1
|
Gleason CE, Dowling NM, Kara F, James TT, Salazar H, Ferrer Simo CA, Harman SM, Manson JE, Hammers DB, Naftolin FN, Pal L, Miller VM, Cedars MI, Lobo RA, Malek-Ahmadi M, Kantarci K. Long-term cognitive effects of menopausal hormone therapy: Findings from the KEEPS Continuation Study. PLoS Med 2024; 21:e1004435. [PMID: 39570992 PMCID: PMC11581397 DOI: 10.1371/journal.pmed.1004435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Findings from Kronos Early Estrogen Prevention Study (KEEPS)-Cog trial suggested no cognitive benefit or harm after 48 months of menopausal hormone therapy (mHT) initiated within 3 years of final menstrual period. To clarify the long-term effects of mHT initiated in early postmenopause, the observational KEEPS Continuation Study reevaluated cognition, mood, and neuroimaging effects in participants enrolled in the KEEPS-Cog and its parent study the KEEPS approximately 10 years after trial completion. We hypothesized that women randomized to transdermal estradiol (tE2) during early postmenopause would show cognitive benefits, while oral conjugated equine estrogens (oCEE) would show no effect, compared to placebo over the 10 years following randomization in the KEEPS trial. METHODS AND FINDINGS The KEEPS-Cog (2005-2008) was an ancillary study to the KEEPS (NCT00154180), in which participants were randomized into 3 groups: oCEE (Premarin, 0.45 mg/d), tE2 (Climara, 50 μg/d) both with micronized progesterone (Prometrium, 200 mg/d for 12 d/mo) or placebo pills and patch for 48 months. KEEPS Continuation (2017-2022), an observational, longitudinal cohort study of KEEPS clinical trial, involved recontacting KEEPS participants approximately 10 years after the completion of the 4-year clinical trial to attend in-person research visits. Seven of the original 9 sites participated in the KEEPS Continuation, resulting in 622 women of original 727 being invited to return for a visit, with 299 enrolling across the 7 sites. KEEPS Continuation participants repeated the original KEEPS-Cog test battery which was analyzed using 4 cognitive factor scores and a global cognitive score. Cognitive data from both KEEPS and KEEPS Continuation were available for 275 participants. Latent growth models (LGMs) assessed whether baseline cognition and cognitive changes during KEEPS predicted cognitive performance at follow-up, and whether mHT randomization modified these relationships, adjusting for covariates. Similar health characteristics were observed at KEEPS randomization for KEEPS Continuation participants and nonparticipants (i.e., women not returning for the KEEPS Continuation). The LGM revealed significant associations between intercepts and slopes for cognitive performance across almost all domains, indicating that cognitive factor scores changed over time. Tests assessing the effects of mHT allocation on cognitive slopes during the KEEPS and across all years of follow-up including the KEEPS Continuation visit were all statistically nonsignificant. The KEEPS Continuation study found no long-term cognitive effects of mHT, with baseline cognition and changes during KEEPS being the strongest predictors of later performance. Cross-sectional comparisons confirmed that participants assigned to mHT in KEEPS (oCEE and tE2 groups) performed similarly on cognitive measures to those randomized to placebo, approximately 10 years after completion of the randomized treatments. These findings suggest that mHT poses no long-term cognitive harm; conversely, it provides no cognitive benefit or protective effects against cognitive decline. CONCLUSIONS In these KEEPS Continuation analyses, there were no long-term cognitive effects of short-term exposure to mHT started in early menopause versus placebo. These data provide reassurance about the long-term neurocognitive safety of mHT for symptom management in healthy, recently postmenopausal women, while also suggesting that mHT does not improve or preserve cognitive function in this population.
Collapse
Affiliation(s)
- Carey E. Gleason
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial VA Hospital, Madison, Wisconsin, United States of America
| | - N. Maritza Dowling
- Department of Acute & Chronic Care, George Washington University, Washington, DC, United States of America
| | - Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Taryn T. James
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Hector Salazar
- Department of Health and Community Systems, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, United States of America
| | - Carola A. Ferrer Simo
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sherman M. Harman
- Phoenix VA Health Care System, Phoenix, Arizona, United States of America
| | - JoAnn E. Manson
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dustin B. Hammers
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | | | - Lubna Pal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, Connecticut, United States of America
| | - Virginia M. Miller
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Marcelle I. Cedars
- Department of Obstetrics and Gynecology, University of California, San Francisco, California, United States of America
| | - Rogerio A. Lobo
- Department of Obstetrics and Gynecology, Columbia University, New York, New York State, United States of America
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
2
|
Singh KA, Soukar J, Zulkifli M, Kersey A, Lokhande G, Ghosh S, Murali A, Garza NM, Kaur H, Keeney JN, Banavath R, Ceylan Koydemir H, Sitcheran R, Singh I, Gohil VM, Gaharwar AK. Atomic vacancies of molybdenum disulfide nanoparticles stimulate mitochondrial biogenesis. Nat Commun 2024; 15:8136. [PMID: 39289340 PMCID: PMC11408498 DOI: 10.1038/s41467-024-52276-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Diminished mitochondrial function underlies many rare inborn errors of energy metabolism and contributes to more common age-associated metabolic and neurodegenerative disorders. Thus, boosting mitochondrial biogenesis has been proposed as a potential therapeutic approach for these diseases; however, currently we have a limited arsenal of compounds that can stimulate mitochondrial function. In this study, we designed molybdenum disulfide (MoS2) nanoflowers with predefined atomic vacancies that are fabricated by self-assembly of individual two-dimensional MoS2 nanosheets. Treatment of mammalian cells with MoS2 nanoflowers increased mitochondrial biogenesis by induction of PGC-1α and TFAM, which resulted in increased mitochondrial DNA copy number, enhanced expression of nuclear and mitochondrial-DNA encoded genes, and increased levels of mitochondrial respiratory chain proteins. Consistent with increased mitochondrial biogenesis, treatment with MoS2 nanoflowers enhanced mitochondrial respiratory capacity and adenosine triphosphate production in multiple mammalian cell types. Taken together, this study reveals that predefined atomic vacancies in MoS2 nanoflowers stimulate mitochondrial function by upregulating the expression of genes required for mitochondrial biogenesis.
Collapse
Affiliation(s)
- Kanwar Abhay Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - John Soukar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Mohammad Zulkifli
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Anna Kersey
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Giriraj Lokhande
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Aparna Murali
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Natalie M Garza
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Harman Kaur
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Justin N Keeney
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Ramu Banavath
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Hatice Ceylan Koydemir
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Raquel Sitcheran
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Irtisha Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA.
| | - Vishal M Gohil
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
3
|
Ali N, Sohail R, Jaffer SR, Siddique S, Kaya B, Atowoju I, Imran A, Wright W, Pamulapati S, Choudhry F, Akbar A, Khawaja UA. The Role of Estrogen Therapy as a Protective Factor for Alzheimer's Disease and Dementia in Postmenopausal Women: A Comprehensive Review of the Literature. Cureus 2023; 15:e43053. [PMID: 37680393 PMCID: PMC10480684 DOI: 10.7759/cureus.43053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
The complete cessation of menstruation for 12 months with associated vasomotor symptoms is termed menopause. Apart from playing a role in reproduction, estrogen significantly affects the central nervous system (CNS). Population-based studies highlighted a substantial difference in the prevalence of dementia between men and women, with Alzheimer-associated dementia being more prevalent in women, indicating that estrogen deficiency might be a risk factor for neurodegenerative diseases. Patients with dementia experience a progressive decline in neurocognitive function, beginning with short-term memory loss that progresses to long-term memory loss and the inability to perform everyday activities, leading ultimately to death. There is currently no cure for dementia, so preventing or slowing the disease's progression is paramount. Accordingly, researchers have widely studied the role of estrogen as a neuroprotective agent. Estrogen prevents dementia by augmenting Hippocampal and prefrontal cortex function, reducing neuroinflammation, preventing degradation of estrogen receptors, decreasing oxidative damage to the brain, and increasing cholinergic and serotonergic function. According to the window phase hypothesis, estrogen's effect on preventing dementia is more pronounced if therapy is started early, during the first five years of menopause. Other studies like The Woman's Health Initiative Memory Study (WHIMS) showed unfavorable effects of estrogen on the brain. This review aims to establish an understanding of the currently available data on estrogen's effect on neurodegeneration, namely, dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- Noor Ali
- Obstetrics and Gynecology, Thumbay University Hospital, Ajman, ARE
- General Physician, Dubai Medical College, DXB, ARE
| | - Rohab Sohail
- Internal Medicine, Quaid-e-Azam Medical College, Bahawalpur, PAK
| | | | - Sadia Siddique
- Gastroenterology, Blackpool Victoria Hospital National Health Services (NHS) Foundation Trust, Blackpool, GBR
| | - Berfin Kaya
- Obstetrics and Gynaecology, Izmir Ataturk Research and Training Hospital, Izmir, TUR
- Obstetrics and Gynaecology, Izmir Kâtip Celebi University, Faculty of Medicine, Izmir, TUR
| | - Inioluwa Atowoju
- Obstetrics and Gynecology, Kharkiv National Medical University, Kharkiv, UKR
| | - Alizay Imran
- Surgery, Windsor University School of Medicine, Chicago, USA
| | - Whitney Wright
- Obstetrics and Gynecology, Texila American University, Georgetown, GUY
| | - Spandana Pamulapati
- Obstetrics and Gynecology, Alluri Sita Rama Raju Academy of Medical Sciences, Eluru, IND
| | - Faiza Choudhry
- Medicine and Surgery, Liaquat University of Medical and Health Sciences, Sindh, PAK
| | - Anum Akbar
- Pediatrics, University of Nebraska Medical Center, Omaha, USA
| | - Uzzam Ahmed Khawaja
- Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Clinical and Translational Research, Dr Ferrer BioPharma, South Miami, USA
| |
Collapse
|
4
|
Young TL, Scieszka D, Begay JG, Lucas SN, Herbert G, Zychowski K, Hunter R, Salazar R, Ottens AK, Erdely A, Gu H, Campen MJ. Aging influence on pulmonary and systemic inflammation and neural metabolomics arising from pulmonary multi-walled carbon nanotube exposure in apolipoprotein E-deficient and C57BL/6 female mice. Inhal Toxicol 2023; 35:86-100. [PMID: 35037817 PMCID: PMC10037439 DOI: 10.1080/08958378.2022.2026538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/03/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Environmental exposures exacerbate age-related pathologies, such as cardiovascular and neurodegenerative diseases. Nanoparticulates, and specifically carbon nanomaterials, are a fast-growing contributor to the category of inhalable pollutants, whose risks to health are only now being unraveled. The current study assessed the exacerbating effect of age on multiwalled-carbon nanotube (MWCNT) exposure in young and old C57BL/6 and ApoE-/- mice. MATERIALS AND METHODS Female C57BL/6 and apolipoprotein E-deficient (ApoE-/-) mice, aged 8 weeks and 15 months, were exposed to 0 or 40 µg MWCNT via oropharyngeal aspiration. Pulmonary inflammation, inflammatory bioactivity of serum, and neurometabolic changes were assessed at 24 h post-exposure. RESULTS Pulmonary neutrophil infiltration was induced by MWCNT in bronchoalveolar lavage fluid in both C57BL/6 and ApoE-/-. Macrophage counts decreased with MWCNT exposure in ApoE-/- mice but were unaffected by exposure in C57BL/6 mice. Older mice appeared to have greater MWCNT-induced total protein in lavage fluid. BALF cytokines and chemokines were elevated with MWCNT exposure, but CCL2, CXCL1, and CXCL10 showed reduced responses to MWCNT in older mice. However, no significant serum inflammatory bioactivity was detected. Cerebellar metabolic changes in response to MWCNT were modest, but age and strain significantly influenced metabolite profiles assessed. ApoE-/- mice and older mice exhibited less robust metabolite changes in response to exposure, suggesting a reduced health reserve. CONCLUSIONS Age influences the pulmonary and neurological responses to short-term MWCNT exposure. However, with only the model of moderate aging (15 months) in this study, the responses appeared modest compared to inhaled toxicant impacts in more advanced aging models.
