1
|
Kim SM, Sultana F, Korkmaz F, Rojekar S, Pallapati A, Ryu V, Lizneva D, Yuen T, Rosen CJ, Zaidi M. Neuroendocrinology of bone. Pituitary 2024; 27:761-777. [PMID: 39096452 DOI: 10.1007/s11102-024-01437-5] [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] [Accepted: 07/26/2024] [Indexed: 08/05/2024]
Abstract
The past decade has witnessed significant advances in our understanding of skeletal homeostasis and the mechanisms that mediate the loss of bone in primary and secondary osteoporosis. Recent breakthroughs have primarily emerged from identifying disease-causing mutations and phenocopying human bone disease in rodents. Notably, using genetically-modified rodent models, disrupting the reciprocal relationship with tropic pituitary hormone and effector hormones, we have learned that pituitary hormones have independent roles in skeletal physiology, beyond their effects exerted through target endocrine glands. The rise of follicle-stimulating hormone (FSH) in the late perimenopause may account, at least in part, for the rapid bone loss when estrogen is normal, while low thyroid-stimulating hormone (TSH) levels may contribute to the bone loss in thyrotoxicosis. Admittedly speculative, suppressed levels of adrenocorticotropic hormone (ACTH) may directly exacerbate bone loss in the setting of glucocorticoid-induced osteoporosis. Furthermore, beyond their established roles in reproduction and lactation, oxytocin and prolactin may affect intergenerational calcium transfer and therefore fetal skeletal mineralization, whereas elevated vasopressin levels in chronic hyponatremic states may increase the risk of bone loss.. Here, we discuss the interaction of each pituitary hormone in relation to its role in bone physiology and pathophysiology.
Collapse
Affiliation(s)
- Se-Min Kim
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Farhath Sultana
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Funda Korkmaz
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Satish Rojekar
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anusha Pallapati
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vitaly Ryu
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daria Lizneva
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tony Yuen
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Mone Zaidi
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
2
|
Butler T, Tey SR, Galvin JE, Perry G, Bowen RL, Atwood CS. Endocrine Dyscrasia in the Etiology and Therapy of Alzheimer's Disease. J Alzheimers Dis 2024; 101:705-713. [PMID: 39240636 DOI: 10.3233/jad-240334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The increase in the incidence of dementia over the last century correlates strongly with the increases in post-reproductive lifespan during this time. As post-reproductive lifespan continues to increase it is likely that the incidence of dementia will also increase unless therapies are developed to prevent, slow or cure dementia. A growing body of evidence implicates age-related endocrine dyscrasia and the length of time that the brain is subjected to this endocrine dyscrasia, as a key causal event leading to the cognitive decline associated with aging and Alzheimer's disease (AD), the major form of dementia in our society. In particular, the elevations in circulating gonadotropins, resulting from the loss of gonadal sex hormone production with menopause and andropause, appear central to the development of AD neuropathology and cognitive decline. This is supported by numerous cell biology, preclinical animal, and epidemiological studies, as well as human clinical studies where suppression of circulating luteinizing hormone and/or follicle-stimulating hormone with either gonadotropin-releasing hormone analogues, or via physiological hormone replacement therapy, has been demonstrated to halt or significantly slow cognitive decline in those with AD. This review provides an overview of past and present studies demonstrating the importance of hypothalamic-pituitary-gonadal hormone balance for normal cognitive functioning, and how targeting age-related endocrine dyscrasia with hormone rebalancing strategies provides an alternative treatment route for those with AD.
Collapse
Affiliation(s)
- Tracy Butler
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sin-Ruow Tey
- JangoBio, LLC, Division of Cell Biology, Fitchburg, WI, USA
| | - James E Galvin
- Departments of Neurology and Psychiatry, Comprehensive Center for Brain Health, University of Miami, Miller School of Medicine, Boca Raton, FL, USA
| | - George Perry
- Department of Neuroscience, Development and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Craig S Atwood
- Geriatric Research, Education and Clinical Center, Veterans Administration Hospital and Department of Medicine, University of Wisconsin, Madison, WI, USA
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
3
|
Yoon BK. The impacts of menopausal hormone therapy on longer-term health consequences of ovarian hormone deficiency. Climacteric 2023; 26:193-197. [PMID: 37011667 DOI: 10.1080/13697137.2023.2173571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
This study on the longer-term health consequences of ovarian hormone deficiency (OHD) received the Henry Burger Prize in 2022. Osteoporosis, cardiovascular disease and dementia are major degenerative diseases that are also causally associated with OHD. Two randomized controlled trials (RCTs) revealed no significant difference in bone mineral density by adding alendronate to ongoing menopausal hormone therapy (MHT) or combining alendronate at MHT initiation. Another RCT pursuing the effects on fracture recurrence and total mortality in women with hip fracture disclosed that MHT with percutaneous estradiol gel (PEG) and micronized progesterone (MP4) was comparable to risedronate. Basic studies reported that 17β-estradiol exerted direct beneficial actions on vascular smooth muscle in cell proliferation, fibrinolysis and apoptosis. A fourth RCT showed that MP4 had a neutral impact on the PEG response of blood pressure and arterial stiffness. A fifth RCT suggested that the combination therapy of conjugated equine estrogen and MP4 was superior to tacrine in preserving activities in daily living in women with Alzheimer's disease. In addition, PEG plus MP4 attenuated cognitive decline in women with mild cognitive impairment in a sixth RCT. Finally, the all-cause mortality in recently menopausal women receiving MHT was updated using an adaptive meta-analysis of four RCTs.
Collapse
Affiliation(s)
- B-K Yoon
- Department of Obstetrics, Gynecology, and Women's Health, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Sharma A, Davies R, Kapoor A, Islam H, Webber L, Jayasena CN. The effect of hormone replacement therapy on cognition and mood. Clin Endocrinol (Oxf) 2023; 98:285-295. [PMID: 36447434 PMCID: PMC11497347 DOI: 10.1111/cen.14856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVES To summarise the available data regarding the effect of hormone replacement therapy (HRT) on cognition and mood in women. BACKGROUND Complaints of impaired cognition and mood are common in the peri-menopausal and menopausal period. There is debate as to whether HRT can ameliorate this phenomenon. DESIGN A literature search of studies using electronic databases was conducted. Both randomised control trials and observational studies were included. PATIENTS Perimenopausal and menopausal women. RESULTS Due to the heterogenicity of results it is challenging to draw firm conclusions. The preparations used in many of the studies are older regimes no longer routinely used clinically. The notion of a 'critical window' for HRT is compelling, suggesting HRT has a positive impact on cognition when administered in the peri-menopausal or early postmenopausal period but may have negative effects on cognition in the older, postmenopausal woman. The evidence would seem to suggest importance of hormonal replacement in woman undergoing a surgical menopause, especially when young. It remains unclear for how long they ought to continue HRT though until at least the natural age of the menopause seems reasonable. Evidence for a positive effect of HRT on mood is more convincing, though possibly more efficacious in the younger age group. The effect of HRT on anxiety is less clear. CONCLUSIONS Further study, particularly focusing on the more contemporaneous HRT preparations, is warranted before evidence-based conclusions can be drawn.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Rhianna Davies
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | | | | | - Lisa Webber
- Department of GynaecologySt. Mary's HospitalLondonUK
| | - Channa N. Jayasena
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| |
Collapse
|
5
|
Is Hormone Replacement Therapy a Risk Factor or a Therapeutic Option for Alzheimer's Disease? Int J Mol Sci 2023; 24:ijms24043205. [PMID: 36834617 PMCID: PMC9964432 DOI: 10.3390/ijms24043205] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accounts for more than half of all dementia cases in the elderly. Interestingly, the clinical manifestations of AD disproportionately affect women, comprising two thirds of all AD cases. Although the underlying mechanisms for these sex differences are not fully elucidated, evidence suggests a link between menopause and a higher risk of developing AD, highlighting the critical role of decreased estrogen levels in AD pathogenesis. The focus of this review is to evaluate clinical and observational studies in women, which have investigated the impact of estrogens on cognition or attempted to answer the prevailing question regarding the use of hormone replacement therapy (HRT) as a preventive or therapeutic option for AD. The articles were retrieved through a systematic review of the databases: OVID, SCOPUS, and PubMed (keywords "memory", "dementia," "cognition," "Alzheimer's disease", "estrogen", "estradiol", "hormone therapy" and "hormone replacement therapy" and by searching reference sections from identified studies and review articles). This review presents the relevant literature available on the topic and discusses the mechanisms, effects, and hypotheses that contribute to the conflicting findings of HRT in the prevention and treatment of age-related cognitive deficits and AD. The literature suggests that estrogens have a clear role in modulating dementia risk, with reliable evidence showing that HRT can have both a beneficial and a deleterious effect. Importantly, recommendation for the use of HRT should consider the age of initiation and baseline characteristics, such as genotype and cardiovascular health, as well as the dosage, formulation, and duration of treatment until the risk factors that modulate the effects of HRT can be more thoroughly investigated or progress in the development of alternative treatments can be made.
