1
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
2
|
Is Hormone Replacement Therapy a Risk Factor or a Therapeutic Option for Alzheimer's Disease? Int J Mol Sci 2023; 24:ijms24043205. [PMID: 36834617 PMCID: PMC9964432 DOI: 10.3390/ijms24043205] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accounts for more than half of all dementia cases in the elderly. Interestingly, the clinical manifestations of AD disproportionately affect women, comprising two thirds of all AD cases. Although the underlying mechanisms for these sex differences are not fully elucidated, evidence suggests a link between menopause and a higher risk of developing AD, highlighting the critical role of decreased estrogen levels in AD pathogenesis. The focus of this review is to evaluate clinical and observational studies in women, which have investigated the impact of estrogens on cognition or attempted to answer the prevailing question regarding the use of hormone replacement therapy (HRT) as a preventive or therapeutic option for AD. The articles were retrieved through a systematic review of the databases: OVID, SCOPUS, and PubMed (keywords "memory", "dementia," "cognition," "Alzheimer's disease", "estrogen", "estradiol", "hormone therapy" and "hormone replacement therapy" and by searching reference sections from identified studies and review articles). This review presents the relevant literature available on the topic and discusses the mechanisms, effects, and hypotheses that contribute to the conflicting findings of HRT in the prevention and treatment of age-related cognitive deficits and AD. The literature suggests that estrogens have a clear role in modulating dementia risk, with reliable evidence showing that HRT can have both a beneficial and a deleterious effect. Importantly, recommendation for the use of HRT should consider the age of initiation and baseline characteristics, such as genotype and cardiovascular health, as well as the dosage, formulation, and duration of treatment until the risk factors that modulate the effects of HRT can be more thoroughly investigated or progress in the development of alternative treatments can be made.
Collapse
|
3
|
Xu M, Chen D, Li H, Wang H, Yang LZ. The Cycling Brain in the Workplace: Does Workload Modulate the Menstrual Cycle Effect on Cognition? Front Behav Neurosci 2022; 16:856276. [PMID: 35722191 PMCID: PMC9201761 DOI: 10.3389/fnbeh.2022.856276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022] Open
Abstract
Recent decades have witnessed increased research efforts to clarify how the menstrual cycle influence females’ cognitive and emotional functions. Despite noticeable progress, the research field faces the challenges of inconsistency and low generalizability of research findings. Females of reproductive ages are a heterogeneous population. Generalizing the results of female undergraduates to women in the workplace might be problematic. Furthermore, the critical cognitive processes for daily life and work deserve additional research efforts for improved ecological validity. Thus, this study investigates cognitive performance across the menstrual cycle using a sample of young nurses with similar duties. We developed a mini-computerized cognitive battery to assess four mental skills critical for nursing work: cognitive flexibility, divided attention, response inhibition, and working memory. Participants completed the cognitive battery at menses, late-follicular, and mid-luteal phases. In addition, they were classified into low- and high workload groups according to their subjective workload ratings. Our results demonstrate a general mid-luteal cognitive advantage. Besides, this study reveals preliminary evidence that workload modulates the menstrual cycle effect on cognition. Only females of low workload manifest the mid-luteal cognitive advantage on divided attention and response inhibition, implying that a suitable workload threshold might be necessary for regular neuro-steroid interactions. Thus, this study advocates the significance of research focusing on the cycling brain under workloads.
Collapse
Affiliation(s)
- Min Xu
- Bengbu Medical College, Bengbu, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Dandan Chen
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Hai Li
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Hongzhi Wang
- Bengbu Medical College, Bengbu, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- *Correspondence: Hongzhi Wang,
| | - Li-Zhuang Yang
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Li-Zhuang Yang,
| |
Collapse
|
4
|
Koebele SV, Poisson ML, Palmer JM, Berns-Leone C, Northup-Smith SN, Peña VL, Strouse IM, Bulen HL, Patel S, Croft C, Bimonte-Nelson HA. Evaluating the Cognitive Impacts of Drospirenone, a Spironolactone-Derived Progestin, Independently and in Combination With Ethinyl Estradiol in Ovariectomized Adult Rats. Front Neurosci 2022; 16:885321. [PMID: 35692432 PMCID: PMC9177129 DOI: 10.3389/fnins.2022.885321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/14/2022] [Indexed: 11/15/2022] Open
Abstract
Oral contraceptives and hormone therapies require a progestogen component to prevent ovulation, curtail uterine hyperplasia, and reduce gynecological cancer risk. Diverse classes of synthetic progestogens, called progestins, are used as natural progesterone alternatives due to progesterone’s low oral bioavailability. Progesterone and several synthetic analogs can negatively impact cognition and reverse some neuroprotective estrogen effects. Here, we investigate drospirenone, a spironolactone-derived progestin, which has unique pharmacological properties compared to other clinically-available progestins and natural progesterone, for its impact on spatial memory, anxiety-like behavior, and brain regions crucial to these cognitive tasks. Experiment 1 assessed three drospirenone doses in young adult, ovariectomized rats, and found that a moderate drospirenone dose benefited spatial memory. Experiment 2 investigated this moderate drospirenone dose with and without concomitant ethinyl estradiol (EE) treatment, the most common synthetic estrogen in oral contraceptives. Results demonstrate that the addition of EE to drospirenone administration reversed the beneficial working memory effects of drospirenone. The hippocampus, entorhinal cortex, and perirhinal cortex were then probed for proteins known to elicit estrogen- and progestin- mediated effects on learning and memory, including glutamate decarboxylase (GAD)65, GAD67, and insulin-like growth factor receptor protein expression, using western blot. EE increased GAD expression in the perirhinal cortex. Taken together, results underscore the necessity to consider the distinct cognitive and neural impacts of clinically-available synthetic estrogen and progesterone analogs, and why they produce unique cognitive profiles when administered together compared to those observed when each hormone is administered separately.
Collapse
Affiliation(s)
- Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Mallori L. Poisson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Justin M. Palmer
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Claire Berns-Leone
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Steven N. Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Veronica L. Peña
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Isabel M. Strouse
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Haidyn L. Bulen
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Corissa Croft
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- *Correspondence: Heather A. Bimonte-Nelson,
| |
Collapse
|
5
|
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: 1.7] [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
|
6
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|
7
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
8
|
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: 4.3] [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
|
9
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2018. [PMID: 29311911 DOI: 10.3389/fnagi.2017.00430/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
10
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2017; 9:430. [PMID: 29311911 PMCID: PMC5743731 DOI: 10.3389/fnagi.2017.00430] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
11
|
Hamson DK, Roes MM, Galea LAM. Sex Hormones and Cognition: Neuroendocrine Influences on Memory and Learning. Compr Physiol 2016; 6:1295-337. [DOI: 10.1002/cphy.c150031] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Tskitishvili E, Pequeux C, Munaut C, Viellevoye R, Nisolle M, Noël A, Foidart JM. Use of estetrol with other steroids for attenuation of neonatal hypoxic-ischemic brain injury: to combine or not to combine? Oncotarget 2016; 7:33722-43. [PMID: 27231853 PMCID: PMC5085115 DOI: 10.18632/oncotarget.9591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Estetrol (E4), estradiol (E2) and progesterone (P4) have important antioxidative and neuroprotective effects in neuronal system. We aimed to study the consequence of combined steroid therapy in neonatal hypoxic-ischemic encephalopathy (HIE). In vitro the effect of E4 combined with other steroids on oxidative stress and the cell viability in primary hippocampal cultures was evaluated by lactate dehydrogenase and cell survival assays. In vivo neuroprotective and therapeutic efficacy of E4 combined with other steroids was studied in HIE model of immature rats. The rat pups rectal temperature, body and brain weights were evaluated.The hippocampus and the cortex were investigated by histo/immunohistochemistry: intact cell number counting, expressions of markers for early gray matter lose, neuro- and angiogenesis were studied. Glial fibrillary acidic protein was evaluated by ELISA in blood samples. In vitro E4 and combinations of high doses of E4 with P4 and/or E2 significantly diminished the LDH activity and upregulated the cell survival.In vivopretreatment or treatment by different combinations of E4 with other steroids had unalike effects on body and brain weight, neuro- and angiogenesis, and GFAP expression in blood. The combined use of E4 with other steroids has no benefit over the single use of E4.
