1
|
Lacasse JM, Heller C, Kheloui S, Ismail N, Raval AP, Schuh KM, Tronson NC, Leuner B. Beyond Birth Control: The Neuroscience of Hormonal Contraceptives. J Neurosci 2024; 44:e1235242024. [PMID: 39358019 PMCID: PMC11450536 DOI: 10.1523/jneurosci.1235-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 10/04/2024] Open
Abstract
Hormonal contraceptives (HCs) are one of the most highly prescribed classes of drugs in the world used for both contraceptive and noncontraceptive purposes. Despite their prevalent use, the impact of HCs on the brain remains inadequately explored. This review synthesizes recent findings on the neuroscience of HCs, with a focus on human structural neuroimaging as well as translational, nonhuman animal studies investigating the cellular, molecular, and behavioral effects of HCs. Additionally, we consider data linking HCs to mood disorders and dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and stress response as a potential mediator. The review also addresses the unique sensitivity of the adolescent brain to HCs, noting significant changes in brain structure and function when HCs are used during this developmental period. Finally, we discuss potential effects of HCs in combination with smoking-derived nicotine on outcomes of ischemic brain damage. Methodological challenges, such as the variability in HC formulations and user-specific factors, are acknowledged, emphasizing the need for precise and individualized research approaches. Overall, this review underscores the necessity for continued interdisciplinary research to elucidate the neurobiological mechanisms of HCs, aiming to optimize their use and improve women's health.
Collapse
Affiliation(s)
- Jesse M Lacasse
- Department of Psychology, Brock University, St Catharines, Ontario L2S 3A1, Canada
- Centre for Neuroscience, Brock University, St Catharines, Ontario L2S 3A1, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario M6J 1H4, Canada
| | - Carina Heller
- Department of Clinical Psychology, Friedrich Schiller University Jena, Jena 07743, Germany
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena 07743, Germany
- German Center for Mental Health (DZPG), Partner Site Jena-Magdeburg-Halle, Jena 07743, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Partner Site Jena-Magdeburg-Halle, Jena 07743, Germany
| | - Sarah Kheloui
- NISE Lab, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Nafissa Ismail
- NISE Lab, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami and Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida 33136
| | - Kristen M Schuh
- Psychology Department, University of Michigan, Ann Arbor, Michigan 48109
| | - Natalie C Tronson
- Psychology Department, University of Michigan, Ann Arbor, Michigan 48109
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
2
|
Conley AC, Vega JN, Johnson JV, Dumas JA, Newhouse PA. Effect of estradiol with or without micronized progesterone on cholinergic-related cognitive performance in postmenopausal women. Front Neurosci 2024; 18:1428675. [PMID: 39184322 PMCID: PMC11342399 DOI: 10.3389/fnins.2024.1428675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Women are at a higher risk of developing Alzheimer's disease (AD), and the decline in estrogens post-menopause is thought of as a factor increasing this risk. Estradiol (E2) is important in supporting cholinergic neuronal integrity, and cholinergic functioning may be negatively impacted following the loss of E2 post-menopause. The use of exogenous E2 has been observed to enhance cholinergically mediated cognitive performance in healthy post-menopausal women, which indicates a potentially protective mechanism. However, E2 is often co-administered with progestin or progesterone to prevent endometrial proliferation. Progesterone/progestins have previously been shown to have a detrimental effect on E2-mediated biological and cognitive effects mediated by cholinergic systems in preclinical models, therefore the present study aimed to assess whether progesterone would modify the effect of E2 to influence cognition during cholinergic blockade. Methods Twenty participants completed 3-months of oral E2 treatment with micronized progesterone (mPRO) or with placebo (PLC) in a repeated-measures within-subjects crossover design, in which they also completed five anticholinergic challenge days per hormone treatment condition. During the challenge participants were administered low or high doses of the nicotinic cholinergic antagonist mecamylamine, the muscarinic cholinergic antagonist scopolamine, or placebo. Following drug administration participants performed cognitive tests sensitive to cholinergic tone, assessing attention, episodic memory, and working memory. Results Significant decrements were found on some tasks when participants were taking E2+mPRO compared to E2 alone. Specifically, under more challenging task conditions and larger anticholinergic doses, participants showed poorer performance on the Critical Flicker Fusion task and the Stroop test and responded more conservatively on the N-back working memory task. Other tasks showed no differences between treatments under cholinergic blockade. Discussion The findings show that mPRO when taken in concert with E2, was detrimental to effortful cognitive performance, in the presence of cholinergic blockade. These results are important for assessing the impact of combined postmenopausal hormone treatment on cognitive performance that is dependent on cholinergic functioning after menopause.
Collapse
Affiliation(s)
- Alexander C. Conley
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jennifer N. Vega
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julia V. Johnson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Julie A. Dumas
- Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Paul A. Newhouse
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, United States
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Tennessee Valley Health System, Nashville, TN, United States
| |
Collapse
|
3
|
Koebele SV, Bernaud VE, Northup-Smith SN, Willeman MN, Strouse IM, Bulen HL, Schrier AR, Newbern JM, DeNardo DF, Mayer LP, Dyer CA, Bimonte-Nelson HA. Gynecological surgery in adulthood imparts cognitive and brain changes in rats: A focus on hysterectomy at short-, moderate-, and long-term intervals after surgery. Horm Behav 2023; 155:105411. [PMID: 37659358 PMCID: PMC11060888 DOI: 10.1016/j.yhbeh.2023.105411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023]
Abstract
Premenopausal hysterectomy is associated with a greater relative risk of dementia. We previously demonstrated cognitive impairments in adult rats six weeks after hysterectomy with ovarian conservation compared with intact sham-controls and other gynecological surgery variations. Here, we investigated whether hysterectomy-induced cognitive impairments are transient or persistent. Adult rats received sham-control, ovariectomy (Ovx), hysterectomy, or Ovx-hysterectomy surgery. Spatial working memory, reference memory, and anxiety-like behavior were tested either six-weeks post-surgery, in adulthood; seven-months post-surgery, in early middle-age; or twelve-months post-surgery, in late middle-age. Hysterectomy in adulthood yielded spatial working memory deficits at short-, moderate-, and long-term post-surgery intervals. Serum hormone levels did not differ between ovary-intact, but differed from Ovx, groups. Hysterectomy had no significant impact on healthy ovarian follicle or corpora lutea counts for any post-surgery timepoint compared with intact sham-controls. Frontal cortex, dorsal hippocampus, and entorhinal cortex were assessed for activity-dependent markers. In entorhinal cortex, there were alterations in FOSB and ΔFOSB expression during the early middle-age timepoint, and phosphorylated ERK1/2 levels at the adult timepoint. Collectively, results suggest a primary role for the uterus in regulating cognition, and that memory-related neural pathways may be modified following gynecological surgery. This is the first preclinical report of long-term effects of hysterectomy with and without ovarian conservation on cognition, endocrine, ovarian, and brain assessments, initiating a comprehensive framework of gynecological surgery effects. Translationally, findings underscore critical needs to decipher how gynecological surgeries, especially those involving the uterus, impact the brain and its functions, the ovaries, and overall aging from a systems perspective.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Mari N Willeman
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Isabel M Strouse
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Haidyn L Bulen
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Ally R Schrier
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Dale F DeNardo
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | | | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA.
| |
Collapse
|
4
|
Bäckström T, Turkmen S, Das R, Doverskog M, Blackburn TP. The GABA system, a new target for medications against cognitive impairment-Associated with neuroactive steroids. J Intern Med 2023; 294:281-294. [PMID: 37518841 DOI: 10.1111/joim.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The prevalence of cognitive dysfunction, dementia, and neurodegenerative disorders such as Alzheimer's disease (AD) is increasing in parallel with an aging population. Distinct types of chronic stress are thought to be instrumental in the development of cognitive impairment in central nervous system (CNS) disorders where cognitive impairment is a major unmet medical need. Increased GABAergic tone is a mediator of stress effects but is also a result of other factors in CNS disorders. Positive GABA-A receptor modulating stress and sex steroids (steroid-PAMs) such as allopregnanolone (ALLO) and medroxyprogesterone acetate can provoke impaired cognition. As such, ALLO impairs memory and learning in both animals and humans. In transgenic AD animal studies, continuous exposure to ALLO at physiological levels impairs cognition and increases degenerative AD pathology, whereas intermittent ALLO injections enhance cognition, indicating pleiotropic functions of ALLO. We have shown that GABA-A receptor modulating steroid antagonists (GAMSAs) can block the acute negative cognitive impairment of ALLO on memory in animal studies and in patients with cognitive impairment due to hepatic encephalopathy. Here we describe disorders affected by steroid-PAMs and opportunities to treat these adverse effects of steroid-PAMs with novel GAMSAs.
Collapse
Affiliation(s)
| | - Sahruh Turkmen
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
| | - Roshni Das
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
- Umecrine Cognition AB, Solna, Sweden
| | | | | |
Collapse
|
5
|
Bernaud VE, Koebele SV, Northup-Smith SN, Willeman MN, Barker C, Schatzki-Lumpkin A, Sanchez MV, Bimonte-Nelson HA. Evaluations of memory, anxiety, and the growth factor IGF-1R after post-surgical menopause treatment with a highly selective progestin. Behav Brain Res 2023; 448:114442. [PMID: 37085118 PMCID: PMC11105077 DOI: 10.1016/j.bbr.2023.114442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Progestogens are a key component of menopausal hormone therapies. While some progestogens can be detrimental to cognition, there is preclinical evidence that progestogens with a strong progesterone-receptor affinity benefit some molecular mechanisms believed to underlie cognitive function. Thus, a progestin that maximizes progesterone-receptor affinity and minimizes affinities to other receptors may be cognitively beneficial. We evaluated segesterone-acetate (SGA), a 19-norprogesterone derivative with a strong progesterone-receptor affinity and no androgenic or estrogenic-receptor activity, hypothesizing that it would enhance cognition. Middle-aged rats underwent Sham or Ovariectomy (Ovx) surgery followed by administration of medroxyprogesterone-acetate (MPA; used as a positive control as we have previously shown MPA-induced cognitive deficits), SGA (low or high dose), or vehicle (one Sham and one Ovx group). Spatial working and reference memory, delayed retention, and anxiety-like behavior were assessed, as were memory- and hormone- related protein assays within the frontal cortex, dorsal hippocampus, and entorhinal cortex. Low-dose SGA impaired spatial working memory, while high-dose SGA had a more extensive detrimental impact, negatively affecting spatial reference memory and delayed retention. Replicating previous findings, MPA impaired spatial reference memory and delayed retention. SGA, but not MPA, alleviated Ovx-induced anxiety-like behaviors. On two working memory measures, IGF-1R expression correlated with better working memory only in rats without hormone manipulation; any hormone manipulation or combination of hormone manipulations used herein altered this relationship. These findings suggest that SGA impairs spatial cognition after surgical menopause, and that surgical menopause with or without progestin administration disrupts relationships between a growth factor critical to neuroplasticity.