Collapse
Affiliation(s)
- Tamara L. Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - David Scieszka
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Jessica G. Begay
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Selita N. Lucas
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | | | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Raul Salazar
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Andrew K. Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA 23298
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, US 85004
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
5
|
Ajayi EI, Molehin OR, Iyoha AE, Tallapragada DS, Oloyede OI, Tikoo KB. Postmortem mitochondrial membrane permeability transition assessment of apoptotic cell death in brain and liver of insulin resistant, ovariectomised rats. IBRO Neurosci Rep 2021; 11:156-163. [PMID: 34939064 PMCID: PMC8664703 DOI: 10.1016/j.ibneur.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
The adverse alterations in mitochondrial functions can affect neuronal function negatively, as they play a crucial role in neuronal plasticity and death. Direct measurements of mitochondrial activity, including membrane potential and ATP production, are not easily achieved in post-mortem brain and liver samples because most organ functions cease to work after death; in fact, with increasing post-mortem intervals (PMI), the brain and liver tissues deteriorate rapidly. Standard procedures of mitochondria isolation, protein determination expressed as BSA equivalent, and spectrophotometric assessment of pore opening at 540 nm were employed. Our results showed that (a) intact mitochondria may be isolated from rat brain and liver of these rats after storage in animal body (in situ) at -20 °C for 7 days (168 h, post-mortem), (b) some population of these mitochondria can still take up exogenous Ca2+ and (c) they can still resist osmotically induced large amplitude swelling in a suitable buffer. The need for mitochondrial purity, structural integrity and abundance for functional studies are common hindrances that can encumber mitochondrial research. Therefore, this study is significant to have shown that PMI up to 7 days did not extensively, diminish MMPT pore status in normal and diabetic, ovariectomised rats. This can be relevant for forensic data mining.
Collapse
Affiliation(s)
- Ebenezer I.O. Ajayi
- Biochemistry Department,Faculty of Basic and Applied Sciences,College of Science, Engineering and Technology, Osun State University, P.M.B. 4494, Osogbo, Nigeria
- Pharmacology/Toxicology Department, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar (Mohali), Punjab 160 062, India
| | - Olorunfemi R. Molehin
- Department of Biochemistry, Faculty of Science, Ekiti State University Ado-Ekiti, P.M.B. 5363, Ado Ekiti 360001, Nigeria
| | - Alex E. Iyoha
- Biochemistry Department,Faculty of Basic and Applied Sciences,College of Science, Engineering and Technology, Osun State University, P.M.B. 4494, Osogbo, Nigeria
| | - Divya S.P. Tallapragada
- Pharmacology/Toxicology Department, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar (Mohali), Punjab 160 062, India
| | - Omotade I. Oloyede
- Department of Biochemistry, Faculty of Science, Ekiti State University Ado-Ekiti, P.M.B. 5363, Ado Ekiti 360001, Nigeria
| | - Kulbhushan B. Tikoo
- Pharmacology/Toxicology Department, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar (Mohali), Punjab 160 062, India
| |
Collapse
|
6
|
Werber T, Bata Z, Vaszine ES, Berente DB, Kamondi A, Horvath AA. The Association of Periodontitis and Alzheimer's Disease: How to Hit Two Birds with One Stone. J Alzheimers Dis 2021; 84:1-21. [PMID: 34511500 DOI: 10.3233/jad-210491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of cognitive impairment in the elderly. Recent evidence suggests that preventive interventional trials could significantly reduce the risk for development of dementia. Periodontitis is the most common dental disease characterized by chronic inflammation and loss of alveolar bone and perialveolar attachment of teeth. Growing number of studies propose a potential link between periodontitis and neurodegeneration. In the first part of the paper, we overview case-control studies analyzing the prevalence of periodontitis among AD patients and healthy controls. Second, we survey observational libraries and cross-sectional studies investigating the risk of cognitive decline in patients with periodontitis. Next, we describe the current view on the mechanism of periodontitis linked neural damage, highlighting bacterial invasion of neural tissue from dental plaques, and periodontitis induced systemic inflammation resulting in a neuroinflammatory process. Later, we summarize reports connecting the four most common periodontal pathogens to AD pathology. Finally, we provide a practical guide for further prevalence and interventional studies on the management of cognitively high-risk patients with and without periodontitis. In this section, we highlight strategies for risk control, patient information, dental evaluation, reporting protocol and dental procedures in the clinical management of patients with a risk for periodontitis and with diagnosed periodontitis. In conclusion, our review summarizes the current view on the association between AD and periodontitis and provides a research and intervention strategy for harmonized interventional trials and for further case-control or cross-sectional studies.
Collapse
Affiliation(s)
- Tom Werber
- Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsofia Bata
- Department of Conservative Dentistry, Semmelweis University, Budapest, Hungary
| | - Eniko Szabo Vaszine
- Department of Conservative Dentistry, Semmelweis University, Budapest, Hungary
| | - Dalida Borbala Berente
- Faculty of Medicine, Semmelweis University, Budapest, Hungary.,Neurocognitive Research Center, National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - Anita Kamondi
- Neurocognitive Research Center, National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary.,Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Andras Attila Horvath
- Neurocognitive Research Center, National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Antidementia Effects of Alternanthera philoxeroides in Ovariectomized Mice Supported by NMR-Based Metabolomic Analysis. Molecules 2021; 26:molecules26092789. [PMID: 34065080 PMCID: PMC8125874 DOI: 10.3390/molecules26092789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
The crude ethanol extract of the whole plant of Alternanthera philoxeroides (Mart.) Griseb was investigated for its potential as antidementia, induced by estrogen deprivation, based on in vitro antioxidant activity, β-amyloid aggregation inhibition and cholinesterase inhibitory activity, as well as in vivo Morris water maze task (MWMT), novel object recognition task (NORT), and Y-maze task. To better understand the effect of the extract, oxidative stress-induced brain membrane damage through lipid peroxidation in the whole brain was also investigated. Additionally, expressions of neuroinflammatory cytokines (IL-1β, IL-6 and TNF-α) and estrogen receptor-mediated facilitation genes such as PI3K and AKT mRNA in the hippocampus and frontal cortex were also evaluated. These effects were confirmed by the determination of its serum metabolites by NMR metabolomic analysis. Both the crude extract of A. philoxeroides and its flavone constituents were found to inhibit β-amyloid (Aβ) aggregation.
Collapse
|
8
|
Kauffman RP, Young C, Castracane VD. Perils of prolonged ovarian suppression and hypoestrogenism in the treatment of breast cancer: Is the risk of treatment worse than the risk of recurrence? Mol Cell Endocrinol 2021; 525:111181. [PMID: 33529690 DOI: 10.1016/j.mce.2021.111181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal breast cancer is usually estrogen receptor positive, and hence, prolonged ovarian suppression by medical or surgical means to prevent recurrence has become standard of management to improve disease-free survival. Ten-year adjuvant tamoxifen therapy is associated with 3.5% fewer recurrences compared to five years. The SOFT trial demonstrated small but statistically significant incremental improvements in long-term disease-free survival by the addition of gonadotropin-releasing hormone analog treatment (triptorelin) to an aromatase inhibitor (exemestane). Profound hypoestrogenism in the premenopausal age group may not be well tolerated due to a host of bothersome side effects (primarily vasomotor symptoms, musculoskeletal complaints, genitourinary syndrome of menopause, and mood disorders). Prolonged hypoestrogenism in younger women is associated with premature development of cardiovascular disease, bone loss, cognitive decline, and all-cause mortality. This paper explores multi-system consequences of prolonged hypoestrogenism in premenopausal women derived from studies of women with and without breast cancer. Pretreatment counseling in estrogen receptor positive breast cancer should emphasize the benefit of prolonged estrogen suppression on breast cancer recurrence and established risks of lifelong hypoestrogenism on quality of life and all-cause mortality. Future genomic research may help identify the best candidates for extended ovarian suppression to avoid treating many women when only a minority benefit.
