1
|
Salinero AE, Abi-Ghanem C, Venkataganesh H, Sura A, Smith RM, Thrasher CA, Kelly RD, Hatcher KM, NyBlom V, Shamlian V, Kyaw NR, Belanger KM, Gannon OJ, Stephens SBZ, Zuloaga DG, Zuloaga KL. Treatment with brain specific estrogen prodrug ameliorates cognitive effects of surgical menopause in mice. Horm Behav 2024; 164:105594. [PMID: 38917776 PMCID: PMC11330726 DOI: 10.1016/j.yhbeh.2024.105594] [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: 01/16/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Menopause is an endocrine shift leading to increased vulnerability for cognitive impairment and dementia risk factors, in part due to loss of neuroprotective circulating estrogens. Systemic replacement of estrogen post-menopause has limitations, including risk for estrogen-sensitive cancers. A promising therapeutic approach therefore might be to deliver estrogen only to the brain. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in ovariectomized mice (a surgical menopause model). We treated mice with the prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17β-estradiol only in the brain. Young and middle-aged C57BL/6 J mice received ovariectomy and subcutaneous implant containing vehicle or DHED and underwent cognitive testing to assess memory after 1-3.5 months of treatment. Low and medium doses of DHED did not alter metabolic status in middle-aged mice. In both age groups, DHED treatment improved spatial memory in ovariectomized mice. Additional testing in middle-aged mice showed that DHED treatment improved working and recognition memory in ovariectomized mice. These results lay the foundation for future studies determining if this intervention is as efficacious in models of dementia with comorbid risk factors.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Christina A Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Katherine M Hatcher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Vanessa NyBlom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA; Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Victoria Shamlian
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Kasey M Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Shannon B Z Stephens
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA.
| |
Collapse
|
2
|
Balu D, Valencia-Olvera AC, Deshpande A, Narayanam S, Konasani S, Pattisapu S, York JM, Thatcher GRJ, LaDu MJ, Tai LM. Estradiol improves behavior in FAD transgenic mice that express APOE3 but not APOE4 after ovariectomy. Front Endocrinol (Lausanne) 2024; 15:1374825. [PMID: 38742194 PMCID: PMC11089251 DOI: 10.3389/fendo.2024.1374825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Increasing evidence suggests that female individuals have a higher Alzheimer's disease (AD) risk associated with post-menopausal loss of circulating estradiol (E2). However, clinical data are conflicting on whether E2 lowers AD risk. One potential contributing factor is APOE. The greatest genetic risk factor for AD is APOE4, a factor that is pronounced in female individuals post-menopause. Clinical data suggests that APOE impacts the response of AD patients to E2 replacement therapy. However, whether APOE4 prevents, is neutral, or promotes any positive effects of E2 is unclear. Therefore, our goal was to determine whether APOE modulates the impact of E2 on behavior and AD pathology in vivo. To that end, mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ42 were ovariectomized at either 4 months (early) or 8 months (late) and treated with vehicle or E2 for 4 months. In E3FAD mice, we found that E2 mitigated the detrimental effect of ovariectomy on memory, with no effect on Aβ in the early paradigm and only improved learning in the late paradigm. Although E2 lowered Aβ in E4FAD mice in the early paradigm, there was no impact on learning or memory, possibly due to higher Aβ pathology compared to E3FAD mice. In the late paradigm, there was no effect on learning/memory and Aβ pathology in E4FAD mice. Collectively, these data support the idea that, in the presence of Aβ pathology, APOE impacts the response to E2 supplementation post-menopause.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ashwini Deshpande
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Saharsh Narayanam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sravya Konasani
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Shreya Pattisapu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Jason M. York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
3
|
Wu CY, Hsieh HH, Huang SM, Chiu SC, Peng SL. Brain alterations in ovariohysterectomized rats revealed by diffusion tensor imaging. Neuroreport 2023; 34:649-654. [PMID: 37506310 DOI: 10.1097/wnr.0000000000001937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
OBJECTIVES Women undergoing hysterectomy with oophorectomy have an increased risk of Alzheimer's disease and Parkinson's disease. However, postoperative neuroimaging data on pathogenic processes in the brain are limited. The aim of this study was to investigate the potential effect of ovariohysterectomy on brain integrity in rat model using diffusion tensor imaging (DTI) technique for the first time. METHODS We enrolled 13 rats each in the control and ovariohysterectomy groups. Rats in the ovariohysterectomy group underwent the ovariohysterectomy at 7 weeks of age, and all rats underwent DTI scans at 9 weeks of age. The DTI-derived parameters, such as fractional anisotropy and mean diffusivity, were compared between the control and ovariohysterectomy groups. RESULTS Compared to the control group, the ovariohysterectomy group showed significantly lower fractional anisotropy in various brain regions, including the corpus callosum, bilateral striatum, and bilateral cortex (all P < 0.05), suggesting neuronal injury in ovariohysterectomized rats. Mean diffusivity did not differ significantly between groups (all P > 0.05). CONCLUSION Rats undergoing ovariohysterectomy had lower fractional anisotropy compared to control in widespread brain regions, suggesting neuronal injury and demyelination. Therefore, neuroimaging should be performed to monitor brain alterations in women after hysterectomy with bilateral oophorectomy in clinical settings.
Collapse
Affiliation(s)
- Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei
| | - Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei
| | - Sheng-Min Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science
- Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
4
|
Salinero AE, Abi-Ghanem C, Venkataganesh H, Sura A, Smith RM, Thrasher CA, Kelly RD, Hatcher KM, NyBlom V, Shamlian V, Kyaw NR, Belanger KM, Gannon OJ, Stephens SB, Zuloaga DG, Zuloaga KL. Brain Specific Estrogen Ameliorates Cognitive Effects of Surgical Menopause in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552687. [PMID: 37609180 PMCID: PMC10441397 DOI: 10.1101/2023.08.09.552687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Menopause is a major endocrinological shift that leads to an increased vulnerability to the risk factors for cognitive impairment and dementia. This is thought to be due to the loss of circulating estrogens, which exert many potent neuroprotective effects in the brain. Systemic replacement of estrogen post-menopause has many limitations, including increased risk for estrogen-sensitive cancers. A more promising therapeutic approach therefore might be to deliver estrogen only to the brain thus limiting adverse peripheral side effects. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in post-menopausal mice. We modeled surgical menopause via bilateral ovariectomy (OVX). We treated mice with the pro-drug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17β-estradiol only in the brain. Young (2.5-month) and middle-aged (11-month-old) female C57BL/6J mice received ovariectomy and a subcutaneous implant containing vehicle (cholesterol) or DHED. At 3.5 months old (young group) and 14.5 months old (middle-aged group), mice underwent behavior testing to assess memory. DHED did not significantly alter metabolic status in middle-aged, post-menopausal mice. In both young and middle-aged mice, the brain-specific estrogen DHED improved spatial memory. Additional testing in middle-aged mice also showed that DHED improved working and recognition memory. These promising results lay the foundation for future studies aimed at determining if this intervention is as efficacious in models of dementia that have comorbid risk factors.
Collapse
Affiliation(s)
- Abigail E. Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Rachel M. Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Christina A. Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Richard D. Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Katherine M. Hatcher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Vanessa NyBlom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Victoria Shamlian
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Kasey M. Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Olivia J. Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Shannon B.Z. Stephens
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| | - Damian G. Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Kristen L. Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue; MC-136, Albany, NY, USA
| |
Collapse
|
5
|
Bhandari A, Kalotra S, Bajaj P, Sunkaria A, Kaur G. Dietary intervention with Tinospora cordifolia improved aging-related decline in locomotor coordination and cerebellar cell survival and plasticity in female rats. Biogerontology 2022; 23:809-824. [PMID: 35767131 DOI: 10.1007/s10522-022-09975-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Reduced bone mineral density, and muscle strength are the hallmark of aging-related motor coordination deficits and related neuropathologies. Since cerebellum regulates motor movements and balance perception of our body, therefore it may be an important target to control the age-related progression of motor dysfunctions. Dry stem powder of Tinospora cordifolia (TCP) was tested as a food supplement to elucidate its activity to attenuate age-associated locomotor dysfunctions. Intact acyclic middle-aged female rats were used in this study as the model system of the transition phase from premenopause to menopause in women along with cycling young adult rats. Normal chow or 30% High Fat Diet (HFD), supplemented with or without TCP was fed to animals for 12 weeks and then tested for locomotor performance on rotarod followed by post-sacrifice protein expression studies. In comparison to young adults, middle-aged animals showed an increase in number of falls and lesser time spent in rotarod performance test, whereas, animals given TCP supplemented feed showed improvement in performance with more pronounced effects observed in normal chow than HFD fed middle-aged rats. Further, due to its multicomponent nature TCP was found to target the expression of various markers of neuroinflammation, apoptosis, cell survival, and synaptic plasticity in the cerebellum region. The current findings suggest that TCP supplementation in the diet may prove to be a potential interventional strategy for the management of frailty and fall-associated morbidities caused by aging-related deterioration of bone mineral density, and muscle strength.