Collapse
Affiliation(s)
- Victoria E Bernaud
- 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
| | - Stephanie V Koebele
- 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
| | - Steven N Northup-Smith
- 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
| | - Mari N Willeman
- 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; TGen Institute, 445 N 5th St, Phoenix, AZ 85004, USA
| | - Charlotte Barker
- 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
| | - Alex Schatzki-Lumpkin
- 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
| | - Maria Valenzuela Sanchez
- 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.
| |
Collapse
|
6
|
Pletzer B, Winkler-Crepaz K, Hillerer K. Progesterone and contraceptive progestin actions on the brain: A systematic review of animal studies and comparison to human neuroimaging studies. Front Neuroendocrinol 2023; 69:101060. [PMID: 36758768 DOI: 10.1016/j.yfrne.2023.101060] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
In this review we systematically summarize the effects of progesterone and synthetic progestins on neurogenesis, synaptogenesis, myelination and six neurotransmitter systems. Several parallels between progesterone and older generation progestin actions emerged, suggesting actions via progesterone receptors. However, existing results suggest a general lack of knowledge regarding the effects of currently used progestins in hormonal contraception regarding these cellular and molecular brain parameters. Human neuroimaging studies were reviewed with a focus on randomized placebo-controlled trials and cross-sectional studies controlling for progestin type. The prefrontal cortex, amygdala, salience network and hippocampus were identified as regions of interest for future preclinical studies. This review proposes a series of experiments to elucidate the cellular and molecular actions of contraceptive progestins in these areas and link these actions to behavioral markers of emotional and cognitive functioning. Emotional effects of contraceptive progestins appear to be related to 1) alterations in the serotonergic system, 2) direct/indirect modulations of inhibitory GABA-ergic signalling via effects on the allopregnanolone content of the brain, which differ between androgenic and anti-androgenic progestins. Cognitive effects of combined oral contraceptives appear to depend on the ethinylestradiol dose.
Collapse
Affiliation(s)
- Belinda Pletzer
- Department of Psychology & Centre for Cognitive Neuroscience, Paris-Lodron-University Salzburg, Salzburg Austria.
| | | | - Katharina Hillerer
- Department of Gynaecology & Obstetrics, Private Medical University, Salzburg, Austria
| |
Collapse
|
7
|
Lacasse JM, Gomez-Perales E, Brake WG. Modeling hormonal contraception in female rats: A framework for studies in behavioral neurobiology. Front Neuroendocrinol 2022; 67:101020. [PMID: 35952797 DOI: 10.1016/j.yfrne.2022.101020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/19/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022]
Abstract
Research on hormonal contraceptives (HC) in animal models is lacking, and as a result, so is our understanding of the impact of HC on the brain and behavior. Here, we provide a review of the pharmacology of HC, as well as the methodology and best practices for designing a model of HC in female rats. We outline specific methodological considerations regarding dosing, route of administration, exposure time/timing, and selecting a control group. We also provide a framework outlining important levels of analysis for thinking about the impact of HC on behavioral and neurobiological outcomes. The purpose of this review is to equip researchers with foundational knowledge, and some basic elements of experimental design for future studies investigating the impact of HC on the brain and behavior of female rats.
Collapse
Affiliation(s)
- Jesse M Lacasse
- Centre for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal H4B 1R6, Canada.
| | - Eamonn Gomez-Perales
- Centre for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal H4B 1R6, Canada
| | - Wayne G Brake
- Centre for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal H4B 1R6, Canada.
| |
Collapse
|
8
|
Bernaud VE, Bulen HL, Peña VL, Koebele SV, Northup-Smith SN, Manzo AA, Valenzuela Sanchez M, Opachich Z, Ruhland AM, Bimonte-Nelson HA. Task-dependent learning and memory deficits in the TgF344-AD rat model of Alzheimer's disease: three key timepoints through middle-age in females. Sci Rep 2022; 12:14596. [PMID: 36028737 PMCID: PMC9418316 DOI: 10.1038/s41598-022-18415-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/10/2022] [Indexed: 11/11/2022] Open
Abstract
The TgF344 rat model of Alzheimer's disease (AD) provides a comprehensive neuropathology presentation, with age-dependent development of tau tangles, amyloid-beta (A[Formula: see text]) plaques, neuronal loss, and increased gliosis. The behavioral trajectory of this model, particularly relating to spatial learning and memory, has yet to be fully characterized. The current experiment evaluated spatial working and reference memory performance, as well as several physiological markers of health, at 3 key age points in female TgF344-AD rats: 6-months, 9-months, and 12-months. At 6 months of age, indications of working and reference memory impairments were observed in transgenic (Tg) rats on the water radial-arm maze, a complex task that requires working and reference memory simultaneously; at 12 months old, Tg impairments were observed for two working memory measures on this task. Notably, no impairments were observed at the 9-month timepoint on this maze. For the Morris maze, a measure of spatial reference memory, Tg rats demonstrated significant impairment relative to wildtype (WT) controls at all 3 age-points. Frontal cortex, entorhinal cortex, and dorsal hippocampus were evaluated for A[Formula: see text]1-42 expression via western blot in Tg rats only. Analyses of A[Formula: see text]1-42 expression revealed age-dependent increases in all 3 regions critical to spatial learning and memory. Measures of physiological health, including heart, uterine, and body weights, revealed unique age-specific outcomes for female Tg rats, with the 9-month timepoint identified as critical for further research within the trajectory of AD-like behavior, physiology, and pathology.
Collapse
Affiliation(s)
- Victoria E Bernaud
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Haidyn L Bulen
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Veronica L Peña
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Stephanie V Koebele
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Steven N Northup-Smith
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Alma A Manzo
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Maria Valenzuela Sanchez
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Zorana Opachich
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Ashley M Ruhland
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA
| | - Heather A Bimonte-Nelson
- Behavioral Neuroscience and Comparative Psychology Division, Department of Psychology, Arizona Alzheimer's Consortium, Arizona State University, 950 S. McAllister Ave., PO Box 871104, Tempe, AZ, 85287, USA.
- Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ, 85014, USA.
| |
Collapse
|
9
|
Concas A, Serra M, Porcu P. How hormonal contraceptives shape brain and behavior: A review of preclinical studies. Front Neuroendocrinol 2022; 66:101017. [PMID: 35843303 DOI: 10.1016/j.yfrne.2022.101017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/13/2022] [Accepted: 07/12/2022] [Indexed: 12/12/2022]
Abstract
Steroid hormones influence different aspects of brain function, including development, neurogenesis, neuronal excitability, and plasticity, thus affecting emotional states, cognition, sociality, and reward. In women, their levels fluctuate across the lifespan and through the reproductive stages but are also altered by exogenous administration of hormonal contraceptives (HC). HC are widely used by women throughout their fertile life both for contraceptive and therapeutic benefits. However, awareness of their effects on brain function and behavior is still poorly appreciated, despite the emerging evidence of their action at the level of the central nervous system. Here, we summarize results obtained in preclinical studies, mostly conducted in intact female rodents, aimed at investigating the neurobiological effects of HC. HC can alter neuroactive hormones, neurotransmitters, neuropeptides, as well as emotional states, cognition, social and sexual behaviors. Animal studies provide insights into the neurobiological effects of HC with the aim to improve women's health and well-being.
Collapse
Affiliation(s)
- Alessandra Concas
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Mariangela Serra
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
10
|
Das R, Ragagnin G, Sjöstedt J, Johansson M, Haage D, Druzin M, Johansson S, Bäckström T. Medroxyprogesterone acetate positively modulates specific GABA A-receptor subtypes - affecting memory and cognition. Psychoneuroendocrinology 2022; 141:105754. [PMID: 35395561 DOI: 10.1016/j.psyneuen.2022.105754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/14/2022] [Accepted: 03/28/2022] [Indexed: 11/28/2022]
Abstract
Medroxyprogesterone acetate (MPA) is a progestin widely used in humans as hormone replacement therapy and at other indications. Many progestin metabolites, as the progesterone metabolite allopregnanolone, have GABAA-receptor modulatory effects and are known to affect memory, learning, appetite, and mood. In women, 4 years chronic treatment with MPA doubles the frequency of dementia and in rats, MPA causes cognitive impairment related to the GABAergic system. Activation of the membrane bound GABAA receptor results in a chloride ion flux that can be studied by whole-cell patch-clamp electrophysiological recordings. The purpose of this study was to clarify the modulatory effects of MPA and specific MPA metabolites, with structures like known GABAA-receptor modulators, on different GABAA-receptor subtypes. An additional aim was to verify the results as steroid effects on GABA response in single cells taken from rat hypothalamus. HEK-293 cell-lines permanently expressing the recombinant human GABAA-receptor subtype α1β2γ2L or α5β3γ2L or α2β3γ2S were created. The MPA metabolites 3α5α-MPA,3β5α-MPA and 3β5β-MPA were synthesised and purified for electrophysiological patch-clamp measurements with a Dynaflow system. The effects of MPA and tetrahydrodeoxycorticosterone were also studied. None of the studied MPA metabolites affected the responses mediated by α1β2γ2L or α5β3γ2L GABAA receptors. Contrary, MPA clearly acted both as a positive modulator and as a direct activator of the α5β3γ2L and α2β3γ2S GABAA receptors. However, in concentrations up to 10 μM, MPA was inactive at the α1β2γ2L GABAA receptor. In the patch-clamp recordings from dissociated cells of the preoptic area in rats, MPA increased the amplitude of responses to GABA. In addition, MPA alone without added GABA, evoked a current response. In conclusion, MPA acts as a positive modulator of specific GABAA receptor subtypes expressed in HEK cells and at native GABA receptors in single cells from the hypothalamic preoptic area.
Collapse
Affiliation(s)
- Roshni Das
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - Gianna Ragagnin
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Jessica Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Maja Johansson
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - David Haage
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Department of Nursing Sciences, Mid Sweden University, Sundsvall, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - Michael Druzin
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Staffan Johansson
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden.
| |
Collapse
|
11
|
Koebele SV, Poisson ML, Palmer JM, Berns-Leone C, Northup-Smith SN, Peña VL, Strouse IM, Bulen HL, Patel S, Croft C, Bimonte-Nelson HA. Evaluating the Cognitive Impacts of Drospirenone, a Spironolactone-Derived Progestin, Independently and in Combination With Ethinyl Estradiol in Ovariectomized Adult Rats. Front Neurosci 2022; 16:885321. [PMID: 35692432 PMCID: PMC9177129 DOI: 10.3389/fnins.2022.885321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/14/2022] [Indexed: 11/15/2022] Open
Abstract
Oral contraceptives and hormone therapies require a progestogen component to prevent ovulation, curtail uterine hyperplasia, and reduce gynecological cancer risk. Diverse classes of synthetic progestogens, called progestins, are used as natural progesterone alternatives due to progesterone’s low oral bioavailability. Progesterone and several synthetic analogs can negatively impact cognition and reverse some neuroprotective estrogen effects. Here, we investigate drospirenone, a spironolactone-derived progestin, which has unique pharmacological properties compared to other clinically-available progestins and natural progesterone, for its impact on spatial memory, anxiety-like behavior, and brain regions crucial to these cognitive tasks. Experiment 1 assessed three drospirenone doses in young adult, ovariectomized rats, and found that a moderate drospirenone dose benefited spatial memory. Experiment 2 investigated this moderate drospirenone dose with and without concomitant ethinyl estradiol (EE) treatment, the most common synthetic estrogen in oral contraceptives. Results demonstrate that the addition of EE to drospirenone administration reversed the beneficial working memory effects of drospirenone. The hippocampus, entorhinal cortex, and perirhinal cortex were then probed for proteins known to elicit estrogen- and progestin- mediated effects on learning and memory, including glutamate decarboxylase (GAD)65, GAD67, and insulin-like growth factor receptor protein expression, using western blot. EE increased GAD expression in the perirhinal cortex. Taken together, results underscore the necessity to consider the distinct cognitive and neural impacts of clinically-available synthetic estrogen and progesterone analogs, and why they produce unique cognitive profiles when administered together compared to those observed when each hormone is administered separately.