Collapse
Affiliation(s)
- Ekaterine Tskitishvili
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Christel Pequeux
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Carine Munaut
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Renaud Viellevoye
- Neonatal Intensive Care Unit, Department of Pediatrics, University of Liege, Liege 1, Belgium
| | - Michelle Nisolle
- Department of Obstetrics and Gynecology, University of Liege, Liege1, Belgium
| | - Agnes Noël
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Jean-Michel Foidart
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| |
Collapse
|
13
|
Graham BM, Daher M. Estradiol and Progesterone have Opposing Roles in the Regulation of Fear Extinction in Female Rats. Neuropsychopharmacology 2016; 41:774-80. [PMID: 26156559 PMCID: PMC4707823 DOI: 10.1038/npp.2015.202] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/05/2015] [Accepted: 07/06/2015] [Indexed: 01/25/2023]
Abstract
Fear extinction, the laboratory basis of exposure therapy for anxiety disorders, fluctuates across the female rat estrous cycle, where extinction is enhanced during proestrus (high estradiol and progesterone), and impaired during metestrus (low estradiol and progesterone). During the estrous cycle increasing levels of estradiol precede and then overlap with increased levels of progesterone. We sought to isolate the impact of these hormonal changes on fear extinction by systematically treating ovariectomized female rats with estradiol alone, or in combination with progesterone. We found that estradiol alone facilitated extinction recall, whereas the effects of progesterone on estradiol-treated rats were biphasic and dependent on the time interval between progesterone administration and extinction training. Progesterone potentiated estradiol's facilitation of extinction recall when extinction training occurred 6 h after progesterone administration. However, progesterone abolished estradiol's facilitation of extinction recall when extinction training occurred 24 h after progesterone administration. Furthermore, in naturally cycling rats, blocking progesterone receptor activation during proestrus (when progesterone levels peak) prevented the impairment in extinction recall in rats extinguished during metestrus. These results suggest that in naturally cycling females whereas cyclical increases in estradiol facilitate fear extinction, cyclical increases in progesterone may lead to fear extinction impairments. As extinction training took place after the hormonal treatments had been metabolized, we propose that genomic mechanisms may at least partly mediate the impact of cyclic fluctuations in sex hormones on fear extinction.
Collapse
Affiliation(s)
- Bronwyn M Graham
- School of Psychology, The University of New South Wales Australia, Sydney, NSW, Australia,School of Psychology, The University of New South Wales Australia, Sydney, NSW 2052, Australia, Tel: +61293853886, Fax: +61293853641, E-mail:
| | - Melissa Daher
- School of Psychology, The University of New South Wales Australia, Sydney, NSW, Australia
| |
Collapse
|
14
|
Giatti S, Garcia-Segura LM, Melcangi RC. New steps forward in the neuroactive steroid field. J Steroid Biochem Mol Biol 2015; 153:127-34. [PMID: 25797031 DOI: 10.1016/j.jsbmb.2015.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/07/2015] [Accepted: 03/17/2015] [Indexed: 12/12/2022]
Abstract
Evidence accumulated in recent years suggests that the systemic treatment with neuroactive steroids, or the pharmacological modulation of its production by brain cells, represent therapeutic options to promote neuroprotection. However, new findings, which are reviewed in this paper, suggest that the factors to be considered for the design of possible therapies based on neuroactive steroids are more complex than previously thought. Thus, although as recently reported, the nervous system regulates neuroactive steroid synthesis and metabolism in adaptation to modifications in peripheral steroidogenesis, the neuroactive steroid levels in the brain do not fully reflect its levels in plasma. Even, in some cases, neuroactive steroid level modifications occurring in the nervous tissues, under physiological and pathological conditions, are in the opposite direction than in the periphery. This suggests that the systemic treatment with these molecules may have unexpected outcomes on neural steroid levels. In addition, the multiple metabolic pathways and signaling mechanisms of neuroactive steroids, which may change from one brain region to another, together with the existence of regional and sex differences in its neural levels are additional sources of complexity that should be clarified. This complexity in the levels and actions of these molecules may explain why in some cases these molecules have detrimental rather than beneficial actions for the nervous system. This article is part of a Special Issue entitled 'Steroid Perspectives'.
Collapse
Affiliation(s)
- Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
15
|
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: 1.9] [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
|
16
|
Pooley AE, Luong M, Hussain A, Nathan BP. Neurite outgrowth promoting effect of 17-β estradiol is mediated through estrogen receptor alpha in an olfactory epithelium culture. Brain Res 2015. [PMID: 26206299 DOI: 10.1016/j.brainres.2015.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Olfactory deficits are observed early in the course of chronic neurological disorders including Alzheimer's disease (AD). Estrogen treatment in post-menopausal women reduced the incidence of olfactory dysfunction, raising the possibility that estrogen treatment can cure olfactory deficits in preclinical stages of AD. In this study, we examined the estradiol׳s effects on neurite outgrowth in explant cultures of mouse olfactory epithelium (OE). We found that neurons in OE cultures treated with 100 pM 17-β estradiol (estradiol) had significantly longer neurite outgrowth than cultures treated with ethanol alone (vehicle). The OE neurons expressed estrogen receptors alpha (ERα) and ER beta (ERβ). Estrogen treatment upregulated both ERα and ERβ expression in OE culture. Treatment of OE cultures with propyl pyrazole triol (PPT), a selective agonist for ERα increased neurite outgrowth to comparable extent as estradiol treatment. In contrast, 2,3-bis-4-hydroxyphenyl (DPN), a specific agonist for ERβ, had no effect on neurite outgrowth. Furthermore, estradiol treatment increased neurite outgrowth in OE cultures derived from ERβ-deficient/knockout mice and wild-type littermates, but not in ERα-deficient/knockout mice. These data suggest that ERα mediates the neurite outgrowth promoting effects of estradiol in OE cultures. We propose that olfactory dysfunction in chronic neurological disorders, where estrogen deficiency is a risk factor, is an indicator of compromised axonal regeneration of olfactory sensory neurons.
Collapse
Affiliation(s)
- Apryl E Pooley
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Minh Luong
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Aseem Hussain
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States
| | - Britto P Nathan
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston IL 61920, United States.
| |
Collapse
|
17
|
Barron AM, Brown MA, Morgan TE, Pike CJ. Impact of continuous versus discontinuous progesterone on estradiol regulation of neuron viability and sprouting after entorhinal cortex lesion in female rats. Endocrinology 2015; 156:1091-9. [PMID: 25514084 PMCID: PMC4330320 DOI: 10.1210/en.2014-1216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Because the estrogen-based hormone therapy (HT) in postmenopausal women typically contains a progestogen component, understanding the interactions between estrogens and progestogens is critical for optimizing the potential neural benefits of HT. An important issue in this regard is the use of continuous vs discontinuous hormone treatments. Although sex steroid hormone levels naturally exhibit cyclic fluctuation, many HT formulations include continuous delivery of hormones. Recent findings from our laboratory and others have shown that coadministration of progesterone (P4) can either attenuate or augment beneficial actions of 17β-estradiol (E2) in experimental models depending in part upon the delivery schedule of P4. In this study, we demonstrate that the P4 delivery schedule in combined E2 and P4 treatments alters degenerative and regenerative outcomes of unilateral entorhinal cortex lesion. We assessed how lesion-induced degeneration of layer II neurons in entorhinal cortex layer and deafferentation in dentate gyrus are affected by ovariectomy and treatments with E2 alone or in combination with either continuous or discontinuous P4. Our results demonstrate the combined efficacy of E2 and P4 is dependent on the administration regimen. Importantly, the discontinuous-combined E2+P4 regimen had the greatest neuroprotective efficacy for both end points. These data extend a growing literature that indicates qualitative differences in the neuroprotective effects of E2 as a function of cotreatment with continuous versus discontinuous P4, the understanding of which has important implications for HT in postmenopausal women.
Collapse
Affiliation(s)
- Anna M Barron
- Davis School of Gerontology (A.M.B., M.A.B., T.E.M., C.J.P.), University of Southern California, Los Angeles, California 90089; Molecular Imaging Center (A.M.B.), National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | | | | | | |
Collapse
|
18
|
Jayaraman A, Pike CJ. Differential effects of synthetic progestagens on neuron survival and estrogen neuroprotection in cultured neurons. Mol Cell Endocrinol 2014; 384:52-60. [PMID: 24424444 PMCID: PMC3954450 DOI: 10.1016/j.mce.2014.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 11/22/2013] [Accepted: 01/03/2014] [Indexed: 11/19/2022]
Abstract
Progesterone and other progestagens are used in combination with estrogens for clinical purposes, including contraception and postmenopausal hormone therapy. Progesterone and estrogens have interactive effects in brain, however interactions between synthetic progestagens and 17β-estradiol (E2) in neurons are not well understood. In this study, we investigated the effects of seven clinically relevant progestagens on estrogen receptor (ER) mRNA expression, E2-induced neuroprotection, and E2-induced BDNF mRNA expression. We found that medroxyprogesterone acetate decreased both ERα and ERβ expression and blocked E2-mediated neuroprotection and BDNF expression. Conversely, levonorgestrel and nesterone increased ERα and or ERβ expression, were neuroprotective, and failed to attenuate E2-mediated increases in neuron survival and BDNF expression. Other progestagens tested, including norethindrone, norethindrone acetate, norethynodrel, and norgestimate, had variable effects on the measured endpoints. Our results demonstrate a range of qualitatively different actions of progestagens in cultured neurons, suggesting significant variability in the neural effects of clinically utilized progestagens.