Collapse
Affiliation(s)
- Anmol Bhandari
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India
| | - Shikha Kalotra
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India
| | - Payal Bajaj
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India
| | - Aditya Sunkaria
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India.
| | - Gurcharan Kaur
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India.
| |
Collapse
|
6
|
Bhandari A, Sunkaria A, Kaur G. Dietary Supplementation With Tinospora cordifolia Improves Anxiety-Type Behavior and Cognitive Impairments in Middle-Aged Acyclic Female Rats. Front Aging Neurosci 2022; 14:944144. [PMID: 35966795 PMCID: PMC9366175 DOI: 10.3389/fnagi.2022.944144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022] Open
Abstract
The midlife transition period in women marks the progressive flattening of neurological health along with increased adiposity, dyslipidemia, frailty, and inflammatory responses mainly attributed to the gradual decline in estrogen levels. Conflicting reports of hormone replacement therapy (HRT) necessitate the exploration of novel therapeutic interventions using bioactive natural products having the least toxicity and a holistic mode of action for the preservation of metabolic homeodynamics with aging in women. The present study was planned to investigate the effects of aging and/or a high-fat diet (HFD) on cognitive impairments and anxiety and further their management by dietary supplement with the Tinospora cordifolia stem powder (TCP). Acyclic female rats were included in this study as the model system of the perimenopause phase of women along with young 3-4 months old rats as controls. Rats were fed on with and without TCP supplemented normal chow or HFD for 12 weeks. Animals fed on a TCP supplemented normal chow showed consistent management of body weight over a 12-week regimen although their calorie intake was much higher in comparison to their age-matched controls. Post-regimen, neurobehavioral tests, such as novel object recognition and elevated plus maze, performed on these animals showed improvement in their learning and memory abilities as well as the anxiety-like behavior. Furthermore, due to the presence of multiple components, TCP was observed to modulate the expression of key marker proteins to ameliorate neuroinflammation and apoptosis and promote cell survival and synaptic plasticity in the hippocampus and the prefrontal cortex (PFC) regions of the brain. These findings suggest that TCP supplementation in diet during the midlife transition period in women may be a potential interventional strategy for the management of menopause-associated anxiety and cognitive impairments and healthy aging.
Collapse
Affiliation(s)
| | | | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
7
|
Fisher VL, Ortiz LS, Powers AR. A computational lens on menopause-associated psychosis. Front Psychiatry 2022; 13:906796. [PMID: 35990063 PMCID: PMC9381820 DOI: 10.3389/fpsyt.2022.906796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
Psychotic episodes are debilitating disease states that can cause extreme distress and impair functioning. There are sex differences that drive the onset of these episodes. One difference is that, in addition to a risk period in adolescence and early adulthood, women approaching the menopause transition experience a second period of risk for new-onset psychosis. One leading hypothesis explaining this menopause-associated psychosis (MAP) is that estrogen decline in menopause removes a protective factor against processes that contribute to psychotic symptoms. However, the neural mechanisms connecting estrogen decline to these symptoms are still not well understood. Using the tools of computational psychiatry, links have been proposed between symptom presentation and potential algorithmic and biological correlates. These models connect changes in signaling with symptom formation by evaluating changes in information processing that are not easily observable (latent states). In this manuscript, we contextualize the observed effects of estrogen (decline) on neural pathways implicated in psychosis. We then propose how estrogen could drive changes in latent states giving rise to cognitive and psychotic symptoms associated with psychosis. Using computational frameworks to inform research in MAP may provide a systematic method for identifying patient-specific pathways driving symptoms and simultaneously refine models describing the pathogenesis of psychosis across all age groups.
Collapse
Affiliation(s)
- Victoria L Fisher
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| | - Liara S Ortiz
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| | - Albert R Powers
- Yale University School of Medicine and the Connecticut Mental Health Center, New Haven, CT, United States
| |
Collapse
|
8
|
Hidalgo-Lopez E, Zeidman P, Harris T, Razi A, Pletzer B. Spectral dynamic causal modelling in healthy women reveals brain connectivity changes along the menstrual cycle. Commun Biol 2021; 4:954. [PMID: 34376799 PMCID: PMC8355156 DOI: 10.1038/s42003-021-02447-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 07/01/2021] [Indexed: 01/01/2023] Open
Abstract
Longitudinal menstrual cycle studies allow to investigate the effects of ovarian hormones on brain organization. Here, we use spectral dynamic causal modelling (spDCM) in a triple network model to assess effective connectivity changes along the menstrual cycle within and between the default mode, salience and executive control networks (DMN, SN, and ECN). Sixty healthy young women were scanned three times along their menstrual cycle, during early follicular, pre-ovulatory and mid-luteal phase. Related to estradiol, right before ovulation the left insula recruits the ECN, while the right middle frontal gyrus decreases its connectivity to the precuneus and the DMN decouples into anterior/posterior parts. Related to progesterone during the mid-luteal phase, the insulae (SN) engage to each other, while decreasing their connectivity to parietal ECN, which in turn engages the posterior DMN. When including the most confident connections in a leave-one out cross-validation, we find an above-chance prediction of the left-out subjects' cycle phase. These findings corroborate the plasticity of the female brain in response to acute hormone fluctuations and may help to further understand the neuroendocrine interactions underlying cognitive changes along the menstrual cycle.
Collapse
Affiliation(s)
- Esmeralda Hidalgo-Lopez
- Department of Psychology and Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria.
| | - Peter Zeidman
- The Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - TiAnni Harris
- Department of Psychology and Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| | - Adeel Razi
- The Wellcome Centre for Human Neuroimaging, University College London, London, UK
- Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Belinda Pletzer
- Department of Psychology and Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria.
| |
Collapse
|
9
|
Blümel JE, Arteaga E, Vallejo MS, Ojeda E, Meza P, Martino M, Rodríguez-Vidal D, Ñañez M, Tserotas K, Rojas J, Rodrígues MA, Espinoza MT, Salinas C, Párraga-Párraga J, Chedraui P. Association of bilateral oophorectomy and menopause hormone therapy with mild cognitive impairment: the REDLINC X study. Climacteric 2021; 25:195-202. [PMID: 34323137 DOI: 10.1080/13697137.2021.1951203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Dementia is a major public health problem. Estrogen is a regulator of the central nervous system and its deficit could be involved in cognitive decline in older women. OBJECTIVE This study aimed to evaluate the association of bilateral oophorectomy, menopause hormone therapy (MHT) and other factors on mild cognitive impairment (MCI). METHOD The case-control study included 941 otherwise healthy postmenopausal women aged 60 years and over from six Latin American countries. Personal and family data were recorded and MCI was assessed using the Montreal Cognitive Assessment test (MoCA). RESULTS Average age, years of education and body mass index were 66.1 ± 5.8 years, 12.4 ± 5.0 years and 26.0 ± 4.3 kg/m2, respectively. A total of 30.2% had undergone bilateral oophorectomy and 40.3% had used MHT. A total of 232 women (24.7%) had MCI. The prevalence of MCI was higher in women with intact ovaries and non-MHT users as compared to MHT users (29.3% vs. 11.7% [odds ratio (OR) 0.32; 95% confidence interval (CI) 0.20-0.51]). Among oophorectomized women, MCI prevalence was higher among non-MHT users as compared to MHT users (45.2% vs. 12.8% [OR 0.18; 95% CI 0.10-0.32]). Logistic regression analysis determined that the variables associated with MCI were age >65 years (OR 1.69; 95% CI 1.20-2.38), parity (having >2 children; OR 1.69; 95% CI 1.21-2.37), bilateral oophorectomy (OR 1.56; 95% CI 1.09-2.24), hypertension (OR 1.41; 95% CI 1.01-1.96), being sexually active (OR 0.56; 95% CI 0.40-0.79), education >12 years (OR 0.46; 95% CI 0.32-0.65) and MHT use (OR 0.31; 95% CI 0.21-0.46). CONCLUSION Age, parity, bilateral oophorectomy and hypertension are independent factors associated with MCI; contrary to this, higher educational level, maintaining sexual activity and using MHT are protective factors.