Collapse
Affiliation(s)
- Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Mallori L. Poisson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Justin M. Palmer
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Claire Berns-Leone
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Steven N. Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Veronica L. Peña
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Isabel M. Strouse
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Haidyn L. Bulen
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Corissa Croft
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- *Correspondence: Heather A. Bimonte-Nelson,
| |
Collapse
|
12
|
Corrê MDS, de Freitas BS, Machado GDB, Pires VN, Bromberg E, Hallak JEC, Zuardi AW, Crippa JAS, Schröder N. Cannabidiol reverses memory impairments and activates components of the Akt/GSK3β pathway in an experimental model of estrogen depletion. Behav Brain Res 2022; 417:113555. [PMID: 34450240 DOI: 10.1016/j.bbr.2021.113555] [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: 05/31/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 11/28/2022]
Abstract
Clinical and preclinical evidence has indicated that estrogen depletion leads to memory impairments and increases the susceptibility to neural damage. Here, we have sought to investigate the effects of Cannabidiol (CBD) a non-psychotomimetic compound from Cannabis sativa, on memory deficits induced by estrogen depletion in rats, and its underlying mechanisms. Adult rats were subjected to bilateral ovariectomy, an established estrogen depletion model in rodents, or sham surgery and allowed to recover for three weeks. After that, they received daily injections of CBD (10 mg/kg) for fourteen days. Rats were tested in the inhibitory avoidance task, a type of emotionally-motivated memory. After behavioral testing they were euthanized, and their hippocampi were isolated for analysis of components of the Akt/GSK3β survival pathway and the antiapoptotic protein Bcl2. Results revealed that ovariectomy impaired avoidance memory, and CBD was able to completely reverse estrogen depletion-induced memory impairment. Ovariectomy also reduced Akt/GSK3β pathway's activation by decreasing the phosphorylation levels of Akt and GSK3β and Bcl2 levels, which were ameliorated by CBD. The present results indicate that CBD leads to a functional recovery accompanied by the Akt/GSK3β survival pathway's activation, supporting its potential as a treatment for estrogen decline-induced deterioration of neural functioning and maintenance.
Collapse
Affiliation(s)
- Márcio da Silveira Corrê
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Faculty of Medicine, Department of Health, Integrated Regional University of Upper Uruguay and Missions, Erechim, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gustavo Dalto Barroso Machado
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Vivian Naziaseno Pires
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Elke Bromberg
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Jaime E C Hallak
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Antônio Waldo Zuardi
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - José Alexandre S Crippa
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
13
|
Koebele SV, Hiroi R, Plumley ZMT, Melikian R, Prakapenka AV, Patel S, Carson C, Kirby D, Mennenga SE, Mayer LP, Dyer CA, Bimonte-Nelson HA. Clinically Used Hormone Formulations Differentially Impact Memory, Anxiety-Like, and Depressive-Like Behaviors in a Rat Model of Transitional Menopause. Front Behav Neurosci 2021; 15:696838. [PMID: 34366807 PMCID: PMC8335488 DOI: 10.3389/fnbeh.2021.696838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/04/2021] [Indexed: 01/29/2023] Open
Abstract
A variety of U.S. Food and Drug Administration-approved hormone therapy options are currently used to successfully alleviate unwanted symptoms associated with the changing endogenous hormonal milieu that occurs in midlife with menopause. Depending on the primary indication for treatment, different hormone therapy formulations are utilized, including estrogen-only, progestogen-only, or combined estrogen plus progestogen options. There is little known about how these formulations, or their unique pharmacodynamics, impact neurobiological processes. Seemingly disparate pre-clinical and clinical findings regarding the cognitive effects of hormone therapies, such as the negative effects associated with conjugated equine estrogens and medroxyprogesterone acetate vs. naturally circulating 17β-estradiol (E2) and progesterone, signal a critical need to further investigate the neuro-cognitive impact of hormone therapy formulations. Here, utilizing a rat model of transitional menopause, we administered either E2, progesterone, levonorgestrel, or combinations of E2 with progesterone or with levonorgestrel daily to follicle-depleted, middle-aged rats. A battery of assessments, including spatial memory, anxiety-like behaviors, and depressive-like behaviors, as well as endocrine status and ovarian follicle complement, were evaluated. Results indicate divergent outcomes for memory, anxiety, and depression, as well as unique physiological profiles, that were dependent upon the hormone regimen administered. Overall, the combination hormone treatments had the most consistently favorable profile for the domains evaluated in rats that had undergone experimentally induced transitional menopause and remained ovary-intact. The collective results underscore the importance of investigating variations in hormone therapy formulation as well as the menopause background upon which these formulations are delivered.
Collapse
Affiliation(s)
- Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Zachary M. T. Plumley
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Ryan Melikian
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Alesia V. Prakapenka
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Catherine Carson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Destiney Kirby
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Sarah E. Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | | | | | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| |
Collapse
|
14
|
Bimonte-Nelson HA, Bernaud VE, Koebele SV. Menopause, hormone therapy and cognition: maximizing translation from preclinical research. Climacteric 2021; 24:373-381. [PMID: 33977823 DOI: 10.1080/13697137.2021.1917538] [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: 10/21/2022]
Abstract
Menopause-associated and hormone-associated cognitive research has a rich history built from varied disciplines and species. This review discusses landmark rodent and human work addressing cognitive outcomes associated with varied experiences of menopause and hormone therapy. Critical variables in menopause and cognitive aging research are considered, including menopause etiology, background hormone milieu and parameters of exposure to estrogens and progestogens. Recent preclinical research has identified that menopause and ovarian hormone fluctuations across many neurobiological systems affect cognitive aging, mapping novel avenues for future research. Preclinical models provide insight into complex interdisciplinary relationships in a systematic and highly controlled fashion. We highlight that acknowledging the strengths and weaknesses for both preclinical and clinical research approaches is vital to accurate interpretation, optimal translation and the direction of future research. There is great value in collaboration and communication across preclinical and clinical realms, especially regarding reciprocal feedback of findings to advance preclinical models, improve experimental designs and enrich basic science translation to the clinic. In searching for biological mechanisms underlying the cognitive consequences of menopause and hormone therapies, it is noteworthy that clinical and preclinical scientists are grounded in the same fundamental goal of optimizing health outcomes for women across the lifespan.
Collapse
Affiliation(s)
- H A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - V E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - S V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| |
Collapse
|
15
|
Medroxyprogesterone opposes estradiol-induced renal damage in midlife ovariectomized Long Evans rats. ACTA ACUST UNITED AC 2021; 27:1411-1419. [PMID: 33109993 DOI: 10.1097/gme.0000000000001675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Our laboratory previously published that long-term administration of estradiol (E2) was detrimental to the kidneys of midlife ovariectomized Long Evans rats, contrasting clinical studies in showing that menopausal hormone therapy is associated with decreased albuminuria. However, it is unknown whether this renal benefit was due to estrogen and/or the combination with progestogen. Therefore, the objective of the current study was to determine the impact of medroxyprogesterone (MPA) on E2-mediated renal damage using a rodent model. METHODS Female Long Evans retired breeders underwent ovariectomy at 11 months of age and were treated for 40 days with subcutaneous E2, E2+MPA or vehicle at doses mimicking that of menopausal hormone therapy (N = 5-7 per group). Systolic blood pressure was measured along with indices of renal damage and function to investigate the impact of MPA on E2-mediated renal outcomes. Renal estrogen receptor alpha and G protein-coupled estrogen receptor transcript copy numbers were measured in all treatment groups through droplet digital PCR. RESULTS Middle-aged female Long Evans rats displayed spontaneous hypertension with similar systolic blood pressures and heart weights between groups. Even though blood pressure was comparable, E2 reduced glomerular filtration rate and increased proteinuria indicating pressure-independent renal damage. Coadministration with MPA prevented E2-induced glomerular filtration rate impairment and proteinuria by promoting renal hypertrophy and preventing renal interstitial fibrosis. Both E2 and E2+MPA reduced renal estrogen receptor alpha (ERα) and increased renal G protein-coupled estrogen receptor mRNA, but neither ERα nor ERß protein was different between groups. CONCLUSION MPA was protective against E2-induced renal damage and dysfunction in middle-aged female Long Evans rats. Assessing the impact of hormone therapy on renal outcomes may be an important clinical factor when considering treatment options for postmenopausal women.
Collapse
|
16
|
Bernaud VE, Hiroi R, Poisson ML, Castaneda AJ, Kirshner ZZ, Gibbs RB, Bimonte-Nelson HA. Age Impacts the Burden That Reference Memory Imparts on an Increasing Working Memory Load and Modifies Relationships With Cholinergic Activity. Front Behav Neurosci 2021; 15:610078. [PMID: 33643006 PMCID: PMC7902531 DOI: 10.3389/fnbeh.2021.610078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/07/2021] [Indexed: 11/19/2022] Open
Abstract
Rodent aging research often utilizes spatial mazes, such as the water radial-arm-maze (WRAM), to evaluate cognition. The WRAM can simultaneously measure spatial working and reference memory, wherein these two memory types are often represented as orthogonal. There is evidence, however, that these two memory forms yield interference at a high working memory load. The current study systematically evaluated whether the presence of a reference memory component impacts handling of an increasing working memory load. Young and aged female rats were tested to assess whether aging impacts this relationship. Cholinergic projections from the basal forebrain to the hippocampus and cortex can affect cognitive outcomes, and are negatively impacted by aging. To evaluate whether age-related changes in working and reference memory profiles are associated with cholinergic functioning, we assessed choline acetyltransferase activity in these behaviorally-tested rats. Results showed that young rats outperformed aged rats on a task testing solely working memory. The addition of a reference memory component deteriorated the ability to handle an increasing working memory load, such that young rats performed similar to their aged counterparts. Aged rats also had challenges when reference memory was present, but in a different context. Specifically, aged rats had difficulty remembering which reference memory arms they had entered within a session, compared to young rats. Further, aged rats that excelled in reference memory also excelled in working memory when working memory demand was high, a relationship not seen in young rats. Relationships between cholinergic activity and maze performance differed by age in direction and brain region, reflecting the complex role that the cholinergic system plays in memory and attentional processes across the female lifespan. Overall, the addition of a reference memory requirement detrimentally impacted the ability to handle working memory information across young and aged timepoints, especially when the working memory challenge was high; these age-related deficits manifested differently with the addition of a reference memory component. This interplay between working and reference memory provides insight into the multiple domains necessary to solve complex cognitive tasks, potentially improving the understanding of complexities of age- and disease- related memory failures and optimizing their respective treatments.