Collapse
|
6
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
7
|
Pratchayasakul W, Arunsak B, Suparan K, Sriwichaiin S, Chunchai T, Chattipakorn N, Chattipakorn SC. Combined caloric restriction and exercise provides greater metabolic and neurocognitive benefits than either as a monotherapy in obesity with or without estrogen deprivation. J Nutr Biochem 2022; 110:109125. [PMID: 35977664 DOI: 10.1016/j.jnutbio.2022.109125] [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: 08/07/2021] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/13/2023]
Abstract
Neurodegeneration, as indicated by brain dysfunction and cognitive decline, is one of the complications associated with obesity and estrogen deprivation. Calorie restriction and exercise regimes improved brain function in neurodegenerative diseases. However, the comparative effects of a combination of calorie restriction with exercise, calorie restriction, and an exercise regime alone on brain/cognitive function in obesity with or without estrogen deprivation have not been investigated. Sixty female rats were fed a normal diet (ND) or a high-fat diet (HFD) for 27 weeks. At week 13, the ND-fed rats underwent a sham operation with sedentary lifestyle, HFD-fed rats were divided into two groups: each having either a sham operation (HFS) or ovariectomy (HFO). At week 20, HFD-fed rats in each group were divided into four subgroups undergoing either a sedentary lifestyle, calorie restriction, exercise regime or a combination of calorie restriction and exercise for 7 weeks. Insulin resistance, cognitive decline and hippocampal pathologies were found in both HFS and HFO rats. HFO rats had higher levels of insulin resistance and hippocampal reactive oxygen species levels than HFS rats. Calorie restriction decreased metabolic disturbance and hippocampal oxidative stress but failed to attenuate cognitive decline in HFS and HFO rats. Exercise attenuated metabolic/hippocampal dysfunctions, resulting in improved cognition only in HFS rats. Combined therapies restored brain function, and cognitive function in HFS and HFO rats. Therefore, a combination of calorie restriction with exercise is probably the greatest lifestyle modification to diminish the brain pathologies and cognitive decline in obesity with or without estrogen deprivation.
Collapse
Affiliation(s)
- Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Kanokphong Suparan
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Diagnostic Sciences, Chiang Mai, Thailand.
| |
Collapse
|
8
|
Arbo BD, Schimith LE, Goulart dos Santos M, Hort MA. Repositioning and development of new treatments for neurodegenerative diseases: Focus on neuroinflammation. Eur J Pharmacol 2022; 919:174800. [DOI: 10.1016/j.ejphar.2022.174800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 11/03/2022]
|
9
|
Stute P, Wienges J, Koller AS, Giese C, Wesemüller W, Janka H, Baumgartner S. Cognitive health after menopause: Does menopausal hormone therapy affect it? Best Pract Res Clin Endocrinol Metab 2021; 35:101565. [PMID: 34538724 DOI: 10.1016/j.beem.2021.101565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dementia is a pandemic chronic non-communicable disease. 10 in 100 women above age 65 will be diagnosed with dementia, primarily Alzheimer's disease (AD). Apart from age and hereditary risk factors, there are multiple acquired risk and protective factors. So far, Menopausal Hormone Therapy (MHT) is not recommended as preventive measure. A systematic literature search on MHT and dementia risk and MHT and cognitive performance in demented women, respectively, was performed. Two recent meta-analyses identified 18 and 16 studies analyzing the impact of MHT on dementia/AD risk. Our systematic literature search identified eight additional original articles. The majority of studies found a risk reducing impact of MHT by 11-33%. However, results may vary depending on MHT type, age at initiation and study design. For example, the Women's Health Initiative Memory Study (WHIMS) reported an approximately 2-fold increased risk of dementia/Alzheimer's disease if MHT comprising conjugated equine estrogens and medroxyprogesterone acetate was initiated in predominantly comorbid postmenopausal women above age 65. In general, MHT displays a beneficial effect on several dementia risk factors and also augments some protective factors. Accordingly, clinicians can be reassured that MHT can be safely prescribed in the context of cognition in women free of dementia. However, MHT is not indicated for cognitive improvement in demented women. International scientific guidelines on MHT and dementia should consider incorporating most recent data.
Collapse
Affiliation(s)
- Petra Stute
- Department of Obstetrics and Gynecology, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland.
| | - Johanna Wienges
- Department of Obstetrics and Gynecology, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Anne-Sophie Koller
- Department of Obstetrics and Gynecology, Inselspital, University of Bern, Friedbühlstrasse 19, 3010, Bern, Switzerland
| | - Christina Giese
- Department of Obstetrics and Gynecology, Evangelisches Diakoniekrankenhaus Freiburg, Wirthstrasse 11, 79110, Freiburg Im Breisgau, Germany.
| | - Wiebke Wesemüller
- Aerzte Am Boulevard, Hauptstrasse 54, 8280, Kreuzlingen, Switzerland.
| | - Heidrun Janka
- Medical Library, University Library Bern, University of Bern, Baltzerstrasse 4, 3012, Bern, Switzerland.
| | - Sabrina Baumgartner
- Department of Obstetrics and Gynecology, University Hospital of Zürich, Frauenklinikstrasse 10, 8091, Zürich, Switzerland.
| |
Collapse
|
10
|
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: 23] [Impact Index Per Article: 5.8] [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
|
11
|
Cardinali CAEF, Martins YA, Torrão AS. Use of Hormone Therapy in Postmenopausal Women with Alzheimer's Disease: A Systematic Review. Drugs Aging 2021; 38:769-791. [PMID: 34342862 DOI: 10.1007/s40266-021-00878-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Around two-thirds of patients with Alzheimer's disease (AD) are women, which could be related to the depletion of female sexual hormones at menopause. The replacement of these hormones with hormone therapy (HT) to possibly decrease AD risk or treat AD patients has generated conflicting results in the literature. OBJECTIVE Our aim was to systematically review the relationship between HT use in postmenopausal women with AD and the risk of developing or treating AD symptoms. DATA SOURCES The PubMed, LILACS, Scopus, Scielo, and Web of Science databases were searched from January 1994 to December 2020 using the descriptors 'Alzheimer Disease OR Alzheimer's Disease' and 'Hormone Replacement Therapy OR Estrogen Replacement Therapy'. STUDY SELECTION Observational and controlled clinical trials including postmenopausal women diagnosed with AD and evaluating HT efficacy were eligible for inclusion. DATA EXTRACTION Extracted data comprise study design, covariates, inclusion criteria for sample selection, AD diagnosis criteria, biases, HT regimen, and cognitive measurement tools used. RESULTS Overall, 25 studies were selected. Among the 14 observational studies, 8 reported an improvement in cognitive function and a decrease in AD risk, especially in younger postmenopausal women. Five observational studies did not demonstrate any association between HT and AD, and one study reported an increase in AD risk, regardless of time of HT initiation. Of the 11 controlled clinical trials included, 7 showed an amelioration in cognitive function after HT. The remaining 4 trials saw no difference between HT and control. CONCLUSION Both observational and controlled clinical trials had methodological issues and discrepancies in inclusion criteria and HT protocols. These inconsistencies made it difficult to establish an association between HT and AD.
Collapse
Affiliation(s)
- Camila A E F Cardinali
- Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, São Paulo, 05508-900, Brazil
| | - Yandara A Martins
- Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, São Paulo, 05508-900, Brazil.