Collapse
Affiliation(s)
- J E Blümel
- Departamento de Medicina Interna Sur, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - E Arteaga
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - M S Vallejo
- Clínica Quilín, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - E Ojeda
- Departamento de Obstetricia y Ginecología, Universidad Andina, Cusco, Perú
| | - P Meza
- Departamento de Medicina Familiar, Universidad San Martin de Porres, Lima, Perú
| | - M Martino
- Departamento de Obstetricia y Ginecología, Universidad Nacional de Rosario, Rosario, Argentina
| | - D Rodríguez-Vidal
- Departamento de Obstetricia y Ginecología, Hospital de Clínicas José de San Martin, Buenos Aires, Argentina
| | - M Ñañez
- Cátedra de Ginecología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - K Tserotas
- Complejo Hospitalario Dr. Arnulfo Arias Madrid, Caja del Seguro Social, Panamá, Panamá
| | - J Rojas
- Departamento de Ginecología Obstetricia, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - M A Rodrígues
- Department of Gynecology and Obstetrics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - M T Espinoza
- Unidad de Climaterio, Clínica Los Ángeles, Cochabamba, Bolivia
| | - C Salinas
- Servicio de Obstetricia y Ginecología, Hospital Ángeles Puebla, Puebla, México
| | - J Párraga-Párraga
- Instituto de Investigación e Innovación en Salud Integral, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador
| | - P Chedraui
- Instituto de Investigación e Innovación en Salud Integral, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador.,Facultad de Ciencias de la Salud, Universidad Católica 'Nuestra Señora de la Asunción', Asunción, Paraguay
| |
Collapse
|
10
|
Kara F, Belloy ME, Voncken R, Sarwari Z, Garima Y, Anckaerts C, Langbeen A, Leysen V, Shah D, Jacobs J, Hamaide J, Bols P, Van Audekerke J, Daans J, Guglielmetti C, Kantarci K, Prevot V, Roßner S, Ponsaerts P, Van der Linden A, Verhoye M. Long-term ovarian hormone deprivation alters functional connectivity, brain neurochemical profile and white matter integrity in the Tg2576 amyloid mouse model of Alzheimer's disease. Neurobiol Aging 2021; 102:139-150. [PMID: 33765427 PMCID: PMC8312737 DOI: 10.1016/j.neurobiolaging.2021.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal bilateral ovariectomy is considered to be one of the risk factors of Alzheimer's disease (AD). However, the underlying mechanisms remain unclear. Here, we aimed to investigate long-term neurological consequences of ovariectomy in a rodent AD model, TG2576 (TG), and wild-type mice (WT) that underwent an ovariectomy or sham-operation, using in vivo MRI biomarkers. An increase in osmoregulation and energy metabolism biomarkers in the hypothalamus, a decrease in white matter integrity, and a decrease in the resting-state functional connectivity was observed in ovariectomized TG mice compared to sham-operated TG mice. In addition, we observed an increase in functional connectivity in ovariectomized WT mice compared to sham-operated WT mice. Furthermore, genotype (TG vs. WT) effects on imaging markers and GFAP immunoreactivity levels were observed, but there was no effect of interaction (Genotype × Surgery) on amyloid-beta-and GFAP immunoreactivity levels. Taken together, our results indicated that both genotype and ovariectomy alters imaging biomarkers associated with AD.
Collapse
Affiliation(s)
- Firat Kara
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Michael E Belloy
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Rick Voncken
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Zahra Sarwari
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yadav Garima
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cynthia Anckaerts
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - An Langbeen
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences and Cognition, UMR-S1172, DistalZ, Lille, France
| | - Disha Shah
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jules Jacobs
- University of Nijmegen, Nijmegen, the Netherlands
| | - Julie Hamaide
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Bols
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Van Audekerke
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences and Cognition, UMR-S1172, DistalZ, Lille, France
| | - Steffen Roßner
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Annemie Van der Linden
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Prakapenka AV, Quihuis AM, Carson CG, Patel S, Bimonte-Nelson HA, Sirianni RW. Poly(lactic-co-glycolic Acid) Nanoparticle Encapsulated 17β-Estradiol Improves Spatial Memory and Increases Uterine Stimulation in Middle-Aged Ovariectomized Rats. Front Behav Neurosci 2021; 14:597690. [PMID: 33424559 PMCID: PMC7793758 DOI: 10.3389/fnbeh.2020.597690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/23/2020] [Indexed: 12/02/2022] Open
Abstract
Hormone therapy that contains 17β-estradiol (E2) is used commonly for treatment of symptoms associated with menopause. E2 treatment has been shown to improve cognitive function following the decrease in ovarian hormones that is characteristic of menopause. However, once in circulation, the majority of E2 is bound to serum hormone binding globulin or albumin, becoming biologically inactive. Thus, therapeutic efficacy of E2 stands to benefit from increased bioavailability via sustained release of the hormone. Here, we focus on the encapsulation of E2 within polymeric nanoparticles composed of poly(lactic-co-glycolic) acid (PLGA). PLGA agent encapsulation offers several delivery advantages, including improved bioavailability and sustained biological activity of encapsulated agents. We hypothesized that delivery of E2 from PLGA nanoparticles would enhance the beneficial cognitive effects of E2 relative to free E2 or non-hormone loaded nanoparticle controls in a rat model of menopause. To test this hypothesis, spatial learning and memory were assessed in middle-aged ovariectomized rats receiving weekly subcutaneous treatment of either oil-control, free (oil-solubilized) E2, blank (non-hormone loaded) PLGA, or E2-loaded PLGA. Unexpectedly, learning and memory differed significantly between the two vehicle control groups. E2-loaded PLGA nanoparticles improved learning and memory relative to its control, while learning and memory were not different between free E2 and its vehicle control. These results suggest that delivery of E2 from PLGA nanoparticles offered cognitive benefit. However, when evaluating peripheral burden, E2-loaded PLGA was found to increase uterine stimulation compared to free E2, which is an undesired outcome, as estrogen exposure increases uterine cancer risk. In sum, a weekly E2 treatment regimen of E2 from PLGA nanoparticles increased cognitive efficacy and was accompanied with an adverse impact on the periphery, effects that may be due to the improved agent bioavailability and sustained biological activity offered by PLGA nanoparticle encapsulation. These findings underscore the risk of non-specific enhancement of E2 delivery and provide a basic framework for the study and development of E2's efficacy as a cognitive therapeutic with the aid of customizable polymeric nano-carriers.
Collapse
Affiliation(s)
- Alesia V Prakapenka
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,School of Life Sciences, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Alicia M Quihuis
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Catherine G Carson
- 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
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Rachael W Sirianni
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Agca C, Klakotskaia D, Stopa EG, Schachtman TR, Agca Y. Ovariectomy Influences Cognition and Markers of Alzheimer's Disease. J Alzheimers Dis 2020; 73:529-541. [PMID: 31796679 DOI: 10.3233/jad-190935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the most devastating and costly diseases, and prevalence of AD increases with age. Furthermore, females are twice as likely to suffer from AD compared to males. The cessation of reproductive steroid hormone production during menopause is hypothesized to cause this difference. Two rodent AD models, APP21 and APP+PS1, and wild type (WT) rats underwent an ovariectomy or sham surgery. Changes in learning and memory, brain histology, amyloid-β (Aβ) deposition, levels of mRNAs involved in Aβ production and clearance, and synaptic and cognitive function were determined. Barnes maze results showed that regardless of ovariectomy status, APP+PS1 rats learned slower and had poor memory retention. Ovariectomy caused learning impairment only in the APP21 rats. High levels of Aβ42 and very low levels of Aβ40 were observed in the brain cortices of APP+PS1 rats indicating limited endogenous PS1. The APP+PS1 rats had 43-fold greater formic acid soluble Aβ42 than Aβ40 at 17 months. Furthermore, levels of formic acid soluble Aβ42 increased 57-fold in ovariectomized APP+PS1 rats between 12 and 17 months of age. The mRNA encoding Grin1 significantly decreased due to ovariectomy whereas levels of Bace1, Chat, and Prkcb all decreased with age. The expression levels of mRNAs involved in Aβ degradation and AβPP cleavage (Neprilysin, Ide, Adam9, and Psenen) were found to be highly correlated with each other as well as hippocampal Aβ deposition. Taken together, these results indicate that both ovariectomy and genotype influence AD markers in a complex manner.
Collapse
Affiliation(s)
- Cansu Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Edward G Stopa
- Departments of Pathology and Neurosurgery, Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
13
|
Koebele SV, Mennenga SE, Poisson ML, Hewitt LT, Patel S, Mayer LP, Dyer CA, Bimonte-Nelson HA. Characterizing the effects of tonic 17β-estradiol administration on spatial learning and memory in the follicle-deplete middle-aged female rat. Horm Behav 2020; 126:104854. [PMID: 32949557 PMCID: PMC8032560 DOI: 10.1016/j.yhbeh.2020.104854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 01/19/2023]
Abstract
17β-estradiol (E2)-containing hormone therapy is a safe, effective way to alleviate unwanted menopause symptoms. Preclinical research has focused upon the role of E2 in learning and memory using a surgically menopausal rodent model whereby the ovaries are removed. Given that most women retain their reproductive tract and undergo a natural menopause transition, it is necessary to understand how exogenous E2 impacts a structurally intact, but follicle-deplete, system. In the current study, 8 month old female rats were administered the ovatoxin 4-vinylcyclohexene diepoxide (VCD), which accelerates ovarian follicular depletion, to model the human menopause transition. After follicular depletion, at 11 months old, rats were administered Vehicle or tonic E2 treatment for 12 days prior to behavioral evaluation on spatial working and reference memory tasks. Results demonstrated that E2 had both enhancing and impairing effects on taxed working memory depending upon the learning or retention phases of the water radial-arm maze, with no impact on reference memory. Relationships between memory scores and circulating estrogen levels were specific to follicle-depleted rats without E2 treatment. Collectively, findings demonstrate the complexity of E2 administration in a follicle-depleted background, with cognitive effects specific to working memory; furthermore, E2 administration altered circulating hormonal milieu and relationships between hormone profiles and memory. In sum, menopausal etiology impacts the parameters of E2 effects on cognition, complementing prior work with other estrogen compounds. Deciphering estrogenic actions in a system wherein the reproductive tract remains intact with follicle-depleted ovaries, thus modeling the majority or menopausal women, is critical for translational perspectives.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Mallori L Poisson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Lauren T Hewitt
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | | | - Cheryl A Dyer
- FYXX Foundation, Flagstaff, AZ, United States of America
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America.
| |
Collapse
|
14
|
Rana AK, Sharma S, Singh D. Differential activation of Gsk-3β in the cortex and the hippocampus induces cognitive and behavioural impairments in middle-aged ovariectomized rat. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2020; 4:100019. [PMID: 35755628 PMCID: PMC9216607 DOI: 10.1016/j.cpnec.2020.100019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/06/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022] Open
Abstract
Glycogen synthase kinase-3 (Gsk-3β) aberration act as a crucial pathogenic factor in several neurological conditions. However its role in menopause associated behavioural impairments is still not unclear. The present study was designed to understand the role of Gsk-3β in the progression of neurobehavioural impairments in middle-aged ovariectomized (ovx) rats. The animals showed a significant impairment in spatial and recognition memory, along with anxiety and depression-like behaviour following 22 weeks of ovx. The genomic expression of ERα, ERβ, Nrf2, HO-1, TNFα, and IL-6 was altered in both the cortex and the hippocampus of ovx rats. Protein expression of p-Gsk-3β(Ser9) was significantly downregulated in the cortex after ovx. However, the hippocampus showed a surprisingly opposite trend in the levels of p-Gsk-3β(Ser9) as that of the cortex. Differential activation of Gsk-3β and its downstream proteins such as β-catenin and p-mTOR were also altered following ovx. The study concluded that differential activation of Gsk-3β, along with oxidative stress and neuroinflammation in the cortex and the hippocampus, leads to the induction of cognitive and behaviour impairments in ovx rats.
