1
|
Kritzer MF, Adler A, Locklear M. Androgen effects on mesoprefrontal dopamine systems in the adult male brain. Neuroscience 2024:S0306-4522(24)00306-3. [PMID: 38977069 DOI: 10.1016/j.neuroscience.2024.07.001] [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: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Epidemiological data show that males are more often and/or more severely affected by symptoms of prefrontal cortical dysfunction in schizophrenia, Parkinson's disease and other disorders in which dopamine circuits associated with the prefrontal cortex are dysregulated. This review focuses on research showing that these dopamine circuits are powerfully regulated by androgens. It begins with a brief overview of the sex differences that distinguish prefrontal function in health and prefrontal dysfunction or decline in aging and/or neuropsychiatric disease. This review article then spotlights data from human subjects and animal models that specifically identify androgens as potent modulators of prefrontal cortical operations and of closely related, functionally critical measures of prefrontal dopamine level or tone. Candidate mechanisms by which androgens dynamically control mesoprefrontal dopamine systems and impact prefrontal states of hypo- and hyper-dopaminergia in aging and disease are then considered. This is followed by discussion of a working model that identifies a key locus for androgen modulation of mesoprefrontal dopamine systems as residing within the prefrontal cortex itself. The last sections of this review critically consider the ways in which the organization and regulation of mesoprefrontal dopamine circuits differ in the adult male and female brain, and highlights gaps where more research is needed.
Collapse
Affiliation(s)
- Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, United States.
| | - Alexander Adler
- Department of Oncology and Immuno-Oncology, Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, United States
| | | |
Collapse
|
2
|
Kang J, Yan J, Yan W. Testosterone ameliorated the behavioural deficits of gonadectomised rats and counteracted free radicals in a dosage-dependent manner. Behav Brain Res 2023; 450:114501. [PMID: 37207980 DOI: 10.1016/j.bbr.2023.114501] [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: 01/27/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Testosterone deficiency may induce behavioural changes in individuals. Oxidative stress resulting from a redox imbalance may be implicated in the initiation and progression of neurobehavioural disorders. However, whether exogenous testosterone intervention in male gonadectomised (GDX) rats ameliorates oxidative stress and plays a neuroprotective role remains unknown. Therefore, we examined this hypothesis by performing sham or gonadectomy surgeries on Sprague-Dawley rats with or without supplementation with different doses of testosterone propionate (TP). Open field and Morris water maze tests were performed, the serum and brain testosterone levels, and oxidative stress markers were analysed. GDX and lower TP doses (0.5mg/kg) induced reduced exploratory and motor behaviours, but impaired spatial learning and memory compared to Sham rats. Administration of physiological TP levels (0.75-1.25mg/kg) to the GDX rats restored the behaviour observed in the intact rats. However, higher TP doses (1.5-3.0mg/kg) induced increased exploratory and motor behaviours but impaired spatial learning and memory. These behavioural impairments were accompanied by a marked decrease in levels of antioxidant enzymes (superoxide dismutase and catalase) and an increase in lipid peroxidation levels in the substantia nigra and hippocampus. These findings indicate that TP administration can alter behavioural performance and induce memory and learning impairment, which may result from changes in redox homeostasis in male GDX animals.
Collapse
Affiliation(s)
- Jie Kang
- Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Jixing Yan
- College of Rehabilitation Sciences, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Wensheng Yan
- Department of Sports Medicine, Hebei Sport University, Shijiazhuang, PR China.