| | - Andréa S Torrão
- Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, São Paulo, 05508-900, Brazil
| |
Collapse
|
12
|
Zhu D, Montagne A, Zhao Z. Alzheimer's pathogenic mechanisms and underlying sex difference. Cell Mol Life Sci 2021; 78:4907-4920. [PMID: 33844047 PMCID: PMC8720296 DOI: 10.1007/s00018-021-03830-w] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
AD is a neurodegenerative disease, and its frequency is often reported to be higher for women than men: almost two-thirds of patients with AD are women. One prevailing view is that women live longer than men on average of 4.5 years, plus there are more women aged 85 years or older than men in most global subpopulations; and older age is the greatest risk factor for AD. However, the differences in the actual risk of developing AD for men and women of the same age is difficult to assess, and the findings have been mixed. An increasing body of evidence from preclinical and clinical studies as well as the complications in estimating incidence support the sex-specific biological mechanisms in diverging AD risk as an important adjunct explanation to the epidemiologic perspective. Although some of the sex differences in AD prevalence are due to differences in longevity, other distinct biological mechanisms increase the risk and progression of AD in women. These risk factors include (1) deviations in brain structure and biomarkers, (2) psychosocial stress responses, (3) pregnancy, menopause, and sex hormones, (4) genetic background (i.e., APOE), (5) inflammation, gliosis, and immune module (i.e., TREM2), and (6) vascular disorders. More studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD. This review presents the most recent data in sex differences in AD-the gateway to precision medicine, therefore, shaping expert perspectives, inspiring researchers to go in new directions, and driving development of future diagnostic tools and treatments for AD in a more customized way.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
- Neuroscience Graduate Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
13
|
Marzban M, Nabipour I, Farhadi A, Ostovar A, Larijani B, Darabi AH, Shabankari E, Gholizade M. Association between anemia, physical performance and cognitive function in Iranian elderly people: evidence from Bushehr Elderly Health (BEH) program. BMC Geriatr 2021; 21:329. [PMID: 34030664 PMCID: PMC8142505 DOI: 10.1186/s12877-021-02285-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
Background and objectives The present study aimed to investigate the relation between anemia and hemoglobin (Hgb) concentration, physical performance, and cognitive function in a large sample of Iranian elderly population. Methods Data were collected from Bushehr elderly health (BEH) program. A total of 3000 persons aged ≥60 years were selected through multistage random sampling. Hemoglobin values lower than 12 and 13 g/dL were considered as anemia for women and men, respectively. The cognitive function was measured using the Mini-cog test and Category fluency test (CFT), and the physical function was measured using handgrip strength (muscle strength), Relative handgrip strength (RHGS), and 4.57-m usual gait speed. Univariate and adjusted multivariate logistic regression and linear regression with Stata MP (version 15) were run, and a p-value of < 0.05 was used as statistically significant for all analyses. Results Among participants, 7.43% were anemic, and 115 (51.57%) simultaneously had anemia and cognitive disorder. There were significant associations between red blood cell count (RBC), hemoglobin (Hgb), platelet count (PLT), and hematocrit percentage (HCT) with cognitive impairment. Additionally, Hgb concentration was significantly associated with all physical measures (Mean handgrip, Relative handgrip, and usual gait speed) and late recall (mini-cog) among the whole participants. This association remained statistically significant after considering multi-cofounders. In contrast, after stratifying the participants by gender, the association between Hgb concentration and usual gait speed was decreased in both men and women; moreover, Hgb association with cognitive measures (category fluency test and late recall) was no longer significant (all p-values > 0.05). Conclusion There was a cross-sectional and significant association between anemia and functional variables (e.g., Relative and mean handgrip) in Iranian elderly population, whereas Semantic memory, Late recall, and walking were more affected by gender.
Collapse
Affiliation(s)
- Maryam Marzban
- Clinical Research Development Center, The Persian Gulf Martyrs, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Iraj Nabipour
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Akram Farhadi
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran. .,The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Afshin Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Darabi
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Elnaz Shabankari
- Department of Nutrition Science, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohamad Gholizade
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.,The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
14
|
GIPSON CD, BIMONTE-NELSON HA. Interactions between reproductive transitions during aging and addiction: promoting translational crosstalk between different fields of research. Behav Pharmacol 2021; 32:112-122. [PMID: 32960852 PMCID: PMC7965232 DOI: 10.1097/fbp.0000000000000591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Discovery of neural mechanisms underlying neuropsychiatric disorders within the aging and addiction fields has been a main focus of the National Institutes of Health. However, there is a dearth of knowledge regarding the biological interactions of aging and addiction, which may have important influences on progression of disease and treatment outcomes in aging individuals with a history of chronic drug use. Thus, there is a large gap in these fields of research, which has slowed progress in understanding and treating substance use disorders (SUDs) as well as age-related diseases, specifically in women who experience precipitous reproductive cycle transitions during aging. The goal of this review is to highlight overlap of SUDs and age-related processes with a specific focus on menopause and smoking, and identify critical gaps. We have narrowed the focus of the review to smoking, as the majority of findings on hormonal and aging influences on drug use have come from this area of research. Further, we highlight female-specific issues such as transitional menopause and exogenous estrogen use. These issues may impact drug use cessation as well as outcomes with aging and age-related neurodegenerative diseases in women. We first review clinical studies for smoking, normal aging, and pathological aging, and discuss the few aging-related studies taking smoking history into account. Conversely, we highlight the dearth of clinical smoking studies taking age as a biological variable into account. Preclinical and clinical literature show that aging, age-related pathological brain disease, and addiction engage overlapping neural mechanisms. We hypothesize that these putative drivers interact in meaningful ways that may exacerbate disease and hinder successful treatment outcomes in such comorbid populations. We highlight areas where preclinical studies are needed to uncover neural mechanisms in aging and addiction processes. Collectively, this review highlights the need for crosstalk between different fields of research to address medical complexities of older adults, and specifically women, who smoke.
Collapse
Affiliation(s)
- Cassandra D. GIPSON
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
- Arizona Alzheimer’s Consortium
| | | |
Collapse
|
15
|
Yare K, Woodward M. Hormone Therapy and Effects on Sporadic Alzheimer’s Disease in Postmenopausal Women: Importance of Nomenclature. J Alzheimers Dis 2020; 73:23-37. [DOI: 10.3233/jad-190896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Katrine Yare
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| | - Michael Woodward
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| |
Collapse
|
16
|
Yoon BK, Chin J, Kim JW, Shin MH, Ahn S, Lee DY, Seo SW, Na DL. Menopausal hormone therapy and mild cognitive impairment: a randomized, placebo-controlled trial. Menopause 2019; 25:870-876. [PMID: 29846283 DOI: 10.1097/gme.0000000000001140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of the study was to explore the therapeutic potential of menopausal hormone therapy (MHT) in women with mild cognitive impairment (MCI). METHODS Thirty-seven postmenopausal women (age range: 57-82 y) with multiple-domain, amnestic subtype MCI were randomly assigned to either placebo (n = 18) or MHT (n = 19) for 24 months (percutaneous estradiol [E2] gel [0.1%, 2 mg/d] and oral micronized progesterone [MP4] [100 mg/d]). All participants received donepezil, and apolipoprotein E genotype was determined. The primary endpoint was general cognitive function: Alzheimer's disease Assessment Scale, cognitive subscale, the Korean version of Mini-Mental State Examination (K-MMSE), and the Korean version of the Montreal Cognitive Assessment (MoCA_K) were performed in-person every 6 months. RESULTS Twenty-one participants (placebo 13, MHT 8) completed the trial (56.8%). Progression rates to dementia were 52.9% (9/17) in the placebo group and 44.4% (8/18) in the MHT group. Within-group analysis showed that all three tests significantly worsened during the trial in the placebo, but not the MHT groups. Analysis adjusted for ε4 allele demonstrated that MHT significantly reduced deterioration of MoCA_K score, a sensitive tool for assessing global cognition in MCI (P = 0.0261). Compared with the control group, both MoCA_K (P = 0.043; mean difference, 3.85; 95% CI, -0.46 to 8.16) and K-MMSE (P = 0.0319; mean difference, 3.26; 95% CI, 0.04-6.48) scores were significantly better at 24 months in the MHT group. CONCLUSIONS Long-term MHT using percutaneous E2 gel and oral MP4 might attenuate cognitive decline in postmenopausal women with MCI.
Collapse
Affiliation(s)
- Byung-Koo Yoon
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Juhee Chin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Won Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Hee Shin
- Department of Social and Preventive Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soohyun Ahn
- Statistics and Data Center, Samsung Medical Center, Seoul, Korea
| | - Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
17
|
Yoon BK. Risks and benefits of menopausal hormone therapy. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2019. [DOI: 10.5124/jkma.2019.62.3.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Byung-Koo Yoon
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Fisher DW, Bennett DA, Dong H. Sexual dimorphism in predisposition to Alzheimer's disease. Neurobiol Aging 2018; 70:308-324. [PMID: 29754747 DOI: 10.1016/j.neurobiolaging.2018.04.004] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and rate of symptom progression, but the mechanisms underlying this sexual divergence are unknown. Although some have suggested this difference in risk is a reflection of the known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities toward the disease. Although a number of potential risk factors have been hypothesized to mediate these differences, none have been definitively verified. In this review, we first summarize the epidemiologic studies of prevalence and incidence of AD among the sexes. Next, we discuss the most likely risk factors to date that interact with biological sex, including (1) genetic factors, (2) sex hormones (3) deviations in brain structure, (4) inflammation and microglia, and (5) and psychosocial stress responses. Overall, though differences in life span are likely to account for part of the divide between the sexes in AD prevalence, the abundance of preclinical and clinical evidence presented here suggests an increase in intrinsic AD risk for women. Therefore, future studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD pathogenesis in both sexes, with the eventual goal of sex-specific prevention and treatment strategies.