Collapse
Affiliation(s)
- Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Mallori L Poisson
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Arthur J Castaneda
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Ziv Z Kirshner
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| |
Collapse
|
17
|
Bengtsson S, Bäckström T, Brinton R, Irwin R, Johansson M, Sjöstedt J, Wang M. GABA-A receptor modulating steroids in acute and chronic stress; relevance for cognition and dementia? Neurobiol Stress 2020; 12:100206. [PMID: 31921942 PMCID: PMC6948369 DOI: 10.1016/j.ynstr.2019.100206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/13/2019] [Accepted: 12/18/2019] [Indexed: 01/10/2023] Open
Abstract
Cognitive dysfunction, dementia and Alzheimer's disease (AD) are increasing as the population worldwide ages. Therapeutics for these conditions is an unmet need. This review focuses on the role of the positive GABA-A receptor modulating steroid allopregnanolone (APα), it's role in underlying mechanisms for impaired cognition and of AD, and to determine options for therapy of AD. On one hand, APα given intermittently promotes neurogenesis, decreases AD-related pathology and improves cognition. On the other, continuous exposure of APα impairs cognition and deteriorates AD pathology. The disparity between these two outcomes led our groups to analyze the mechanisms underlying the difference. We conclude that the effects of APα depend on administration pattern and that chronic slightly increased APα exposure is harmful to cognitive function and worsens AD pathology whereas single administrations with longer intervals improve cognition and decrease AD pathology. These collaborative assessments provide insights for the therapeutic development of APα and APα antagonists for AD and provide a model for cross laboratory collaborations aimed at generating translatable data for human clinical trials.
Collapse
Affiliation(s)
- S.K.S. Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - T. Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - R. Brinton
- Center for Innovation in Brain Science, Professor Departments of Pharmacology and Neurology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R.W. Irwin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA
| | - M. Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - J. Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - M.D. Wang
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| |
Collapse
|
18
|
Porter KN, Sarkar SN, Dakhlallah DA, Vannoy ME, Quintana DD, Simpkins JW. Medroxyprogesterone Acetate Impairs Amyloid Beta Degradation in a Matrix Metalloproteinase-9 Dependent Manner. Front Aging Neurosci 2020; 12:92. [PMID: 32317959 PMCID: PMC7155169 DOI: 10.3389/fnagi.2020.00092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/18/2020] [Indexed: 12/25/2022] Open
Abstract
Despite the extensive use of hormonal methods as either contraception or menopausal hormone therapy (HT), there is very little known about the potential effects of these compounds on the cellular processes of the brain. Medroxyprogesterone Acetate (MPA) is a progestogen used globally in the hormonal contraceptive, Depo Provera, by women in their reproductive prime and is a major compound found in HT formulations used by menopausal women. MPA promotes changes in the circulating levels of matrix metalloproteinases (MMPs), such as MMP-9, in the endometrium, yet limited literature studying the effects of MPA on neurons and astroglia cells has been conducted. Additionally, the dysregulation of MMPs has been implicated in the pathology of Alzheimer's disease (AD), where inhibiting the secretion of MMP-9 from astroglia reduces the proteolytic degradation of amyloid-beta. Thus, we hypothesize that exposure to MPA disrupts proteolytic degradation of amyloid-beta through the downregulation of MMP-9 expression and subsequent secretion. To assess the effect of progestins on MMP-9 and amyloid-beta, in vitro, C6 rat glial cells were exposed to MPA for 48 h and then the enzymatic, secretory, and amyloid-beta degrading capacity of MMP-9 was assessed from the conditioned culture medium. We found that MPA treatment inhibited transcription of MMP-9, which resulted in a subsequent decrease in the production and secretion of MMP-9 protein, in part through the glucocorticoid receptor. Additionally, we investigated the consequences of amyloid beta-degrading activity and found that MPA treatment decreased proteolytic degradation of amyloid-beta. Our results suggest MPA suppresses amyloid-beta degradation in an MMP-9-dependent manner, in vitro, and potentially compromises the clearance of amyloid-beta in vivo.
Collapse
Affiliation(s)
- Keyana N. Porter
- Department of Pharmaceutical and Pharmacological Sciences, West Virginia University School of Pharmacy, Morgantown, WV, United States
| | - Saumyendra N. Sarkar
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Duaa A. Dakhlallah
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Mya E. Vannoy
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Dominic D. Quintana
- Department of Neuroscience, Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - James W. Simpkins
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, United States
- Department of Neuroscience, Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
19
|
The brain as a target of hormonal contraceptives: Evidence from animal studies. Front Neuroendocrinol 2019; 55:100799. [PMID: 31614151 DOI: 10.1016/j.yfrne.2019.100799] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022]
Abstract
Hormonal contraceptives are frequently prescribed drugs among women, mainly for their reversible contraceptive purposes but also for beneficial effects in some gynecological pathologies. Despite extensive studies aimed at elucidating the physical effects of hormonal contraceptives and ameliorating some unwanted outcomes, little is known yet about the effects of these drugs on brain function and related behavior, which are known to be modulated by endogenous steroid hormones. We describe the current literature on preclinical studies in animals undertaken to investigate effects of hormonal contraceptives on brain function and behavior. These studies suggest that hormonal contraceptives influence neurohormones, neurotransmitters, neuropeptides, and emotional, cognitive, social and sexual behaviors. Animals allow examination of the basic biological mechanisms of these drugs, devoid of the psychological aspect often associated to hormonal contraceptives' use in women. Understanding the neurobiological effects of these drugs may improve women's health and may help women making informed choices on hormonal contraception.
Collapse
|
20
|
Prakapenka AV, Hiroi R, Quihuis AM, Carson C, Patel S, Berns-Leone C, Fox C, Sirianni RW, Bimonte-Nelson HA. Contrasting effects of individual versus combined estrogen and progestogen regimens as working memory load increases in middle-aged ovariectomized rats: one plus one does not equal two. Neurobiol Aging 2018; 64:1-14. [PMID: 29316527 PMCID: PMC5820186 DOI: 10.1016/j.neurobiolaging.2017.11.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 02/03/2023]
Abstract
Most estrogen-based hormone therapies are administered in combination with a progestogen, such as Levonorgestrel (Levo). Individually, the estrogen 17β-estradiol (E2) and Levo can improve cognition in preclinical models. However, although these hormones are often given together clinically, the impact of the E2 + Levo combination on cognitive function has yet to be methodically examined. Thus, we investigated E2 + Levo treatment on a cognitive battery in middle-aged, ovariectomized rats. When administered alone, E2 and Levo treatments each enhanced spatial working memory relative to vehicle treatment, whereas the E2 + Levo combination impaired high working memory load performance relative to E2 only and Levo only treatments. There were no effects on spatial reference memory. Mitogen-activated protein kinases/extracellular signal-regulated kinases pathway activation, which is involved in memory formation and estrogen-induced memory effects, was evaluated in 5 brain regions implicated in learning and memory. A distinct relationship was seen in the E2-only treatment group between mitogen-activated protein kinases/extracellular signal-regulated kinases pathway activation in the frontal cortex and working memory performance. Collectively, the results indicate that the differential neurocognitive effects of combination versus sole treatments are vital considerations as we move forward as a field to develop novel, and to understand currently used, exogenous hormone regimens across the lifespan.
Collapse
Affiliation(s)
- Alesia V Prakapenka
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA; Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Alicia M Quihuis
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Catie Carson
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Claire Berns-Leone
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Carly Fox
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Rachael W Sirianni
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
21
|
Koebele SV, Mennenga SE, Hiroi R, Quihuis AM, Hewitt LT, Poisson ML, George C, Mayer LP, Dyer CA, Aiken LS, Demers LM, Carson C, Bimonte-Nelson HA. Cognitive changes across the menopause transition: A longitudinal evaluation of the impact of age and ovarian status on spatial memory. Horm Behav 2017; 87:96-114. [PMID: 27793768 PMCID: PMC5479707 DOI: 10.1016/j.yhbeh.2016.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 09/19/2016] [Accepted: 10/23/2016] [Indexed: 11/16/2022]
Abstract
Cognitive changes that occur during mid-life and beyond are linked to both aging and the menopause transition. Studies in women suggest that the age at menopause onset can impact cognitive status later in life; yet, little is known about memory changes that occur during the transitional period to the postmenopausal state. The 4-vinylcyclohexene diepoxide (VCD) model simulates transitional menopause in rodents by depleting the immature ovarian follicle reserve and allowing animals to retain their follicle-deplete ovarian tissue, resulting in a profile similar to the majority of perimenopausal women. Here, Vehicle or VCD treatment was administered to ovary-intact adult and middle-aged Fischer-344 rats to assess the trajectory of cognitive change across time with normal aging and aging with transitional menopause via VCD-induced follicular depletion, as well as to evaluate whether age at the onset of follicular depletion plays a role in cognitive outcomes. Animals experiencing the onset of menopause at a younger age exhibited impaired spatial memory early in the transition to a follicle-deplete state. Additionally, at the mid- and post- follicular depletion time points, VCD-induced follicular depletion amplified an age effect on memory. Overall, these findings suggest that age at the onset of menopause is a critical parameter to consider when evaluating learning and memory across the transition to reproductive senescence. From a translational perspective, this study illustrates how age at menopause onset might impact cognition in menopausal women, and provides insight into time points to explore for the window of opportunity for hormone therapy during the menopause transition period. Hormone therapy during this critical juncture might be especially efficacious at attenuating age- and menopause- related cognitive decline, producing healthy brain aging profiles in women who retain their ovaries throughout their lifespan.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Alicia M Quihuis
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Lauren T Hewitt
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Mallori L Poisson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Christina George
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Loretta P Mayer
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Cheryl A Dyer
- Senestech, Inc., 3140 N Caden Court, Flagstaff, AZ 86004, United States
| | - Leona S Aiken
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States
| | - Laurence M Demers
- The Pennsylvania State University College of Medicine, The M. S. Hershey Medical Center, Hershey, PA 17033, United States
| | - Catherine Carson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
22
|
Koebele SV, Bimonte-Nelson HA. The endocrine-brain-aging triad where many paths meet: female reproductive hormone changes at midlife and their influence on circuits important for learning and memory. Exp Gerontol 2016; 94:14-23. [PMID: 27979770 DOI: 10.1016/j.exger.2016.12.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 01/15/2023]
Abstract
Female mammals undergo natural fluctuations in sex steroid hormone levels throughout life. These fluctuations span from early development, to cyclic changes associated with the menstrual or estrous cycle and pregnancy, to marked hormone flux during perimenopause, and a final decline at reproductive senescence. While the transition to reproductive senescence is not yet fully understood, the vast majority of mammals experience this spontaneous, natural phenomenon with age, which has broad implications for long-lived species. Indeed, this post-reproductive life stage, and its transition, involves significant and enduring physiological changes, including considerably altered sex steroid hormone and gonadotropin profiles that impact multiple body systems, including the brain. The endocrine-brain-aging triad is especially noteworthy, as many paths meet and interact. Many of the brain regions affected by aging are also sensitive to changes in ovarian hormone levels, and aging and reproductive senescence are both associated with changes in memory performance. This review explores how menopause is related to cognitive aging, and discusses some of the key neural systems and molecular factors altered with age and reproductive hormone level changes, with an emphasis on brain regions important for learning and memory.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
23
|
Braden BB, Andrews MG, Acosta JI, Mennenga SE, Lavery C, Bimonte-Nelson HA. A comparison of progestins within three classes: Differential effects on learning and memory in the aging surgically menopausal rat. Behav Brain Res 2016; 322:258-268. [PMID: 27368418 DOI: 10.1016/j.bbr.2016.06.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/03/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION For decades, progestins have been included in hormone therapies (HT) prescribed to women to offset the risk of unopposed estrogen-induced endometrial hyperplasia. However, the potential effects on cognition of subcategories of clinically used progestins have been largely unexplored. METHODS In two studies, the present investigation evaluated the cognitive effects of norethindrone acetate (NETA), levonorgestrel (LEVO), and medroxyprogesterone acetate (MPA) on the water radial-arm maze (WRAM) and Morris water maze (MM) in middle-aged ovariectomized rats. RESULTS In Study 1, six-weeks of a high-dose NETA treatment impaired learning and delayed retention on the WRAM, and impaired reference memory on the MM. Low-dose NETA treatment impaired delayed retention on the WRAM. In Study 2, high-dose NETA treatment was reduced to four-weeks and compared to MPA and LEVO. As previously shown, MPA impaired working memory performance during the lattermost portion of testing, at the highest working memory load, impaired delayed retention on the WRAM, and impaired reference memory on the MM. NETA also impaired performance on these WRAM and MM measures. Interestingly, LEVO did not impair performance, but instead enhanced learning on the WRAM. CONCLUSIONS The current study corroborates previous evidence that the most commonly prescribed FDA-approved progestin for HT, MPA, impairs learning and memory in the ovariectomized middle-aged rat. When progestins from two different additional subcategories were investigated, NETA impaired learning and memory similarly to MPA, but LEVO enhanced learning. Future research is warranted to determine LEVO's potential as an ideal progestin for optimal health in women, including for cognition.