Collapse
Affiliation(s)
- Anil Kumar Rana
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
| | - Supriya Sharma
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
| |
Collapse
|
15
|
Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci 2020; 21:535-550. [PMID: 32879508 PMCID: PMC8302223 DOI: 10.1038/s41583-020-0362-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 12/24/2022]
Abstract
Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
16
|
Koebele SV, Nishimura KJ, Bimonte-Nelson HA, Kemmou S, Ortiz JB, Judd JM, Conrad CD. A long-term cyclic plus tonic regimen of 17β-estradiol improves the ability to handle a high spatial working memory load in ovariectomized middle-aged female rats. Horm Behav 2020; 118:104656. [PMID: 31862208 PMCID: PMC7286486 DOI: 10.1016/j.yhbeh.2019.104656] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/30/2023]
Abstract
The influence of estrogens on modifying cognition has been extensively studied, revealing that a wide array of factors can significantly impact cognition, including, but not limited to, subject age, estrogen exposure duration, administration mode, estrogen formulation, stress history, and progestogen presence. Less known is whether long-term, extended exposure to estrogens would benefit or otherwise impact cognition. The present study examined the effects of 17β-estradiol (E2) exposure for seven months, beginning in late adulthood and continuing into middle age, using a regimen of cyclic exposure (bi-monthly subcutaneous injection of 10 μg E2), or Cyclic+Tonic exposure (bi-monthly subcutaneous injection of 10 μg E2 + Silastic capsules of E2) in ovariectomized female Fischer-344-CDF rats. Subjects were tested on a battery of learning and memory tasks. All groups learned the water radial-arm maze (WRAM) and Morris water maze tasks in a similar fashion, regardless of hormone treatment regimen. In the asymptotic phase of the WRAM, rats administered a Cyclic+Tonic E2 regimen showed enhanced performance when working memory was taxed compared to Vehicle and Cyclic E2 groups. Assessment of spatial memory on object placement and object recognition was not possible due to insufficient exploration of objects; however, the Cyclic+Tonic group showed increased total time spent exploring all objects compared to Vehicle-treated animals. Overall, these data demonstrate that long-term Cyclic+Tonic E2 exposure can result in some long-term cognitive benefits, at least in the spatial working memory domain, in a surgically menopausal rat model.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ, United States of America
| | - Kenji J Nishimura
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ, United States of America
| | - Salma Kemmou
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America
| | - J Bryce Ortiz
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America
| | - Jessica M Judd
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America.
| |
Collapse
|
17
|
Zhang W, Wu H, Xu Q, Chen S, Sun L, Jiao C, Wang L, Fu F, Feng Y, Qian X, Chen X. Estrogen modulation of pain perception with a novel 17β-estradiol pretreatment regime in ovariectomized rats. Biol Sex Differ 2020; 11:2. [PMID: 31918752 PMCID: PMC6953313 DOI: 10.1186/s13293-019-0271-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/21/2019] [Indexed: 11/10/2022] Open
Abstract
Estrogen plays substantial roles in pain modulation; however, studies concerning sex hormones and nociception often yield confusing results. The discrepancy could be a result of lack of consensus to regard estrogen as a variable when working with animal models; thus, the influence of hormones’ fluctuations on nociception has continually been neglected. In the present study, we designed a novel hormone substitution model to aid us to evaluate the effects of estrogen’s long-term alterations on ovariectomy (OVX)-induced mechanical hyperalgesia and the expression of estrogen receptors(ERs). OVX rats were implanted with slow-release estrogen pellets at differently arranged time points and doses, such that a gradual elevation or decrease of serum estrogen levels following a relatively stable period of estrogen replacement was achieved in rats. Our results demonstrated that gradual estrogen depletion rather than elevation following the stable period of estrogen substitution in OVX rats alleviated OVX-induced mechanical hyperalgesia in a dose-independent manner, and the opposite estrogen increase or decrease paradigms differently regulate the expression of spinal ERs. Specifically, in rats rendered to continuously increased serum estrogen, the early phase estrogen-induced anti-nociception effect in OVX rats was eliminated, which was accompanied by an over-activation of ERα and a strong depression of ERβ, while in the OVX rats subject to gradual decrease of estrogen replacement, both ERα and ERβ increased modestly compared with the OVX group. Thus, the present study demonstrated that estrogen increase or decrease modulate nociception differently through change of spinal ERs.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Sheng Chen
- Zhejiang University School of Medicine, 866th Yuhangtang Road, Hangzhou, 310058, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Luyang Wang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Feng Fu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Ying Feng
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xiaowei Qian
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China.
| |
Collapse
|
18
|
Xu Q, Sun L, Zhang W, Chen S, Wu H, Jiao C, Fu F, Feng Y, Yao H, Chen Q, Chen X. Effect of chronic pretreatment with 17β-estradiol and/or progesterone on the nociceptive response to uterine cervical distension in a rat model. Eur J Pharmacol 2019; 865:172791. [PMID: 31712057 DOI: 10.1016/j.ejphar.2019.172791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 10/25/2022]
Abstract
It is widely known that visceral pain is more prevalent in women than in men, and this phenomenon is interpreted as a consequence of the gonadal hormone modulation of pain perception and transduction. Uterine cervical distension might cause obstetric and gynecologic pain with clinical relevance to visceral pain. In this study, we focused on the roles of 17β-estradiol and progesterone in visceral nociception with the use of a rat model of uterine cervical distension. Female ovariectomized rats were injected with 17β-estradiol (E2) or progesterone (P4) for 21 days, after which visceral pain-induced spinal c-fos expression and visceromotor reflex changes were compared between ovariectomized and hormone-substituted groups. We found that uterine cervical distension induced a drastic increase in spinal c-fos expression and visceromotor reflex activity, and ovariectomy inhibited the increase in c-fos expression induced by visceral pain; this inhibition was reversed by estrogen but not progesterone replacement. This study demonstrates that estrogen is involved in uterine cervical nociception, while progesterone plays less of a significant role.
Collapse
Affiliation(s)
- Qi Xu
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenxin Zhang
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Chen
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Fu
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Feng
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huaqi Yao
- Department of Anesthesia, Maternity and Child Care Hospital, Huzhou, China
| | - Qing Chen
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Pertesi S, Coughlan G, Puthusseryppady V, Morris E, Hornberger M. Menopause, cognition and dementia - A review. Post Reprod Health 2019; 25:200-206. [PMID: 31690174 DOI: 10.1177/2053369119883485] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is increasing evidence that menopausal changes can have an impact on women’s cognition and potentially, the future development of dementia. In particular, the role of reduced levels of estrogen in postmenopausal changes has been linked to an increased risk of developing dementia in observational studies. Not surprisingly, this has led to several clinical trials investigating whether postmenopausal hormone replacement therapy can potentially delay/avoid cognitive changes and subsequently, the onset of dementia. However, the evidence of these trials has been mixed, with some showing positive effects while others show no or even negative effects. In the current review, we investigate this controversy further by reviewing the existing studies and trials in cognition and dementia. Based on the current evidence, we conclude that previous approaches may have used a mixture of women with different genetic risk factors for dementia which might explain these contradicting findings. Therefore, it is recommended that future interventional studies take a more personalised approach towards hormone replacement therapy use in postmenopausal women, by taking into account the women’s genetic status for dementia risk.