Collapse
Affiliation(s)
- Robert P Kauffman
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA.
| | - Christina Young
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| | - V Daniel Castracane
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| |
Collapse
|
9
|
Zhou Y, Xu B, Yu H, Zhao W, Song X, Liu Y, Wang K, Peacher N, Zhao X, Zhang HT. Biochanin A Attenuates Ovariectomy-Induced Cognition Deficit via Antioxidant Effects in Female Rats. Front Pharmacol 2021; 12:603316. [PMID: 33815102 PMCID: PMC8010695 DOI: 10.3389/fphar.2021.603316] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/19/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Impairment of memory and cognition is one of the major symptoms in women with postmenopausal disorders due to estrogen deficiency, which accounts for the much higher prevalence of Alzheimer’s disease in females. Biochanin A (BCA), a natural phytoestrogen, has been reported to protect neurons against ischemic brain injury. However, the neuroprotective effects of BCA in the postmenopausal-like model of ovariectomized (OVX) rats remain to be investigated. Methods: All the rats except for the sham group underwent the resection of bilateral ovaries. Seven days after the OVX surgery, rats were randomly divided into six groups: sham, OVX, OVX + BCA (5 mg/kg), OVX + BCA (20 mg/kg), OVX + BCA (60 mg/kg), and OVX + estradiol (E2; 0.35 mg/kg), which were administrated daily by gavage for 12 weeks. Learning and memory were examined using the Morris water-maze test before the end of the experiment. Morphological changes of the rat hippocampus were observed by HE staining and electron microscopy. Malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) in the hippocampus were measured. The effect of BCA on cell viability was measured in the presence of hydrogen peroxide (H2O2) using CCK8. Flow cytometry was used to measure neuronal apoptosis and reactive oxygen species (ROS) induced by H2O2. Expression of Bcl-2, Bax, and Caspase-3 was determined by Western blotting using hippocampal tissues and primary cultures of hippocampal neurons. Results: Chronic treatment with BCA mimicked the ability of E2 to reverse the deficit of learning and memory in the Morris water-maze test in OVX rats. BCA normalized OVX-induced morphological changes as revealed by HE staining and electron microscopy. In addition, BCA significantly decreased the levels of MDA, the biomarker of oxidative damage, and increased the activity of the intracellular antioxidant enzymes SOD and GSH-Px in OVX rats. Further, in primary cultures of hippocampal neurons, BCA reversed H2O2-induced decreases in cell viability and accumulation of ROS. Finally, BCA reversed OVX- or H2O2-induced increases in Bax and Caspase-3 and decreases in Bcl-2 in the hippocampus and primary cultures of hippocampal neurons. Conclusion: These results suggest that BCA improves memory through its neuroprotective properties in the brain under the circumstance of estrogen deficiency and can be used for treatment of memory loss in postmenopausal women.
Collapse
Affiliation(s)
- Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Bingbing Xu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yan Liu
- The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Kainan Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Nikoli Peacher
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
10
|
Steinman J, Sun HS, Feng ZP. Microvascular Alterations in Alzheimer's Disease. Front Cell Neurosci 2021; 14:618986. [PMID: 33536876 PMCID: PMC7849053 DOI: 10.3389/fncel.2020.618986] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with continual decline in cognition and ability to perform routine functions such as remembering familiar places or understanding speech. For decades, amyloid beta (Aβ) was viewed as the driver of AD, triggering neurodegenerative processes such as inflammation and formation of neurofibrillary tangles (NFTs). This approach has not yielded therapeutics that cure the disease or significant improvements in long-term cognition through removal of plaques and Aβ oligomers. Some researchers propose alternate mechanisms that drive AD or act in conjunction with amyloid to promote neurodegeneration. This review summarizes the status of AD research and examines research directions including and beyond Aβ, such as tau, inflammation, and protein clearance mechanisms. The effect of aging on microvasculature is highlighted, including its contribution to reduced blood flow that impairs cognition. Microvascular alterations observed in AD are outlined, emphasizing imaging studies of capillary malfunction. The review concludes with a discussion of two therapies to protect tissue without directly targeting Aβ for removal: (1) administration of growth factors to promote vascular recovery in AD; (2) inhibiting activity of a calcium-permeable ion channels to reduce microglial activation and restore cerebral vascular function.
Collapse
Affiliation(s)
- Joe Steinman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Song X, Wu J, Zhou Y, Feng L, Yuan JM, Pan A, Koh WP. Reproductive and hormonal factors and risk of cognitive impairment among Singapore Chinese women. Am J Obstet Gynecol 2020; 223:410.e1-410.e23. [PMID: 32112728 DOI: 10.1016/j.ajog.2020.02.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Epidemiologic studies suggest that declining estrogen levels in menopause may play an important role in the pathogenesis of dementia and contribute to increased risk of cognitive impairment in women. Most previous studies have been conducted in Western populations to investigate the relationship of the length of reproductive periods and use of hormone-replacement therapy with risk of cognitive function and dementia, but the findings are inconclusive. Relevant evidence among Asian populations is limited. OBJECTIVES To evaluate the association between reproductive and hormonal factors and the risk of cognitive impairment in Chinese women with natural menopause. STUDY DESIGN The Singapore Chinese Health Study is a population-based study that recruited participants aged 45-74 years between 1993 and 1998, and the current study included 8222 women from this cohort who had natural menopause, complete data on reproductive factors and hormonal therapies at baseline (1993-1998), follow-up 1 (1999-2004) and follow-up 2 interviews (2006-2010), and cognitive function evaluated at ages 61-96 years using the Singapore Modified Mini-Mental State Examination during the follow-up 3 visits (2014-2016). Multivariable logistic regression models were used to estimate odds ratios and 95% confidence intervals for the risk of cognitive impairment. RESULTS Compared with women with menopause at 50-54 years of age, the odds ratios (95% confidence interval) were 1.67 (1.32-2.11), 1.24 (1.08-1.44), and 1.06 (0.87- 1.29) for women who experienced menopause before 45 years, at 45-49 years of age, and after 54 years, respectively. Compared with women with 35-39 reproductive years from menarche to menopause, the odds ratios (95% confidence interval) were 1.28 (1.11-1.48) for women with <35 reproductive years. Furthermore, compared with women who had 1-2 children, the odds ratios (95% confidence interval) were 1.27 (1.04-1.55) for women who had more than 5 children, and the risk increased significantly by 5% per child birth (odds ratio, 1.05; 95% confidence interval, 1.01-1.09). Compared with those who had never used oral contraceptives, women with short-term use (≤5 years) of oral contraceptives had 26% lower odds of having cognitive impairment (odds ratio, 0.74; 95% confidence interval, 0.63-0.87), whereas the association was not statistically significant for those used for more than 5 years (odds ratio, 0.87; 95% confidence interval, 0.68-1.13). Women who used hormone-replacement therapy had a 39% lower odd of getting cognitive impairment compared with nonusers (odds ratio, 0.61; 95% confidence interval, 0.46-0.80). CONCLUSION Our data suggest that shorter reproductive years and greater parity were associated with a greater risk of cognitive impairment in late life, whereas the use of oral contraceptives and hormone-replacement therapy was associated with decreased risk. As the population ages, understanding how these factors affect late-life cognitive function in women may help health professionals develop preventive measures targeting lifetime estrogen exposure from endogenous or exogenous sources.
Collapse
|
12
|
Apolipoprotein E4 genotype in combination with poor metabolic profile is associated with reduced cognitive performance in healthy postmenopausal women: implications for late onset Alzheimer's disease. Menopause 2020; 26:7-15. [PMID: 29975287 DOI: 10.1097/gme.0000000000001160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE We hypothesized the association of metabolic profile on cognition in postmenopausal women will be greater among ApoE4 carriers compared with noncarriers. METHODS Metabolic biomarkers and measures of global cognition, executive functions, and verbal memory, collected among postmenopausal females, were used in this analysis. Clustering analyses of metabolic biomarkers revealed three phenotypes: healthy, predominantly hypertensive, and poor metabolic with (borderline normal laboratory values). General linear models tested whether an association of metabolic cluster with cognition differed by ApoE4 genotype. RESULTS In the total sample of 497 women, verbal memory was lower in the poor metabolic cluster (P = 0.04). Among ApoE4+ women, performance in all cognitive domains was lowest in the poor metabolic cluster. Differences in executive functions among metabolic clusters were detected only in ApoE4+ women (P value for interaction = 0.003). CONCLUSIONS In a general population of postmenopausal women, association between poor metabolic profile with reduction in cognitive performance is more apparent in women who carry an ApoE4 allele. These data indicate a window of opportunity for interventions to reverse the trajectory of the preclinical phase of Alzheimer's disease.
Collapse
|
13
|
Yare K, Woodward M. Hormone Therapy and Effects on Sporadic Alzheimer’s Disease in Postmenopausal Women: Importance of Nomenclature. J Alzheimers Dis 2020; 73:23-37. [DOI: 10.3233/jad-190896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Katrine Yare
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| | - Michael Woodward
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| |
Collapse
|
14
|
Ghosh S, Durgvanshi S, Agarwal S, Raghunath M, Sinha JK. Current Status of Drug Targets and Emerging Therapeutic Strategies in the Management of Alzheimer's Disease. Curr Neuropharmacol 2020; 18:883-903. [PMID: 32348223 PMCID: PMC7569315 DOI: 10.2174/1570159x18666200429011823] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/09/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease affecting the elderly. AD is associated with a progressive decline in memory and cognitive abilities, drastic changes in behavioural patterns and other psychiatric manifestations. It leads to a significant decline in the quality of life at personal, household as well as national level. Although AD was described about hundred years back and multiple theories have been proposed, its exact pathophysiology is unknown. There is no cure for AD and the life expectancy of AD patients remains low at 3-9 years. An accurate understanding of the molecular mechanism(s) involved in the pathogenesis of AD is imperative to devise a successful treatment strategy. This review explains and summarises the current understanding of different therapeutic strategies based on various molecular pathways known to date. Different strategies based on anti-amyloid pathology, glutamatergic pathway, anti-tau, neuroprotection through neurotrophic factors and cholinergic neurotransmission have been discussed. Further, the use of anti-inflammatory drugs, nutraceuticals, and dietary interventions has also been explained in the management of AD. It further describes different pharmacological and dietary interventions being used in treating and/or managing AD. Additionally, this article provides a thorough review of the literature for improving the therapeutic paradigm of AD.