Collapse
Affiliation(s)
- Anusha Jayaraman
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
19
|
Sfikakis A, Pitychoutis PM, Tsouma A, Messari I, Papadopoulou-Daifoti Z. Effects of testosterone and estradiol on stress-induced adrenal and hippocampal weight changes in female rats. Hormones (Athens) 2014; 13:119-30. [PMID: 24722133 DOI: 10.1007/bf03401327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To examine the impact of circulating testosterone (T) and the T/Estradiol (T/Ediol) ratio on chronic stress-induced changes of adrenal and hippocampal weight during proestrus (PE) and estrus (E) in female rats. DESIGN Stress was composed of repeated vaginal smear screening (VSS) and measured by the emotional reactivity score (ERS). Adrenal and hippocampal weight and the T, Ediol and T/Ediol ratio were assessed in PE and E controls as well as 20 h after sham or left adrenalectomy performed on diestrus-2 (DE-2) and PE, respectively. T was measured in ovariectomized (OVX) rats treated with estradiol benzoate (EB) or vehicle (VEH) and in non-OVX EB-treated rats. RESULTS In OVX rats EB treatment increased adrenal weight and T levels. After separation of VEH- and EB-treated rats into the low and high T-range (below and above the mean, respectively), it was observed that higher T was accompanied by higher adrenal weight in EB- compared to VEH-treated rats only in the low T-range. Non-OVX EB-treated rats with high T had lower adrenal weight compared to low T. Cycling rats assigned to the high T-range presented higher T/Ediol ratio but similar ERS and Ediol levels compared to rats in the low T-range, and were characterized by reduced adrenal weight, higher hippocampal weight and prevalence of PE versus E. CONCLUSIONS High T and high T/Ediol ratios are prominent in PE compared to E and exert a protective effect on hippocampal neuronal degeneration after similar chronic stress through T-mediated lessening of stress response thus counteracting the stress-promoting effects of Ediol.
Collapse
Affiliation(s)
- Anastasia Sfikakis
- Department of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Pothitos M Pitychoutis
- Department of Biology & Center for Tissue Regeneration and Engineering (TREND), University of Dayton, Dayton, USA
| | - Aikaterini Tsouma
- Department of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Messari
- Department of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Zeta Papadopoulou-Daifoti
- Department of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Baudry M, Bi X, Aguirre C. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience 2013; 239:280-94. [PMID: 23142339 PMCID: PMC3628409 DOI: 10.1016/j.neuroscience.2012.10.051] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/23/2012] [Accepted: 10/26/2012] [Indexed: 01/01/2023]
Abstract
17ß-Estradiol and progesterone exert a number of physiological effects throughout the brain due to interactions with several types of receptors belonging to the traditional family of intracellular hormonal receptors as well as to membrane-bound receptors. In particular, both hormones elicit rapid modifications of neuronal excitability that have been postulated to underlie their effects on synaptic plasticity and learning and memory. Likewise, both hormones have been shown to be neuroprotective under certain conditions, possibly due to the activation of pro-survival pathways and the inhibition of pro-apoptotic cascades. Because of the similarities in their cellular effects, there have been a number of questions raised by numerous observations that progesterone inhibits the effects of estrogen. In this manuscript, we first review the interactions between 17ß-estradiol (E2) and progesterone (P4) in synaptic plasticity, and conclude that, while E2 exerts a clear and important role in long-term potentiation of synaptic transmission in hippocampal neurons, the role of P4 is much less clear, and could be accounted by the direct or indirect regulation of GABAA receptors. We then discuss the neuroprotective roles of both hormones, in particular against excitotoxicity. In this case, the neuroprotective effects of these hormones are very similar to those of the neurotrophic factor BDNF. Interestingly, P4 antagonizes the effects of E2, possibly through the regulation of estrogen receptors or of proteins associated with the receptors or interactions with signaling pathways activated by E2. Overall, this review emphasizes the existence of common molecules and pathways that participate in the regulation of both synaptic plasticity and neurodegeneration.
Collapse
Affiliation(s)
- M Baudry
- GCBS and COMP, Western University of Health Sciences, Pomona, CA, USA.
| | | | | |
Collapse
|
21
|
Abstract
Numerous studies aimed at identifying the role of estrogen on the brain have used the ovariectomized rodent as the experimental model. And while estrogen intervention in these animals has, at least partially, restored cholinergic, neurotrophin and cognitive deficits seen in the ovariectomized animal, it is worth considering that the removal of the ovaries results in the loss of not only circulating estrogen but of circulating progesterone as well. As such, the various deficits associated with ovariectomy may be attributed to the loss of progesterone as well. Similarly, one must also consider the fact that the human menopause results in the precipitous decline of not just circulating estrogens, but in circulating progesterone as well and as such, the increased risk for diseases such as Alzheimer's disease during the postmenopausal period could also be contributed by this loss of progesterone. In fact, progesterone has been shown to exert neuroprotective effects, both in cell models, animal models and in humans. Here, we review the evidence that supports the neuroprotective effects of progesterone and discuss the various mechanisms that are thought to mediate these protective effects. We also discuss the receptor pharmacology of progesterone's neuroprotective effects and present a conceptual model of progesterone action that supports the complementary effects of membrane-associated and classical intracellular progesterone receptors. In addition, we discuss fundamental differences in the neurobiology of progesterone and the clinically used, synthetic progestin, medroxyprogesterone acetate that may offer an explanation for the negative findings of the combined estrogen/progestin arm of the Women's Health Initiative-Memory Study (WHIMS) and suggest that the type of progestin used may dictate the outcome of either pre-clinical or clinical studies that addresses brain function.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, Center FOR HER, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107, USA.
| | | |
Collapse
|
22
|
|
23
|
Progesterone, brain-derived neurotrophic factor and neuroprotection. Neuroscience 2012; 239:84-91. [PMID: 23036620 DOI: 10.1016/j.neuroscience.2012.09.056] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/20/2012] [Accepted: 09/23/2012] [Indexed: 01/06/2023]
Abstract
While the effects of progesterone in the CNS, like those of estrogen, have generally been considered within the context of reproductive function, growing evidence supports its importance in regulating non-reproductive functions including cognition and affect. In addition, progesterone has well-described protective effects against numerous insults in a variety of cell models, animal models and in humans. While ongoing research in several laboratories continues to shed light on the mechanism(s) by which progesterone and its related progestins exert their effects in the CNS, our understanding is still incomplete. Among the key mediators of progesterone's beneficial effects is the family of growth factors called neurotrophins. Here, we review the mechanisms by which progesterone regulates one important member of the neurotrophin family, brain-derived neurotrophic factor (BDNF), and provides support for its pivotal role in the protective program elicited by progesterone in the brain.
Collapse
|
24
|
Diaz Brinton R. Minireview: translational animal models of human menopause: challenges and emerging opportunities. Endocrinology 2012; 153:3571-8. [PMID: 22778227 PMCID: PMC3404353 DOI: 10.1210/en.2012-1340] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/31/2012] [Indexed: 02/02/2023]
Abstract
Increasing importance is placed on the translational validity of animal models of human menopause to discern risk vs. benefit for prediction of outcomes after therapeutic interventions and to develop new therapeutic strategies to promote health. Basic discovery research conducted over many decades has built an extensive body of knowledge regarding reproductive senescence across mammalian species upon which to advance animal models of human menopause. Modifications to existing animal models could rapidly address translational gaps relevant to clinical issues in human menopausal health, which include the impact of 1) chronic ovarian hormone deprivation and hormone therapy, 2) clinically relevant hormone therapy regimens (cyclic vs. continuous combined), 3) clinically relevant hormone therapy formulations, and 4) windows of opportunity and optimal duration of interventions. Modifications in existing animal models to more accurately represent human menopause and clinical interventions could rapidly provide preclinical translational data to predict outcomes regarding unresolved clinical issues relevant to women's menopausal health. Development of the next generation of animal models of human menopause could leverage advances in identifying genotypic variations in estrogen and progesterone receptors to develop personalized menopausal care and to predict outcomes of interventions for protection against or vulnerability to disease. Key to the success of these models is the close coupling between the translational target and the range of predictive validity. Preclinical translational animal models of human menopause need to keep pace with changes in clinical practice. With focus on predictive validity and strategic use of advances in genetic and epigenetic science, new animal models of human menopause have the opportunity to set new directions for menopausal clinical care for women worldwide.