| |
Collapse
|
3
|
Branigan GL, Torrandell‐Haro G, Soto M, Gelmann EP, Vitali F, Rodgers KE, Brinton RD. Androgen-targeting therapeutics mitigate the adverse effect of GnRH agonist on the risk of neurodegenerative disease in men treated for prostate cancer. Cancer Med 2022; 11:2687-2698. [PMID: 35293700 PMCID: PMC9249980 DOI: 10.1002/cam4.4650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/18/2022] [Accepted: 02/23/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Prostate cancer and multiple neurodegenerative diseases (NDD) share an age-associated pattern of onset. Therapy of prostate cancer is known to impact cognitive function. The objective of this study was to determine the impact of multiple classes of androgen-targeting therapeutics (ATT) on the risk of NDD. METHODS A retrospective cohort study of men aged 45 and older with prostate within the US-based Mariner claims data set between January 1 and 27, 2021. A propensity score approach was used to minimize measured and unmeasured selection bias. Disease risk was determined using Kaplan-Meier survival analyses. RESULTS Of the 1,798,648 men with prostate cancer, 209,722 met inclusion criteria. Mean (SD) follow-up was 6.4 (1.8) years. In the propensity score-matched population, exposure to ATT was associated with a minimal increase in NDD incidence (relative risk [RR], 1.07; 95% CI, 1.05-1.10; p < 0.001). However, GnRH agonists alone were associated with significantly increased NDD risk (RR, 1.47; 95% CI, 1.30-1.66; p <0.001). Abiraterone, commonly administered with GnRH agonists and low-dose prednisone, was associated with a significantly decreased risk (RR, 0.77; 95% CI, 0.68-0.87; p < 0.001) of any NDD. CONCLUSIONS Among patients with prostate cancer, GnRH agonist exposure was associated with an increased NDD risk. Abiraterone acetate reduced the risks of Alzheimer's disease and Parkinson's disease conferred by GnRH agonists, whereas the risk for ALS was reduced by androgen receptor inhibitors. Outcomes of these analyses contribute to addressing controversies in the field and indicate that GnRH agonism may be a predictable instigator of risk for NDD with opportunities for risk mitigation in combination with another ATT.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Medical Scientist Training ProgramUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Georgina Torrandell‐Haro
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Edward P. Gelmann
- Department of Medicine, Division of Hematology and OncologyUniversity of Arizona College of Medicine and University of Arizona Cancer CenterTucsonArizonaUSA
| | - Francesca Vitali
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of NeurologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Center for Biomedical Informatics and BiostatisticsUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen E. Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Department of NeurologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| |
Collapse
|
4
|
Tan S, Porter T, Bucks RS, Weinborn M, Milicic L, Brown A, Rainey-Smith SR, Taddei K, Ames D, Masters CL, Maruff P, Savage G, Rowe CC, Villemagne VL, Brown B, Sohrabi HR, Laws SM, Martins RN. Androgen receptor CAG repeat length as a moderator of the relationship between free testosterone levels and cognition. Horm Behav 2021; 131:104966. [PMID: 33714752 DOI: 10.1016/j.yhbeh.2021.104966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 11/23/2022]
Abstract
Age-related decrease in testosterone levels is a potential risk factor for cognitive decline in older men. However, observational studies and clinical trials have reported inconsistent results on the effects of testosterone on individual cognitive domains. Null findings may be attributed to factors that studies have yet to consider. In particular, individual variations in polyglutamine (CAG) length in the androgen receptor (AR) gene could alter androgenic activity in brain regions associated with cognitive processes including memory and executive functions. However, the role of AR CAG repeat length as a moderator of the relationship between testosterone levels and cognition has not been investigated. Therefore, we aimed to examine the relationship between baseline calculated free testosterone (cFT) levels, change in cFT levels over 18 months and CAG repeat length on cognitive performance in memory, executive function, language, attention and processing speed domains. These relationships were examined in 304 cognitively normal older male participants of the Australian Imaging, Biomarkers and Lifestyle (AIBL) Study of Ageing. In the attention and processing speed domain, a short CAG repeat length appears to exacerbate the effects of low baseline cFT levels that are also lower than expected at follow-up. These results highlight that individual variations in AR CAG repeat length should be considered in future studies and clinical trials that examine the complex relationship between testosterone and cognition.
Collapse
Affiliation(s)
- Sherilyn Tan
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Nedlands, Western Australia, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Romola S Bucks
- School of Psychological Science, University of Western Australia, Nedlands, Western Australia, Australia
| | - Michael Weinborn
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Nedlands, Western Australia, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Ailsa Brown
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Nedlands, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Kevin Taddei
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - David Ames
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, Victoria, Australia; National Ageing Research Institute (NARI), Parkville, Victoria, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Cooperative Research Centre for Mental Health, Carlton, Victoria, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; CogState Ltd, Melbourne, Victoria, Australia
| | - Greg Savage
- Department of Biomedical Sciences, Faculty of Medicine and Health Science, Macquarie University, Sydney, New South Wales, Australia
| | - Christopher C Rowe
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Nuclear Medicine & Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Victor L Villemagne
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Nuclear Medicine & Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Belinda Brown
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Healthy Ageing, College of Science, Health, Engineering and Education (SHEE), Murdoch University, Murdoch, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Healthy Ageing, College of Science, Health, Engineering and Education (SHEE), Murdoch University, Murdoch, Western Australia, Australia; Department of Biomedical Sciences, Faculty of Medicine and Health Science, Macquarie University, Sydney, New South Wales, Australia.
| | - Simon M Laws
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia.
