with short retention intervals, but did not prevent deficits in spatial memory. Acute estradiol significantly increased the number of surviving CA1 neurons, assessed either at 7 days after ischemia or after the completion of behavioral testing 9 days after ischemia. In contrast, chronic estradiol did not reduce CA1 cell death 9 days after ischemia. Thus, long-term estradiol at near physiological levels and acute estradiol administered after ischemic insult improve functional recovery after global ischemia. These findings have important implications for intervention in the neurological sequellae associated with global ischemia.
Collapse
Affiliation(s)
- Maria Gulinello
- Albert Einstein College of Medicine, Department of Neuroscience, 1300 Morris Park Avenue, Room F113, Bronx, NY 10461, USA.
| | | | | | | | | |
Collapse
|