Collapse
Affiliation(s)
- Roberta Diaz Brinton
- Pharmacology and Pharmaceutical Sciences, University of Southern California, School of Pharmacy, 1985 Zonal Avenue, PSC-502, Los Angeles, California 90033, USA.
| |
Collapse
|
25
|
Bian C, Zhu K, Yang L, Lin S, Li S, Su B, Zhang J. Gonadectomy differentially regulates steroid receptor coactivator-1 and synaptic proteins in the hippocampus of adult female and male C57BL/6 mice. Synapse 2012; 66:849-57. [PMID: 22623226 DOI: 10.1002/syn.21574] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/14/2012] [Indexed: 02/06/2023]
Abstract
Hippocampus is one of the most important structures that mediates learning and memory, cognition, and mental behaviors and profoundly regulated by sex hormones in a sex-specific manner, but the mechanism of underlying sex differences regulation is still unclear. We have previously reported that in the male and female mice, steroid receptor coactivator-1 (SRC-1) and some key synaptic proteins share similar developmental profile in the hippocampus, but how circulating sex hormones affect hippocampal SRC-1 as well as these synaptic proteins remain unclear. In this study, we examined how gonad sex hormones regulate hippocampal SRC-1, synaptophysin, PSD-95, and AMPA receptor subtype GluR1 by using immunohistochemistry and Western blot. The results showed that in the female mice, ovariectomy affected hippocampal SRC-1 and GluR1 were only detected at 2 weeks post operation, then it recovered to sham level; synaptophysin was unaffected at any timepoint examined; significant decrease of PSD-95 was only detected at 4 weeks post operation. However, in the male hippocampus, SRC-1 and PSD-95 were decreased from one week and lasted to 4 weeks after orchidectomy, GluR1 decreased from 2 weeks after orchidectomy, but synaptophysin remained unchanged as in the females. Correlation analysis showed the profiles of SRC-1 were positively correlated with GluR1 of the females, PSD-95 and GluR1 of the males, respectively. The above results suggested a distinct regulatory mode between female and male gonad hormones in the regulation of hippocampal SRC-1 and synaptic proteins, which may be one of the mechanisms contributing to the dimorphism of hippocampus during development and ageing.
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Lazaroni TL, Raslan ACS, Fontes WR, de Oliveira ML, Bader M, Alenina N, Moraes MF, dos Santos RA, Pereira GS. Angiotensin-(1–7)/Mas axis integrity is required for the expression of object recognition memory. Neurobiol Learn Mem 2012; 97:113-23. [DOI: 10.1016/j.nlm.2011.10.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 09/28/2011] [Accepted: 10/17/2011] [Indexed: 12/22/2022]
|
27
|
Abstract
A promising strategy to delay and perhaps prevent Alzheimer's disease (AD) is to identify the age-related changes that put the brain at risk for the disease. A significant normal age change known to result in tissue-specific dysfunction is the depletion of sex hormones. In women, menopause results in a relatively rapid loss of estradiol and progesterone. In men, aging is associated with a comparatively gradual yet significant decrease in testosterone. We review a broad literature that indicates age-related losses of estrogens in women and testosterone in men are risk factors for AD. Both estrogens and androgens exert a wide range of protective actions that improve multiple aspects of neural health, suggesting that hormone therapies have the potential to combat AD pathogenesis. However, translation of experimental findings into effective therapies has proven challenging. One emerging treatment option is the development of novel hormone mimetics termed selective estrogen and androgen receptor modulators. Continued research of sex hormones and their roles in the aging brain is expected to yield valuable approaches to reducing the risk of AD.
Collapse
Affiliation(s)
- Anna M. Barron
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Japan
| | - Christian J. Pike
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
28
|
Barron AM, Pike CJ. Sex hormones, aging, and Alzheimer's disease. Front Biosci (Elite Ed) 2012. [PMID: 22201929 DOI: 10.2741/434] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A promising strategy to delay and perhaps prevent Alzheimer's disease (AD) is to identify the age-related changes that put the brain at risk for the disease. A significant normal age change known to result in tissue-specific dysfunction is the depletion of sex hormones. In women, menopause results in a relatively rapid loss of estradiol and progesterone. In men, aging is associated with a comparatively gradual yet significant decrease in testosterone. We review a broad literature that indicates age-related losses of estrogens in women and testosterone in men are risk factors for AD. Both estrogens and androgens exert a wide range of protective actions that improve multiple aspects of neural health, suggesting that hormone therapies have the potential to combat AD pathogenesis. However, translation of experimental findings into effective therapies has proven challenging. One emerging treatment option is the development of novel hormone mimetics termed selective estrogen and androgen receptor modulators. Continued research of sex hormones and their roles in the aging brain is expected to yield valuable approaches to reducing the risk of AD.
Collapse
Affiliation(s)
- Anna M Barron
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | | |
Collapse
|
29
|
Nakamagoe M, Tabuchi K, Nishimura B, Hara A. Effects of neuroactive steroids on cochlear hair cell death induced by gentamicin. Steroids 2011; 76:1443-50. [PMID: 21856322 DOI: 10.1016/j.steroids.2011.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 07/27/2011] [Accepted: 07/27/2011] [Indexed: 01/22/2023]
Abstract
As neuroactive steroids, sex steroid hormones have non-reproductive effects. We previously reported that 17β-estradiol (βE2) had protective effects against gentamicin (GM) ototoxicity in the cochlea. In the present study, we examined whether the protective action of βE2 on GM ototoxicity is mediated by the estrogen receptor (ER) and whether other estrogens (17α-estradiol (αE2), estrone (E1), and estriol (E3)) and other neuroactive steroids, dehydroepiandrosterone (DHEA) and progesterone (P), have similar protective effects. The basal turn of the organ of Corti was dissected from Sprague-Dawley rats and cultured in a medium containing 100 μM GM for 48h. The effects of βE2 and ICI 182,780, a selective ER antagonist, were examined. In addition, the effects of other estrogens, DHEA and P were tested using this culture system. Loss of outer hair cells induced by GM exposure was compared among groups. βE2 exhibited a protective effect against GM ototoxicity, but its protective effect was antagonized by ICI 182,780. αE2, E1, and E3 also protected hair cells against gentamicin ototoxicity. DHEA showed a protective effect; however, the addition of ICI 182,780 did not affect hair cell loss. P did not have any effect on GM-induced outer hair cell death. The present findings suggest that estrogens and DHEA are protective agents against GM ototoxicity. The results of the ER antagonist study also suggest that the protective action of βE2 is mediated via ER but that of DHEA is not related to its conversion to estrogen and binding to ER. Further studies on neuroactive steroids may lead to new insights regarding cochlear protection.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/adverse effects
- Cell Death/drug effects
- Cochlea/cytology
- Cytoprotection/drug effects
- Dehydroepiandrosterone/pharmacology
- Estradiol/pharmacology
- Estrogens/pharmacology
- Gene Expression Regulation/drug effects
- Gentamicins/adverse effects
- Hair Cells, Auditory/cytology
- Hair Cells, Auditory/drug effects
- Hair Cells, Auditory/metabolism
- Hair Cells, Auditory, Outer/cytology
- Hair Cells, Auditory, Outer/drug effects
- Hair Cells, Auditory, Outer/metabolism
- Progesterone/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/metabolism
- Steroids/pharmacology
Collapse
Affiliation(s)
- Mariko Nakamagoe
- Department of Otolaryngology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | |
Collapse
|
30
|
Frye CA, Walf A. Progesterone, administered before kainic acid, prevents decrements in cognitive performance in the Morris Water Maze. Dev Neurobiol 2011; 71:142-52. [PMID: 20715152 DOI: 10.1002/dneu.20832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The nature of progesterone (P₄)'s neuroprotective effects is of interest. We investigated effects of P₄ when administered before, or after, kainic acid, which produces ictal activity and damage to the hippocampus, to mediate effects on spatial performance. The hypothesis was that P₄, compared with vehicle, would reduce decrements in Morris Water Maze performance induced by kainic acid. Experiment 1: We examined the effects of kainic acid on plasma stress hormone, corticosterone, and progestogen (P₄ and its metabolites) levels in plasma and the hippocampus after subcutaneous (s.c.) P₄ administration to ovariectomized rats. Rats administered kainic acid had the highest corticosterone levels immediately following injection. P₄ is 5α-reduced to dihydroprogesterone (DHP) and subsequently metabolized to 5α-pregnan-3α-ol-20-one (3α,5α-THP) by 3α-hydroxysteroid dehydrogenase. The regimen of P₄ used produced circulating and hippocampal levels of P₄, DHP, and 3α,5α-THP within a physiological range, which declined at 14 hours postinjection and were not altered by kainic acid. Experiment 2: The physiological P₄ regimen was administered to rats before, or after, kainic acid-induced seizures, and later effects on water maze performance were compared with that of rats administered vehicle. Rats administered kainic acid had significantly poorer performance in the water maze (i.e., increased latencies and distances to the hidden platform) than did rats administered vehicle. Administration of P₄ before, but not after, kainic acid prevented these performance deficits. Thus, these data suggest that a physiological regimen of P₄ can prevent some of the deficits in water maze performance produced by kainic acid.