Collapse
Affiliation(s)
- S Pertesi
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - G Coughlan
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - E Morris
- Norfolk and Norwich University Hospital, Norwich, UK
| | - M Hornberger
- Norwich Medical School, University of East Anglia, Norwich, UK.,Norfolk and Suffolk Foundation Trust, Norwich, UK
| |
Collapse
|
20
|
Long T, Yao JK, Li J, Kirshner ZZ, Nelson D, Dougherty GG, Gibbs RB. Estradiol and selective estrogen receptor agonists differentially affect brain monoamines and amino acids levels in transitional and surgical menopausal rat models. Mol Cell Endocrinol 2019; 496:110533. [PMID: 31394142 PMCID: PMC6717664 DOI: 10.1016/j.mce.2019.110533] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/18/2019] [Accepted: 08/03/2019] [Indexed: 10/26/2022]
Abstract
Estrogens have many beneficial effects in the brain. Previously, we evaluated the effects of two models of menopause (surgical vs. transitional) on multiple monoaminergic endpoints in different regions of the adult rat brain in comparison with levels in gonadally intact rats. Here we evaluated the effects of estrogen receptor (ER) agonist treatments in these same two models of menopause. Neurochemical endpoints were evaluated in the hippocampus (HPC), frontal cortex (FCX), and striatum (STR) of adult ovariectomized (OVX) rats and in rats that underwent selective and gradual ovarian follicle depletion by daily injection of 4-vinylcyclohexene-diepoxide (VCD), after 1- and 6-weeks treatment with 17β-estradiol (E2), or with selective ERα (PPT), ERβ (DPN), or GPR30 (G-1) agonists. Endpoints included serotonin (5-HT) and 5-Hydroxyindoleacetic acid, dopamine (DA), 3,4-Dihydroxyphenylacetic acid and homovanillic acid, norepinephrine (NE) and epinephrine, as well as the amino acids tryptophan (TRP) and tyrosine (TYR). Significant differences between the models were detected. OVX rats were much more sensitive to ER agonist treatments than VCD-treated rats. Significant differences between brain regions also were detected. Within OVX rats, more agonist effects were detected in the HPC than in any other region. One interesting finding was the substantial decrease in TRP and TYR detected in the HPC and FCX in response to agonist treatments, particularly in OVX rats. This is on top of the substantial decreases in TRP and TYR previously reported one week after OVX or VCD-treatments in comparison with gonadally intact controls. Other interesting findings included increases in the levels of 5-HT, DA, and NE in the HPC of OVX rats treated with DPN, increases in DA detected in the FCX of OVX rats treated with any of the ER agonists, and increases in 5-HT and DA detected in the STR of OVX rats treated with E2. Many effects that were observed after 1-week of treatment were no longer observed after 6-weeks of treatment, demonstrating that effects were temporary despite continued agonist treatment. Collectively, the results demonstrate significant differences in the effects of ER agonists on monoaminergic endpoints in OVX vs. VCD-treated rats that also were brain region-specific and time dependent. The fact that agonist treatments had lesser effects in VCD treated rats than in OVX rats may help to explain reports of lesser effects of estrogen replacement on cognitive performance in women that have undergone transitional vs. surgical menopause.
Collapse
Affiliation(s)
- Tao Long
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Jeffrey K Yao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Junyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ziv Z Kirshner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Doug Nelson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - George G Dougherty
- Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
21
|
Swenson S, Hamilton J, Robison L, Thanos PK. Chronic aerobic exercise: Lack of effect on brain CB1 receptor levels in adult rats. Life Sci 2019; 230:84-88. [DOI: 10.1016/j.lfs.2019.05.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 01/25/2023]
|
22
|
Sandini TM, Reis-Silva TM, Moreira N, Bernardi MM, Lebrun I, Spinosa HDS. Effects of isoflavones on behavior, estradiol, glutamate, and GABA levels in intact middle-aged female rats. Nutr Neurosci 2018. [DOI: 10.1080/1028415x.2018.1447296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Thaísa Meira Sandini
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago Marinho Reis-Silva
- Department of Neuroscience and Behavior, Psychology Institute, University of São Paulo, São Paulo, Brazil
| | - Natalia Moreira
- Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Maria Martha Bernardi
- Post-Graduate Program of Environmental and Experimental Pathology and Post-Graduate Program of Dentistry, Paulista University, São Paulo, Brazil
| | - Ivo Lebrun
- Laboratory of Biochemistry and Biophysics, Institute Butantan, São Paulo, Brazil
| | | |
Collapse
|
23
|
Bhatta S, Blair JA, Casadesus G. Luteinizing Hormone Involvement in Aging Female Cognition: Not All Is Estrogen Loss. Front Endocrinol (Lausanne) 2018; 9:544. [PMID: 30319538 PMCID: PMC6165885 DOI: 10.3389/fendo.2018.00544] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/28/2018] [Indexed: 01/29/2023] Open
Abstract
Pervasive age-related dysfunction in hypothalamic-pituitary-gonadal (HPG) axis is associated with cognitive impairments in aging as well as pathogenesis of age-related neurodegenerative diseases such as the Alzheimer's disease (AD). As a major regulator of the HPG axis, the steroid hormone estrogen has been widely studied for its role in regulation of memory. Although estrogen modulates both cognition as well as cognition associated morphological components in a healthy state, the benefits of estrogen replacement therapy on cognition and disease seem to diminish with advancing age. Emerging data suggests an important role for luteinizing hormone (LH) in CNS function, which is another component of the HPG axis that becomes dysregulated during aging, particularly in menopause. The goal of this review is to highlight the current existing literature on LH and provide new insights on possible mechanisms of its action.
Collapse
Affiliation(s)
- Sabina Bhatta
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jeffrey A. Blair
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
- Department of Biological Sciences, Kent State University, Kent, OH, United States
- *Correspondence: Gemma Casadesus
| |
Collapse
|
24
|
Engler-Chiurazzi EB, Brown CM, Povroznik JM, Simpkins JW. Estrogens as neuroprotectants: Estrogenic actions in the context of cognitive aging and brain injury. Prog Neurobiol 2017; 157:188-211. [PMID: 26891883 PMCID: PMC4985492 DOI: 10.1016/j.pneurobio.2015.12.008] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/06/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022]
Abstract
There is ample empirical evidence to support the notion that the biological impacts of estrogen extend beyond the gonads to other bodily systems, including the brain and behavior. Converging preclinical findings have indicated a neuroprotective role for estrogen in a variety of experimental models of cognitive function and brain insult. However, the surprising null or even detrimental findings of several large clinical trials evaluating the ability of estrogen-containing hormone treatments to protect against age-related brain changes and insults, including cognitive aging and brain injury, led to hesitation by both clinicians and patients in the use of exogenous estrogenic treatments for nervous system outcomes. That estrogen-containing therapies are used by tens of millions of women for a variety of health-related applications across the lifespan has made identifying conditions under which benefits with estrogen treatment will be realized an important public health issue. Here we provide a summary of the biological actions of estrogen and estrogen-containing formulations in the context of aging, cognition, stroke, and traumatic brain injury. We have devoted special attention to highlighting the notion that estrogen appears to be a conditional neuroprotectant whose efficacy is modulated by several interacting factors. By developing criteria standards for desired beneficial peripheral and neuroprotective outcomes among unique patient populations, we can optimize estrogen treatments for attenuating the consequences of, and perhaps even preventing, cognitive aging and brain injury.
Collapse
Affiliation(s)
- E B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| | - C M Brown
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Neurobiology and Anatomy, West Virginia University, Morgantown, WV 26506, United States.
| | - J M Povroznik
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Pediatrics, West Virginia University, Morgantown, WV 26506, United States.
| | - J W Simpkins
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
25
|
Gervais NJ, Mong JA, Lacreuse A. Ovarian hormones, sleep and cognition across the adult female lifespan: An integrated perspective. Front Neuroendocrinol 2017; 47:134-153. [PMID: 28803147 PMCID: PMC7597864 DOI: 10.1016/j.yfrne.2017.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 12/22/2022]
Abstract
Loss of ovarian function in women is associated with sleep disturbances and cognitive decline, which suggest a key role for estrogens and/or progestins in modulating these symptoms. The effects of ovarian hormones on sleep and cognitive processes have been studied in separate research fields that seldom intersect. However, sleep has a considerable impact on cognitive function. Given the tight connections between sleep and cognition, ovarian hormones may influence selective aspects of cognition indirectly, via the modulation of sleep. In support of this hypothesis, a growing body of evidence indicates that the development of sleep disorders following menopause contributes to accelerated cognitive decline and dementia in older women. This paper draws from both the animal and human literature to present an integrated view of the effects of ovarian hormones on sleep and cognition across the adult female lifespan.
Collapse
Affiliation(s)
- Nicole J Gervais
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, 135 Hicks Way, Amherst, MA 01003, United States.
| | - Jessica A Mong
- Department of Pharmacology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, United States
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, 135 Hicks Way, Amherst, MA 01003, United States
| |
Collapse
|
26
|
Age-dependent regulation of obesity and Alzheimer-related outcomes by hormone therapy in female 3xTg-AD mice. PLoS One 2017; 12:e0178490. [PMID: 28575011 PMCID: PMC5456100 DOI: 10.1371/journal.pone.0178490] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/14/2017] [Indexed: 12/31/2022] Open
Abstract
Depletion of ovarian hormones at menopause is associated with increased Alzheimer's disease (AD) risk. Hormone loss also increases central adiposity, which promotes AD development. One strategy to improve health outcomes in postmenopausal women is estrogen-based hormone therapy (HT), though its efficacy is controversial. The window of opportunity hypothesis posits that HT is beneficial only if initiated near the onset of menopause. Here, we tested this hypothesis by assessing the efficacy of HT against diet-induced obesity and AD-related pathology in female 3xTg-AD mice at early versus late middle-age. HT protected against obesity and reduced β-amyloid burden only at early middle-age. One mechanism that contributes to AD pathogenesis is microglial activation, which is increased by obesity and reduced by estrogens. In parallel to its effects on β-amyloid accumulation, we observed that HT reduced morphological evidence of microglial activation in early but not late middle-age. These findings suggest that HT may be effective during human perimenopause in reducing indices of obesity and AD-related pathology, a conclusion consistent with the window of opportunity hypothesis.