Collapse
Affiliation(s)
- B Blair Braden
- Department of Psychology, Arizona State University, Tempe, AZ, United States; Arizona Alzheimer's Consortium, United States
| | - Madeline G Andrews
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Jazmin I Acosta
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Courtney Lavery
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States; Arizona Alzheimer's Consortium, United States.
| |
Collapse
|
24
|
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]
|
25
|
Koebele SV, Bimonte-Nelson HA. Modeling menopause: The utility of rodents in translational behavioral endocrinology research. Maturitas 2016; 87:5-17. [PMID: 27013283 DOI: 10.1016/j.maturitas.2016.01.015] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 01/25/2016] [Indexed: 01/31/2023]
Abstract
The human menopause transition and aging are each associated with an increase in a variety of health risk factors including, but not limited to, cardiovascular disease, osteoporosis, cancer, diabetes, stroke, sexual dysfunction, affective disorders, sleep disturbances, and cognitive decline. It is challenging to systematically evaluate the biological underpinnings associated with the menopause transition in the human population. For this reason, rodent models have been invaluable tools for studying the impact of gonadal hormone fluctuations and eventual decline on a variety of body systems. While it is essential to keep in mind that some of the mechanisms associated with aging and the transition into a reproductively senescent state can differ when translating from one species to another, animal models provide researchers with opportunities to gain a fundamental understanding of the key elements underlying reproduction and aging processes, paving the way to explore novel pathways for intervention associated with known health risks. Here, we discuss the utility of several rodent models used in the laboratory for translational menopause research, examining the benefits and drawbacks in helping us to better understand aging and the menopause transition in women. The rodent models discussed are ovary-intact, ovariectomy, and 4-vinylcylohexene diepoxide for the menopause transition. We then describe how these models may be implemented in the laboratory, particularly in the context of cognition. Ultimately, we aim to use these animal models to elucidate novel perspectives and interventions for maintaining a high quality of life in women, and to potentially prevent or postpone the onset of negative health consequences associated with these significant life changes during aging.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
26
|
Bengtsson SKS, Johansson M, Bäckström T. Long-term continuous allopregnanolone elevation causes memory decline and hippocampus shrinkage, in female wild-type B6 mice. Horm Behav 2016; 78:160-167. [PMID: 26497250 DOI: 10.1016/j.yhbeh.2015.10.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 10/02/2015] [Accepted: 10/20/2015] [Indexed: 11/20/2022]
Abstract
Chronic stress in various forms increases the risk for cognitive dysfunction, dementia and Alzheimer's disease. While the pathogenesis behind these findings is unknown, growing evidence suggests that chronic increase in neurosteroid levels, such as allopregnanolone, is part of the mechanism. We treated wild-type C57BL/6J mice with allopregnanolone for 5months, using osmotic pumps. This treatment led to moderately increased levels of allopregnanolone, equivalent to that of mild chronic stress. After an interval of no treatment for 1month, female mice showed impaired learning and memory function in the Morris water maze (MWM) in combination with diminished hippocampus weight and increased cerebellum weight, both correlating to MWM performance. Male mice showed a minor reduction in memory function and no differences in brain structure. We conclude that chronic allopregnanolone elevation can lead to cognitive dysfunction and negative brain alterations. We suggest that allopregnanolone could play a key role in the pathogenesis of stress-induced cognitive disturbances and perhaps dementia.
Collapse
Affiliation(s)
- Sara K S Bengtsson
- Umeå Neurosteroid Research Center, Umeå University, Department of Clinical Sciences, Umeå, Sweden.
| | - Maja Johansson
- Umeå Neurosteroid Research Center, Umeå University, Department of Clinical Sciences, Umeå, Sweden; Umecrine Cognition AB, Umeå, Sweden
| | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Umeå University, Department of Clinical Sciences, Umeå, Sweden
| |
Collapse
|
27
|
Gaudet CM, Lim YP, Stonestreet BS, Threlkeld SW. Effects of age, experience and inter-alpha inhibitor proteins on working memory and neuronal plasticity after neonatal hypoxia-ischemia. Behav Brain Res 2016; 302:88-99. [PMID: 26778784 DOI: 10.1016/j.bbr.2016.01.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/23/2015] [Accepted: 01/05/2016] [Indexed: 01/15/2023]
Abstract
Neonatal cerebral hypoxia-ischemia (HI) commonly results in cognitive and sensory impairments. Early behavioral experience has been suggested to improve cognitive and sensory outcomes in children and animal models with perinatal neuropathology. In parallel, we previously showed that treatment with immunomodulator Inter-alpha Inhibitor Proteins (IAIPs) improves cellular and behavioral outcomes in neonatal HI injured rats. The purpose of the current study was to evaluate the influences of early experience and typical maturation in combination with IAIPs treatment on spatial working and reference memory after neonatal HI injury. A second aim was to determine the effects of these variables on hippocampal CA1 neuronal morphology. Subjects were divided into two groups that differed with respect to the time when exposed to eight arm radial water maze testing: Group one was tested as juveniles (early experience, Postnatal day (P) 36-61) and adults (P88-113), and Group two was tested in adulthood only (P88-113; without early experience). Three treatment conditions were included in each experience group (HI+Vehicle, HI+IAIPs, and Sham subjects). Incorrect arm entries (errors) were compared between treatment and experience groups across three error types (reference memory (RM), working memory incorrect (WMI), working memory correct (WMC)). Early experience led to improved working memory performance regardless of treatment. Combining IAIPs intervention with early experience provided a long-term behavioral advantage on the WMI component of the task in HI animals. Anatomically, early experience led to a decrease in the average number of basal dendrites per CA1 pyramidal neuron for IAIP treated subjects and a significant reduction in basal dendritic length in control subjects, highlighting the importance of pruning in typical early life learning. Our results support the hypothesis that early behavioral experience combined with IAIPs improve outcome on a relativity demanding cognitive task, beyond that of a single intervention strategy, and appears to facilitate neuronal plasticity following neonatal brain injury.
Collapse
Affiliation(s)
- Cynthia M Gaudet
- Department of Biology, Rhode Island College, 600 Mount Pleasant Ave., Providence, RI 02904, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., 349 Eddy Street, Providence, RI 02903, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905, USA
| | - Steven W Threlkeld
- Department of Psychology, Rhode Island College, 600 Mount Pleasant Ave. Providence, RI 02904, USA.
| |
Collapse
|
28
|
Berent-Spillson A, Briceno E, Pinsky A, Simmen A, Persad CC, Zubieta JK, Smith YR. Distinct cognitive effects of estrogen and progesterone in menopausal women. Psychoneuroendocrinology 2015; 59:25-36. [PMID: 26010861 PMCID: PMC4490102 DOI: 10.1016/j.psyneuen.2015.04.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 04/03/2015] [Accepted: 04/27/2015] [Indexed: 01/03/2023]
Abstract
The effects of postmenopausal hormone treatment on cognitive outcomes are inconsistent in the literature. Emerging evidence suggests that cognitive effects are influenced by specific hormone formulations, and that progesterone is more likely to be associated with positive outcomes than synthetic progestin. There are very few studies of unopposed progesterone in postmenopausal women, and none that use functional neuroimaging, a sensitive measure of neurobiological function. In this study of 29 recently postmenopausal women, we used functional MRI and neuropsychological measures to separately assess the effects of estrogen or progesterone treatment on visual and verbal cognitive function. Women were randomized to receive 90 days of either estradiol or progesterone counterbalanced with placebo. After each treatment arm, women were given a battery of verbal and visual cognitive function and working memory tests, and underwent functional MRI including verbal processing and visual working memory tasks. We found that both estradiol and progesterone were associated with changes in activation patterns during verbal processing. Compared to placebo, women receiving estradiol treatment had greater activation in the left prefrontal cortex, a region associated with verbal processing and encoding. Progesterone was associated with changes in regional brain activation patterns during a visual memory task, with greater activation in the left prefrontal cortex and right hippocampus compared to placebo. Both treatments were associated with a statistically non-significant increase in number of words remembered following the verbal task performed during the fMRI scanning session, while only progesterone was associated with improved neuropsychological measures of verbal working memory compared to placebo. These results point to potential cognitive benefits of both estrogen and progesterone.