Collapse
Affiliation(s)
- Cheryl A Frye
- Department of Psychology, The University at Albany-SUNY, 1400 Washington Avenue, Albany, New York 12222, USA.
| | | |
Collapse
|
31
|
Yao J, Chen S, Cadenas E, Brinton RD. Estrogen protection against mitochondrial toxin-induced cell death in hippocampal neurons: antagonism by progesterone. Brain Res 2011; 1379:2-10. [PMID: 21134358 PMCID: PMC3200366 DOI: 10.1016/j.brainres.2010.11.090] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 11/06/2010] [Accepted: 11/29/2010] [Indexed: 02/06/2023]
Abstract
Previously we demonstrated that mitochondrial dysfunction plays a critical role in the pathogenesis of Alzheimer's disease. Further, we have shown that the neuroprotective effects of 17β-estradiol (E2) are dependent upon mitochondrial function. In the current study, we sought to identify mitochondrial sites of E2 action that mediate neuroprotection by assessing the efficacy of E2 to protect neurons against inhibitors of mitochondrial respiration which target specific complexes within the respiratory chain. Subsequently, the impact of progesterone (P4) on E2-induced prevention against mitochondrial toxins was investigated. Mitochondrial inhibitors, rotenone, 3-NPA, antimycin, KCN, and oligomycin, exhibited concentration dependent toxicity in primary hippocampal neurons. The concentration inducing 30% cell death (LD30) was selected for analyses assessing the neuroprotective efficacy of ovarian hormones (E2 and P4). Pretreatment of hippocampal neurons with E2 significantly protected against 3-NPA (7.5mM) and antimycin (125 μM) induced cell death and was moderately neuroprotective against rotenone (3 μM). E2 was ineffective against KCN and oligomycin-induced cell death. Pretreatment with P4 was without effect against these mitochondrial inhibitors. Co-administration of P4 with E2 abolished E2 induced neuroprotection against 3-NPA and antimycin. Additional metabolic analyses indicated that E2 and P4 separately increased mitochondrial respiratory capacity whereas the co-administration of E2 and P4 resulted in diminished mitochondrial respiration. These findings indicate that E2 protects against mitochondrial toxins that target complexes I, II and III whereas P4 was without effect. The data also predict that continuous combined co-administration of estrogen and progesterone common to many hormone therapy regimens is unlikely to prevent the deficits in mitochondrial function.
Collapse
Affiliation(s)
- Jia Yao
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
32
|
Zhang D, Guo Q, Bian C, Zhang J, Cai W, Su B. Expression of Steroid Receptor Coactivator-1 Was Regulated by Postnatal Development but Not Ovariectomy in the Hippocampus of Rats. Dev Neurosci 2011; 33:57-63. [DOI: 10.1159/000322978] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/20/2010] [Indexed: 11/19/2022] Open
|
33
|
Aguirre C, Jayaraman A, Pike C, Baudry M. Progesterone inhibits estrogen-mediated neuroprotection against excitotoxicity by down-regulating estrogen receptor-β. J Neurochem 2010; 115:1277-87. [PMID: 20977477 DOI: 10.1111/j.1471-4159.2010.07038.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
While both 17β-estradiol (E2) and progesterone (P4) are neuroprotective in several experimental paradigms, P4 also counteracts E2 neuroprotective effects. We recently reported that a 4-h treatment of cultured hippocampal slices with P4 following a prolonged (20 h) treatment with E2 eliminated estrogenic neuroprotection against NMDA toxicity and induction of brain-derived neurotrophic factor (BDNF) expression. In the present study, we evaluated the effects of the same treatment on levels of estrogen receptors, ERα and ERβ, and BDNF using a similar paradigm. E2 treatment resulted in elevated ERβ mRNA and protein levels, did not modify ERα mRNA, but increased ERα protein levels, and increased BDNF mRNA levels. P4 reversed E2-elicited increases in ERβ mRNA and protein levels, in ERα protein levels, and in BDNF mRNA levels. Experiments with an ERβ-specific antagonist, PHTPP, and specific agonists of ERα and ERβ, propylpyrazoletriol and diarylpropionitrile, respectively, indicated that E2-mediated neuroprotection against NMDA toxicity was, at least in part, mediated via ERβ receptor. In support of this conclusion, E2 did not protect against NMDA toxicity in cultured hippocampal slices from ERβ-/- mice. Thus, E2-mediated neuroprotection against NMDA toxicity may be because of estrogenic induction of BDNF via its ERβ receptor, and P4-mediated inhibition of E2 neuroprotective effects treatment to P4-induced down-regulation of ERβ and BDNF.
Collapse
Affiliation(s)
- Claudia Aguirre
- Neuroscience Program, University of Southern California, Los Angeles, CA 90089-2520, USA
| | | | | | | |
Collapse
|
34
|
Carroll JC, Rosario ER, Villamagna A, Pike CJ. Continuous and cyclic progesterone differentially interact with estradiol in the regulation of Alzheimer-like pathology in female 3xTransgenic-Alzheimer's disease mice. Endocrinology 2010; 151:2713-22. [PMID: 20410196 PMCID: PMC2875823 DOI: 10.1210/en.2009-1487] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Depletion of estrogens and progesterone at menopause has been linked to an increased risk for the development of Alzheimer's disease (AD) in women. A currently controversial literature indicates that although treatment of postmenopausal women with hormone therapy (HT) may reduce the risk of AD, several parameters of HT may limit its potential efficacy and perhaps, even exacerbate AD risk. One such parameter is continuous vs. cyclic delivery of the progestogen component of HT. Recent experimental evidence suggests that continuous progesterone can attenuate neural actions of estradiol (E(2)). In the present study, we compared the effects of continuous and cyclic progesterone treatment in the presence and absence of E(2) in ovariectomized 3xTg-AD mice, a transgenic mouse model of AD. We found that ovariectomy-induced hormone depletion increases AD-like pathology in female 3xTg-AD mice, including accumulation of beta-amyloid, tau hyperphosphorylation, and impaired hippocampal-dependent behavior. E(2) treatment alone prevents the increases in pathology. Continuous progesterone did not affect beta-amyloid levels when delivered alone but blocked the Abeta-lowering action of E(2). In contrast, cyclic progesterone significantly reduced beta-amyloid levels by itself and enhanced rather than inhibited the E(2) effects. These results provide new insight into the neural interactions between E(2) and progesterone that may prove valuable in optimizing HT regimens in postmenopausal women.
Collapse
Affiliation(s)
- Jenna C Carroll
- University of Southern California, 3715 McClintock Avenue, Los Angeles, California 90089-0191.
| | | | | | | |
Collapse
|
35
|
Henderson VW, Brinton RD. Menopause and mitochondria: windows into estrogen effects on Alzheimer's disease risk and therapy. PROGRESS IN BRAIN RESEARCH 2010; 182:77-96. [PMID: 20541661 PMCID: PMC5776041 DOI: 10.1016/s0079-6123(10)82003-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabolic derangements and oxidative stress are early events in Alzheimer's disease pathogenesis. Multi-faceted effects of estrogens include improved cerebral metabolic profile and reduced oxidative stress through actions on mitochondria, suggesting that a woman's endogenous and exogenous estrogen exposures during midlife and in the late post-menopause might favourably influence Alzheimer risk and symptoms. This prediction finds partial support in the clinical literature. As expected, early menopause induced by oophorectomy may increase cognitive vulnerability; however, there is no clear link between age at menopause and Alzheimer risk in other settings, or between natural menopause and memory loss. Further, among older post-menopausal women, initiating estrogen-containing hormone therapy increases dementia risk and probably does not improve Alzheimer's disease symptoms. As suggested by the 'critical window' or 'healthy cell' hypothesis, better outcomes might be expected from earlier estrogen exposures. Some observational results imply that effects of hormone therapy on Alzheimer risk are indeed modified by age at initiation, temporal proximity to menopause, or a woman's health. However, potential methodological biases warrant caution in interpreting observational findings. Anticipated results from large, ongoing clinical trials [Early Versus Late Intervention Trial with Estradiol (ELITE), Kronos Early Estrogen Prevention Study (KEEPS)] will help settle whether midlife estrogen therapy improves midlife cognitive skills but not whether midlife estrogen exposures modify late-life Alzheimer risk. Estrogen effects on mitochondria adumbrate the potential relevance of estrogens to Alzheimer's disease. However, laboratory models are inexact embodiments of Alzheimer pathogenesis and progression, making it difficult to surmise net effects of estrogen exposures. Research needs include better predictors of adverse cognitive outcomes, biomarkers for risks associated with hormone therapy, and tools for monitoring brain function and disease progression.