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Cooperative Research Centre for Mental Health, Carlton, Victoria, Australia; Department of Biomedical Sciences, Faculty of Medicine and Health Science, Macquarie University, Sydney, New South Wales, Australia.
| | | |
Collapse
|
5
|
Tozzi A, Bellingacci L, Pettorossi VE. Rapid Estrogenic and Androgenic Neurosteroids Effects in the Induction of Long-Term Synaptic Changes: Implication for Early Memory Formation. Front Neurosci 2020; 14:572511. [PMID: 33192257 PMCID: PMC7653679 DOI: 10.3389/fnins.2020.572511] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/21/2020] [Indexed: 11/17/2022] Open
Abstract
Mounting experimental evidence demonstrate that sex neuroactive steroids (neurosteroids) are essential for memory formation. Neurosteroids have a profound impact on the function and structure of neural circuits and their local synthesis is necessary for the induction of both long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission and for neural spine formation in different areas of the central nervous system (CNS). Several studies demonstrated that in the hippocampus, 17β-estradiol (E2) is necessary for inducing LTP, while 5α-dihydrotestosterone (DHT) is necessary for inducing LTD. This contribution has been proven by administering sex neurosteroids in rodent models and by using blocking agents of their synthesis or of their specific receptors. The general opposite role of sex neurosteroids in synaptic plasticity appears to be dependent on their different local availability in response to low or high frequency of synaptic stimulation, allowing the induction of bidirectional synaptic plasticity. The relevant contribution of these neurosteroids to synaptic plasticity has also been described in other brain regions involved in memory processes such as motor learning, as in the case of the vestibular nuclei, the cerebellum, and the basal ganglia, or as the emotional circuit of the amygdala. The rapid effects of sex neurosteroids on neural synaptic plasticity need the maintenance of a tonic or phasic local steroid synthesis determined by neural activity but might also be influenced by circulating hormones, age, and gender. To disclose the exact mechanisms how sex neurosteroids participate in finely tuning long-term synaptic changes and spine remodeling, further investigation is required.
Collapse
Affiliation(s)
- Alessandro Tozzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Laura Bellingacci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
6
|
Conner MR, Adeyemi OM, Anderson BJ, Kritzer MF. Domain-specific contributions of biological sex and sex hormones to what, where and when components of episodic-like memory in adult rats. Eur J Neurosci 2020; 52:2705-2723. [PMID: 31943448 DOI: 10.1111/ejn.14676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/21/2022]
Abstract
Episodic memory involves the integration and recall of discrete events that include information about what happened, where it happened and when it occurred. Episodic memory function is critical to daily life, and its dysfunction is both a first identifiable indicator and an enduring core feature of cognitive decline in ageing and in neuropsychiatric disorders including Alzheimer's disease and schizophrenia. Available evidence from human studies suggests that biological sex and sex hormones modulate episodic memory function in health and disease. However, knowledge of how this occurs is constrained by the limited availability and underutilization of validated animal models in investigating hormone impacts on episodic-like memory function. Here, adult female, adult male and gonadally manipulated adult male rats were tested on the what-where-when episodic-like memory task to determine whether rats model human sex differences in episodic memory and how the hormonal milieu impacts episodic-like memory processes in this species. These studies revealed salient ways in which rats model human sex differences in episodic memory, including a male advantage in spatial episodic memory performance. They also identified domain-specific roles for oestrogens and androgens in modulating what, where and when discriminations in male rats that were unlike those engaged in corresponding novel object recognition and novel object location tasks. These studies thus identify rats and the what-where-when task as suitable for investigating the neuroendocrine bases of episodic-like memory, and provide new information about the unique contributions that sex and sex hormones make to this complex mnemonic process.