Collapse
Affiliation(s)
| | | | | | | | - Jitendra Kumar Sinha
- Address correspondence to this author at the Amity Institute of Neuropsychology and Neurosciences (AINN), Amity University UP, Sector-125, Noida 201303, India; Tel: +91-120-4392971, +91-8919679822; Emails: ,
| |
Collapse
|
15
|
Khamphukdee C, Monthakantirat O, Chulikhit Y, Buttachon S, Lee M, Silva AMS, Sekeroglu N, Kijjoa A. Chemical Constituents and Antidepressant-Like Effects in Ovariectomized Mice of the Ethanol Extract of Alternanthera philoxeroides. Molecules 2018; 23:E2202. [PMID: 30200295 PMCID: PMC6225253 DOI: 10.3390/molecules23092202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 01/30/2023] Open
Abstract
The previously unreported flavone glycoside, demethyltorosaflavone B (2) and the E-propenoic acid substituted flavone, torosaflavone E (3a), were isolated together with nine previously reported metabolites, including indole-3-carbaldehyde, oleanonic acid, vanillic acid, p-hydroxybenzoic acid, altheranthin (1a), alternanthin B (1b), demethyltorosaflavone D (3b), luteolin 8-C-E-propenoic acid (4) and chrysoeriol 7-O-rhamnoside (5), from the ethanol extract of the aerial part of Althernanthera philoxeroides. The crude ethanol extract was evaluated for its in vitro estrogenic activity in MCF-7 breast cancer cell line. The crude ethanol extract was also investigated in vivo for its antidepressant-like effects on ovariectomized mice using tail suspension and forced swimming tests, while its effect on the locomotor activity was evaluated by a Y-maze test. The effect of the crude extract on the serum corticosterone level, size and volume of uterus of the ovariectomized mice were also investigated. The expression of the mouse cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF) and β-actin mRNAs in hippocampus and frontal cortex was also evaluated, using semiquantitative reverse transcription-polymerase chain reaction. The crude extract and the isolated compounds 1a, 1b, 3a, 3b and 5, were evaluated for their inhibitory effects on monoamine oxidases (MAOs)-A and -B.
Collapse
Affiliation(s)
- Charinya Khamphukdee
- Graduate School of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Orawan Monthakantirat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Yaowared Chulikhit
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Suradet Buttachon
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal.
| | - Michael Lee
- Department of Chemistry, University of Leicester, University Road, Leicester LE 7 RH, UK.
| | - Artur M S Silva
- Departamento de Química & QOPNA, Universidade de Aveiro, 3810-193 Aveiro, Portugal.
| | - Nazim Sekeroglu
- Department of Food Engineering, Faculty of Architecture and Engineering, Kilis 7 Aralık University, Kilis 79000, Turkey.
| | - Anake Kijjoa
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
16
|
Díaz M, Fabelo N, Ferrer I, Marín R. “Lipid raft aging” in the human frontal cortex during nonpathological aging: gender influences and potential implications in Alzheimer's disease. Neurobiol Aging 2018; 67:42-52. [DOI: 10.1016/j.neurobiolaging.2018.02.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/06/2018] [Accepted: 02/22/2018] [Indexed: 01/27/2023]
|
17
|
Yao Q, Feng M, Yang B, Long Z, Luo S, Luo M, He G, Wang K. Effects of ovarian hormone loss on neuritic plaques and autophagic flux in the brains of adult female APP/PS1 double-transgenic mice. Acta Biochim Biophys Sin (Shanghai) 2018; 50:447-455. [PMID: 29617703 PMCID: PMC5946928 DOI: 10.1093/abbs/gmy032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/07/2018] [Indexed: 01/07/2023] Open
Abstract
Epidemiologic studies have demonstrated that women account for two-thirds of Alzheimer’s disease (AD) cases, for which the decline in circulating gonadal hormone is considered to be one of the major risk factors. In addition, ovarian hormone deficiency may affect β-amyloid (Aβ) deposition, which has a close relationship with autophagic flux. In this study, we investigated the impact of short-term or long-term ovarian hormone deprivation on two mouse models, the non-transgenic (wild-type) and the APP/PS1 double-transgenic AD (2×TgAD) model. Autophagy-related proteins (Beclin1, LC3, and p62) and lysosome-related proteins were detected to evaluate Aβ deposition and autophagy. Our results showed that in the group with short-term depletion of ovarian hormones by ovariectomy (ovx), Beclin1, Cathepsin B (Cath-B), and LAMP1 levels were significantly decreased, while the levels of LC3-II and p62 were increased. In the long-term group, however, there was a sharp decline in Beclin1, LC3-II, Cath-B, and LAMP1 expression but not in p62 expression which is increased. It is worthwhile to note that the occurrence of neuritic plaque-induced ovarian hormone loss increased both the Aβ level and neuritic plaque deposition in 2×TgAD mice. Therefore, autophagy may play an important role in the pathogenesis of female AD, which is also expected to help post-menopausal patients with AD.
Collapse
Affiliation(s)
- Qiuhui Yao
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Min Feng
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Bo Yang
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Shifang Luo
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Min Luo
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Kejian Wang
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Correspondence address. Tel: +86-23-68485763; Fax: +86-23-68485000; E-mail:
| |
Collapse
|
18
|
Gaignard P, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Role of Sex Hormones on Brain Mitochondrial Function, with Special Reference to Aging and Neurodegenerative Diseases. Front Aging Neurosci 2017; 9:406. [PMID: 29270123 PMCID: PMC5725410 DOI: 10.3389/fnagi.2017.00406] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/24/2017] [Indexed: 01/18/2023] Open
Abstract
The mitochondria have a fundamental role in both cellular energy supply and oxidative stress regulation and are target of the effects of sex steroids, particularly the neuroprotective ones. Aging is associated with a decline in the levels of different steroid hormones, and this decrease may underline some neural dysfunctions. Besides, modifications in mitochondrial functions associated with aging processes are also well documented. In this review, we will discuss studies that describe the modifications of brain mitochondrial function and of steroid levels associated with physiological aging and with neurodegenerative diseases. A special emphasis will be placed on describing and discussing our recent findings concerning the concomitant study of mitochondrial function (oxidative phosphorylation, oxidative stress) and brain steroid levels in both young (3-month-old) and aged (20-month-old) male and female mice.
Collapse
Affiliation(s)
- Pauline Gaignard
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Patrice Thérond
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Abdelhamid Slama
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
19
|
Toufexis D, King SB, Michopoulos V. Socially Housed Female Macaques: a Translational Model for the Interaction of Chronic Stress and Estrogen in Aging. Curr Psychiatry Rep 2017; 19:78. [PMID: 28905316 DOI: 10.1007/s11920-017-0833-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Estrogen's role in cognitive aging remains unclear. Despite evidence implicating stress in pathological aging, the interaction of stress with estrogen on cognition in older women has received little attention, and few animal models exist with which to examine this interaction. RECENT FINDINGS We present evidence that aging socially subordinate female macaques that experience chronic psychosocial stress constitute a suitable model to investigate this. First, we review studies showing that estrogen modulates cognition in animal models, as well as studies demonstrating that estrogen's action on certain types of cognition is impaired by stress. Next, we discuss data showing that middle-aged socially subordinate female macaques exhibit distinct stress-induced phenotypes, and review our investigations indicating that estrogen modulates behavior and physiology differently in subordinate female monkeys. We conclude that socially housed female macaques represent a translational animal model for investigating the interplay of chronic stress and estrogen on cognitive aging in women.
Collapse
Affiliation(s)
- Donna Toufexis
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA.,Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - S Bradley King
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA
| | - Vasiliki Michopoulos
- Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA. .,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Moreira AC, Silva AM, Branco AF, Baldeiras I, Pereira GC, Seiça R, Santos MS, Sardão VA. Phytoestrogen coumestrol improves mitochondrial activity and decreases oxidative stress in the brain of ovariectomized Wistar-Han rats. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
21
|
Hormone therapy at early post-menopause increases cognitive control-related prefrontal activity. Sci Rep 2017; 7:44917. [PMID: 28322310 PMCID: PMC5359606 DOI: 10.1038/srep44917] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/14/2017] [Indexed: 12/15/2022] Open
Abstract
Clinical data have been equivocal and controversial as to the benefits to the brain and cognition of hormone therapy (HT) in postmenopausal women. Recent reevaluation of the role of estrogens proposed that HT may effectively prevent the deleterious effects of aging on cognition, and reduces the risks of dementia, including Alzheimer's disease, if initiated early at the beginning of menopause. Yet, little is known about the effects of HT on brain activation related to cognitive control, the ability to make flexible decisions in relation to internal goals. Here, we used fMRI to directly test for a modulation of sequential 17β estradiol (2 mg/day) plus oral progesterone (100 mg/day) on task switching-related brain activity in women at early postmenopause. The results showed that HT enhanced dorsolateral prefrontal cortex recruitment during task switching. Between-subjects correlation analyses revealed that women who engaged more the dorsolateral prefrontal cortex showed higher task switching performance after HT administration. These results suggest that HT, when taken early at the beginning of postmenopause, may have beneficial effect on cognitive control prefrontal mechanisms. Together, these findings demonstrate that HT can prevent the appearance of reduced prefrontal cortex activity, a neurophysiological measure observed both in healthy aging and early dementia.
Collapse
|
22
|
Wang X, Huang Y, Yuan S, Tamadon A, Ma S, Feng Y. The Role of Hippocampal Estradiol Receptor- α in a Perimenopausal Affective Disorders-Like Rat Model and Attenuating of Anxiety by Electroacupuncture. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:4958312. [PMID: 28044085 PMCID: PMC5156811 DOI: 10.1155/2016/4958312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/16/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Hormone replacement therapy is the principal treatment for perimenopausal affective disorders which can cause severe side effects. The present study compared the effects of electroacupuncture (EA) and estradiol treatment on perimenopausal affective disorders at the behavioral and cellular levels. In this randomized experimental in vivo study, adult female rats were divided into intact, ovariectomy, chronic unpredictable stress (CUS), and ovariectomy and CUS combination groups. After week 6, all groups were subdivided to three subgroups of control, EA, and estradiol treatment. The behavioral parameters in the open field and the elevated plus maze tests were assessed before and after treatments. Alterations of serum steroid hormones and changes of estradiol receptor-α (ER-α) immunofluorescence neurons in the hippocampus sections were evaluated. EA treatment caused more antianxiety effects than estradiol treatment in CUS group (P < 0.05). Notably, estradiol and EA treatments had better significant behavioral effects when the models were not estrogen-deficient. Importantly, within each group, compared to the control group, the numbers of ER-α-positive neurons were significantly larger in EA subgroups. Therefore, EA had antianxiety effects on perimenopausal affective disorders caused by CUS but not by estrogen deficiency and upregulation of hippocampus ER-α neurons may contribute to its mechanism of action.