Collapse
Affiliation(s)
- Victor W Henderson
- Department of Health Research & Policy (Epidemiology), Stanford University, Stanford, CA, USA.
| | | |
Collapse
|
36
|
Abstract
OBJECTIVE To review the relation in midlife and beyond between estrogen exposures and episodic memory in women. BACKGROUND Episodic memory performance declines with usual aging, and impairments in episodic memory often portend the development of Alzheimer disease. In the laboratory, estradiol influences hippocampal function and animal learning. However, it is controversial whether estrogens affect memory after a woman's reproductive years. METHOD Focused literature review, including a summary of a systematic search of clinical trials of estrogens in which outcomes included an objective measure of episodic memory. RESULTS The natural menopause transition is not associated with the objective changes in episodic memory. Strong clinical trial evidence indicates that initiating estrogen-containing hormone therapy after the age of about 60 years does not benefit episodic memory. Clinical trial findings in middle-aged women before the age of 60 years are limited by smaller sample sizes and shorter treatment durations, but these also do not indicate substantial memory effects. Limited short-term evidence, however, suggests that estrogens may improve verbal memory after surgical menopause. Although hormone therapy initiation in old age increases dementia risk, observational studies raise the question of an early critical window during which midlife estrogen therapy reduces late-life Alzheimer disease. However, almost no data address whether midlife estrogen therapy affects episodic memory in old age. CONCLUSIONS Episodic memory is not substantially impacted by the natural menopause transition or improved by the use of estrogen-containing hormone therapy after the age of 60 years. Further research is needed to determine whether outcomes differ after surgical menopause or whether episodic memory later in life is modified by midlife estrogenic exposures.
Collapse
Affiliation(s)
- Victor W Henderson
- Departments of Health Research and Policy (Epidemiology), and of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305-5405, USA.
| |
Collapse
|
37
|
Frick KM, Fernandez SM, Harburger LL. A new approach to understanding the molecular mechanisms through which estrogens affect cognition. Biochim Biophys Acta Gen Subj 2009; 1800:1045-55. [PMID: 19913600 DOI: 10.1016/j.bbagen.2009.11.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 10/23/2009] [Accepted: 11/02/2009] [Indexed: 12/23/2022]
Abstract
Traditional approaches to the study of hormones and cognition have been primarily observational or correlational in nature. Because this work does not permit causal relationships to be identified, very little is known about the specific molecules and cellular events through which hormones affect cognitive function. In this review, we propose a new approach to study hormones and memory, where the systematic blocking of cellular events can reveal which such events are necessary for hormones to influence memory consolidation. The discussion will focus on the modulation of the hippocampus and hippocampal memory by estrogens, given the extensive literature on this subject, and will illustrate how the application of this approach is beginning to reveal important new information about the molecular mechanisms through which estrogens modulate memory consolidation. The clinical relevance of this work will also be discussed.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, Yale University, New Haven, CT 06520, USA.
| | | | | |
Collapse
|
38
|
Saldanha CJ, Duncan KA, Walters BJ. Neuroprotective actions of brain aromatase. Front Neuroendocrinol 2009; 30:106-18. [PMID: 19450619 PMCID: PMC2700852 DOI: 10.1016/j.yfrne.2009.04.016] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 04/02/2009] [Accepted: 04/14/2009] [Indexed: 12/16/2022]
Abstract
The steroidal regulation of vertebrate neuroanatomy and neurophysiology includes a seemingly unending list of brain areas, cellular structures and behaviors modulated by these hormones. Estrogens, in particular have emerged as potent neuromodulators, exerting a range of effects including neuroprotection and perhaps neural repair. In songbirds and mammals, the brain itself appears to be the site of injury-induced estrogen synthesis via the rapid transcription and translation of aromatase (estrogen synthase) in astroglia. This induction seems to occur regardless of the nature and location of primary brain damage. The induced expression of aromatase apparently elevates local estrogen levels enough to interfere with apoptotic pathways, thereby decreasing secondary degeneration and ultimately lessening the extent of damage. There is even evidence suggesting that aromatization may affect injury-induced cytogenesis. Thus, aromatization in the brain appears to confer neuroprotection by an array of mechanisms that involve the deceleration and acceleration of degeneration and repair, respectively. We are only beginning to understand the factors responsible for the injury-induced transcription of aromatase in astroglia. In contrast, much of the manner in which local and circulating estrogens may achieve their neuroprotective effects has been elucidated. However, gaps in our knowledge include issues about the cell-specific regulation of aromatase expression, steroidal influences of aromatization distinct from estrogen formation, and questions about the role of constitutive aromatase in neuroprotection. Here we describe the considerable consensus and some interesting differences in knowledge gained from studies conducted on diverse animal models, experimental paradigms and preparations towards understanding the neuroprotective actions of brain aromatase.
Collapse
Affiliation(s)
- Colin J Saldanha
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015, United States.
| | | | | |
Collapse
|
39
|
Pike CJ, Carroll JC, Rosario ER, Barron AM. Protective actions of sex steroid hormones in Alzheimer's disease. Front Neuroendocrinol 2009; 30:239-58. [PMID: 19427328 PMCID: PMC2728624 DOI: 10.1016/j.yfrne.2009.04.015] [Citation(s) in RCA: 373] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/19/2022]
Abstract
Risk for Alzheimer's disease (AD) is associated with age-related loss of sex steroid hormones in both women and men. In post-menopausal women, the precipitous depletion of estrogens and progestogens is hypothesized to increase susceptibility to AD pathogenesis, a concept largely supported by epidemiological evidence but refuted by some clinical findings. Experimental evidence suggests that estrogens have numerous neuroprotective actions relevant to prevention of AD, in particular promotion of neuron viability and reduction of beta-amyloid accumulation, a critical factor in the initiation and progression of AD. Recent findings suggest neural responsiveness to estrogen can diminish with age, reducing neuroprotective actions of estrogen and, consequently, potentially limiting the utility of hormone therapies in aged women. In addition, estrogen neuroprotective actions are also modulated by progestogens. Specifically, continuous progestogen exposure is associated with inhibition of estrogen actions whereas cyclic delivery of progestogens may enhance neural benefits of estrogen. In recent years, emerging literature has begun to elucidate a parallel relationship of sex steroid hormones and AD risk in men. Normal age-related testosterone loss in men is associated with increased risk to several diseases including AD. Like estrogen, testosterone has been established as an endogenous neuroprotective factor that not only increases neuronal resilience against AD-related insults, but also reduces beta-amyloid accumulation. Androgen neuroprotective effects are mediated both directly by activation of androgen pathways and indirectly by aromatization to estradiol and initiation of protective estrogen signaling mechanisms. The successful use of hormone therapies in aging men and women to delay, prevent, and or treat AD will require additional research to optimize key parameters of hormone therapy and may benefit from the continuing development of selective estrogen and androgen receptor modulators.
Collapse
Affiliation(s)
- Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
40
|
Dorsal hippocampal progesterone infusions enhance object recognition in young female mice. Pharmacol Biochem Behav 2009; 93:177-82. [PMID: 19477194 DOI: 10.1016/j.pbb.2009.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 05/12/2009] [Accepted: 05/17/2009] [Indexed: 11/21/2022]
Abstract
The effects of progesterone on memory are not nearly as well studied as the effects of estrogens. Although progesterone can reportedly enhance spatial and/or object recognition in female rodents when given immediately after training, previous studies have injected progesterone systemically, and therefore, the brain regions mediating this enhancement are not clear. As such, this study was designed to determine the role of the dorsal hippocampus in mediating the beneficial effect of progesterone on object recognition. Young ovariectomized C57BL/6 mice were trained in a hippocampal-dependent object recognition task utilizing two identical objects, and then immediately or 2 h afterwards, received bilateral dorsal hippocampal infusions of vehicle or 0.01, 0.1, or 1.0 microg/microl water-soluble progesterone. Forty-eight hours later, object recognition memory was tested using a previously explored object and a novel object. Relative to the vehicle group, memory for the familiar object was enhanced in all groups receiving immediate infusions of progesterone. Progesterone infusion delayed 2 h after training did not affect object recognition. These data suggest that the dorsal hippocampus may play a critical role in progesterone-induced enhancement of object recognition.