Collapse
Affiliation(s)
- Meagan R Conner
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, USA.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | | | - Brenda J Anderson
- Department of Psychology, Stony Brook University, Stony Brook, NY, USA
| | - Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
7
|
Ahmad MH, Fatima M, Mondal AC. Role of Hypothalamic-Pituitary-Adrenal Axis, Hypothalamic-Pituitary-Gonadal Axis and Insulin Signaling in the Pathophysiology of Alzheimer's Disease. Neuropsychobiology 2019; 77:197-205. [PMID: 30605907 DOI: 10.1159/000495521] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/13/2018] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD), the commonest progressive neurodegenerative disorder of the brain, is clinically characterized by the formation of extracellular amyloid plaques and intracellular neurofibrillary tangles. Recent studies suggest a relationship between the endocrinal dysregulation and the neuronal loss during the AD pathology. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and hypothalamic-pituitary-gonadal (HPG) axis regulating circulating levels of glucocorticoid hormones has been implicated in the pathophysiology of AD. Likewise, dysregulated insulin signaling, impaired glucose uptake and insulin resistance are some of the prime factors in the onset/progression of AD. In this review, we have discussed the changes in HPA and HPG axes, implicated insulin resistance/signaling and glucose regulation during the onset/progression of AD. Therefore, simultaneous detection of these endocrinal markers in the early or presymptomatic stages may help in the early diagnosis of AD. This evidence for implicated endocrinal functions supports the fact that modulation of endocrinal pathways can be used as therapeutic targets for AD. Future studies need to determine how the induction or inhibition of endocrinal targets could be used for predictable neuroprotection in AD therapies.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mahino Fatima
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India,
| |
Collapse
|
8
|
Carr JS, Bonham LW, Morgans AK, Ryan CJ, Yokoyama JS, Geier EG. Genetic Variation in the Androgen Receptor and Measures of Plasma Testosterone Levels Suggest Androgen Dysfunction in Alzheimer's Disease. Front Neurosci 2018; 12:529. [PMID: 30131669 PMCID: PMC6090298 DOI: 10.3389/fnins.2018.00529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/13/2018] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease (AD) prevalence varies by sex, suggesting that sex chromosomes, sex hormones and/or their signaling could potentially modulate AD risk and progression. Low testosterone levels are reported in men with AD. Further, variation in the androgen receptor (AR) gene has been associated with AD risk and cognitive impairment. We assessed measures of plasma testosterone levels as a biomarker of AD in male participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. Baseline testosterone levels were significantly different between clinical diagnosis groups [cognitively normal controls, mild cognitive impairment (MCI), or AD], with the lowest testosterone levels in men with AD. Lower baseline testosterone levels were associated with higher baseline clinical severity. Change in testosterone levels between baseline and 1-year follow-up varied by diagnosis; MCI had the greatest decreases in testosterone levels between baseline and 1-year follow-up. Despite differences by clinical diagnosis, there was no association between plasma testosterone and CSF biomarkers of AD pathology. We also tested single nucleotide polymorphisms (SNPs) in AR for association with AD risk in a separate cohort from ADNI and found 26 SNPs associated with risk for AD. The top associated SNP is predicted to be an expression quantitative trait locus for AR in multiple tissues, including brain, with the AD-associated risk allele predicted to confer lower AR expression. Our findings suggest a link between the androgen pathway and AD through Aβ/tau independent pathways. These effects may be most pronounced during conversion from MCI to dementia.
Collapse
Affiliation(s)
- Jessie S Carr
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Luke W Bonham
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States.,School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Alicia K Morgans
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Charles J Ryan
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Ethan G Geier
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | | |
Collapse
|
9
|
Jia JX, Yan XS, Cai ZP, Song W, Huo DS, Zhang BF, Wang H, Yang ZJ. The effects of phenylethanoid glycosides, derived from Herba cistanche, on cognitive deficits and antioxidant activities in male SAMP8 mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2017; 80:1180-1186. [PMID: 28880744 DOI: 10.1080/15287394.2017.1367097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cognitive deficits are closely associated with hippocampal synaptic changes. Phenylethanoid glycosides (PhG), derived from Herba cistanche, are known to exert protective effects on cognitive deficits in Alzheimer's disease (AD); however, the underlying mechanisms of this herbal extract on cognitive performance remain unclear. The aim of this study was thus to examine the protective mechanism attributed to PhG on cognitive deficits in an AD senescence accelerated mouse prone 8 (SAMP8) model. Cognitive deficit parameters examined included (1) Morris water maze (MWM) assessing cognitive performance and (2) quantification of dendritic spine density in hippocampal CA1 region by Golgi staining, a molecular biomarker of synaptic function. In addition, levels of malondialdehyde (MDA) and activities of superoxide dismutase (SOD) and gluthathione peroxidase (GSH-Px) were determined to examine the potential role of oxidant processes in cognitive dysfunction. Data showed that PhG significantly decreased escape latency and path length, associated with a rise in the percentage of time spent in the target quadrant and number of platform crossings. In addition, PhG significantly increased dendritic spine density in the hippocampal CA1 region accompanied by elevated expression levels of synaptophysin (SYN) and post synaptic density 95 (PSD-95), reduced MDA content, and elevated the activities of SOD and GSH-Px. Data suggest that the ability of PhG to ameliorate cognitive deficits in SAMP8 mice may be related to promotion in synaptic plasticity involving antioxidant processes.