Collapse
Affiliation(s)
- Xun Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongheng Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shiwen Yuan
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Amin Tamadon
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Shulan Ma
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
Grimm A, Mensah-Nyagan AG, Eckert A. Alzheimer, mitochondria and gender. Neurosci Biobehav Rev 2016; 67:89-101. [DOI: 10.1016/j.neubiorev.2016.04.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
|
24
|
Rettberg JR, Dang H, Hodis HN, Henderson VW, St John JA, Mack WJ, Brinton RD. Identifying postmenopausal women at risk for cognitive decline within a healthy cohort using a panel of clinical metabolic indicators: potential for detecting an at-Alzheimer's risk metabolic phenotype. Neurobiol Aging 2016; 40:155-163. [PMID: 26973115 DOI: 10.1016/j.neurobiolaging.2016.01.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 01/08/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
Abstract
Detecting at-risk individuals within a healthy population is critical for preventing or delaying Alzheimer's disease. Systems biology integration of brain and body metabolism enables peripheral metabolic biomarkers to serve as reporters of brain bioenergetic status. Using clinical metabolic data derived from healthy postmenopausal women in the Early versus Late Intervention Trial with Estradiol (ELITE), we conducted principal components and k-means clustering analyses of 9 biomarkers to define metabolic phenotypes. Metabolic clusters were correlated with cognitive performance and analyzed for change over 5 years. Metabolic biomarkers at baseline generated 3 clusters, representing women with healthy, high blood pressure, and poor metabolic phenotypes. Compared with healthy women, poor metabolic women had significantly lower executive, global and memory cognitive performance. Hormone therapy provided metabolic benefit to women in high blood pressure and poor metabolic phenotypes. This panel of well-established clinical peripheral biomarkers represents an initial step toward developing an affordable, rapidly deployable, and clinically relevant strategy to detect an at-risk phenotype of late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Jamaica R Rettberg
- Neuroscience Department, University of Southern California, Los Angeles, CA, USA
| | - Ha Dang
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Howard N Hodis
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Victor W Henderson
- Division of Epidemiology, Department of Health Research and Policy, Stanford University, Stanford, CA, USA; Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jan A St John
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wendy J Mack
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberta Diaz Brinton
- Neuroscience Department, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
25
|
McCarrey AC, Resnick SM. Postmenopausal hormone therapy and cognition. Horm Behav 2015; 74:167-72. [PMID: 25935728 PMCID: PMC4573348 DOI: 10.1016/j.yhbeh.2015.04.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/17/2015] [Accepted: 04/23/2015] [Indexed: 01/17/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Prior to the publication of findings from the Women's Health Initiative (WHI) in 2002, estrogen-containing hormone therapy (HT) was used to prevent age-related disease, especially cardiovascular disease, and to treat menopausal symptoms such as hot flushes and sleep disruptions. Some observational studies of HT in midlife and aging women suggested that HT might also benefit cognitive function, but randomized clinical trials have produced mixed findings in terms of health and cognitive outcomes. This review focuses on hormone effects on cognition and risk for dementia in naturally menopausal women as well as surgically induced menopause, and highlights findings from the large-scale WHI Memory Study (WHIMS) which, contrary to expectation, showed increased dementia risk and poorer cognitive outcomes in older postmenopausal women randomized to HT versus placebo. We consider the 'critical window hypothesis', which suggests that a window of opportunity may exist shortly after menopause during which estrogen treatments are most effective. In addition, we highlight emerging evidence that potential adverse effects of HT on cognition are most pronounced in women who have other health risks, such as lower global cognition or diabetes. Lastly, we point towards implications for future research and clinical treatments.
Collapse
Affiliation(s)
- Anna C McCarrey
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, 21224, USA.
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, 21224, USA.
| |
Collapse
|
26
|
Hara Y, Waters EM, McEwen BS, Morrison JH. Estrogen Effects on Cognitive and Synaptic Health Over the Lifecourse. Physiol Rev 2015; 95:785-807. [PMID: 26109339 PMCID: PMC4491541 DOI: 10.1152/physrev.00036.2014] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Estrogen facilitates higher cognitive functions by exerting effects on brain regions such as the prefrontal cortex and hippocampus. Estrogen induces spinogenesis and synaptogenesis in these two brain regions and also initiates a complex set of signal transduction pathways via estrogen receptors (ERs). Along with the classical genomic effects mediated by activation of ER α and ER β, there are membrane-bound ER α, ER β, and G protein-coupled estrogen receptor 1 (GPER1) that can mediate rapid nongenomic effects. All key ERs present throughout the body are also present in synapses of the hippocampus and prefrontal cortex. This review summarizes estrogen actions in the brain from the standpoint of their effects on synapse structure and function, noting also the synergistic role of progesterone. We first begin with a review of ER subtypes in the brain and how their abundance and distributions are altered with aging and estrogen loss (e.g., ovariectomy or menopause) in the rodent, monkey, and human brain. As there is much evidence that estrogen loss induced by menopause can exacerbate the effects of aging on cognitive functions, we then review the clinical trials of hormone replacement therapies and their effectiveness on cognitive symptoms experienced by women. Finally, we summarize studies carried out in nonhuman primate models of age- and menopause-related cognitive decline that are highly relevant for developing effective interventions for menopausal women. Together, we highlight a new understanding of how estrogen affects higher cognitive functions and synaptic health that go well beyond its effects on reproduction.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Elizabeth M Waters
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Bruce S McEwen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - John H Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| |
Collapse
|
27
|
Sex differences in liver toxicity-do female and male human primary hepatocytes react differently to toxicants in vitro? PLoS One 2015; 10:e0122786. [PMID: 25849576 PMCID: PMC4388670 DOI: 10.1371/journal.pone.0122786] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/18/2015] [Indexed: 01/10/2023] Open
Abstract
There is increasing amount of evidence for sex variation in drug efficiency and toxicity profiles. Women are more susceptible than men to acute liver injury from xenobiotics. In general, this is attributed to sex differences at a physiological level as well as differences in pharmacokinetics and pharmacodynamics, but neither of these can give a sufficient explanation for the diverse responses to xenobiotics. Existing data are mainly based on animal models and limited data exist on in vitro sex differences relevant to humans. To date, male and female human hepatocytes have not yet been compared in terms of their responses to hepatotoxic drugs. We investigated whether sex-specific differences in acute hepatotoxicity can be observed in vitro by comparing hepatotoxic drug effects in male and female primary human hepatocytes. Significant sex-related differences were found for certain parameters and individual drugs, showing an overall higher sensitivity of female primary hepatocytes to hepatotoxicants. Moreover, our work demonstrated that high content screening is feasible with pooled primary human hepatocytes in suspension.
Collapse
|
28
|
Grimm A, Schmitt K, Lang UE, Mensah-Nyagan AG, Eckert A. Improvement of neuronal bioenergetics by neurosteroids: implications for age-related neurodegenerative disorders. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2427-38. [PMID: 25281013 DOI: 10.1016/j.bbadis.2014.09.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 01/09/2023]
Abstract
The brain has high energy requirements to maintain neuronal activity. Consequently impaired mitochondrial function will lead to disease. Normal aging is associated with several alterations in neurosteroid production and secretion. Decreases in neurosteroid levels might contribute to brain aging and loss of important nervous functions, such as memory. Up to now, extensive studies only focused on estradiol as a promising neurosteroid compound that is able to ameliorate cellular bioenergetics, while the effects of other steroids on brain mitochondria are poorly understood or not investigated at all. Thus, we aimed to characterize the bioenergetic modulating profile of a panel of seven structurally diverse neurosteroids (progesterone, estradiol, estrone, testosterone, 3α-androstanediol, DHEA and allopregnanolone), known to be involved in brain function regulation. Of note, most of the steroids tested were able to improve bioenergetic activity in neuronal cells by increasing ATP levels, mitochondrial membrane potential and basal mitochondrial respiration. In parallel, they modulated redox homeostasis by increasing antioxidant activity, probably as a compensatory mechanism to a slight enhancement of ROS which might result from the rise in oxygen consumption. Thereby, neurosteroids appeared to act via their corresponding receptors and exhibited specific bioenergetic profiles. Taken together, our results indicate that the ability to boost mitochondria is not unique to estradiol, but seems to be a rather common mechanism of different steroids in the brain. Thus, neurosteroids may act upon neuronal bioenergetics in a delicate balance and an age-related steroid disturbance might be involved in mitochondrial dysfunction underlying neurodegenerative disorders.
Collapse
Affiliation(s)
- Amandine Grimm
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland; Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland; Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Karen Schmitt
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland; Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland
| | - Undine E Lang
- Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000 Strasbourg, France
| | - Anne Eckert
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular & Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland; Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, CH-4012 Basel, Switzerland.
| |
Collapse
|
29
|
Conroy SK, McDonald BC, Ahles TA, West JD, Saykin AJ. Chemotherapy-induced amenorrhea: a prospective study of brain activation changes and neurocognitive correlates. Brain Imaging Behav 2014; 7:491-500. [PMID: 23793983 DOI: 10.1007/s11682-013-9240-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chemotherapy-induced amenorrhea (CIA) often occurs in pre- and peri-menopausal BC patients, and while cancer/chemotherapy and abrupt estrogen loss have separately been shown to affect cognition and brain function, studies of the cognitive effects of CIA are equivocal, and its effects on brain function are unknown. Functional MRI (fMRI) during a working memory task was used to prospectively assess the pattern of brain activation and deactivation prior to and 1 month after chemotherapy in BC patients who experienced CIA (n = 9), post-menopausal BC patients undergoing chemotherapy (n = 9), and pre- and post-menopausal healthy controls (n = 6 each). Neurocognitive testing was also performed at both time points. Repeated measures general linear models were used to assess statistical significance, and age was a covariate in all analyses. We observed a group-by-time interaction in the combined magnitudes of brain activation and deactivation (p = 0.006): the CIA group increased in magnitude from baseline to post-treatment while other groups maintained similar levels over time. Further, the change in brain activity magnitude in CIA was strongly correlated with change in processing speed neurocognitive testing score (r = 0.837 p = 0.005), suggesting this increase in brain activity reflects effective cognitive compensation. Our results demonstrate prospectively that the pattern of change in brain activity from pre- to post-chemotherapy varies according to pre-treatment menopausal status. Cognitive correlates add to the potential clinical significance of these findings. These findings have implications for risk appraisal and development of prevention or treatment strategies for cognitive changes in CIA.