Collapse
Affiliation(s)
- Daniel W Fisher
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Hongxin Dong
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
19
|
Hestiantoro A, Wiwie M, Shadrina A, Ibrahim N, Purba JS. FSH to estradiol ratio can be used as screening method for mild cognitive impairment in postmenopausal women. Climacteric 2017; 20:577-582. [DOI: 10.1080/13697137.2017.1377696] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- A. Hestiantoro
- Department of Obstetrics and Gynecology, Reproductive Immunoendocrinology Division, Faculty of Medicine Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - M. Wiwie
- Department of Psychiatry, Faculty of Medicine Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - A. Shadrina
- Department of Obstetrics and Gynecology, Reproductive Immunoendocrinology Division, Faculty of Medicine Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - N. Ibrahim
- Department of Physiology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - J. S. Purba
- Department of Neurology, Faculty of Medicine Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
20
|
Neuroinflammation, immune system and Alzheimer disease: searching for the missing link. Aging Clin Exp Res 2017; 29:821-831. [PMID: 27718173 DOI: 10.1007/s40520-016-0637-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/29/2016] [Indexed: 12/19/2022]
Abstract
Due to an increasingly aging population, Alzheimer disease (AD) represents a crucial issue for the healthcare system because of its widespread prevalence and the burden of its care needs. Several hypotheses on AD pathogenesis have been proposed and current therapeutical strategies have shown limited effectiveness. In the last decade, more evidence has supported a role for neuroinflammation and immune system dysregulation in AD. It remains unclear whether astrocytes, microglia and immune cells influence disease onset, progression or both. Amyloid-β peptides that aggregate extracellularly in the typical neuritic plaques generate a constant inflammatory environment. This causes a prolonged activation of microglial and astroglial cells that potentiate neuronal damage and provoke the alteration of the blood brain barrier (BBB), damaging the permeability of blood vessels. Recent data support the role of the BBB as a link between neuroinflammation, the immune system and AD. Hence, a thorough investigation of the neuroinflammatory and immune system pathways that impact neurodegeneration and novel exciting findings such as microglia-derived microvesicles, inflammasomes and signalosomes will ultimately enhance our understanding of the pathological process. Eventually, we should proceed with caution in defining a causal or consequential role of neuroinflammation in AD, but rather focus on identifying its exact pathological contribution.
Collapse
|
21
|
Prakapenka AV, Bimonte-Nelson HA, Sirianni RW. Engineering poly(lactic-co-glycolic acid) (PLGA) micro- and nano-carriers for Controlled Delivery of 17β-Estradiol. Ann Biomed Eng 2017; 45:1697-1709. [PMID: 28634732 PMCID: PMC5599155 DOI: 10.1007/s10439-017-1859-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/23/2017] [Indexed: 12/29/2022]
Abstract
With menopause, circulating levels of 17β-estradiol (E2) markedly decrease. E2-based hormone therapy is prescribed to alleviate symptoms associated with menopause. E2 is also recognized for its beneficial effects in the central nervous system (CNS), such as enhanced cognitive function following abrupt hormonal loss associated with ovariectomy. For women with an intact uterus, an opposing progestogen component is required to decrease the risk of developing endometrial hyperplasia. While adding an opposing progestogen attenuates these detrimental effects on the uterus, it can attenuate the beneficial effects of E2 in the CNS. Poly(lactic-co-glycolic acid) (PLGA) micro- and nano- carriers (MNCs) have been heavily investigated for their ability to enhance the therapeutic activity of hydrophobic agents following exogenous administration, including E2. Multiple PLGA MNC formulation parameters, such as composition, molecular weight, and type of solvent used, can be altered to systematically manipulate the pharmacokinetic and pharmacodynamic profiles of encapsulated agents. Thus, there is an opportunity to enhance the therapeutic activity of E2 in the CNS through controlled delivery from PLGA MNCs. The aim of this review is to consider the fate of exogenously administered E2 and discuss how PLGA MNCs and route of administration can be used as strategies for controlled E2 delivery.
Collapse
Affiliation(s)
- Alesia V Prakapenka
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Rd., Phoenix, AZ, 85013, USA
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Rachael W Sirianni
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Rd., Phoenix, AZ, 85013, USA.
| |
Collapse
|
22
|
Osmanovic-Barilar J, Salkovic-Petrisi M. Evaluating the Role of Hormone Therapy in Postmenopausal Women with Alzheimer’s Disease. Drugs Aging 2016; 33:787-808. [DOI: 10.1007/s40266-016-0407-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
23
|
The Potential of Gonadal Hormone Signalling Pathways as Therapeutics for Dementia. J Mol Neurosci 2016; 60:336-348. [PMID: 27525638 DOI: 10.1007/s12031-016-0813-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022]
Abstract
Dementia is an ever-expanding problem facing an ageing society. Currently, there is a sharp paucity of treatment strategies. It has long been known that sex hormones, namely 17β-estradiol and testosterone, possess neuroprotective- and cognitive-enhancing qualities. However, certain lacunae in the knowledge underlying their molecular mechanisms have delayed their use as treatment strategies in dementia. With recent advancements in pharmacology and molecular biology, especially in the development of safer selective oestrogen receptor modulators and the recent discovery of the small-molecule brain-derived neurotrophic factor receptor agonist, 7,8-dihydroxyflavone, the exploitation of these signalling pathways for clinical use has become possible. This review aims to adumbrate the evidence and hurdles underscoring the use of sex hormones in the treatment of dementia as well as discussing some direction that is required to advance the translation of evidence into practise.
Collapse
|
24
|
Atwood CS, Bowen RL. A Unified Hypothesis of Early- and Late-Onset Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2016; 47:33-47. [PMID: 26402752 DOI: 10.3233/jad-143210] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Early-onset familial Alzheimer's disease (EOFAD) and late-onset sporadic AD (LOSAD) both follow a similar pathological and biochemical course that includes: neuron and synapse loss and dysfunction, microvascular damage, microgliosis, extracellular amyloid-β deposition, tau phosphorylation, formation of intracellular neurofibrillary tangles, endoreduplication and related cell cycle events in affected brain regions. Any mechanistic explanation of AD must accommodate these biochemical and neuropathological features for both forms of the disease. In this insight paper we provide a unifying hypothesis for EOFAD and LOSAD that proposes that the aberrant re-entry of terminally differentiated, post-mitotic neurons into the cell division cycle is a common pathway that explains both early and late-onset forms of AD. Cell cycle abnormalities appear very early in the disease process, prior to the appearance of plaques and tangles, and explain the biochemical (e.g. tau phosphorylation), neuropathological (e.g. neuron hypertrophy; polypoidy) and cognitive changes observed in EOFAD and LOSAD. Genetic mutations in AβPP, PSEN1, and PSEN2 that alter amyloid-β precursor protein and Notch processing drive reactivation of the cell cycle in EOFAD, while age-related reproductive endocrine dyscrasia that upregulates mitogenic TNF signaling and AβPP processing toward the amyloidogenic pathway drives reactivation of the cell cycle in LOSAD. In essence, AβPP and presenilin mutations initiate early, what endocrine dyscrasia initiates later: aberrant cell cycle re-entry of post-mitotic neurons leading to neurodegeneration and cognitive decline in AD. Inhibition of cell cycle re-entry in post-mitotic neurons may be a useful therapeutic strategy to prevent, slow or halt disease progression.