Collapse
Affiliation(s)
- Jian-Xin Jia
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - Xu-Sheng Yan
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - Zhi-Ping Cai
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - Wei Song
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - Dong-Sheng Huo
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - Bai-Feng Zhang
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| | - He Wang
- b School of Health Sciences , University of Newcastle , Newcastle , Australia
| | - Zhan-Jun Yang
- a Department of Human Anatomy , Baotou Medical College , Inner Mongolia , China
| |
Collapse
|
10
|
Presence of Androgen Receptor Variant in Neuronal Lipid Rafts. eNeuro 2017; 4:eN-NWR-0109-17. [PMID: 28856243 PMCID: PMC5575139 DOI: 10.1523/eneuro.0109-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 11/21/2022] Open
Abstract
Fast, nongenomic androgen actions have been described in various cell types, including neurons. However, the receptor mediating this cell membrane–initiated rapid signaling remains unknown. This study found a putative androgen receptor splice variant in a dopaminergic N27 cell line and in several brain regions (substantia nigra pars compacta, entorhinal cortex, and hippocampus) from gonadally intact and gonadectomized (young and middle-aged) male rats. This putative splice variant protein has a molecular weight of 45 kDa and lacks an N-terminal domain, indicating it is homologous to the human AR45 splice variant. Interestingly, AR45 was highly expressed in all brain regions examined. In dopaminergic neurons, AR45 is localized to plasma membrane lipid rafts, a microdomain involved in cellular signaling. Further, AR45 protein interacts with membrane-associated G proteins Gαq and Gαo. Neither age nor hormone levels altered AR45 expression in dopaminergic neurons. These results provide the first evidence of AR45 protein expression in the brain, specifically plasma membrane lipid rafts. AR45 presence in lipid rafts indicates that it may function as a membrane androgen receptor to mediate fast, nongenomic androgen actions.
Collapse
|
11
|
Xu P, Xu SP, Wang KZ, Lu C, Zhang HX, Pan RL, Qi C, Yang YY, Li YH, Liu XM. Cognitive-enhancing effects of hydrolysate of polygalasaponin in SAMP8 mice. J Zhejiang Univ Sci B 2017; 17:503-14. [PMID: 27381727 DOI: 10.1631/jzus.b1500321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The aim of the study is to evaluate the cognitive-enhancing effects of hydrolysate of polygalasaponin (HPS) on senescence accelerate mouse P8 (SAMP8) mice, an effective Alzheimer's disease (AD) model, and to research the relevant mechanisms. METHODS The cognitive-enhancing effects of HPS on SAMP8 mice were assessed using Morris water maze (MWM) and step-through passive avoidance tests. Then N-methyl-D-aspartate (NMDA) receptor subunit expression for both the cortex and hippocampus of mice was observed using Western blotting. RESULTS HPS (25 and 50 mg/kg) improved the escape rate and decreased the escape latency and time spent in the target quadrant for the SAMP8 mice in the MWM after oral administration of HPS for 10 d. Moreover, it decreased error times in the passive avoidance tests. Western blotting showed that HPS was able to reverse the levels of NMDAR1 and NMDAR2B expression in the cortex or hippocampus of model mice. CONCLUSIONS The present study suggested that HPS can improve cognitive deficits in SAMP8 mice, and this mechanism might be associated with NMDA receptor (NMDAR)-related pathways.