Collapse
Affiliation(s)
- Alison Berent-Spillson
- University of Michigan, Psychiatry Department, MBNI, 205 Zina Pitcher Place, Ann Arbor, MI 48109, USA.
| | - Emily Briceno
- University of Michigan, Psychiatry Department, Neuropsychology Division, 2101 Commonwealth Blvd, Suite C, Ann Arbor, MI 48105, USA.
| | - Alana Pinsky
- University of Michigan Medical School, 1301 Catherine, Ann Arbor, MI, 48109, USA.
| | - Angela Simmen
- University of Michigan, Obstetrics and Gynecology Department, L4000 Womens SPC, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA.
| | - Carol C. Persad
- University of Michigan, Psychiatry Department, Neuropsychology Division, 2101 Commonwealth Blvd, Suite C, Ann Arbor, MI 48105, USA
| | - Jon-Kar Zubieta
- University of Michigan, Psychiatry Department, MBNI, 205 Zina Pitcher Place, Ann Arbor, MI 48109, USA.
| | - Yolanda R. Smith
- University of Michigan, Obstetrics and Gynecology Department, L4000 Womens SPC, 1500 E. Medical Center Dr, Ann Arbor, MI 48109, USA,Corresponding author: Alison Berent-Spillson, 1-734-615-4252
| |
Collapse
|
29
|
Braden BB, Kingston ML, Koenig EN, Lavery CN, Tsang CWS, Bimonte-Nelson HA. The GABAA antagonist bicuculline attenuates progesterone-induced memory impairments in middle-aged ovariectomized rats. Front Aging Neurosci 2015; 7:149. [PMID: 26321945 PMCID: PMC4536389 DOI: 10.3389/fnagi.2015.00149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/20/2015] [Indexed: 01/24/2023] Open
Abstract
In women, high levels of natural progesterone have been associated with detrimental cognitive effects via the “maternal amnesia” phenomenon as well as in controlled experiments. In aged ovariectomized (Ovx) rats, progesterone has been shown to impair cognition and impact the GABAergic system in cognitive brain regions. Here, we tested whether the GABAergic system is a mechanism of progesterone’s detrimental cognitive effects in the Ovx rat by attempting to reverse progesterone-induced impairments via concomitant treatment with the GABAA antagonist, bicuculline. Thirteen month old rats received Ovx plus daily vehicle, progesterone, bicuculline, or progesterone+bicuculline injections beginning 2 weeks prior to testing. The water radial-arm maze was used to evaluate spatial working and reference memory. During learning, rats administered progesterone made more working memory errors than those administered vehicle, and this impairment was reversed by the addition of bicuculline. The progesterone impairment was transient and all animals performed similarly by the end of regular testing. On the last day of testing, a 6 hour delay was administered to evaluate memory retention. Progesterone-treated rats were the only group to increase working memory errors with the delay relative to baseline performance; again, the addition of bicuculline prevented the progesterone-induced impairment. The vehicle, bicuculline, and progesterone+bicuculline groups were not impaired by the delay. The current rodent findings corroborate prior research reporting progesterone-induced detriments on cognition in women and in the aging Ovx rat. Moreover, the data suggest that the progesterone-induced cognitive impairment is, in part, related to the GABAergic system. Given that progesterone is included in numerous clinically-prescribed hormone therapies and contraceptives (e.g., micronized), and as synthetic analogs, further research is warranted to better understand the parameters and mechanism(s) of progesterone-induced cognitive impairments.
Collapse
Affiliation(s)
- B Blair Braden
- Human Brain Mapping Laboratory, Neuroimaging, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center Phoenix, AZ, USA ; Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA ; Arizona Alzheimer's Consortium Phoenix, AZ, USA
| | - Melissa L Kingston
- Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA
| | - Elizabeth N Koenig
- Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA
| | - Courtney N Lavery
- Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA
| | - Candy W S Tsang
- Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA
| | - Heather A Bimonte-Nelson
- Memory and Aging Laboratory, Department of Psychology, Arizona State University Tempe, AZ, USA ; Arizona Alzheimer's Consortium Phoenix, AZ, USA
| |
Collapse
|
30
|
Koebele SV, Bimonte-Nelson HA. Trajectories and phenotypes with estrogen exposures across the lifespan: What does Goldilocks have to do with it? Horm Behav 2015; 74:86-104. [PMID: 26122297 PMCID: PMC4829405 DOI: 10.1016/j.yhbeh.2015.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/14/2015] [Accepted: 06/04/2015] [Indexed: 01/04/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Estrogens impact the organization and activation of the mammalian brain in both sexes, with sex-specific critical windows. Throughout the female lifespan estrogens activate brain substrates previously organized by estrogens, and estrogens can induce non-transient brain and behavior changes into adulthood. Therefore, from early life through the transition to reproductive senescence and beyond, estrogens are potent modulators of the brain and behavior. Organizational, reorganizational, and activational hormone events likely impact the trajectory of brain profiles during aging. A "brain profile," or quantitative brain measurement for research purposes, is typically a snapshot in time, but in life a brain profile is anything but static--it is in flux, variable, and dynamic. Akin to this, the only thing continuous and consistent about hormone exposures across a female's lifespan is that they are noncontinuous and inconsistent, building and rebuilding on past exposures to create a present brain and behavioral landscape. Thus, hormone variation is especially rich in females, and is likely the destiny for maximal responsiveness in the female brain. The magnitude and direction of estrogenic effects on the brain and its functions depend on a myriad of factors; a "Goldilocks" phenomenon exists for estrogens, whereby if the timing, dose, and regimen for an individual are just right, markedly efficacious effects present. Data indicate that exogenously-administered estrogens can bestow beneficial cognitive effects in some circumstances, especially when initiated in a window of opportunity such as the menopause transition. Could it be that the age-related reduction in efficacy of estrogens reflects the closure of a late-in-life critical window occurring around the menopause transition? Information from classic and contemporary works studying organizational/activational estrogen actions, in combination with acknowledging the tendency for maximal responsiveness to cyclicity, will elucidate ways to extend sensitivity and efficacy into post-menopause.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA.
| |
Collapse
|
31
|
Walf AA, Koonce CJ, Frye CA. Progestogens' effects and mechanisms for object recognition memory across the lifespan. Behav Brain Res 2015; 294:50-61. [PMID: 26235328 DOI: 10.1016/j.bbr.2015.07.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/16/2015] [Accepted: 07/28/2015] [Indexed: 12/11/2022]
Abstract
This review explores the effects of female reproductive hormones, estrogens and progestogens, with a focus on progesterone and allopregnanolone, on object memory. Progesterone and its metabolites, in particular allopregnanolone, exert various effects on both cognitive and non-mnemonic functions in females. The well-known object recognition task is a valuable experimental paradigm that can be used to determine the effects and mechanisms of progestogens for mnemonic effects across the lifespan, which will be discussed herein. In this task there is little test-decay when different objects are used as targets and baseline valance for objects is controlled. This allows repeated testing, within-subjects designs, and longitudinal assessments, which aid understanding of changes in hormonal milieu. Objects are not aversive or food-based, which are hormone-sensitive factors. This review focuses on published data from our laboratory, and others, using the object recognition task in rodents to assess the role and mechanisms of progestogens throughout the lifespan. Improvements in object recognition performance of rodents are often associated with higher hormone levels in the hippocampus and prefrontal cortex during natural cycles, with hormone replacement following ovariectomy in young animals, or with aging. The capacity for reversal of age- and reproductive senescence-related decline in cognitive performance, and changes in neural plasticity that may be dissociated from peripheral effects with such decline, are discussed. The focus here will be on the effects of brain-derived factors, such as the neurosteroid, allopregnanolone, and other hormones, for enhancing object recognition across the lifespan.
Collapse
Affiliation(s)
- Alicia A Walf
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA; The Center for Life Sciences Research, The University at Albany-SUNY, Albany, NY 12222, USA; Institute of Arctic Biology, The University of Alaska-Fairbanks, Fairbanks, Alaska 99775, USA; The University of Alaska-Fairbanks, IDeA Network of Biomedical Excellence (INBRE), Fairbanks, Alaska 99775, USA; Cognitive Science Department, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Carolyn J Koonce
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA; Institute of Arctic Biology, The University of Alaska-Fairbanks, Fairbanks, Alaska 99775, USA; The University of Alaska-Fairbanks, IDeA Network of Biomedical Excellence (INBRE), Fairbanks, Alaska 99775, USA
| | - Cheryl A Frye
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA; Dept. of Biological Sciences, The University at Albany-SUNY, Albany, NY 12222, USA; The Center for Neuroscience, The University at Albany-SUNY, Albany, NY 12222, USA; The Center for Life Sciences Research, The University at Albany-SUNY, Albany, NY 12222, USA; Department of Chemistry and Biochemistry, The University of Alaska-Fairbanks, Fairbanks, Alaska 99775, USA; Institute of Arctic Biology, The University of Alaska-Fairbanks, Fairbanks, Alaska 99775, USA; The University of Alaska-Fairbanks, IDeA Network of Biomedical Excellence (INBRE), Fairbanks, Alaska 99775, USA.
| |
Collapse
|
32
|
Mennenga SE, Koebele SV, Mousa AA, Alderete TJ, Tsang CWS, Acosta JI, Camp BW, Demers LM, Bimonte-Nelson HA. Pharmacological blockade of the aromatase enzyme, but not the androgen receptor, reverses androstenedione-induced cognitive impairments in young surgically menopausal rats. Steroids 2015; 99:16-25. [PMID: 25159107 PMCID: PMC4398574 DOI: 10.1016/j.steroids.2014.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 07/02/2014] [Indexed: 02/05/2023]
Abstract
Androstenedione, the main circulating ovarian hormone present after menopause, has been shown to positively correlate with poor spatial memory in an ovary-intact rodent model of follicular depletion, and to impair spatial memory when administered exogenously to surgically menopausal ovariectomized rats. Androstenedione can be converted directly to estrone via the aromatase enzyme, or to testosterone. The current study investigated the hormonal mechanism underlying androstenedione-induced cognitive impairments. Young adult ovariectomized rats were given either androstenedione, androstenedione plus the aromatase inhibitor anastrozole to block conversion to estrone, androstenedione plus the androgen receptor blocker flutamide to block androgen receptor activity, or vehicle treatment, and were then administered a battery of learning and memory maze tasks. Since we have previously shown that estrone administration to ovariectomized rats impaired cognition, we hypothesized that androstenedione's conversion to estrone underlies, in part, its negative cognitive impact. Here, androstenedione administration impaired spatial reference and working memory. Further, androstenedione did not induce memory deficits when co-administered with the aromatase inhibitor, anastrozole, whereas pharmacological blockade of the androgen receptor failed to block the cognitive impairing effects of androstenedione. Anastrozole alone did not impact performance on any cognitive measure. The current data support the tenet that androstenedione impairs memory through its conversion to estrone, rather than via actions on the androgen receptor. Studying the effects of aromatase and estrogen metabolism is critical to elucidating how hormones impact women's health across the lifespan, and results hold important implications for understanding and optimizing the hormone milieu from the many endogenous and exogenous hormone exposures across the lifetime.