Collapse
|
27
|
Koebele SV, Mennenga SE, Hiroi R, Quihuis AM, Hewitt LT, Poisson ML, George C, Mayer LP, Dyer CA, Aiken LS, Demers LM, Carson C, Bimonte-Nelson HA. Cognitive changes across the menopause transition: A longitudinal evaluation of the impact of age and ovarian status on spatial memory. Horm Behav 2017; 87:96-114. [PMID: 27793768 PMCID: PMC5479707 DOI: 10.1016/j.yhbeh.2016.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 09/19/2016] [Accepted: 10/23/2016] [Indexed: 11/16/2022]
Abstract
Cognitive changes that occur during mid-life and beyond are linked to both aging and the menopause transition. Studies in women suggest that the age at menopause onset can impact cognitive status later in life; yet, little is known about memory changes that occur during the transitional period to the postmenopausal state. The 4-vinylcyclohexene diepoxide (VCD) model simulates transitional menopause in rodents by depleting the immature ovarian follicle reserve and allowing animals to retain their follicle-deplete ovarian tissue, resulting in a profile similar to the majority of perimenopausal women. Here, Vehicle or VCD treatment was administered to ovary-intact adult and middle-aged Fischer-344 rats to assess the trajectory of cognitive change across time with normal aging and aging with transitional menopause via VCD-induced follicular depletion, as well as to evaluate whether age at the onset of follicular depletion plays a role in cognitive outcomes. Animals experiencing the onset of menopause at a younger age exhibited impaired spatial memory early in the transition to a follicle-deplete state. Additionally, at the mid- and post- follicular depletion time points, VCD-induced follicular depletion amplified an age effect on memory. Overall, these findings suggest that age at the onset of menopause is a critical parameter to consider when evaluating learning and memory across the transition to reproductive senescence. From a translational perspective, this study illustrates how age at menopause onset might impact cognition in menopausal women, and provides insight into time points to explore for the window of opportunity for hormone therapy during the menopause transition period. Hormone therapy during this critical juncture might be especially efficacious at attenuating age- and menopause- related cognitive decline, producing healthy brain aging profiles in women who retain their ovaries throughout their lifespan.
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
| | - Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Alicia M Quihuis
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Lauren T Hewitt
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Mallori L Poisson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Christina George
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Loretta P Mayer
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Cheryl A Dyer
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Leona S Aiken
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States
| | - Laurence M Demers
- The Pennsylvania State University College of Medicine, The M. S. Hershey Medical Center, Hershey, PA 17033, United States
| | - Catherine Carson
- 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
|
28
|
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
|
29
|
Pinceti E, Shults CL, Rao YS, Pak TR. Differential Effects of E2 on MAPK Activity in the Brain and Heart of Aged Female Rats. PLoS One 2016; 11:e0160276. [PMID: 27487271 PMCID: PMC4972350 DOI: 10.1371/journal.pone.0160276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
Aging and the coincident loss of circulating estrogens at menopause lead to increased risks for neurological and cardiovascular pathologies. Clinical studies show that estrogen therapy (ET) can be beneficial in mitigating these negative effects, in both the brain and heart, when it is initiated shortly after the perimenopausal transition. However, this same therapy is detrimental when initiated >10 years postmenopause. Importantly, the molecular mechanisms underlying this age-related switch in ET efficacy are unknown. Estrogen receptors (ERs) mediate the neuroprotective and cardioprotective functions of estrogens by modulating gene transcription or, non-genomically, by activating second messenger signaling pathways, such as mitogen activated protein kinases (MAPK). These kinases are critical regulators of cell signaling pathways and have widespread downstream effects. Our hypothesis is that age and estrogen deprivation following menopause alters the expression and activation of the MAPK family members p38 and ERK in the brain and heart. To test this hypothesis, we used a surgically induced model of menopause in 18 month old rats through bilateral ovariectomy (OVX) followed by an acute dose of 17β-estradiol (E2) administered at varying time points post-OVX (1 week, 4 weeks, 8 weeks, or 12 weeks). Age and E2 treatment differentially regulated kinase activity in both the brain and heart, and the effects were also brain region specific. MAPK signaling plays an integral role in aging, and the aberrant regulation of those signaling pathways might be involved in age-related disorders. Clinical studies show benefits of ET during early menopause but detrimental effects later, which might be reflective of changes in kinase expression and activation status.
Collapse
Affiliation(s)
- Elena Pinceti
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Cody L. Shults
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Yathindar S. Rao
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
| | - Toni R. Pak
- Department of Cell and Molecular Physiology, Health Science Division, Loyola University Chicago, Maywood, Illinois, United States of America
- * E-mail:
| |
Collapse
|
30
|
Marriott L, Wenk G. Neurobiological Consequences of Long-Term Estrogen Therapy. CURRENT DIRECTIONS IN PSYCHOLOGICAL SCIENCE 2016. [DOI: 10.1111/j.0963-7214.2004.00301.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Postmenopausal women demonstrate an increased incidence of Alzheimer's disease (AD). Epidemiological evidence suggests that estrogen replacement therapy (ERT) may reduce the risk or delay the onset of AD, yet recent clinical trials found no cognitive benefits of ERT in women with mild to moderate AD. This review suggests that the timing of estrogen administration may explain these conflicting results. Chronic administration has neurobiological consequences that can affect neural and immune function, but a therapy designed to mimic the natural cycle of fluctuating hormones may more effectively slow the progression of AD in postmenopausal women.
Collapse
Affiliation(s)
- L.K. Marriott
- Division of Neural Systems, Memory & Aging, Arizona Research Laboratories, University of Arizona
| | - G.L. Wenk
- Division of Neural Systems, Memory & Aging, Arizona Research Laboratories, University of Arizona
| |
Collapse
|
31
|
Ortiz-Pérez A, Espinosa-Raya J, Picazo O. An enriched environment and 17-beta estradiol produce similar pro-cognitive effects on ovariectomized rats. Cogn Process 2016; 17:15-25. [DOI: 10.1007/s10339-015-0746-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 12/11/2015] [Indexed: 02/01/2023]
|
32
|
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
|
33
|
Frick KM, Kim J, Tuscher JJ, Fortress AM. Sex steroid hormones matter for learning and memory: estrogenic regulation of hippocampal function in male and female rodents. Learn Mem 2015; 22:472-93. [PMID: 26286657 PMCID: PMC4561402 DOI: 10.1101/lm.037267.114] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/09/2015] [Indexed: 01/24/2023]
Abstract
Ample evidence has demonstrated that sex steroid hormones, such as the potent estrogen 17β-estradiol (E2), affect hippocampal morphology, plasticity, and memory in male and female rodents. Yet relatively few investigators who work with male subjects consider the effects of these hormones on learning and memory. This review describes the effects of E2 on hippocampal spinogenesis, neurogenesis, physiology, and memory, with particular attention paid to the effects of E2 in male rodents. The estrogen receptors, cell-signaling pathways, and epigenetic processes necessary for E2 to enhance memory in female rodents are also discussed in detail. Finally, practical considerations for working with female rodents are described for those investigators thinking of adding females to their experimental designs.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
34
|
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
|
35
|
Marques DA, de Carvalho D, da Silva GSF, Szawka RE, Anselmo-Franci JA, Bícego KC, Gargaglioni LH. Ventilatory, metabolic, and thermal responses to hypercapnia in female rats: effects of estrous cycle, ovariectomy, and hormonal replacement. J Appl Physiol (1985) 2015; 119:61-8. [DOI: 10.1152/japplphysiol.00254.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/23/2015] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to examine how estrous cycle, ovariectomy, and hormonal replacement affect the respiratory [ventilation (V̇e), tidal volume, and respiratory frequency], metabolic (V̇o2), and thermoregulatory (body temperature) responses to hypercapnia (7% CO2) in female Wistar rats. The parameters were measured in rats during different phases of the estrous cycle, and also in ovariectomized (OVX) rats supplemented with 17β-estradiol (OVX+E2), with a combination of E2 and progesterone (OVX+E2P), or with corn oil (OVX+O, vehicle). All experiments were conducted on day 8 after ovariectomy. The intact animals did not present alterations during normocapnia or under hypercapnia in V̇e, tidal volume, respiratory frequency, V̇o2, and V̇e/V̇o2 in the different phases of the estrous cycle. However, body temperature was higher in female rats on estrus. Hormonal replacement did not change the ventilatory, thermoregulatory, or metabolic parameters during hypercapnia, compared with the OVX animals. Nevertheless, OVX+E2, OVX+E2P, and OVX+O presented lower hypercapnic ventilatory responses compared with intact females on the day of estrus. Also, rats in estrus showed higher V̇e and V̇e/V̇o2 during hypercapnia than OVX animals. The data suggest that other gonadal factors, besides E2 and P, are possibly involved in these responses.