Collapse
|
41
|
Harburger LL, Saadi A, Frick KM. Dose-dependent effects of post-training estradiol plus progesterone treatment on object memory consolidation and hippocampal extracellular signal-regulated kinase activation in young ovariectomized mice. Neuroscience 2009; 160:6-12. [PMID: 19223011 DOI: 10.1016/j.neuroscience.2009.02.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 02/11/2009] [Accepted: 02/12/2009] [Indexed: 10/21/2022]
Abstract
Previous work from our laboratory has shown that the ability of estradiol to enhance object memory consolidation in young ovariectomized mice is dependent on dorsal hippocampal activation of the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) signaling pathway [Fernandez SM, Lewis MC, Pechenino AS, Harburger LL, Orr PT, Gresack JE, Schafe GE, Frick KM (2008) Estradiol-induced enhancement of object memory consolidation involves hippocampal extracellular signal-regulated kinase activation and membrane-bound estrogen receptors. J Neurosci 28:8660-8667]. However, it is unclear if estradiol modulates memory or ERK activation similarly in the presence of progesterone. Therefore, the present study investigated effects of combined estradiol and progesterone treatment on object memory consolidation and dorsal hippocampal ERK activation in young ovariectomized C57BL/6 mice. Object memory was tested in a novel object recognition task. Immediately after training, mice received intraperiotoneal (i.p.) injections of vehicle, 17beta-estradiol (E(2); 0.2 mg/kg), or E(2) plus 5, 10, or 20 mg/kg progesterone (P). Forty-eight hours later, mice receiving E(2) alone or E(2) plus 10 or 20 mg/kg P exhibited significantly enhanced memory for the novel object relative to chance, whereas those receiving vehicle or E(2) plus 5 mg/kg P spent no more time than chance with the novel object. Two weeks later, ERK phosphorylation was measured in the dorsal hippocampus 1 h after i.p. injection of vehicle, E(2), or E(2) plus P. Consistent with our previous work [Fernandez SM, Lewis MC, Pechenino AS, Harburger LL, Orr PT, Gresack JE, Schafe GE, Frick KM (2008) Estradiol-induced enhancement of object memory consolidation involves hippocampal extracellular signal-regulated kinase activation and membrane-bound estrogen receptors. J Neurosci 28:8660-8667], E(2) alone significantly increased phospho-p42 ERK protein levels in the dorsal hippocampus relative to vehicle controls. In contrast, no combination of E(2) and P affected dorsal hippocampal phospho-ERK levels. These data indicate that, unlike E(2) alone, the beneficial effects of combined E(2) plus P treatment on memory are not associated with ERK activation in the dorsal hippocampus 1 h after treatment, and suggest that E(2) alone and combined E(2) plus P may influence ERK activation in different time frames or enhance memory through different mechanisms.
Collapse
Affiliation(s)
- L L Harburger
- Department of Psychology, Stern College for Women, Yeshiva University, New York, NY 10016, USA
| | | | | |
Collapse
|
42
|
Aguirre CC, Baudry M. Progesterone reverses 17beta-estradiol-mediated neuroprotection and BDNF induction in cultured hippocampal slices. Eur J Neurosci 2009; 29:447-54. [PMID: 19175406 DOI: 10.1111/j.1460-9568.2008.06591.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Due to the many similarities in mechanisms of action, targets and effects, progesterone (P4), estrogen and neurotrophins have been implicated in synaptic plasticity as well as in neuroprotection and neurodegeneration. In this study, we examined the interactions between 17beta-estradiol (E2) and P4 and brain-derived neurotrophic factor (BDNF) on both plasticity and excitotoxicity in rat cultured hippocampal slices. First, we evaluated the neuroprotective effects of E2 and P4 against N-methyl-D-aspartate (NMDA) toxicity in cultured rat hippocampal slices. As previously reported, pretreatment with 10 nm E2 (24 h) was neuroprotective against NMDA toxicity. However, P4 (10 nm) added 20 h after E2 treatment for 4 h reversed its protective effect. In addition, the same E2 treatment resulted in an increase in BDNF protein levels as well as in activation of its receptor, TrkB, while addition of P4 attenuated E2-mediated increase in BDNF and TrkB levels. Furthermore, E2-mediated neuroprotection was eliminated by a BDNF scavenger, TrkB-Fc. Our results indicate that E2 neuroprotective effects are mediated through the BDNF pathway and that, under certain conditions, P4 antagonizes the protective effect of estrogen.
Collapse
Affiliation(s)
- Claudia C Aguirre
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089-2520, USA
| | | |
Collapse
|
43
|
Wong AM, Rozovsky I, Arimoto JM, Du Y, Wei M, Morgan TE, Finch CE. Progesterone influence on neurite outgrowth involves microglia. Endocrinology 2009; 150:324-32. [PMID: 18772232 PMCID: PMC2630906 DOI: 10.1210/en.2008-0988] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Progesterone (P4) antagonizes estradiol (E2) in synaptic remodeling in the hippocampus during the rat estrous cycle. To further understand how P4 modulates synaptic plasticity, we used entorhinal cortex lesions, which induce E2-dependent neurite sprouting in the hippocampus. In young ovariectomized rats, the E2-dependent entorhinal cortex lesion-induced sprouting was attenuated by concurrent treatment with P4 and E2. Microglial activation also showed the E2-P4 antagonism. These findings extend reports on the estrous cycle synaptic remodeling without lesions by showing the P4-E2 antagonism during simultaneous treatment with both E2 and P4. Glial mechanisms were analyzed with the wounding-in-a-dish model of cocultured glia and embryonic d-18 cortical neurons from rat. In cocultures of mixed glia (astrocytes plus 30% microglia), P4 antagonized the E2-dependent neurite outgrowth (number and length) and neuron viability in the presence of E2, as observed in vivo. However, removal of microglia (astrocyte-neuron coculture) abolished the antagonism of E2 by P4 on neuron sprouting. The P4 receptor antagonists ORG-31710 and RU-486 blocked the antagonism of P4 on E2-dependent sprouting. These findings suggest a new role for microglia in P4 antagonism of E2 in neuronal plasticity and show its dependence on progesterone receptors. These findings are also relevant to the inclusion of progestins in hormone therapy, which is controversial in relation to cognitive declines during aging and in Alzheimer's disease.
Collapse
Affiliation(s)
- Angela M Wong
- Biogerontology Division, Davis School of Gerontology, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Jayaraman A, Pike CJ. Progesterone attenuates oestrogen neuroprotection via downregulation of oestrogen receptor expression in cultured neurones. J Neuroendocrinol 2009; 21:77-81. [PMID: 19094096 PMCID: PMC2692678 DOI: 10.1111/j.1365-2826.2008.01801.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Recent findings indicate that progesterone can attenuate the beneficial neural effects of oestrogen. In the present study, we investigated the hypothesis that progesterone can modulate oestrogen actions by regulating the expression and activity of oestrogen receptors, ERalpha and ERbeta. Our studies in cultured neurones demonstrate that progesterone decreases the expression of both ERalpha and ERbeta and, as a consequence, also reduces both ER-dependent transcriptional activity and neuroprotection. These results identify a potential mechanism by which progesterone antagonises neural oestrogen actions, a finding that may have important implications for hormone therapy in postmenopausal women.
Collapse
Affiliation(s)
- A Jayaraman
- Neuroscience Graduate Programme and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191, USA
| | | |
Collapse
|
45
|
Waters EM, Torres-Reveron A, McEwen BS, Milner TA. Ultrastructural localization of extranuclear progestin receptors in the rat hippocampal formation. J Comp Neurol 2008; 511:34-46. [PMID: 18720413 DOI: 10.1002/cne.21826] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Progesterone's effects on hippocampus-dependent behavior and synaptic connectivity maybe mediated through the progestin receptor (PR). Although estrogen induces PR mRNA and cytosolic PR in the hippocampus, nuclear PR immunoreactivity is undetectable by light microscopy, suggesting that PR is present at extranuclear sites. To determine whether this is the case, we used immunoelectron microscopy to examine PR distribution in the hippocampal formation of proestrus rats. Ultrastructural analysis revealed that PR labeling is present in extranuclear profiles throughout the CA1 and CA3 regions and dentate gyrus, and, in contrast to light microscopic findings, in nuclei of a few pyramidal and subgranular zone cells. Most neuronal PR labeling is extranuclear and is divided between pre- and postsynaptic compartments; approximately 30% of labeled profiles were axon terminals and 30% were dendrites and dendritic spines. In most laminae, except in CA3 stratum lucidum, about 15% of PR-immunoreactive profiles were unmyelinated axons. In stratum lucidum, where the mossy fiber axons course, more than 50% of PR-labeled profiles were axonal. The remaining 25% of PR-labeled profiles were glia, some resembling astrocytes. PR labeling is strongly dependent on estrogen priming, insofar as few PR-labeled profiles were detected in ovariectomized, oil-replaced females. Synapses formed by PR-labeled terminals were predominantly asymmetric, consistent with a role for progesterone in directly regulating excitatory transmission. These findings suggest that some of progesterone's actions in the hippocampal formation may be mediated by direct and rapid actions on extranuclear PRs and that PRs are well positioned to regulate progesterone-induced changes at synapses.