Collapse
Affiliation(s)
- Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Abeer A Mousa
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States
| | - Tanya J Alderete
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States
| | - Candy W S Tsang
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States
| | - Jazmin I Acosta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Bryan W Camp
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Laurence M Demers
- The Pennsylvania State University College of Medicine, The M. S. Hershey Medical Center, Hershey, PA 17033, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
33
|
Gleason CE, Dowling NM, Wharton W, Manson JE, Miller VM, Atwood CS, Brinton EA, Cedars MI, Lobo RA, Merriam GR, Neal-Perry G, Santoro NF, Taylor HS, Black DM, Budoff MJ, Hodis HN, Naftolin F, Harman SM, Asthana S. Effects of Hormone Therapy on Cognition and Mood in Recently Postmenopausal Women: Findings from the Randomized, Controlled KEEPS-Cognitive and Affective Study. PLoS Med 2015; 12:e1001833; discussion e1001833. [PMID: 26035291 PMCID: PMC4452757 DOI: 10.1371/journal.pmed.1001833] [Citation(s) in RCA: 308] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 04/22/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Menopausal hormone therapy (MHT) reportedly increases the risk of cognitive decline in women over age 65 y. It is unknown whether similar risks exist for recently postmenopausal women, and whether MHT affects mood in younger women. The ancillary Cognitive and Affective Study (KEEPS-Cog) of the Kronos Early Estrogen Prevention Study (KEEPS) examined the effects of up to 4 y of MHT on cognition and mood in recently postmenopausal women. METHODS AND FINDINGS KEEPS, a randomized, double-blinded, placebo-controlled clinical trial, was conducted at nine US academic centers. Of the 727 women enrolled in KEEPS, 693 (95.3%) participated in the ancillary KEEPS-Cog, with 220 women randomized to receive 4 y of 0.45 mg/d oral conjugated equine estrogens (o-CEE) plus 200 mg/d micronized progesterone (m-P) for the first 12 d of each month, 211 women randomized to receive 50 μg/d transdermal estradiol (t-E2) plus 200 mg/d m-P for the first 12 d of each month, and 262 women randomized to receive placebo pills and patches. Primary outcomes included the Modified Mini-Mental State examination; four cognitive factors: verbal learning/memory, auditory attention/working memory, visual attention/executive function, and speeded language/mental flexibility; and a mood measure, the Profile of Mood States (POMS). MHT effects were analyzed using linear mixed-effects (LME) models, which make full use of all available data from each participant, including those with missing data. Data from those with and without full data were compared to assess for potential biases resulting from missing observations. For statistically significant results, we calculated effect sizes (ESs) to evaluate the magnitude of changes. On average, participants were 52.6 y old, and 1.4 y past their last menstrual period. By month 48, 169 (24.4%) and 158 (22.8%) of the 693 women who consented for ancillary KEEPS-Cog were lost to follow-up for cognitive assessment (3MS and cognitive factors) and mood evaluations (POMS), respectively. However, because LME models make full use all available data, including data from women with missing data, 95.5% of participants were included in the final analysis (n = 662 in cognitive analyses, and n = 661 in mood analyses). To be included in analyses, women must have provided baseline data, and data from at least one post-baseline visit. The mean length of follow-up was 2.85 y (standard deviation [SD] = 0.49) for cognitive outcomes and 2.76 (SD = 0.57) for mood outcomes. No treatment-related benefits were found on cognitive outcomes. For mood, model estimates indicated that women treated with o-CEE showed improvements in depression and anxiety symptoms over the 48 mo of treatment, compared to women on placebo. The model estimate for the depression subscale was -5.36 × 10(-2) (95% CI, -8.27 × 10(-2) to -2.44 × 10(-2); ES = 0.49, p < 0.001) and for the anxiety subscale was -3.01 × 10(-2) (95% CI, -5.09 × 10(-2) to -9.34 × 10(-3); ES = 0.26, p < 0.001). Mood outcomes for women randomized to t-E2 were similar to those for women on placebo. Importantly, the KEEPS-Cog results cannot be extrapolated to treatment longer than 4 y. CONCLUSIONS The KEEPS-Cog findings suggest that for recently postmenopausal women, MHT did not alter cognition as hypothesized. However, beneficial mood effects with small to medium ESs were noted with 4 y of o-CEE, but not with 4 y of t-E2. The generalizability of these findings is limited to recently postmenopausal women with low cardiovascular risk profiles. TRIAL REGISTRATION ClinicalTrials.gov NCT00154180 and NCT00623311.
Collapse
Affiliation(s)
- Carey E. Gleason
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, United States of America
- * E-mail:
| | - N. Maritza Dowling
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Whitney Wharton
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Alzheimer’s Disease Research Center, Atlanta, Georgia, United States of America
| | - JoAnn E. Manson
- Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Virginia M. Miller
- Departments of Surgery and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Craig S. Atwood
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, United States of America
| | - Eliot A. Brinton
- Utah Foundation for Biomedical Research, Salt Lake City, Utah, United States of America
| | - Marcelle I. Cedars
- Obstetrics & Gynecology, University of California at San Francisco, San Francisco, California, United States of America
| | - Rogerio A. Lobo
- Obstetrics & Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - George R. Merriam
- VA Puget Sound Health Care System, Tacoma, Washington, United States of America
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Tacoma, Washington, United States of America
| | - Genevieve Neal-Perry
- Neuroscience and Obstetrics & Gynecology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Nanette F. Santoro
- Obstetrics & Gynecology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Hugh S. Taylor
- Obstetrics & Gynecology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Dennis M. Black
- Epidemiology & Biostatistics, University of California at San Francisco, San Francisco, California, United States of America
| | - Matthew J. Budoff
- Division of Cardiology, Los Angeles Biomedical Research Institute at Harbor–UCLA Medical Center, Torrance, California, United States of America
| | - Howard N. Hodis
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, California, United States of America
| | - Frederick Naftolin
- Obstetrics & Gynecology, New York University School of Medicine, New York, New York, United States of America
| | - S. Mitchell Harman
- Kronos Longevity Research Institute, Phoenix, Arizona, United States of America
- Division of Endocrinology, Phoenix VA Medical Center, Phoenix, Arizona, United States of America
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, United States of America
| |
Collapse
|
34
|
Mennenga SE, Gerson JE, Koebele SV, Kingston ML, Tsang CWS, Engler-Chiurazzi EB, Baxter LC, Bimonte-Nelson HA. Understanding the cognitive impact of the contraceptive estrogen Ethinyl Estradiol: tonic and cyclic administration impairs memory, and performance correlates with basal forebrain cholinergic system integrity. Psychoneuroendocrinology 2015; 54:1-13. [PMID: 25679306 PMCID: PMC4433884 DOI: 10.1016/j.psyneuen.2015.01.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 11/23/2022]
Abstract
Ethinyl Estradiol (EE), a synthetic, orally bio-available estrogen, is the most commonly prescribed form of estrogen in oral contraceptives, and is found in at least 30 different contraceptive formulations currently prescribed to women as well as hormone therapies prescribed to menopausal women. Thus, EE is prescribed clinically to women at ages ranging from puberty to reproductive senescence. Here, in two separate studies, the cognitive effects of cyclic or tonic EE administration following ovariectomy (Ovx) were evaluated in young female rats. Study I assessed the cognitive effects of low and high doses of EE, delivered tonically via a subcutaneous osmotic pump. Study II evaluated the cognitive effects of low, medium, and high doses of EE administered via a daily subcutaneous injection, modeling the daily rise and fall of serum EE levels with oral regimens. Study II also investigated the impact of low, medium and high doses of EE on the basal forebrain cholinergic system. The low and medium doses utilized here correspond to the range of doses currently used in clinical formulations, and the high dose corresponds to doses prescribed to a generation of women between 1960 and 1970, when oral contraceptives first became available. We evaluate cognition using a battery of maze tasks tapping several domains of spatial learning and memory as well as basal forebrain cholinergic integrity using immunohistochemistry and unbiased stereology to estimate the number of choline acetyltransferase (ChAT)-producing cells in the medial septum and vertical/diagonal bands. At the highest dose, EE treatment impaired multiple domains of spatial memory relative to vehicle treatment, regardless of administration method. When given cyclically at the low and medium doses, EE did not impact working memory, but transiently impaired reference memory during the learning phase of testing. Of the doses and regimens tested here, only EE at the highest dose impaired several domains of memory; tonic delivery of low EE, a dose that corresponds to the most popular doses used in the clinic today, did not impact cognition on any measure. Both medium and high injection doses of EE reduced the number of ChAt-immunoreactive cells in the basal forebrain, and cell population estimates in the vertical/diagonal bands negatively correlated with working memory errors.
Collapse
Affiliation(s)
- Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Julia E Gerson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Melissa L Kingston
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Candy W S Tsang
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Elizabeth B Engler-Chiurazzi
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA
| | - Leslie C Baxter
- Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA; Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, USA.
| |
Collapse
|
35
|
Liu J, Lin H, Huang Y, Liu Y, Wang B, Su F. Cognitive effects of long-term dydrogesterone treatment used alone or with estrogen on rat menopausal models of different ages. Neuroscience 2015; 290:103-14. [DOI: 10.1016/j.neuroscience.2015.01.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 11/16/2022]
|
36
|
Talboom JS, West SG, Engler-Chiurazzi EB, Enders CK, Crain I, Bimonte-Nelson HA. Learning to remember: cognitive training-induced attenuation of age-related memory decline depends on sex and cognitive demand, and can transfer to untrained cognitive domains. Neurobiol Aging 2014; 35:2791-2802. [PMID: 25104561 DOI: 10.1016/j.neurobiolaging.2014.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 05/27/2014] [Accepted: 06/10/2014] [Indexed: 01/09/2023]
Abstract
Aging is associated with progressive changes in learning and memory. A potential approach to attenuate age-related cognitive decline is cognitive training. In this study, adult male and female rats were given either repeated exposure to a T-maze, or no exposure to any maze, and then tested on a final battery of cognitive tasks. Two groups of each sex were tested from 6 to 18 months old on the same T-maze; Group one received a version testing spatial reference memory, and Group two received only the procedural testing components with minimal cognitive demand. Groups three and four of each sex had no maze exposure until the final battery, and were comprised of aged or young rats, respectively. The final maze battery included the practiced T-maze plus two novel tasks, one with a similar, and one with a different, memory type to the practice task. Group five of each sex was not maze tested, serving as an aged control for the effects of maze testing on neurotrophin protein levels in cognitive brain regions. Results showed that adult intermittent cognitive training enhanced performance on the practice task when aged in both sexes, that cognitive training benefits transferred to novel tasks only in females, and that cognitive demand was necessary for these effects, since rats receiving only the procedural testing components showed no improvement on the final maze battery. Further, for both sexes, rats that showed faster learning when young demonstrated better memory when aged. Age-related increases in neurotrophin concentrations in several brain regions were revealed, which were related to performance on the training task only in females. This longitudinal study supports the tenet that cognitive training can help one remember later in life, with broader enhancements and associations with neurotrophins in females.
Collapse
Affiliation(s)
- Joshua S Talboom
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Stephen G West
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Elizabeth B Engler-Chiurazzi
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Craig K Enders
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Ian Crain
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
37
|
Seven A, Yüksel B, Kılıç S, Esen H, Keskin U, Ulubay M, Ozekinci M. Effect of injectable medroxyprogesterone acetate and etonogestrel implants on GABA-A and serotonin receptors in white and gray matter of the brain: experimental study in rats. Gynecol Endocrinol 2014; 30:320-4. [PMID: 24460500 DOI: 10.3109/09513590.2014.880417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to evaluate the time-dependent effect of progesterone-only contraceptives on the brain and to obtain an improved understanding of mood disorders experienced under this medication. A total of 66 Wistar albino rats were divided into three groups: etonogestrel (ENG) implant (group 1, n = 30); depot medroxyprogesterone acetate (MPA)-injectable (group 2, n = 30); and control (group 3, n = 6) groups. Groups 1 and 2 were each divided into five subgroups, which were examined every 10 d for up to 50 d after medication administration, to evaluate its time-dependent effect. There was no difference in terms of gamma-aminobutyric acid (GABA) and serotonin immunohistochemical staining in white and gray matter among the subgroups of group 1. In group 2, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on day 50, when compared to the control group (p = 0.041). When the subgroups of group 2 were compared, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on days 40 and 50 when compared to day 10. In conclusion, we showed that ENG and MPA have no effect on apoptosis and GABA-A receptors in the brain. We also showed that MPA has time-dependent effects on serotonin receptors, which may be a possible mechanism involved in mood disorders during long-term usage of injectable progesterone-only contraceptives.