Collapse
Affiliation(s)
- Craig S Atwood
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI, USA.,School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | | |
Collapse
|
25
|
Hamson DK, Roes MM, Galea LAM. Sex Hormones and Cognition: Neuroendocrine Influences on Memory and Learning. Compr Physiol 2016; 6:1295-337. [DOI: 10.1002/cphy.c150031] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Atwood CS, Bowen RL. The endocrine dyscrasia that accompanies menopause and andropause induces aberrant cell cycle signaling that triggers re-entry of post-mitotic neurons into the cell cycle, neurodysfunction, neurodegeneration and cognitive disease. Horm Behav 2015; 76:63-80. [PMID: 26188949 PMCID: PMC4807861 DOI: 10.1016/j.yhbeh.2015.06.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 12/26/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Sex hormones are physiological factors that promote neurogenesis during embryonic and fetal development. During childhood and adulthood these hormones support the maintenance of brain structure and function via neurogenesis and the formation of dendritic spines, axons and synapses required for the capture, processing and retrieval of information (memories). Not surprisingly, changes in these reproductive hormones that occur with menopause and during andropause are strongly correlated with neurodegeneration and cognitive decline. In this connection, much evidence now indicates that Alzheimer's disease (AD) involves aberrant re-entry of post-mitotic neurons into the cell cycle. Cell cycle abnormalities appear very early in the disease, prior to the appearance of plaques and tangles, and explain the biochemical, neuropathological and cognitive changes observed with disease progression. Intriguingly, a recent animal study has demonstrated that induction of adult neurogenesis results in the loss of previously encoded memories while decreasing neurogenesis after memory formation during infancy mitigated forgetting. Here we review the biochemical, epidemiological and clinical evidence that alterations in sex hormone signaling associated with menopause and andropause drive the aberrant re-entry of post-mitotic neurons into an abortive cell cycle that leads to neurite retraction, neuron dysfunction and neuron death. When the reproductive axis is in balance, gonadotropins such as luteinizing hormone (LH), and its fetal homolog, human chorionic gonadotropin (hCG), promote pluripotent human and totipotent murine embryonic stem cell and neuron proliferation. However, strong evidence supports menopausal/andropausal elevations in the LH:sex steroid ratio as driving aberrant mitotic events. These include the upregulation of tumor necrosis factor; amyloid-β precursor protein processing towards the production of mitogenic Aβ; and the activation of Cdk5, a key regulator of cell cycle progression and tau phosphorylation (a cardinal feature of both neurogenesis and neurodegeneration). Cognitive and biochemical studies confirm the negative consequences of a high LH:sex steroid ratio on dendritic spine density and human cognitive performance. Prospective epidemiological and clinical evidence in humans supports the premise that rebalancing the ratio of circulating gonadotropins:sex steroids reduces the incidence of AD. Together, these data support endocrine dyscrasia and the subsequent loss of cell cycle control as an important etiological event in the development of neurodegenerative diseases including AD, stroke and Parkinson's disease.
Collapse
Affiliation(s)
- Craig S Atwood
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA; Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI 53705, USA; School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, 6027 WA, Australia.
| | - Richard L Bowen
- OTB Research, 217 Calhoun St, Unit 1, Charleston, SC 29401, USA
| |
Collapse
|
27
|
Burnham VL, Thornton JE. Luteinizing hormone as a key player in the cognitive decline of Alzheimer's disease. Horm Behav 2015; 76:48-56. [PMID: 26031357 DOI: 10.1016/j.yhbeh.2015.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/10/2015] [Accepted: 05/25/2015] [Indexed: 12/30/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Alzheimer's disease is one of the most prevalent and costly neurological diseases in the world. Although decades of research have focused on understanding Alzheimer's disease pathology and progression, there is still a great lack of clinical treatments for those who suffer from it. One of the factors most commonly associated with the onset of Alzheimer's disease is a decrease in levels of gonadal hormones, such as estrogens and androgens. Despite the correlational and experimental data which support the role of these hormones in the etiology of Alzheimer's disease, clinical trials involving their reintroduction through hormone therapy have had varied results and these gonadal hormones often have accompanying health risks. More recently, investigation has turned toward other hormones in the hypothalamic-pituitary-gonadal axis that are disrupted by age-related decreases in gonadal hormones. Specifically, luteinizing hormone, which is increased with age in both men and women (in response to removal of negative feedback), has surfaced as a potentially powerful player in the risk and onset of Alzheimer's disease. Mounting evidence in basic research and epidemiological studies supports the role of elevated luteinizing hormone in exacerbating age-related cognitive decline in both males and females. This review summarizes the recent developments involving luteinizing hormone in increasing the cognitive deficits and molecular pathology characteristic of Alzheimer's disease.
Collapse
Affiliation(s)
- Veronica L Burnham
- Department of Neuroscience, Oberlin College, 119 Woodland St, Oberlin, OH 44074, USA
| | - Janice E Thornton
- Department of Neuroscience, Oberlin College, 119 Woodland St, Oberlin, OH 44074, USA.
| |
Collapse
|
28
|
Pooley AE, Luong M, Hussain A, Nathan BP. Neurite outgrowth promoting effect of 17-β estradiol is mediated through estrogen receptor alpha in an olfactory epithelium culture. Brain Res 2015. [PMID: 26206299 DOI: 10.1016/j.brainres.2015.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Olfactory deficits are observed early in the course of chronic neurological disorders including Alzheimer's disease (AD). Estrogen treatment in post-menopausal women reduced the incidence of olfactory dysfunction, raising the possibility that estrogen treatment can cure olfactory deficits in preclinical stages of AD. In this study, we examined the estradiol׳s effects on neurite outgrowth in explant cultures of mouse olfactory epithelium (OE). We found that neurons in OE cultures treated with 100 pM 17-β estradiol (estradiol) had significantly longer neurite outgrowth than cultures treated with ethanol alone (vehicle). The OE neurons expressed estrogen receptors alpha (ERα) and ER beta (ERβ). Estrogen treatment upregulated both ERα and ERβ expression in OE culture. Treatment of OE cultures with propyl pyrazole triol (PPT), a selective agonist for ERα increased neurite outgrowth to comparable extent as estradiol treatment. In contrast, 2,3-bis-4-hydroxyphenyl (DPN), a specific agonist for ERβ, had no effect on neurite outgrowth. Furthermore, estradiol treatment increased neurite outgrowth in OE cultures derived from ERβ-deficient/knockout mice and wild-type littermates, but not in ERα-deficient/knockout mice. These data suggest that ERα mediates the neurite outgrowth promoting effects of estradiol in OE cultures. We propose that olfactory dysfunction in chronic neurological disorders, where estrogen deficiency is a risk factor, is an indicator of compromised axonal regeneration of olfactory sensory neurons.
Collapse
Affiliation(s)
- Apryl E Pooley
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Minh Luong
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Aseem Hussain
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Britto P Nathan
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States.
| |
Collapse
|
29
|
Henderson VW. Alzheimer's disease: review of hormone therapy trials and implications for treatment and prevention after menopause. J Steroid Biochem Mol Biol 2014; 142:99-106. [PMID: 23727128 PMCID: PMC3830600 DOI: 10.1016/j.jsbmb.2013.05.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 05/08/2013] [Accepted: 05/18/2013] [Indexed: 12/27/2022]
Abstract
Hormonal changes associated with the menopausal transition and postmenopause have the potential to influence processes linked to Alzheimer's disease symptoms and pathogenesis, but effects of menopause on Alzheimer risk can be addressed only indirectly. Nine randomized clinical trials of estrogen-containing hormone therapy in Alzheimer's disease patients were identified by a systematic literature search. Findings suggest that hormone therapy does not improve cognitive symptoms of women with Alzheimer's disease. No clinical trials of hormone therapy address Alzheimer prevention, but one clinical trial provides moderate evidence that continuous, combined estrogen plus progestogen initiated at age 65 years or older increases the risk of dementia. The timing, or critical window, hypothesis suggests that hormone therapy initiated at a younger age in closer temporal proximity to menopause may reduce the risk of Alzheimer's disease. This hypothesis is supported by observational research but is not addressed by clinical trial data. Unrecognized confounding is of concern in interpreting observational results, and research that helps resolve this issue will have important public health implications. Well-designed cohort studies, convergent evidence from appropriate laboratory models, and long-term clinical trials using surrogate biomarkers of brain function and neural pathology could provide relevant answers. Other estrogenic compounds are of theoretical interest with respect to Alzheimer treatment and risk. Effects of selective estrogen receptor modulators such as raloxifene may differ from those of estrogens; potential effects of phytoestrogens are not well studied. This article is part of a Special Issue entitled 'Menopause'.
Collapse
Affiliation(s)
- Victor W Henderson
- Department of Health Research & Policy (Epidemiology), Stanford University, Stanford, CA, USA; Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
30
|
Fischer B, Gleason C, Asthana S. Effects of hormone therapy on cognition and mood. Fertil Steril 2014; 101:898-904. [PMID: 24680649 DOI: 10.1016/j.fertnstert.2014.02.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/10/2014] [Accepted: 02/12/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Results of the Women's Health Initiative (WHI) and Women's Health Initiative Memory Study (WHIMS) suggested that hormone therapy (HT) may be detrimental to cognitive health. This article reviews clinical studies that address issues relevant to those results. DESIGN Literature review. INTERVENTION(S) A search of Pubmed and Web of Science was conducted using the search terms HT and cognition, HT and mood. Clinical and observational studies were selected if they were published after the year 2000. Theories of HT mechanisms of action, pharmacology, biology, and observational and clinical trials are discussed. RESULT(S) Although observational and clinical trials show conflicting findings, methodologic considerations must be acknowledged. HT formulation and dose, route of administration, timing of initiation, length of treatment, and health of participants all contribute to inconsistencies in results. Transdermal estradiol and micronized progesterone administered at time of menopause are generally associated with cognitive and affective benefit. CONCLUSION(S) At the present time, results from existing studies are equivocal regarding the benefits of HT on cognition and affect. Future studies, such as the Kronos Early Estrogen Prevention Study (KEEPS), should address methodologic inconsistencies to provide clearer answers to this important question.