Collapse
Affiliation(s)
- Danuzia A. Marques
- Department of Animal Morphology and Physiology, São Paulo State University, UNESP FCAV at Jaboticabal, São Paulo, Brazil
| | | | - Glauber S. F. da Silva
- Department of Animal Morphology and Physiology, São Paulo State University, UNESP FCAV at Jaboticabal, São Paulo, Brazil
| | - Raphael E. Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Brazil; and
| | - Janete A. Anselmo-Franci
- Department of Morphology, Stomatology and Physiology, Dental School of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Kênia C. Bícego
- Department of Animal Morphology and Physiology, São Paulo State University, UNESP FCAV at Jaboticabal, São Paulo, Brazil
| | - Luciane H. Gargaglioni
- Department of Animal Morphology and Physiology, São Paulo State University, UNESP FCAV at Jaboticabal, São Paulo, Brazil
| |
Collapse
|
36
|
Abstract
Estradiol effects on memory depend on hormone levels and the interaction of different estrogen receptors within neural circuits. Estradiol induces gene transcription and rapid membrane signaling mediated by estrogen receptor-alpha (ERα), estrogen receptor-beta (ERβ), and a recently characterized G-protein coupled estrogen receptor, each with distinct distributions and ability to influence estradiol-dependent signaling. Investigations using receptor specific agonists suggest that all three receptors rapidly activate kinase-signaling and have complex dose-dependent influences on memory. Research employing receptor knockout mice demonstrate that ERα maintains transcription and memory as estradiol levels decline. This work indicates a regulatory role of ERβ in transcription and cognition, which depends on estradiol levels and the function of ERα. The regulatory role of ERβ is due in part to ERβ acting as a negative regulator of ERα-mediated transcription. Vector-mediated expression of estrogen receptors in the hippocampus provides an innovative research approach and suggests that memory depends on the relative expression of ERα and ERβ interacting with estradiol levels. Notably, the ability of estradiol to improve cognition declines with advanced age along with decreased expression of estrogen receptors. Thus, it will be important for future research to determine the mechanisms that regulate estrogen receptor expression during aging.
Collapse
Affiliation(s)
- Linda A Bean
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lara Ianov
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
37
|
The Effects of Ethinylestradiol and Progestins (“the pill”) on Cognitive Function in Pre-menopausal Women. Neurochem Res 2014; 39:2288-300. [DOI: 10.1007/s11064-014-1444-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/18/2014] [Accepted: 09/23/2014] [Indexed: 11/27/2022]
|
38
|
Long-term effects of ageing and ovariectomy on aversive and recognition memory and DNA damage in the hippocampus of female rats. Acta Neuropsychiatr 2014; 26:161-9. [PMID: 25142192 DOI: 10.1017/neu.2013.48] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study investigated the influence of ageing - in particular the decrease of gonadal hormone levels during the ageing process - on the memory and the levels of DNA damage in the hippocampus of female rats. METHODS Three groups of female Wistar rats were investigated: Group I consisted of non-ovariectomised, adult animals (6 months old); Group II consisted of non-ovariectomised, aged animals (18 months old); and Group III consisted of ovariectomised, aged animals (18 months old). The memory of the animals in these groups was examined via novel object recognition and inhibitory avoidance tests. The hippocampus tissue samples of all animals were obtained via biopsy and used to quantify the DNA damage using a Comet Assay. RESULTS According to our findings, the process of ageing results in a change during the behavioural tests. To prevent genotoxic damage to the hippocampus caused by the ageing process, lowered hormone levels seem to be part of a protective biochemical mechanism in the body of rats. Animals that were previously submitted to an ovariectomy adapted better to these lower levels of hormones. CONCLUSION Our results indicate that ovariectomy can provide beneficial long-term effects on the memory. However, this could be specific to the kind of memory examined, as the aversive memory deficits caused by ageing were not affected by ovariectomy.
Collapse
|
39
|
Li LH, Wang ZC, Yu J, Zhang YQ. Ovariectomy results in variable changes in nociception, mood and depression in adult female rats. PLoS One 2014; 9:e94312. [PMID: 24710472 PMCID: PMC3978042 DOI: 10.1371/journal.pone.0094312] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/15/2014] [Indexed: 01/08/2023] Open
Abstract
Decline in the ovarian hormones with menopause may influence somatosensory, cognitive, and affective processing. The present study investigated whether hormonal depletion alters the nociceptive, depressive-like and learning behaviors in experimental rats after ovariectomy (OVX), a common method to deplete animals of their gonadal hormones. OVX rats developed thermal hyperalgesia in proximal and distal tail that was established 2 weeks after OVX and lasted the 7 weeks of the experiment. A robust mechanical allodynia was also occurred at 5 weeks after OVX. In the 5th week after OVX, dilute formalin (5%)-induced nociceptive responses (such as elevating and licking or biting) during the second phase were significantly increased as compared to intact and sham-OVX females. However, chronic constriction injury (CCI) of the sciatic nerve-induced mechanical allodynia did not differ as hormonal status (e.g. OVX and ovarian intact). Using formalin-induced conditioned place avoidance (F-CPA), which is believed to reflect the pain-related negative emotion, we further found that OVX significantly attenuated F-CPA scores but did not alter electric foot-shock-induced CPA (S-CPA). In the open field and forced swimming test, there was an increase in depressive-like behaviors in OVX rats. There was no detectable impairment of spatial performance by Morris water maze task in OVX rats up to 5 weeks after surgery. Estrogen replacement retrieved OVX-induced nociceptive hypersensitivity and depressive-like behaviors. This is the first study to investigate the impacts of ovarian removal on nociceptive perception, negative emotion, depressive-like behaviors and spatial learning in adult female rats in a uniform and standard way.
Collapse
Affiliation(s)
- Li-Hong Li
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Zhe-Chen Wang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical Colloge, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
40
|
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
|
41
|
Estrogen-dependent changes in estrogen receptor-β mRNA expression in middle-aged female rat brain. Brain Res 2013; 1543:49-57. [PMID: 24239930 DOI: 10.1016/j.brainres.2013.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 01/21/2023]
Abstract
During aging, estrogen production and circulating levels of estrogen are markedly decreased in females. Although several differences exist in the process of reproductive aging between women and female rats, the results of many studies suggest that the female rat, especially the middle-aged or aged ovariectomized female, is an important animal model of hormone loss in women. In target tissues including the brain, the actions of estrogen are mediated mainly via the alpha and beta subtypes of the estrogen receptor (ER-α and ER-β). Estrogen treatment is known to change the expression of ER-α mRNA and protein in specific regions of the brain in middle-aged female rodents. In contrast, we do not know if estrogen regulates the expression of ER-β in the brain at this stage of life. In the present study, we performed in situ hybridization on brain sections of ovariectomized and estrogen-treated middle-aged female rats to reveal the effects of estrogen on the expression of ER-β throughout the brain. Our results showed that estrogen treatment decreased the number of ER-β mRNA-positive cells in the mitral cell and external plexiform layers of the olfactory bulb, central amygdaloid nucleus, medial geniculate nucleus, posterior hypothalamic nucleus, suprachiasmatic nucleus, and reticular part of the substantia nigra. As compared to the results of previous studies of young females, our data revealed that the regions in which expression of ER-β mRNA expression is affected by estrogen differ in middle age. These results suggest that the effects of estrogen on ER-β expression change with age.
Collapse
|
42
|
Frick KM. Epigenetics, oestradiol and hippocampal memory consolidation. J Neuroendocrinol 2013; 25:1151-62. [PMID: 24028406 PMCID: PMC3943552 DOI: 10.1111/jne.12106] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 08/13/2013] [Accepted: 09/05/2013] [Indexed: 02/02/2023]
Abstract
Epigenetic alterations of histone proteins and DNA are essential for hippocampal synaptic plasticity and cognitive function, and contribute to the aetiology of psychiatric disorders and neurodegenerative diseases. Hippocampal memory formation depends on histone alterations and DNA methylation, and increasing evidence suggests that the regulation of these epigenetic processes by modulatory factors, such as environmental enrichment, stress and hormones, substantially influences memory function. Recent work from our laboratory suggests that the ability of the sex-steroid hormone 17β-oestradiol (E2 ) to enhance novel object recognition memory consolidation in young adult female mice is dependent on histone H3 acetylation and DNA methylation in the dorsal hippocampus. Our data also suggest that enzymes mediating DNA methylation and histone acetylation work in concert to regulate the effects of E2 on memory consolidation. These findings shed light on the epigenetic mechanisms that influence hormonal modulation of cognitive function, and may have important implications for understanding how hormones influence cognition in adulthood and ageing. The present review provides a brief overview of the literature on epigenetics and memory, describes in detail our findings demonstrating that epigenetic alterations regulate E2 -induced memory enhancement in female mice, and discusses future directions for research on the epigenetic regulation of E2 -induced memory enhancement.
Collapse
Affiliation(s)
- Karyn M. Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| |
Collapse
|
43
|
Stouffer EM, Barry JL. A sex difference in the onset of the latent learning impairment in rats. Dev Psychobiol 2013; 56:1134-41. [PMID: 24122647 DOI: 10.1002/dev.21168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/06/2013] [Indexed: 11/10/2022]
Abstract
The current study examined a sex difference in the onset of a latent learning impairment in Sprague-Dawley rats. Forty rats (20 male, 20 female) were tested on the Latent Cue Preference (LCP) task at 3 or 11 months of age. Additionally, 19 female rats were tested at 14 or 18 months of age. All rats were given four training trials in the LCP task using a three-compartment box, during which the rats explored a water-paired compartment and an unpaired compartment (each with a different visual cue) on consecutive days. Rats were then water-deprived for 23 hr and given a compartment preference test, in which more time spent in the water-paired compartment demonstrated latent learning. Results showed that 11-month old males and 18-month old females showed impaired latent learning, but 11- and 14-month old females showed intact latent learning, which may possibly be due to the neuroprotective effects of estrogen.