Collapse
Affiliation(s)
- Susan K Conroy
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Suite 4100, Indianapolis, IN, 46202, USA
| | | | | | | | | |
Collapse
|
30
|
Women with the Alzheimer's risk marker ApoE4 lose Aβ-specific CD4⁺ T cells 10-20 years before men. Transl Psychiatry 2014; 4:e414. [PMID: 25072319 PMCID: PMC4119219 DOI: 10.1038/tp.2014.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/21/2014] [Accepted: 05/22/2014] [Indexed: 12/13/2022] Open
Abstract
Adaptive immunity to self-antigens causes autoimmune disorders, such as multiple sclerosis, psoriasis and type 1 diabetes; paradoxically, T- and B-cell responses to amyloid-β (Aβ) reduce Alzheimer's disease (AD)-associated pathology and cognitive impairment in mouse models of the disease. The manipulation of adaptive immunity has been a promising therapeutic approach for the treatment of AD, although vaccine and anti-Aβ antibody approaches have proven difficult in patients, thus far. CD4(+) T cells have a central role in regulating adaptive immune responses to antigens, and Aβ-specific CD4(+) T cells have been shown to reduce AD pathology in mouse models. As these cells may facilitate endogenous mechanisms that counter AD, an evaluation of their abundance before and during AD could provide important insights. Aβ-CD4see is a new assay developed to quantify Aβ-specific CD4(+) T cells in human blood, using dendritic cells derived from human pluripotent stem cells. In tests of >50 human subjects Aβ-CD4see showed an age-dependent decline of Aβ-specific CD4(+) T cells, which occurs earlier in women than men. In aggregate, men showed a 50% decline in these cells by the age of 70 years, but women reached the same level before the age of 60 years. Notably, women who carried the AD risk marker apolipoproteinE-ɛ4 (ApoE4) showed the earliest decline, with a precipitous drop between 45 and 52 years, when menopause typically begins. Aβ-CD4see requires a standard blood draw and provides a minimally invasive approach for assessing changes in Aβ biology that may reveal AD-related changes in physiology by a decade. Furthermore, CD4see probes can be modified to target any peptide, providing a powerful new tool to isolate antigen-specific CD4(+) T cells from human subjects.
Collapse
|
31
|
Rocca WA, Grossardt BR, Shuster LT. Oophorectomy, estrogen, and dementia: a 2014 update. Mol Cell Endocrinol 2014; 389:7-12. [PMID: 24508665 PMCID: PMC4040304 DOI: 10.1016/j.mce.2014.01.020] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/23/2014] [Accepted: 01/23/2014] [Indexed: 10/25/2022]
Abstract
Current evidence suggests that estrogen may have beneficial, neutral, or detrimental effects on the brain depending on age, type of menopause (natural versus induced), or stage of menopause (early versus late), consistent with the timing hypothesis. Three studies have now compared women who underwent bilateral oophorectomy before menopause with referent women and consistently showed an increased risk of cognitive decline and dementia. These studies suggest a sizeable neuroprotective effect of estrogen naturally produced by the ovaries before age 50 years. In this article, we focus on neuroprotection as related to cognitive decline and dementia. Several case-control studies and cohort studies also showed neuroprotective effects in women who received estrogen treatment (ET) in the early postmenopausal stage (most commonly at ages 50-60 years). The majority of women in those observational studies had undergone natural menopause and were treated for the relief of menopausal symptoms. However, the clinical trials by the Women's Health Initiative showed that women who initiated ET alone or in combination with a progestin in the late postmenopausal stage (ages 65-79 years) experienced an increased risk of dementia and cognitive decline regardless of the type of menopause. Three observational studies have now formally tested the timing hypothesis, and showed that the neuroprotective or harmful effects of estrogen depend on age at the time of initiation of treatment and on stage of menopause. Therefore, women who undergo bilateral oophorectomy before the onset of menopause or women who experience premature or early natural menopause should be considered for hormonal treatment until the average age of natural menopause (around age 50 years). Recommendations for the use of ET by women who experience natural menopause at typical ages remain less certain, and more research is needed.
Collapse
Affiliation(s)
- Walter A Rocca
- Division of Epidemiology, Department of Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA; Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Brandon R Grossardt
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lynne T Shuster
- Women's Health Clinic, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Scott EL, Zhang QG, Vadlamudi RK, Brann DW. Premature menopause and risk of neurological disease: basic mechanisms and clinical implications. Mol Cell Endocrinol 2014; 389:2-6. [PMID: 24462786 PMCID: PMC4040297 DOI: 10.1016/j.mce.2014.01.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/14/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
Since basic scientific studies in the 1990s revealed dramatic gender differences in neurological damage from cerebral ischemia, significant evidence has accumulated for a neuroprotective role of ovarian-derived 17β-Estradiol (E2). Intriguingly, observational studies have further suggested that early and prolonged loss of ovarian E2 (premature menopause) leads to a doubled lifetime risk for dementia and a fivefold increased risk of mortality from neurological disorders, but some controversy remains. Here, we briefly summarize and analyze clinical cohort studies assessing the detrimental neurological outcomes of premature menopause. Furthermore, we discuss current basic science studies elucidating the molecular mechanisms underlying the enhanced risk of neurological disease in prematurely menopausal women and the "window of opportunity" for estrogen benefit. Finally, we highlight four critical issues in the field that require collaboration between basic scientists and clinicians for successful resolution, with the ultimate goal of maintaining optimal neurological health in prematurely menopausal women.
Collapse
Affiliation(s)
- Erin L Scott
- University System of Georgia MD/PhD Program, Georgia Regents University, Augusta, GA 30912, USA; Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA 30912, USA
| | - Quan-Guang Zhang
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Darrell W Brann
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
33
|
Brand MD. The role of mitochondria in longevity and healthspan. LONGEVITY & HEALTHSPAN 2014; 3:7. [PMID: 24855560 PMCID: PMC4030464 DOI: 10.1186/2046-2395-3-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 05/06/2014] [Indexed: 12/21/2022]
Abstract
The role of mitochondria in aging and disease remains contentious more than 40 years after the mitochondrial free radical theory of aging was first proposed. As part of a wider cross-journal series on contemporary mitochondrial biology, Longevity & Healthspan presents a thematic series of four reviews that discuss the evidence for and against the modern incarnations of the theory, and examine the relevance of mitochondrial membrane phospholipid unsaturation and the interactions of mitochondria with sex hormones.
Collapse
Affiliation(s)
- Martin D Brand
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA
| |
Collapse
|
34
|
Velarde MC. Mitochondrial and sex steroid hormone crosstalk during aging. LONGEVITY & HEALTHSPAN 2014; 3:2. [PMID: 24495597 PMCID: PMC3922316 DOI: 10.1186/2046-2395-3-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/08/2014] [Indexed: 12/21/2022]
Abstract
Decline in circulating sex steroid hormones accompanies several age-associated pathologies which may influence human healthspan. Mitochondria play important roles in biosynthesis of sex steroid hormones, and these hormones can also regulate mitochondrial function. Understanding the cross talk between mitochondria and sex steroid hormones may provide insights into the pathologies associated with aging. The aim of this review is to summarize the current knowledge regarding the interplay between mitochondria and sex steroid hormones during the aging process. The review describes the effect of mitochondria on sex steroid hormone production in the gonads, and then enumerates the contribution of sex steroid hormones on mitochondrial function in hormone responsive cells. Decline in sex steroid hormones and accumulation of mitochondrial damage may create a positive feedback loop that contributes to the progressive degeneration in tissue function during aging. The review further speculates whether regulation between mitochondrial function and sex steroid hormone action can potentially influence healthspan.
Collapse
Affiliation(s)
- Michael C Velarde
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.
| |
Collapse
|
35
|
|
36
|
Hara Y, Yuk F, Puri R, Janssen WGM, Rapp PR, Morrison JH. Presynaptic mitochondrial morphology in monkey prefrontal cortex correlates with working memory and is improved with estrogen treatment. Proc Natl Acad Sci U S A 2014; 111:486-91. [PMID: 24297907 PMCID: PMC3890848 DOI: 10.1073/pnas.1311310110] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Humans and nonhuman primates are vulnerable to age- and menopause-related decline in working memory, a cognitive function reliant on the energy-demanding recurrent excitation of neurons within Brodmann's Area 46 of the dorsolateral prefrontal cortex (dlPFC). Here, we tested the hypothesis that the number and morphology (straight, curved, or donut-shaped) of mitochondria in dlPFC presynaptic boutons are altered with aging and menopause in rhesus monkeys (Macaca mulatta) and that these metrics correlate with delayed response (DR) accuracy, a well-characterized measure of dlPFC-dependent working memory. Although presynaptic bouton density or size was not significantly different across groups distinguished by age or menses status, DR accuracy correlated positively with the number of total and straight mitochondria per dlPFC bouton. In contrast, DR accuracy correlated inversely with the frequency of boutons containing donut-shaped mitochondria, which exhibited smaller active zone areas and fewer docked synaptic vesicles than those with straight or curved mitochondria. We then examined the effects of estrogen administration to test whether a treatment known to improve working memory influences mitochondrial morphology. Aged ovariectomized monkeys treated with vehicle displayed significant working memory impairment and a concomitant 44% increase in presynaptic donut-shaped mitochondria, both of which were reversed with cyclic estradiol treatment. Together, our data suggest that hormone replacement therapy may benefit cognitive aging, in part by promoting mitochondrial and synaptic health in the dlPFC.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Frank Yuk
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Rishi Puri
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - William G. M. Janssen
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Peter R. Rapp
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224
| | - John H. Morrison
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
- Department of Geriatrics and Palliative Medicine, and
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| |
Collapse
|
37
|
Bernal-Mondragón C, Rivas-Arancibia S, Kendrick KM, Guevara-Guzmán R. Estradiol prevents olfactory dysfunction induced by A-β 25-35 injection in hippocampus. BMC Neurosci 2013; 14:104. [PMID: 24059981 PMCID: PMC3849069 DOI: 10.1186/1471-2202-14-104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 09/19/2013] [Indexed: 01/09/2023] Open
Abstract
Background Some neurodegenerative diseases, such as Alzheimer and Parkinson, present an olfactory impairment in early stages, and sometimes even before the clinical symptoms begin. In this study, we assess the role of CA1 hippocampus (structure highly affected in Alzheimer disease) subfield in the rats’ olfactory behavior, and the neuroprotective effect of 17 beta estradiol (E2) against the oxidative stress produced by the injection of amyloid beta 25–35. Results 162 Wistar rats were ovariectomized and two weeks after injected with 2 μl of amyloid beta 25–35 (A-β25–35) in CA1 subfield. Olfactory behavior was evaluated with a social recognition test, odor discrimination, and search tests. Oxidative stress was evaluated with FOX assay and Western Blot against 4-HNE, Fluoro Jade staining was made to quantify degenerated neurons; all these evaluations were performed 24 h, 8 or 15 days after A-β25–35 injection. Three additional groups treated with 17 beta estradiol (E2) were also evaluated. The injection of A-β25–35 produced an olfactory impairment 24 h and 8 days after, whereas a partial recovery of the olfactory behavior was observed at 15 days. A complete prevention of the olfactory impairment was observed with the administration of E2 two weeks before the amyloid injection (A-β25–35 24 h + E2) and one or two weeks after (groups 8 A-β +E2 and 15 A-β +E2 days, respectively); a decrease of the oxidative stress and neurodegeneration were also observed. Conclusions Our finding shows that CA1 hippocampus subfield plays an important role in the olfactory behavior of the rat. The oxidative stress generated by the administration of A-β25–35 is enough to produce an olfactory impairment. This can be prevented with the administration of E2 before and after amyloid injection. This suggests a possible therapeutic use of estradiol in Alzheimer’s disease.