Collapse
Affiliation(s)
- Barbara Fischer
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Affairs Hospital, Madison, Wisconsin.
| | - Carey Gleason
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Affairs Hospital, Madison, Wisconsin; Wisconsin Alzheimer's Disease Research Center (ADRC), Madison, Wisconsin; Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Sanjay Asthana
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Affairs Hospital, Madison, Wisconsin; Wisconsin Alzheimer's Disease Research Center (ADRC), Madison, Wisconsin; Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
31
|
Moreira PI, Zhu X, Nunomura A, Smith MA, Perry G. Therapeutic options in Alzheimer’s disease. Expert Rev Neurother 2014; 6:897-910. [PMID: 16784412 DOI: 10.1586/14737175.6.6.897] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) places an enormous burden on individuals, families and society. Consequently, a tremendous effort is being devoted to the development of drugs that prevent or delay neurodegeneration. Current pharmacological treatments are based on the use of acetylcholinesterase inhibitors or memantine, a N-methyl-D-aspartate channel blocker. However, new therapeutic approaches, including those more closely targeted to the pathogenesis of the disease, are being developed. These potentially disease-modifying therapeutics include secretase inhibitors, cholesterol-lowering drugs, amyloid-beta immunotherapy, nonsteroidal anti-inflammatory drugs, hormonal modulation and the use of antioxidants. The possibility that oxidative stress is a primary event in AD indicates that antioxidant-based therapies are perhaps the most promising weapons against this devastating neurodegenerative disorder.
Collapse
Affiliation(s)
- Paula I Moreira
- Case Western Reserve University, Department of Pathology, Cleveland, Ohio 44106, USA.
| | | | | | | | | |
Collapse
|
32
|
Rettberg JR, Yao J, Brinton RD. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol 2014; 35:8-30. [PMID: 23994581 PMCID: PMC4024050 DOI: 10.1016/j.yfrne.2013.08.001] [Citation(s) in RCA: 350] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/09/2013] [Accepted: 08/10/2013] [Indexed: 01/12/2023]
Abstract
Estrogen is a fundamental regulator of the metabolic system of the female brain and body. Within the brain, estrogen regulates glucose transport, aerobic glycolysis, and mitochondrial function to generate ATP. In the body, estrogen protects against adiposity, insulin resistance, and type II diabetes, and regulates energy intake and expenditure. During menopause, decline in circulating estrogen is coincident with decline in brain bioenergetics and shift towards a metabolically compromised phenotype. Compensatory bioenergetic adaptations, or lack thereof, to estrogen loss could determine risk of late-onset Alzheimer's disease. Estrogen coordinates brain and body metabolism, such that peripheral metabolic state can indicate bioenergetic status of the brain. By generating biomarker profiles that encompass peripheral metabolic changes occurring with menopause, individual risk profiles for decreased brain bioenergetics and cognitive decline can be created. Biomarker profiles could identify women at risk while also serving as indicators of efficacy of hormone therapy or other preventative interventions.
Collapse
Affiliation(s)
- Jamaica R Rettberg
- Neuroscience Department, University of Southern California, Los Angeles, CA 90033, United States
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, United States
| | - Roberta Diaz Brinton
- Neuroscience Department, University of Southern California, Los Angeles, CA 90033, United States; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, United States; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
33
|
Tschanz JT, Norton MC, Zandi PP, Lyketsos CG. The Cache County Study on Memory in Aging: factors affecting risk of Alzheimer's disease and its progression after onset. Int Rev Psychiatry 2013; 25:673-85. [PMID: 24423221 PMCID: PMC4089882 DOI: 10.3109/09540261.2013.849663] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Cache County Study on Memory in Aging is a longitudinal, population-based study of Alzheimer's disease (AD) and other dementias. Initiated in 1995 and extending to 2013, the study has followed over 5,000 elderly residents of Cache County, Utah (USA) for over twelve years. Achieving a 90% participation rate at enrolment, and spawning two ancillary projects, the study has contributed to the literature on genetic, psychosocial and environmental risk factors for AD, late-life cognitive decline, and the clinical progression of dementia after its onset. This paper describes the major study contributions to the literature on AD and dementia.
Collapse
Affiliation(s)
- JoAnn T. Tschanz
- Department of Psychology, Utah State University,Center for Epidemiologic Studies, Utah State University
| | - Maria C. Norton
- Center for Epidemiologic Studies, Utah State University,Department of Family, Consumer & Human Development, Utah State University
| | - Peter P. Zandi
- Bloomberg School of Public Health, Johns Hopkins University
| | | |
Collapse
|
34
|
Acosta JI, Hiroi R, Camp BW, Talboom JS, Bimonte-Nelson HA. An update on the cognitive impact of clinically-used hormone therapies in the female rat: models, mazes, and mechanisms. Brain Res 2013; 1514:18-39. [PMID: 23333453 PMCID: PMC3739440 DOI: 10.1016/j.brainres.2013.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/09/2013] [Indexed: 01/05/2023]
Abstract
In women, ovarian hormone loss associated with menopause has been related to cognitive decline. Hormone therapy (HT) may ameliorate some of these changes. Understanding the cognitive impact of female steroids, including estrogens, progestogens, and androgens, is key to discovering treatments that promote brain health in women. The preclinical literature has presented elegant and methodical experiments allowing a better understanding of parameters driving the cognitive consequences of ovarian hormone loss and HT. Animal models have been a valuable tool in this regard, and will be vital to future discoveries. Here, we provide an update on the literature evaluating the impact of female steroid hormones on cognition, and the putative mechanisms mediating these effects. We focus on preclinical work that was done with an eye toward clinical realities. Parameters that govern the cognitive efficacy of HT, from what we know thus far, include but are not limited to: type, dose, duration, and route of HT, age at HT initiation, timing of HT relative to ovarian hormone loss, memory type examined, menopause history, and hormone receptor status. Researchers have identified intricate relationships between some of these factors by studying their individual effects on cognition. As of late, there is increased focus on studying interactions between these variables as well as multiple hormone types when administered concomitantly. This is key to translating preclinical data to the clinic, wherein women typically have concurrent exposure to endogenous ovarian hormones as well as exogenous combination HTs, which include both estrogens and progestins. Gains in understanding the parameters of HT effects on cognition provide exciting novel avenues that can inform clinical treatments, eventually expanding the window of opportunity to optimally enhance cognition and brain health in aging women. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- J I Acosta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | |
Collapse
|
35
|
Mennenga S, Bimonte-Nelson H. Translational cognitive endocrinology: designing rodent experiments with the goal to ultimately enhance cognitive health in women. Brain Res 2013; 1514:50-62. [PMID: 23391594 PMCID: PMC3936018 DOI: 10.1016/j.brainres.2013.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/12/2013] [Indexed: 02/08/2023]
Abstract
Understanding the cognitive impact of endogenously derived, and exogenously administered, hormone alterations is necessary for developing hormone treatments to support healthy brain function in women, especially during aging. The increasing number of studies in the burgeoning area of translational cognitive neuroendocrinology has revealed numerous factors that influence the extent and direction of female steroid effects on cognition. Here, we discuss the decision processes underlying the design of rodent hormone manipulation experiments evaluating learning and memory. It is noted that even when beginning with a clear hypothesis-driven question, there are numerous factors to consider in order to solidify a sound experimental design that will yield clean, interpretable results. Decisions and considerations include: age of animals at hormone administration and test, ovariectomy implementation, when to administer hormones relative to ovarian hormone loss, how and whether to monitor the estrous cycle if animals are ovary-intact, dose of hormone, administration route of hormone, hormone treatment confirmation protocols, handling procedures required for hormone administration and treatment confirmation, cognitive domains to be tested and which mazes should be utilized to test these cognitive domains, and control measures to be used. A balanced view of optimal design and realistic experimental practice and protocol is presented. The emerging results from translational cognitive neuroendocrinology studies have been diverse, but also enlightening and exciting as we realize the broad scope and powerful nature of ovarian hormone effects on the brain and its function. We must design, implement, and interpret hormone and cognition experiments with sensitivity to these tenets, acknowledging and respecting the breadth and depth of the impact gonadal hormones have on brain functioning and its rich plasticity. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- S.E. Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer’s Consortium, USA
| | - H.A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer’s Consortium, USA
| |
Collapse
|
36
|
Trippier PC, Labby KJ, Hawker DD, Mataka JJ, Silverman RB. Target- and mechanism-based therapeutics for neurodegenerative diseases: strength in numbers. J Med Chem 2013; 56:3121-47. [PMID: 23458846 PMCID: PMC3637880 DOI: 10.1021/jm3015926] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of new therapeutics for the treatment of neurodegenerative pathophysiologies currently stands at a crossroads. This presents an opportunity to transition future drug discovery efforts to target disease modification, an area in which much still remains unknown. In this Perspective we examine recent progress in the areas of neurodegenerative drug discovery, focusing on some of the most common targets and mechanisms: N-methyl-d-aspartic acid (NMDA) receptors, voltage gated calcium channels (VGCCs), neuronal nitric oxide synthase (nNOS), oxidative stress from reactive oxygen species, and protein aggregation. These represent the key players identified in neurodegeneration and are part of a complex, intertwined signaling cascade. The synergistic delivery of two or more compounds directed against these targets, along with the design of small molecules with multiple modes of action, should be explored in pursuit of more effective clinical treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul C. Trippier
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Kristin Jansen Labby
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Dustin D. Hawker
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Jan J. Mataka
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Richard B. Silverman
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA
| |
Collapse
|
37
|
Hogervorst E. Estrogen and the brain: does estrogen treatment improve cognitive function? ACTA ACUST UNITED AC 2013; 19:6-19. [PMID: 27951525 DOI: 10.1177/1754045312473873] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2012] [Indexed: 11/15/2022]
Abstract
In this paper we describe potential reasons for the discrepancies between data from basic sciences and observational studies and those of large treatment studies investigating the association between brain function and sex steroids. Observational studies which often showed positive associations between hormone use and cognition can be affected by 'recall bias' and 'healthy user bias', while outcomes of treatment studies were hypothesized to be modified by age at treatment, age at or type of menopause, health status, addition of a progestogen or type of estrogen treatment. However, meta-analyses of data from treatment studies negate many of these hypotheses showing at best mainly short-term (up to 6 months) positive effects of estrogen treatment on cognition regardless of age. This positive effect may reverse, particularly in older women with prolonged hormone treatment, which was predominantly seen after addition of progestogen. Medroxyprogesterone acetate seemed to have worse effects on cognition than other types of progestogen in these long-term studies. Estradiol with or without a progestogen was three times more likely to have positive effects on cognition than conjugated equine estrogens. However, two-thirds of studies showed no associations at all which may be an underestimate given the possibility of publication bias. We briefly review alternative treatments, such as testosterone and soy-derived supplements, but currently insufficient data are available for conclusive comments. Women who have undergone surgical menopause or who undergo natural menopause before age 47 may benefit most from hormone treatment and a special case may need to be made for this group. Long-term safety studies for this group are urgently needed.