Collapse
Affiliation(s)
- Eric M Stouffer
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E 2nd Street, Bloomsburg, PA, 17815.
| | | |
Collapse
|
44
|
Continuously delivered ovarian steroids do not alter dendritic spine density or morphology in macaque dorsolateral prefrontal cortical neurons. Neuroscience 2013; 255:219-25. [PMID: 24120552 DOI: 10.1016/j.neuroscience.2013.09.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 11/22/2022]
Abstract
Aged ovariectomized (OVX) female monkeys, a model for menopause in humans, show a decline in spine density in the dorsolateral prefrontal cortex (dlPFC) and diminished performance in cognitive tasks requiring this brain region. Previous studies in our laboratory have shown that long-term cyclic treatment with 17β-estradiol (E) produces an increase in spine density and in the proportion of thinner spines in layer III pyramidal neurons in the dlPFC of both young and aged OVX rhesus monkeys. Here we used 3D reconstruction of Lucifer yellow-loaded neurons to investigate whether clinically relevant schedules of hormone therapy would produce similar changes in prefrontal cortical neuronal morphology as long-term cyclic E treatment in young female monkeys. We found that continuously delivered E, with or without a cyclic progesterone treatment, did not alter spine density or morphology in the dlPFC of young adult OVX rhesus monkeys. We also found that the increased density of thinner spines evident in the dlPFC 24h after E administration in the context of long-term cyclic E therapy is no longer detectable 20days after E treatment. When compared with the results of our previously published investigations, our results suggest that cyclic fluctuations in serum E levels may cause corresponding fluctuations in the density of thin spines in the dlPFC. By contrast, continuous administration of E does not support sustained increases in thin spine density. Physiological fluctuations in E concentration may be necessary to maintain the morphological sensitivity of the dlPFC to E.
Collapse
|
45
|
Maki P. Is timing everything? New insights into why the effect of estrogen therapy on memory might be age dependent. Endocrinology 2013; 154:2570-2. [PMID: 23873768 PMCID: PMC3713207 DOI: 10.1210/en.2013-1598] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Pauline Maki
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612–7327, USA.
| |
Collapse
|
46
|
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
|
47
|
Acosta JI, Hiroi R, Camp BW, Talboom JS, Bimonte-Nelson HA. An update on the cognitive impact of clinically-used hormone therapies in the female rat: models, mazes, and mechanisms. Brain Res 2013; 1514:18-39. [PMID: 23333453 PMCID: PMC3739440 DOI: 10.1016/j.brainres.2013.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/09/2013] [Indexed: 01/05/2023]
Abstract
In women, ovarian hormone loss associated with menopause has been related to cognitive decline. Hormone therapy (HT) may ameliorate some of these changes. Understanding the cognitive impact of female steroids, including estrogens, progestogens, and androgens, is key to discovering treatments that promote brain health in women. The preclinical literature has presented elegant and methodical experiments allowing a better understanding of parameters driving the cognitive consequences of ovarian hormone loss and HT. Animal models have been a valuable tool in this regard, and will be vital to future discoveries. Here, we provide an update on the literature evaluating the impact of female steroid hormones on cognition, and the putative mechanisms mediating these effects. We focus on preclinical work that was done with an eye toward clinical realities. Parameters that govern the cognitive efficacy of HT, from what we know thus far, include but are not limited to: type, dose, duration, and route of HT, age at HT initiation, timing of HT relative to ovarian hormone loss, memory type examined, menopause history, and hormone receptor status. Researchers have identified intricate relationships between some of these factors by studying their individual effects on cognition. As of late, there is increased focus on studying interactions between these variables as well as multiple hormone types when administered concomitantly. This is key to translating preclinical data to the clinic, wherein women typically have concurrent exposure to endogenous ovarian hormones as well as exogenous combination HTs, which include both estrogens and progestins. Gains in understanding the parameters of HT effects on cognition provide exciting novel avenues that can inform clinical treatments, eventually expanding the window of opportunity to optimally enhance cognition and brain health in aging women. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- J I Acosta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | |
Collapse
|
48
|
Chisholm NC, Juraska JM. Factors influencing the cognitive and neural effects of hormone treatment during aging in a rodent model. Brain Res 2013; 1514:40-9. [PMID: 23419893 DOI: 10.1016/j.brainres.2013.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 02/06/2013] [Accepted: 02/11/2013] [Indexed: 12/18/2022]
Abstract
Whether hormone treatment alters brain structure or has beneficial effects on cognition during aging has recently become a topic of debate. Although previous research has indicated that hormone treatment benefits memory in menopausal women, several newer studies have shown no effect or detrimental effects. These inconsistencies emphasize the need to evaluate the role of hormones in protecting against age-related cognitive decline in an animal model. Importantly, many studies investigating the effects of estrogen and progesterone on cognition and related brain regions have used young adult animals, which respond differently than aged animals. However, when only the studies that have examined the effects of hormone treatment in an aging model are reviewed, there are still varied behavioral and neural outcomes. This article reviews some of the important factors that can influence the behavioral and neural outcomes of hormone treatment including the type of estrogen administered, whether or not estrogen is combined with progesterone and if so, the type of progesterone used, as well as the route, mode, and length of treatment. How these factors influence cognitive outcomes highlights the importance of study design and avoiding generalizations from a small number of studies. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Psychology, University of Illinois at Urbana - Champaign, Champaign, IL 61820, USA.
| | | |
Collapse
|
49
|
Daniel JM. Estrogens, estrogen receptors, and female cognitive aging: the impact of timing. Horm Behav 2013; 63:231-7. [PMID: 22587940 DOI: 10.1016/j.yhbeh.2012.05.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 10/28/2022]
Abstract
Estrogens have been shown to be protective agents against neurodegeneration and associated cognitive decline in aging females. However, clinical data have been equivocal as to the benefits to the brain and cognition of estrogen therapy in postmenopausal women. One factor that is proposed to be critical in determining the efficacy of hormone therapy is the timing of its initiation. The critical period or window of opportunity hypothesis proposes that following long-term ovarian hormone deprivation, the brain and cognition become insensitive to exogenously administered estrogens. In contrast, if estrogens are administered during a critical period near the time of cessation of ovarian function, they will exert beneficial effects. The focus of the current review is the examination of evidence from rodent models investigating the critical period hypothesis. A growing body of experimental data indicates that beneficial effects of 17β-estradiol (estradiol) on cognition and on cholinergic function and hippocampal plasticity, both of which have been linked to the ability of estradiol to exert beneficial effects on cognition, are attenuated if estradiol is administered following a period of long-term ovarian hormone deprivation. Further, emerging data implicate loss of estrogen receptor alpha (ERα) in the brain resulting from long-term hormone deprivation as a basis for the existence of the critical period. A unifying model is proposed by which the presence or absence of estrogens during a critical period following the cessation of ovarian function permanently alters the system resulting in decreased or increased risk, respectively, of neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Program in Neuroscience, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
50
|
Pompili A, Arnone B, Gasbarri A. Estrogens and memory in physiological and neuropathological conditions. Psychoneuroendocrinology 2012; 37:1379-96. [PMID: 22309827 DOI: 10.1016/j.psyneuen.2012.01.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 01/13/2012] [Accepted: 01/13/2012] [Indexed: 12/22/2022]
Abstract
Ovarian hormones can influence brain regions crucial to higher cognitive functions, such as learning and memory, acting at structural, cellular and functional levels, and modulating neurotransmitter systems. Among the main effects of estrogens, the protective role that they may play against the deterioration of cognitive functions occurring with normal aging is of essential importance. In fact, during the last century, there has been a 30 years increase in female life expectancy, from 50 to 83 years; however, the mean age of spontaneous menopause remains stable, 50-51 years, with variability related to race and ethnicity. Therefore, women are now spending a greater fraction of their lives in a hypoestrogenic state. Although many cognitive functions seem to be unaffected by normal aging, age-related impairments are particularly evident in tasks involving working memory (WM), whose deficits are a recognized feature of Alzheimer's disease (AD). Many studies conducted over the past two decades showed that the female gonadal hormone estradiol can influence performance of learning and memory tasks, both in animal and humans. There is a great deal of evidence, mostly from animal models, that estrogens can facilitate or enhance performance on WM tasks; therefore, it is very important to clarify their role on this type of memory. To this aim, in this review we briefly describe the most relevant neurobiological bases of estrogens, that can explain their effects on cognitive functioning, and then we summarize the results of works conducted in our laboratory, both on animals and humans, utilizing the menstrual/estrous cycle as a useful noninvasive model. Finally, we review the possible role of estrogens in neuropathological conditions, such as AD and schizophrenia.
Collapse
Affiliation(s)
- Assunta Pompili
- Department of Biomedical Sciences and Technologies, University of L'Aquila, L'Aquila, Italy.
| | | | | |
Collapse
|