Collapse
Affiliation(s)
- Carlos Bernal-Mondragón
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Apdo, Postal 70250, D,F, México, Delegación Coyoacán 04510, Mexico.
| | | | | | | |
Collapse
|
38
|
O'Brien J, Jackson JW, Grodstein F, Blacker D, Weuve J. Postmenopausal hormone therapy is not associated with risk of all-cause dementia and Alzheimer's disease. Epidemiol Rev 2013; 36:83-103. [PMID: 24042430 DOI: 10.1093/epirev/mxt008] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The relationship of postmenopausal hormone therapy with all-cause dementia and Alzheimer's disease dementia has been controversial. Given continued interest in the role of hormone therapy in chronic disease prevention and the emergence of more prospective studies, we conducted a systematic review to identify all epidemiologic studies meeting prespecified criteria reporting on postmenopausal hormone therapy use and risk of Alzheimer's disease or dementia. A systematic search of Medline and Embase through December 31, 2012, returned 15 articles meeting our criteria. Our meta-analysis of any versus never use did not support the hypothesis that hormone therapy reduces risk of Alzheimer's disease (summary estimate = 0.88, 95% confidence interval: 0.66, 1.16). Exclusion of trial findings did not change this estimate. There were not enough all-cause dementia results for a separate meta-analysis, but when we combined all-cause dementia results (n = 3) with Alzheimer's disease results (n = 7), the summary estimate remained null (summary estimate = 0.94, 95% confidence interval: 0.71, 1.26). The limited explorations of timing of use-both duration and early initiation-did not yield consistent findings. Our findings support current recommendations that hormone therapy should not be used for dementia prevention. We discuss trends in hormone therapy research that could explain our novel findings and highlight areas where additional data are needed.
Collapse
Affiliation(s)
- Jacqueline O'Brien
- Abbreviations: CEE, conjugated equine estrogen; CEE+MPA, conjugated equine estrogen plus medroxyprogesterone acetate combined; CI, confidence interval; MPA, medroxyprogesterone acetate; RR, relative risk; WHI, Women's Health Initiative; WHIMS, Women's Health Initiative Memory Study
| | | | | | | | | |
Collapse
|
39
|
Baxter MG, Roberts MT, Gee NA, Lasley BL, Morrison JH, Rapp PR. Multiple clinically relevant hormone therapy regimens fail to improve cognitive function in aged ovariectomized rhesus monkeys. Neurobiol Aging 2013; 34:1882-90. [PMID: 23369546 PMCID: PMC3622837 DOI: 10.1016/j.neurobiolaging.2012.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 12/10/2012] [Accepted: 12/22/2012] [Indexed: 10/27/2022]
Abstract
Preclinical studies in aged, surgically-menopausal rhesus monkeys have revealed powerful benefits of intermittent estrogen injections on prefrontal cortex-dependent working memory, together with corresponding effects on dendritic spine morphology in the prefrontal cortex. This contrasts with the inconsistent effects of hormone therapy (HT) reported in clinical studies in women. Factors contributing to this discrepancy could include differences in the formulation and sequence of HT regimens, resulting in different neurobiological outcomes. The current study evaluated, in aging surgically menopausal rhesus monkeys, the cognitive effects of 4 HT regimens modeled directly on human clinical practice, including continuous estrogen treatment opposed by progesterone. None of the regimens tested produced any cognitive effect, despite yielding physiologically relevant serum hormone levels, as intended. These findings have implications for the design of regimens that might optimize the benefits of hormone treatment for healthy aging, and suggest that common HT protocols used by women may fail to result in substantial cognitive benefit, at least via direct effects on the prefrontal cortex.
Collapse
Affiliation(s)
- Mark G Baxter
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
40
|
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: 7.3] [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
|
41
|
Ishunina TA, Sluiter AA, Swaab DF, Verwer RWH. Transcriptional activity of human brain estrogen receptor-α splice variants: evidence for cell type-specific regulation. Brain Res 2013; 1500:1-9. [PMID: 23333800 DOI: 10.1016/j.brainres.2012.12.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 12/19/2012] [Accepted: 12/27/2012] [Indexed: 12/19/2022]
Abstract
Estrogen receptor α (ERα) isoforms with complex types of alternative splicing are naturally present in the human brain and may affect canonical receptor signaling. In the present study we investigated transcriptional activity of common ERα splice variants from this group with different molecular defects: MB1 (intron retention), TADDI (small deletion between exons 3 and 4 with an insert), the Δ (deletion) 3(⁎)-7(*)/819 (complete skipping of exons 4, 5 and 6 and partial deletion of exons 3 and 7) and the Δ3-6 (lacking exons 3, 4, 5 and 6) in HeLa and M17 cells upon stimulation with (17β)estradiol or insulin-like growth factor 1 (IGF-1). In HeLa cells, all these splice variants showed the dominant negative function that was more pronounced for the TADDI. In M17 cells the dominant negative variants appeared to be the MB1 and the Δ3-6, whereas TADDI turned out to be a clearly dominant positive variant. In M17 cells mRNA levels of Δ3-6 and Δ3(*)-7(*)/819 variants increased following (17β)estradiol administration. In Hela cells (17β)estradiol up-regulated the IGF-1 receptor mRNA levels in cultures transfected with MB1, TADDI and Δ3(*)-7(*)/819. Our data demonstrate that ERα splice variants show differential levels of the transcriptional activity in a cell type-specific way and that IGF-1 signaling pathways are differentially employed in a cell-type specific manner depending on the level of the discrete ERα splice variants expressed. Functional properties of various ERα splice variants and their cell type-specificity should, thus, be considered as potential confounders of estrogen therapy effects on the brain.
Collapse
Affiliation(s)
- T A Ishunina
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Hedges VL, Ebner TJ, Meisel RL, Mermelstein PG. The cerebellum as a target for estrogen action. Front Neuroendocrinol 2012; 33:403-11. [PMID: 22975197 PMCID: PMC3496070 DOI: 10.1016/j.yfrne.2012.08.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 08/20/2012] [Accepted: 08/24/2012] [Indexed: 12/22/2022]
Abstract
This review focuses on the effects of estrogens upon the cerebellum, a brain region long ignored as a site of estrogen action. Highlighted are the diverse effects of estradiol within the cerebellum, emphasizing the importance of estradiol signaling in cerebellar development, modulation of synaptic neurotransmission in the adult, and the potential influence of estrogens on various health and disease states. We also provide new data, consistent with previous studies, in which locally synthesized estradiol modulates cerebellar glutamatergic neurotransmission, providing one underlying mechanism by which the actions of estradiol can affect this brain region.
Collapse
Affiliation(s)
- Valerie L Hedges
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | | | | | | |
Collapse
|
43
|
Simpkins JW, Singh M, Brock C, Etgen AM. Neuroprotection and estrogen receptors. Neuroendocrinology 2012; 96:119-30. [PMID: 22538356 PMCID: PMC6507404 DOI: 10.1159/000338409] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/18/2012] [Indexed: 11/19/2022]
Abstract
This review is intended to assess the state of current knowledge on the role of estrogen receptors (ERs) in the neuroprotective effects of estrogens in models for acute neuronal injury and death. We evaluate the overall evidence that estrogens are neuroprotective in acute injury and critically assess the role of ERα, ERβ, GPR 30, and nonreceptor-mediated mechanisms in these robust neuroprotective effects of this ovarian steroid hormone. We conclude that all three receptors, as well as nonreceptor-mediated mechanisms can be involved in neuroprotection, depending on the model used, the level of estrogen administrated, and the mode of administration of the steroid. Also, the signaling pathways used by both ER-dependent and ER-independent mechanisms to exert neuroprotection are considered. Finally, further studies that are needed to parse out the relative contribution of receptor versus nonreceptor-mediated signaling are discussed.