Collapse
Affiliation(s)
- Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10065, USA.
| | | | | | | |
Collapse
|
46
|
Carroll JC, Rosario ER, Pike CJ. Progesterone blocks estrogen neuroprotection from kainate in middle-aged female rats. Neurosci Lett 2008; 445:229-32. [PMID: 18790007 DOI: 10.1016/j.neulet.2008.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 08/26/2008] [Accepted: 09/04/2008] [Indexed: 01/18/2023]
Abstract
The neuroprotective effects of estrogen in young adult rodents are well established. Less well understood is how estrogen neuroprotection is affected by aging and interactions with progesterone. In this study, we investigated the effects of estrogen and continuous progesterone, both alone and in combination, on hippocampal neuron survival following kainate lesion in 14-month-old female rats entering reproductive senescence. Our results show that ovariectomy-induced hormone depletion did not significantly affect the extent of kainate-induced neuron loss. Treatment of ovariectomized rats with estrogen significantly reduced neuron loss, however this effect was blocked by co-administration of continuous progesterone. Treatment of ovariectomized rats with progesterone alone did not significantly affect kainate toxicity. These results provide new insight into factors that regulate estrogen neuroprotection, which has important implications for hormone therapy in postmenopausal women.
Collapse
Affiliation(s)
- Jenna C Carroll
- Neuroscience Graduate Program, Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | | | | |
Collapse
|
47
|
Brinton RD, Thompson RF, Foy MR, Baudry M, Wang J, Finch CE, Morgan TE, Pike CJ, Mack WJ, Stanczyk FZ, Nilsen J. Progesterone receptors: form and function in brain. Front Neuroendocrinol 2008; 29:313-39. [PMID: 18374402 PMCID: PMC2398769 DOI: 10.1016/j.yfrne.2008.02.001] [Citation(s) in RCA: 464] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Accepted: 02/08/2008] [Indexed: 12/13/2022]
Abstract
Emerging data indicate that progesterone has multiple non-reproductive functions in the central nervous system to regulate cognition, mood, inflammation, mitochondrial function, neurogenesis and regeneration, myelination and recovery from traumatic brain injury. Progesterone-regulated neural responses are mediated by an array of progesterone receptors (PR) that include the classic nuclear PRA and PRB receptors and splice variants of each, the seven transmembrane domain 7TMPRbeta and the membrane-associated 25-Dx PR (PGRMC1). These PRs induce classic regulation of gene expression while also transducing signaling cascades that originate at the cell membrane and ultimately activate transcription factors. Remarkably, PRs are broadly expressed throughout the brain and can be detected in every neural cell type. The distribution of PRs beyond hypothalamic borders, suggests a much broader role of progesterone in regulating neural function. Despite the large body of evidence regarding progesterone regulation of reproductive behaviors and estrogen-inducible responses as well as effects of progesterone metabolite neurosteroids, much remains to be discovered regarding the functional outcomes resulting from activation of the complex array of PRs in brain by gonadally and/or glial derived progesterone. Moreover, the impact of clinically used progestogens and developing selective PR modulators for targeted outcomes in brain is a critical avenue of investigation as the non-reproductive functions of PRs have far-reaching implications for hormone therapy to maintain neurological health and function throughout menopausal aging.
Collapse
Affiliation(s)
- Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA 90089, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Carroll JC, Pike CJ. Selective estrogen receptor modulators differentially regulate Alzheimer-like changes in female 3xTg-AD mice. Endocrinology 2008; 149:2607-11. [PMID: 18276750 PMCID: PMC2329277 DOI: 10.1210/en.2007-1346] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen-based hormone therapy (HT) in postmenopausal women may reduce the risk of Alzheimer's disease (AD), although HT remains controversial. One key concern with HT is the potential of adverse outcomes such as breast and uterine cancer. A promising strategy to maximize HT benefits and minimize HT risks is the use of selective estrogen receptor modulators (SERMs) that exert tissue-specific estrogenic effects. To begin investigating the SERM approach in reducing the risk of AD, we investigated whether AD-like neuropathology in the 3xTg-AD mouse model of AD is regulated by the SERMs propylpyrazole triol (PPT) and diarylpropionitrile (DPN) that exhibit relative specificity for estrogen receptor-alpha and -beta, respectively. Consistent with our previous observations, we found that ovariectomy-induced hormone depletion in adult female 3xTg-AD mice significantly increased accumulation of beta-amyloid protein (Abeta) and decreased hippocampal-dependent behavioral performance. Treatment with 17beta-estradiol (E2) prevented the ovariectomized-induced worsening of both pathologies. PPT treatment was similar to E2 in terms of reducing Abeta accumulation in hippocampus, subiculum, and amygdala but comparatively less effective in frontal cortex. In contrast, DPN did not significantly reduce Abeta accumulation in hippocampus and subiculum, was partially effective in frontal cortex, and nearly as effective as E2 in amygdala. Furthermore, PPT but not DPN mimicked the ability of E2 to improve behavioral performance. These findings provide initial evidence of beneficial actions of SERMs in a mouse model of AD and support continued investigation of SERMs as an alternative to estrogen-based HT in reducing the risk of AD in postmenopausal women.
Collapse
Affiliation(s)
- Jenna C Carroll
- Neuroscience Graduate Program and Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, California 90089-0191, USA
| | | |
Collapse
|
49
|
Harden CL. Hormone replacement therapy: will it affect seizure control and AED levels? Seizure 2008; 17:176-80. [PMID: 18187348 DOI: 10.1016/j.seizure.2007.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Interest in the years of reproductive changes for women with epilepsy (WWE), specifically perimenopause, menopause and postmenopause has been emerging in the epilepsy community. This article discusses evidence for changes in seizure frequency during perimenopause and postmenopause. Further, a catamenial epilepsy pattern during the reproductive years may be a hallmark for the observed seizure frequency change during these years; that is, an increase at perimenopause but a decrease at menopause. This finding implies that a subset of WWE are particularly susceptible to endogenous reproductive hormonal changes. An adverse effect on seizure frequency with the use of hormone replacement therapy (HRT) during postmenopause for WWE was reported in questionnaires, and was later borne out in a clinical trial. The laboratory counterpart of this human trial, HRT in ovariectomized rodent seizure models, shows that estrogen and progesterone are neuroprotective and do not uniformly increase seizure frequency. Possible reasons for the discrepancy between "the lab and the clinic" are presented. Strategies for managing HRT in symptomatic postmenopausal WWE using estrogenic and progestogenic compounds that may be less likely to promote seizures are discussed.
Collapse
Affiliation(s)
- Cynthia L Harden
- Comprehensive Epilepsy Center, Department of Neurology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
50
|
Abstract
Estrogen depletion in postmenopausal women is a significant risk factor for the development of Alzheimer's disease (AD), and estrogen-based hormone therapy may reduce this risk. However, the effects of progesterone both alone and in combination with estrogen on AD neuropathology remain unknown. In this study, we used the triple transgenic mouse model of AD (3xTg-AD) to investigate the individual and combined effects of estrogen and progesterone on beta-amyloid (Abeta) accumulation, tau hyperphosphorylation, and hippocampal-dependent behavioral impairments. In gonadally intact female 3xTg-AD mice, AD-like neuropathology was apparent by 3 months of age and progressively increased through age 12 months, a time course that was paralleled by behavioral impairment. Ovariectomy-induced depletion of sex steroid hormones in adult female 3xTg-AD mice significantly increased Abeta accumulation and worsened memory performance. Treatment of ovariectomized 3xTg-AD mice with estrogen, but not progesterone, prevented these effects. When estrogen and progesterone were administered in combination, progesterone blocked the beneficial effect of estrogen on Abeta accumulation but not on behavioral performance. Interestingly, progesterone significantly reduced tau hyperphosphorylation when administered both alone and in combination with estrogen. These results demonstrate that estrogen and progesterone independently and interactively regulate AD-like neuropathology and suggest that an optimized hormone therapy may be useful in reducing the risk of AD in postmenopausal women.
Collapse
|