Collapse
|
38
|
Associations between sex hormones and cognitive and neuropsychiatric manifestations in vascular dementia (VaD). Arch Gerontol Geriatr 2013; 56:85-90. [DOI: 10.1016/j.archger.2012.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 09/27/2012] [Accepted: 10/02/2012] [Indexed: 11/18/2022]
|
39
|
Tan JW, Tham CL, Israf DA, Lee SH, Kim MK. Neuroprotective effects of biochanin A against glutamate-induced cytotoxicity in PC12 cells via apoptosis inhibition. Neurochem Res 2012; 38:512-8. [PMID: 23224778 DOI: 10.1007/s11064-012-0943-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/02/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023]
Abstract
L-Glutamate plays a crucial role in neuronal cell death, which is known to be associated with various neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases. In this study, we investigated the protective effects of biochanin A, a phytoestrogen compound found mainly in Trifolium pratense, against L-glutamate-induced cytotoxicity in a PC12 cell line. Exposure of the cells to 10 mM L-glutamate was found to significantly increase cell viability loss and apoptosis, whereas pretreatment with various concentrations of biochanin A attenuated the cytotoxic effects of L-glutamate. Specifically, the pretreatment led to not only decreases in the release of lactate dehydrogenase, the number of apoptotic cells, and the activity of caspase-3 but also an increase in the total glutathione level in the L-glutamate-treated PC12 cells. These results indicate that biochanin A may be able to exert neuroprotective effects against L-glutamate-induced cytotoxicity. Furthermore, our findings also imply that biochanin A may act as an antiapoptotic agent in order to perform its protective function.
Collapse
Affiliation(s)
- Ji Wei Tan
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | | | | | | | | |
Collapse
|
40
|
Clark I, Atwood C, Bowen R, Paz-Filho G, Vissel B. Tumor necrosis factor-induced cerebral insulin resistance in Alzheimer's disease links numerous treatment rationales. Pharmacol Rev 2012; 64:1004-26. [PMID: 22966039 DOI: 10.1124/pr.112.005850] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The evident limitations of the amyloid theory of the pathogenesis of Alzheimer's disease are increasingly putting alternatives in the spotlight. We argue here that a number of independently developing approaches to therapy-including specific and nonspecific anti-tumor necrosis factor (TNF) agents, apolipoprotein E mimetics, leptin, intranasal insulin, the glucagon-like peptide-1 mimetics and glycogen synthase kinase-3 (GSK-3) antagonists-are all part of an interlocking chain of events. All these approaches inform us that inflammation and thence cerebral insulin resistance constitute the pathway on which to focus for a successful clinical outcome in treating this disease. The key link in this chain presently absent is a recognition by Alzheimer's research community of the long-neglected history of TNF induction of insulin resistance. When this is incorporated into the bigger picture, it becomes evident that the interventions we discuss are not competing alternatives but equally valid approaches to correcting different parts of the same pathway to Alzheimer's disease. These treatments can be expected to be at least additive, and conceivably synergistic, in effect. Thus the inflammation, insulin resistance, GSK-3, and mitochondrial dysfunction hypotheses are not opposing ideas but stages of the same fundamental, overarching, pathway of Alzheimer's disease pathogenesis. The insight this provides into progenitor cells, including those involved in adult neurogenesis, is a key part of this approach. This pathway also has therapeutic implications for other circumstances in which brain TNF is pathologically increased, such as stroke, traumatic brain injury, and the infectious disease encephalopathies.
Collapse
Affiliation(s)
- Ian Clark
- Division of Medical Science and Biochemistry, Research School of Biology, Australian National University, Canberra ACT, Australia.
| | | | | | | | | |
Collapse
|
41
|
Fukui H, Arai A, Toyoshima K. Efficacy of music therapy in treatment for the patients with Alzheimer's disease. Int J Alzheimers Dis 2012; 2012:531646. [PMID: 23056992 PMCID: PMC3463939 DOI: 10.1155/2012/531646] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 08/21/2012] [Accepted: 08/26/2012] [Indexed: 12/25/2022] Open
Abstract
We report that music therapy is effective in the treatment of Alzheimer's disease. We found that the secretion of 17β-estradiol and testosterone, hormones that are supposed to have preventive effects on Alzheimer's disease, is significantly increased by music therapy. During the sessions, patients with Alzheimer's disease were allowed to listen to music and songs with verbal contact from the therapist. It was found that problematic behaviors such as poriomania (fugue) had decreased. Music therapy has the potential as an alternative treatment for adverse hormone replacement therapy.
Collapse
Affiliation(s)
- H. Fukui
- Department of Education, Nara University of Education, Takabatake, Nara 630-8528, Japan
| | - A. Arai
- Ongakunomori Nonprofit Organization, 1-12 Kobocho, Saidaiji, Nara 631-0827, Japan
| | - K. Toyoshima
- Department of Education, Nara University of Education, Takabatake, Nara 630-8528, Japan
| |
Collapse
|
42
|
Clark IA, Atwood CS. Is TNF a link between aging-related reproductive endocrine dyscrasia and Alzheimer's disease? J Alzheimers Dis 2012; 27:691-9. [PMID: 21891866 DOI: 10.3233/jad-2011-110887] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This commentary addresses a novel mechanism by which aging-related changes in reproductive hormones could mediate their action in the brain. It presents the evidence that dyotic endocrine signals modulate the expression of tumor necrosis factor (TNF) and related cytokines, and that these cytokines are a functionally important downstream link mediating neurodegeneration and dysfunction. This convergence of dyotic signaling on TNF-mediated degeneration and dysfunction has important implications for understanding the pathophysiology of AD, stroke, and traumatic brain disease, and also for the treatment of these diseases.
Collapse
Affiliation(s)
- Ian A Clark
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | | |
Collapse
|
43
|
Abstract
Extant research findings allow several conclusions regarding the relationship between estrogen and cognitive functioning across the female life span. First, performance on tests of verbal memory fluctuates in concert with physiological changes in ovarian hormone production during the menstrual cycle and during pregnancy and the postpartum period. Estrogen therapy (ET) prevents the decrease in verbal memory when administered immediately following the surgical removal of both ovaries in premenopausal women. Some, but relatively little evidence is available to support the idea that ET, initiated at the time of a natural or a surgical menopause for a few years, may protect against cognitive decline 30 years later and more research in this area is urgently needed. Finally, the evidence to date strongly suggests that the initiation of ET decades after the menopause has occurred does not protect against cognitive decline or dementia. Taken together, these findings support the so-called "window of opportunity" hypothesis which holds that ET will be neuroprotective only when administered closely in time to a natural or surgical menopause.