Collapse
Affiliation(s)
- Ali Seven
- Department of Obstetrics and Gynaecology, Dumlupinar University, Faculty of Medicine , Kutahya , Turkey
| | | | | | | | | | | | | |
Collapse
|
38
|
Koonce CJ, Frye CA. Female mice with deletion of Type One 5α-reductase have reduced reproductive responding during proestrus and after hormone-priming. Pharmacol Biochem Behav 2014; 122:20-9. [PMID: 24650589 DOI: 10.1016/j.pbb.2014.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 03/06/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022]
Abstract
The capacity to form progesterone (P₄)'s 5α-reduced metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP; a.k.a. allopregnanolone), in the brain may be related to facilitation of lordosis among estrogen-primed (E₂) mice. We investigated this idea further by comparing effects of endogenous and exogenous progestogens in mice that are deficient in the Type One 5α-reductase enzyme (5α-reductase knockout mice; 5α-RKO), and their wildtype counterparts for sexual behavior. Comparisons were made following administration of progestogens that are expected to increase 3α,5α-THP or not. Sexual receptivity of 5α-RKO mice and their wildtype counterparts was examined when mice were naturally-cycling (Experiment 1); ovariectomized (OVX), E₂-primed (10 μg, subcutaneous; SC) and administered P₄ (0, 125, 250, or 500 μg SC; Experiment 2); and OVX, E₂-primed and administered P₄, medroxyprogesterone acetate (MPA, 4 mg/kg, SC, which does not convert to 3α,5α-THP) or 3α,5α-THP (4 mg/kg, SC; Experiment 3). The percentage of mounts that elicited lordosis (lordosis quotient) or aggression/rejection behavior (aggression quotient), as well as the quality of lordosis (lordosis rating), was scored. Wildtype, but not 5α-RKO, mice in behavioral estrus demonstrated significantly greater lordosis quotients and lordosis ratings, but similar aggression quotients, compared to their diestrous counterparts. Among OVX and E₂-primed mice, P₄ facilitated lordosis of wildtype, but not 5α-RKO, mice. MPA neither facilitated lordosis of wildtype, nor 5α-RKO mice. 3α,5α-THP administered to wildtype or 5α-RKO mice increased lordosis quotients and lordosis ratings and decreased aggression quotients. 3α,5α-THP levels in the midbrain, one brain region important for sexual behavior, were increased during behavioral estrus, with P4 administered to WT, but not 5α-RKO mice, and 3α,5α-THP administered to WT and 5α-RKO mice. MPA did not increase 3α,5α-THP. Thus, deletion of Type One 5α-reductase among female mice may attenuate reproductive responding during the estrous cycle and after hormone-priming.
Collapse
Affiliation(s)
- Carolyn J Koonce
- Department of Psychology, University at Albany-SUNY, Albany, NY, United States; Institute of Arctic Biology, University of Alaska-Fairbanks, Fairbanks, AK, United States; IDeA Network of Biomedical Excellence (INBRE), University of Alaska-Fairbanks, Fairbanks, AK, United States
| | - Cheryl A Frye
- Department of Psychology, University at Albany-SUNY, Albany, NY, United States; Department of Biological Sciences, University at Albany-SUNY, Albany, NY, United States; The Centers for Neuroscience and Life Sciences Research, University at Albany-SUNY, Albany, NY, United States; Department of Chemistry & Biochemistry, University of Alaska-Fairbanks, Fairbanks, AK, United States; Institute of Arctic Biology, University of Alaska-Fairbanks, Fairbanks, AK, United States; IDeA Network of Biomedical Excellence (INBRE), University of Alaska-Fairbanks, Fairbanks, AK, United States.
| |
Collapse
|
39
|
Mott NN, Pak TR. Estrogen signaling and the aging brain: context-dependent considerations for postmenopausal hormone therapy. ISRN ENDOCRINOLOGY 2013; 2013:814690. [PMID: 23936665 PMCID: PMC3725729 DOI: 10.1155/2013/814690] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/21/2013] [Indexed: 02/08/2023]
Abstract
Recent clinical studies have spurred rigorous debate about the benefits of hormone therapy (HT) for postmenopausal women. Controversy first emerged based on a sharp increase in the risk of cardiovascular disease in participants of the Women's Health Initiative (WHI) studies, suggesting that decades of empirical research in animal models was not necessarily applicable to humans. However, a reexamination of the data from the WHI studies suggests that the timing of HT might be a critical factor and that advanced age and/or length of estrogen deprivation might alter the body's ability to respond to estrogens. Dichotomous estrogenic effects are mediated primarily by the actions of two high-affinity estrogen receptors alpha and beta (ER α & ER β ). The expression of the ERs can be overlapping or distinct, dependent upon brain region, sex, age, and exposure to hormone, and, during the time of menopause, there may be changes in receptor expression profiles, post-translational modifications, and protein:protein interactions that could lead to a completely different environment for E2 to exert its effects. In this review, factors affecting estrogen-signaling processes will be discussed with particular attention paid to the expression and transcriptional actions of ER β in brain regions that regulate cognition and affect.
Collapse
Affiliation(s)
- Natasha N. Mott
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| | - Toni R. Pak
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Effects of long-term treatment with estrogen and medroxyprogesterone acetate on synapse number in the medial prefrontal cortex of aged female rats. Menopause 2012; 19:804-11. [PMID: 22617337 DOI: 10.1097/gme.0b013e31824d1fc4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study investigated the effects of long-term hormone treatment, including the most commonly prescribed progestin, medroxyprogesterone acetate, during aging on synaptophysin-labeled boutons, a marker of synapses, in the medial prefrontal cortex (mPFC) of rats. METHODS Female Long Evans hooded rats were ovariectomized at middle age (12-13 mo) and were placed in one of four groups: no replacement (n = 5), 17β-estradiol alone (n = 6), estradiol and progesterone (n = 7), or estradiol and medroxyprogesterone acetate (n = 4). Estradiol was administered in the drinking water and progestogens were administered via subcutaneous pellets that were replaced every 90 days. After 7 months of hormone replacement, the animals were euthanized, and the brains were stained for synaptophysin, a membrane component of synaptic vesicles. The density of synaptophysin-labeled boutons was quantified in the mPFC using unbiased stereology and multiplied by the volume of the mPFC to obtain the total number. RESULTS Animals receiving estradiol and medroxyprogesterone acetate had significantly more synaptophysin-labeled boutons in the mPFC than did animals not receiving replacement (P < 0.03) and those receiving estradiol and progesterone (P < 0.02). In addition, there was a nonsignificant trend for animals receiving estradiol alone to have more synapses than those receiving estradiol and progesterone. CONCLUSIONS This study is the first to examine the effects of estradiol and medroxyprogesterone acetate during rat aging on cortical synaptic number. Estradiol with medroxyprogesterone acetate, but not progesterone, resulted in a greater number of synapses in the mPFC during aging than did no replacement.
Collapse
|
43
|
Camp BW, Gerson JE, Tsang CWS, Villa SR, Acosta JI, Blair Braden B, Hoffman AN, Conrad CD, Bimonte-Nelson HA. High serum androstenedione levels correlate with impaired memory in the surgically menopausal rat: a replication and new findings. Eur J Neurosci 2012; 36:3086-95. [PMID: 22758646 DOI: 10.1111/j.1460-9568.2012.08194.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
After natural menopause in women, androstenedione becomes the primary hormone secreted by the residual follicle-depleted ovaries. In two independent studies, in rodents that had undergone ovarian follicular depletion, we found that higher endogenous serum androstenedione levels correlated with increased working memory errors. This led to the hypothesis that higher androstenedione levels impair memory. The current study directly tested this hypothesis, examining the cognitive effects of exogenous androstenedione administration in rodents. Middle-aged ovariectomised rats received vehicle or one of two doses of androstenedione. Rats were tested on a spatial working and reference memory maze battery including the water-radial arm maze, Morris water maze (MM) and delay match-to-sample task. Androstenedione at the highest dose impaired reference memory as well as the ability to maintain performance as memory demand was elevated. This was true for both high temporal demand memory retention of one item of spatial information, as well as the ability to handle multiple items of spatial working memory information. We measured glutamic acid decarboxylase (GAD) protein in multiple brain regions to determine whether the gamma-aminobutyric acid (GABA) system relates to androstenedione-induced memory impairments. Results showed that higher entorhinal cortex GAD levels were correlated with worse MM performance, irrespective of androstenedione treatment. These findings suggest that androstenedione, the main hormone produced by the follicle-depleted ovary, is detrimental to working memory, reference memory and memory retention. Furthermore, while spatial reference memory performance might be related to the GABAergic system, it does not appear to be altered with androstenedione administration, at least at the doses used in the current study.
Collapse
Affiliation(s)
- Bryan W Camp
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chisholm NC, Juraska JM. Long-term replacement of estrogen in combination with medroxyprogesterone acetate improves acquisition of an alternation task in middle-aged female rats. Behav Neurosci 2011; 126:128-36. [PMID: 22141470 DOI: 10.1037/a0026461] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Studies have shown that ovarian hormones protect against some of the cognitive deficits associated with aging. Although much of the literature in rodents has focused on hippocampal dependent tasks, studies suggest that tasks dependent on the prefrontal cortex are also influenced by ovarian hormones. The present study investigated the effects of ovarian hormone treatment during aging on a delayed alternation t-maze. Female Long Evans hooded rats were ovariectomized at middle age (11-12 months) and placed in 1 of 5 treatment groups: no replacement, chronic estradiol (E(2)), cyclic E(2), chronic E(2) and progesterone, or chronic E(2) and medroxyprogesterone acetate (MPA). Following 6 months of hormone treatment, animals were trained to alternate in a t-maze. After reaching criterion, a series of delays from 5 to 90 s were introduced in random order. Rats receiving E(2) with MPA reached criterion significantly faster than animals not receiving treatment and those who received chronic or cyclic E(2) only. There was a nonsignificant trend for animals receiving E(2) and progesterone to reach criterion in fewer sessions than animals receiving E(2) only. Mode of administration, cyclic or chronic, did not affect performance. Hormones did not affect performance on the delayed alternation. This study, in combination with previous research, indicates that hormone effects cannot be generalized across tasks, age, or duration, and long-term estrogen in combination with MPA can be beneficial for some tasks.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Psychology, University of Illinois at Urbana, Champaign, IL 61820, USA.
| | | |
Collapse
|