Collapse
Affiliation(s)
- James W. Simpkins
- Department of Pharmacology & Neuroscience, Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Meharvan Singh
- Department of Pharmacology & Neuroscience, Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Courtney Brock
- Department of Pharmacology & Neuroscience, Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Anne M. Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer Building, Room 113, Bronx, NY 10461
| |
Collapse
|
44
|
Yao J, Irwin R, Chen S, Hamilton R, Cadenas E, Brinton RD. Ovarian hormone loss induces bioenergetic deficits and mitochondrial β-amyloid. Neurobiol Aging 2012; 33:1507-21. [PMID: 21514693 PMCID: PMC3181273 DOI: 10.1016/j.neurobiolaging.2011.03.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 02/15/2011] [Accepted: 03/01/2011] [Indexed: 01/08/2023]
Abstract
Previously, we demonstrated that reproductive senescence was associated with mitochondrial deficits comparable to those of female triple-transgenic Alzheimer's mice (3xTgAD). Herein, we investigated the impact of chronic ovarian hormone deprivation and 17β-estradiol (E2) replacement on mitochondrial function in nontransgenic (nonTg) and 3xTgAD female mouse brain. Depletion of ovarian hormones by ovariectomy (OVX) in nontransgenic mice significantly decreased brain bioenergetics, and induced mitochondrial dysfunction and oxidative stress. In 3xTgAD mice, OVX significantly exacerbated mitochondrial dysfunction and induced mitochondrial β-amyloid and β-amyloid (Aβ)-binding-alcohol-dehydrogenase (ABAD) expression. Treatment with E2 at OVX prevented OVX-induced mitochondrial deficits, sustained mitochondrial bioenergetic function, decreased oxidative stress, and prevented mitochondrial β-amyloid and ABAD accumulation. In vitro, E2 increased maximal mitochondrial respiration in neurons and basal and maximal respiration in glia. Collectively, these data demonstrate that ovarian hormone loss induced a mitochondrial phenotype comparable to a transgenic female model of Alzheimer's disease (AD), which was prevented by E2. These findings provide a plausible mechanism for increased risk of Alzheimer's disease in premenopausally oophorectomized women while also suggesting a therapeutic strategy for prevention.
Collapse
Affiliation(s)
- Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
| | - Ronald Irwin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
| | - Shuhua Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
| | - Ryan Hamilton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
| | - Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| |
Collapse
|
45
|
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: 185] [Impact Index Per Article: 14.2] [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
|
46
|
Alzheimer's disease, oestrogen and mitochondria: an ambiguous relationship. Mol Neurobiol 2012; 46:151-60. [PMID: 22678467 PMCID: PMC3443477 DOI: 10.1007/s12035-012-8281-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/21/2012] [Indexed: 11/02/2022]
Abstract
Hormonal deficit in post-menopausal women has been proposed to be one risk factor in Alzheimer's disease (AD) since two thirds of AD patients are women. However, large treatment trials showed negative effects of long-term treatment with oestrogens in older women. Thus, oestrogen treatment after menopause is still under debate, and several hypotheses trying to explain the failure in outcome are under discussion. Concurrently, it was shown that amyloid-beta (Aβ) peptide, the main constituent of senile plaques, as well as abnormally hyperphosphorylated tau protein, the main component of neurofibrillary tangles, can modulate the level of neurosteroids which notably represent neuroactive steroids synthetized within the nervous system, independently of peripheral endocrine glands. In this review, we summarize the role of neurosteroids especially that of oestrogen in AD and discuss their potentially neuroprotective effects with specific regard to the role of oestrogens on the maintenance and function of mitochondria, important organelles which are highly vulnerable to Aβ- and tau-induced toxicity. We also discuss the role of Aβ-binding alcohol dehydrogenase (ABAD), a mitochondrial enzyme able to bind Aβ peptide thereby modifying mitochondrial function as well as oestradiol levels suggesting possible modes of interaction between the three, and the potential therapeutic implication of inhibiting Aβ-ABAD interaction.
Collapse
|
47
|
Benedusi V, Meda C, Della Torre S, Monteleone G, Vegeto E, Maggi A. A lack of ovarian function increases neuroinflammation in aged mice. Endocrinology 2012; 153:2777-88. [PMID: 22492304 PMCID: PMC3359599 DOI: 10.1210/en.2011-1925] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although several lines of evidence have indicated that menopause is associated with increased susceptibility to neurological disorders, the mechanisms involved in this phenomenon remain to be elucidated. Because neuroinflammation is a common feature of a number of brain diseases, we hypothesized that the cessation of ovarian functions and the consequent decrease in estrogen receptor (ER)-mediated antiinflammatory activity may represent a trigger for postmenopausal brain dysfunctions. The aim of the present study was to investigate the effects of aging and surgical menopause on the activity of ER in neuroinflammation. The present study shows that ER genes are expressed in the hippocampus, but ER transcriptional activity decreases significantly beginning at 12 months of age in intact and ovariectomized mice. With ovariectomy, we observe an age-dependent accumulation of mRNA encoding inflammatory mediators (e.g. TNFα, IL1β, and macrophage inflammatory protein-2) and changes in the morphology of astroglia and microglia. In addition, we show that aging itself is coupled with an exaggerated response to acute inflammatory stimuli with a major accumulation of TNFα, IL1β, macrophage inflammatory protein-2, and macrophage chemoattractant protein-1 mRNA in response to lipopolysaccharide administration. The response to acute inflammatory stimuli appears to be differentially modulated by the duration of hormone deprivation in 12-month-old mice. Taken together, the present results show that aging is associated with decreased ER activity, despite continuous ER synthesis, and that age-dependent neuroinflammation is strongly influenced by hormone deprivation.
Collapse
Affiliation(s)
- Valeria Benedusi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | | | | | | | | | | |
Collapse
|
48
|
Zamora R, Bartholow J, Green E, Morgan CD, Murphy C. Adiposity measures predict olfactory processing speed in older adult carriers of the apolipoprotein E ε4 allele. Clin Neurophysiol 2012; 123:918-24. [PMID: 22055839 PMCID: PMC3691270 DOI: 10.1016/j.clinph.2011.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 08/25/2011] [Accepted: 09/01/2011] [Indexed: 10/15/2022]
Abstract
OBJECTIVE The current study investigated the relationship between adiposity and P3 latency. METHODS Fifty-one adults in two age groups (18-25 and 65+) participated. Odor stimuli were delivered via olfactometer as participants focused on a computer screen. Each stimulus was followed by presentation on the screen of four odor identification choices. Participants attempted identification by button press. Olfactory event-related potentials (OERPs) were recorded. BMI and waist circumference were measured as indicators of adiposity. RESULTS In bivariate analyses with all participants included, positive correlations for P3 latency with both BMI and waist circumference were observed, indicating that as adiposity increased latencies also increased. When each age group was separately examined, correlations between adiposity measures and latency remained statistically significant for older adults. Furthermore, ApoE ε4 allele status was examined. Latencies remained positively correlated with adiposity in older adult ε4 carriers; but not in non-carriers. CONCLUSIONS This study indicates that adiposity predicts olfactory processing speed in older adults, specifically in ε4 carriers. SIGNIFICANCE The results suggest that olfactory processing speed may be a useful measure for detecting and following the effects of adiposity on brain integrity and cognitive function in those at genetic risk for AD.
Collapse
Affiliation(s)
- R Zamora
- San Diego State University, Department of Psychology, USA
| | | | | | | | | |
Collapse
|
49
|
Yao J, Brinton RD. Estrogen regulation of mitochondrial bioenergetics: implications for prevention of Alzheimer's disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 64:327-71. [PMID: 22840752 PMCID: PMC3970844 DOI: 10.1016/b978-0-12-394816-8.00010-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with a complex and progressive pathological phenotype characterized first by hypometabolism and impaired mitochondrial bioenergetics followed by pathological burden. Increasing evidence indicates an antecedent and potentially causal role of mitochondrial bioenergetic deficits and brain hypometabolism coupled with increased mitochondrial oxidative stress in AD pathogenesis. Compromised aerobic glycolysis pathway coupled with oxidative stress is first accompanied by a shift toward a ketogenic pathway that eventually progresses into fatty acid oxidation (FAO) pathways and leads to white matter degeneration and overproduction and mitochondrial accumulation of β-amyloid. Estrogen-induced signaling pathways converge upon the mitochondria to enhance mitochondrial function and to sustain aerobic glycolysis coupled with citric acid cycle-driven oxidative phosphorylation to potentiate ATP (Adenosine triphosphate) generation. In addition to potentiated mitochondrial bioenergetics, estrogen also enhances neural survival and health through maintenance of calcium homeostasis, promotion of antioxidant defense against free radicals, efficient cholesterol trafficking, and beta amyloid clearance. Significantly, the convergence of E2 mechanisms of action onto mitochondria is also a potential point of vulnerability when activated in diseased neurons that exacerbates degeneration through increased load on dysregulated calcium homeostasis. The "healthy cell bias of estrogen action" hypothesis examines the role that regulating mitochondrial function and bioenergetics play in promoting neural health and the mechanistic crossroads that lead to divergent outcomes following estrogen exposure. As the continuum of neurological health progresses from healthy to unhealthy, so too do the benefits of estrogen or hormone therapy.
Collapse
Affiliation(s)
- Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
50
|
Abstract
Oestradiol exerts a profound influence upon multiple brain circuits. For the most part, these effects are mediated by oestrogen receptor (ER)α. We review here the roles of ERβ, the other ER isoform, in mediating rodent oestradiol-regulated anxiety, aggressive and sexual behaviours, the control of gonadotrophin secretion, and adult neurogenesis. Evidence exists for: (i) ERβ located in the paraventricular nucleus underpinning the suppressive influence of oestradiol on the stress axis and anxiety-like behaviour; (ii) ERβ expressed in gonadotrophin-releasing hormone neurones contributing to oestrogen negative-feedback control of gonadotrophin secretion; (iii) ERβ controlling the offset of lordosis behaviour; (iv) ERβ suppressing aggressive behaviour in males; (v) ERβ modulating responses to social stimuli; and (vi) ERβ in controlling adult neurogenesis. This review highlights two major themes; first, ERβ and ERα are usually tightly inter-related in the oestradiol-dependent control of a particular brain function. For example, even though oestradiol feedback to control reproduction occurs principally through ERα-dependent mechanisms, modulatory roles for ERβ also exist. Second, the roles of ERα and ERβ within a particular neural network may be synergistic or antagonistic. Examples of the latter include the role of ERα to enhance, and ERβ to suppress, anxiety-like and aggressive behaviours. Splice variants such as ERβ2, acting as dominant negative receptors, are of further particular interest because their expression levels may reflect preceeding oestradiol exposure of relevance to oestradiol replacement therapy. Together, this review highlights the predominant modulatory, but nonetheless important, roles of ERβ in mediating the many effects of oestradiol upon adult brain function.
Collapse
Affiliation(s)
- R. J. Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - S. Ogawa
- Laboratory of Behavioral Neuroendocrinology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - J. M. Wang
- Department of Pathology, Pharmacology and Toxicology, Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - A. E. Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| |
Collapse
|