Collapse
Affiliation(s)
- Barbara B Sherwin
- Department of Psychology, McGill University, Montreal, Quebec, Canada H3A 1B1.
| |
Collapse
|
44
|
Barron AM, Pike CJ. Sex hormones, aging, and Alzheimer's disease. Front Biosci (Elite Ed) 2012. [PMID: 22201929 DOI: 10.2741/434] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A promising strategy to delay and perhaps prevent Alzheimer's disease (AD) is to identify the age-related changes that put the brain at risk for the disease. A significant normal age change known to result in tissue-specific dysfunction is the depletion of sex hormones. In women, menopause results in a relatively rapid loss of estradiol and progesterone. In men, aging is associated with a comparatively gradual yet significant decrease in testosterone. We review a broad literature that indicates age-related losses of estrogens in women and testosterone in men are risk factors for AD. Both estrogens and androgens exert a wide range of protective actions that improve multiple aspects of neural health, suggesting that hormone therapies have the potential to combat AD pathogenesis. However, translation of experimental findings into effective therapies has proven challenging. One emerging treatment option is the development of novel hormone mimetics termed selective estrogen and androgen receptor modulators. Continued research of sex hormones and their roles in the aging brain is expected to yield valuable approaches to reducing the risk of AD.
Collapse
Affiliation(s)
- Anna M Barron
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | | |
Collapse
|
45
|
Verbal episodic memory and endogenous estradiol: an association in patients with mild cognitive impairment and Alzheimer's disease. Curr Gerontol Geriatr Res 2011; 2011:673012. [PMID: 22216027 PMCID: PMC3246697 DOI: 10.1155/2011/673012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 09/05/2011] [Accepted: 09/19/2011] [Indexed: 12/02/2022] Open
Abstract
In the continuum of patients with Alzheimer's disease (AD), mild cognitive impairment (MCI), and normal controls, a possible association of verbal memory and endogenous estradiol (E2) levels was investigated. Verbal episodic memory was measured with a german version of the California verbal memory test (CVLT). Results were controlled for apolipoprotein E (ApoE) phenotype. We studied 37 controls, 32 MCIs and 117 ADs. Groups differed in all trials of the CVLT (P < 0.001) and in E2 levels (P < 0.001). E2 levels differed significantly between groups only among females (P < 0.001). In females correcting for age and ApoE, there was an overall correlation between CVLT delayed recall and level of E2 (P = 0.025). Stepwise regression analyses found E2 level to be a significant predictor for CVLT delayed recall (P < 0.001). It may be concluded that lower E2 levels occur more in the course of the disease than may be considered as a risk factor per se.
Collapse
|
46
|
Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Effects of estrogen in preventing neuronal insulin resistance in hippocampus of obese rats are different between genders. Life Sci 2011; 89:702-7. [DOI: 10.1016/j.lfs.2011.08.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 06/27/2011] [Accepted: 08/09/2011] [Indexed: 11/16/2022]
|
47
|
Cognitive response to estradiol in postmenopausal women is modified by high cortisol. Neurobiol Aging 2011; 33:829.e9-20. [PMID: 21855173 DOI: 10.1016/j.neurobiolaging.2011.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/28/2011] [Accepted: 07/07/2011] [Indexed: 01/11/2023]
Abstract
Estradiol has potent favorable effects on brain function and behavior in animals while in human trials, the results are inconsistent. A number of potential mediating variables influencing response to estradiol have been proposed to account for this variability, 1 of which includes stress. We conducted a placebo-controlled study to examine joint and independent effects of estradiol and elevated levels of the stress hormone cortisol on cognition and biomarkers of aging and neurodegenerative disease. Thirty-nine healthy postmenopausal women (56-84 years) received 0.10 mg/dL of transdermal 17β-estradiol (E2) or placebo for 8 weeks. During the last 4 days of the trial, subjects also received 90 mg/day (30 mg 3×/day) of oral hydrocortisone (CORT) to induce stress-level elevations in cortisol, or a matched placebo. The 4 groups thus included placebo (placebo patch/placebo pill), CORT-alone (placebo patch/hydrocortisone), E2-alone (estradiol patch/placebo pill), and E2+CORT (estradiol patch/hydrocortisone). Eight weeks of E2 increased plasma estradiol by 167%, and 4 days of CORT increased plasma cortisol by 119%. Overall, E2 had favorable effects on verbal memory (p = 0.03), working memory (p = 0.02), and selective attention (p = 0.04), and the magnitude of these effects was attenuated for E2+CORT. E2-alone and E2+CORT had opposing effects on plasma levels of the amyloid-β (Aβ) biomarker (Aβ40/42 ratio, p < 0.05), with the more favorable response observed for E2-alone. CORT-induced increases in insulin-like growth factor-1 were blunted by E2 coadministration. Our findings indicate that cognitive and physiological responses to estradiol are adversely affected by elevated stress hormone levels of cortisol in healthy postmenopausal women.
Collapse
|
48
|
Di Stefano A, Iannitelli A, Laserra S, Sozio P. Drug delivery strategies for Alzheimer's disease treatment. Expert Opin Drug Deliv 2011; 8:581-603. [PMID: 21391862 DOI: 10.1517/17425247.2011.561311] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Current Alzheimer's disease (AD) therapy is based on the administration of the drugs donepezil, galantamine, rivastigmine and memantine. Until disease-modifying therapies become available, further research is needed to develop new drug delivery strategies to ensure ease of administration and treatment persistence. AREAS COVERED In addition to the conventional oral formulations, a variety of drug delivery strategies applied to the treatment of AD are reviewed in this paper, with a focus on strategies leading to simplified dosage regimens and to providing new pharmacological tools. Alternatives include extended release, orally disintegrating or sublingual formulations, intranasal or short- and long-acting intramuscular or transdermal forms, and nanotechnology-based delivery systems. EXPERT OPINION The advent of new research on molecular mechanisms of AD pathogenesis has outlined new strategies for therapeutic intervention; these include the stimulation of α-secretase cleavage, the inhibition of γ-secretase activity, the use of non-steroidal anti-inflammatory drugs, neuroprotection based on antioxidant therapy, the use of estrogens, NO synthetase inhibitors, and natural agents such as polyphenols. Unfortunately, these compounds might not help patients with end stage AD, but might hopefully slow or stop the disease process in its early stage. Nanotechnologies may prove to be a promising contribution in future AD drug delivery strategies, in particular drug carrier nano- or microsystems, which can limit the side effects of anti-Alzheimer drugs.
Collapse
Affiliation(s)
- Antonio Di Stefano
- G d'Annunzio University, School of Pharmacy, Department of Drug Sciences, Via dei Vestini 31, 66100 Chieti, Italy.
| | | | | | | |
Collapse
|
49
|
Quinlan MG, Duncan A, Loiselle C, Graffe N, Brake WG. Latent inhibition is affected by phase of estrous cycle in female rats. Brain Cogn 2011; 74:244-8. [PMID: 20817338 DOI: 10.1016/j.bandc.2010.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 08/06/2010] [Accepted: 08/11/2010] [Indexed: 11/28/2022]
Abstract
Estrogen has been shown to have a strong modulatory influence on several types of cognition in both women and female rodents. Latent inhibition is a task in which pre-exposure to a neutral stimulus, such as a tone, later impedes the association of that stimulus with a particular consequence, such as a shock. Previous work from our lab demonstrates that high levels of estradiol (E2) administered to ovariectomized (OVX) female rats abolishes latent inhibition when compared to female rats with low levels of E2 or male rats. To determine if this E2-induced impairment also occurs with the natural variations of ovarian hormones during the estrous cycle, this behavior was investigated in cycling female rats. In addition, pre-pubertal male and female rats were also tested in this paradigm to determine if the previously described sex differences are activational or organizational in nature. In a latent inhibition paradigm using a tone and a shock, adult rats were conditioned during different points of the estrous cycle. Rats conditioned during proestrus, a period of high E2 levels, exhibited attenuated latent inhibition when compared to rats conditioned during estrus or metestrus, periods associated with low levels of E2. Moreover, this effect is not seen until puberty indicating it is dependent on the surge of hormones at puberty. This study confirms recent findings that high E2 interferes with latent inhibition and is the first to show this is based in the activational actions of hormones.
Collapse
Affiliation(s)
- Matthew G Quinlan
- Center for Studies in Behavioral Neurobiology (CSBN), Department of Psychology, Concordia University, Montreal, QC, Canada H4B 1R6
| | | | | | | | | |
Collapse
|
50
|
Östrogene und Alzheimer-Demenz. GYNAKOLOGISCHE ENDOKRINOLOGIE 2011. [DOI: 10.1007/s10304-010-0389-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|