Collapse
Affiliation(s)
- Pan Xu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Shu-Ping Xu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Ke-Zhu Wang
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Cong Lu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Hong-Xia Zhang
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Rui-le Pan
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Chang Qi
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yan-Yan Yang
- Science and Technology on Human Factors Engineering Laboratory, Astronaut Centre of China, Beijing 100193, China
| | - Ying-Hui Li
- Science and Technology on Human Factors Engineering Laboratory, Astronaut Centre of China, Beijing 100193, China
| | - Xin-Min Liu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
12
|
Nikmahzar E, Jahanshahi M, Ghaemi A, Naseri GR, Moharreri AR, Lotfinia AA. Hippocampal serotonin-2A receptor-immunoreactive neurons density increases after testosterone therapy in the gonadectomized male mice. Anat Cell Biol 2016; 49:259-272. [PMID: 28127501 PMCID: PMC5266105 DOI: 10.5115/acb.2016.49.4.259] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/06/2016] [Accepted: 12/09/2016] [Indexed: 12/15/2022] Open
Abstract
The change of steroid levels may also exert different modulatory effects on the number and class of serotonin receptors present in the plasma membrane. The effects of chronic treatment of testosterone for anxiety were examined and expression of 5-HT2A serotonergic receptor, neuron, astrocyte, and dark neuron density in the hippocampus of gonadectomized male mice was determined. Thirty-six adult male NMRI mice were randomly divided into six groups: intact-no testosterone treatment (No T), gonadectomy (GDX)-No T, GDX-Vehicle, GDX-6.25 mg/kg testosterone (T), GDX-12.5 mg/kg T, and GDX-25 mg/kg T. Anxiety-related behavior was evaluated using elevated plus maze apparatus. The animals were anesthetized after 48 hours after behavioral testing, and decapitated and micron slices were prepared for immunohistochemical as well as histopathological assessment. Subcutaneous injection of testosterone (25 mg/kg) may induce anxiogenic-like behavior in male mice. In addition, immunohistochemical data reveal reduced expression of 5-HT2A serotonergic receptor after gonadectomy in all areas of the hippocampus. However, treatment with testosterone could increase the mean number of dark neurons as well as immunoreactive neurons in CA1 and CA3 area, dose dependently. The density of 5-HT2A receptor-immunoreactive neurons may play a crucial role in the induction of anxiety like behavior. As reduction in such receptor expression have shown to significantly enhance anxiety behaviors. However, replacement of testosterone dose dependently enhances the number of 5-HT2A receptor-immunoreactive neurons and interestingly also reduced anxiety like behaviors.
Collapse
Affiliation(s)
- Emsehgol Nikmahzar
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Department of Anatomy, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amir Ghaemi
- Shefa Neuroscience Research Center, Tehran, Iran
| | - Gholam Reza Naseri
- Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Reza Moharreri
- Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | |
Collapse
|
13
|
Xu P, Wang K, Lu C, Dong L, Gao L, Yan M, Aibai S, Liu X. Protective effect of lavender oil on scopolamine induced cognitive deficits in mice and H 2O 2 induced cytotoxicity in PC12 cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 193:408-415. [PMID: 27558947 DOI: 10.1016/j.jep.2016.08.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/10/2016] [Accepted: 08/20/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lavender essential oil (LO), an aromatic liquid extracted from Lavandula angustifolia Mill., has been traditionally used in the treatments of many nervous system diseases, and recently LO also reported to be effective for the Alzheimer's disease (AD). AIM OF THE STUDY The improvement effect of lavender oil (LO) on the scopolamine-induced cognitive deficits in mice and H2O2 induced cytotoxicity in PC12 cells have been evaluated. The relevant mechanism was also researched from the perspective of antioxidant effect and cholinergic system modulation. MATERIALS AND METHODS Cognitive deficits were induced in C57BL/6J mice treated with scopolamine (1mg/kg, i.p.) and were assessed by Morris water maze (MWM) and step-through passive avoidance tests. Then their hippocampus were removed for biochemical assays (acetylcholinesterase (AChE), superoxide dismutase (SOD), glutathione peroxidase (GPX) and malondialdehyde (MDA)). In vitro, the cytotoxicity were induced by 4h exposure to H2O2 in PC12 and evaluated by cell viability (MTT), lactate dehydrogenase (LDH) level, nitric oxide (NO) release, reactive oxygen species (ROS) production and mitochondrial membrane potential (MMP). RESULTS The results demonstrated that LO (100mg/kg) could improve the cognitive performance of scopolamine induced mice in behavioral tests. Meanwhile, it significantly decreased the AChE activity, MDA level, and increase SOD and GPX activities of the model. Moreover, LO (12μg/mL) protected PC12 cells from H2O2 induced cytotoxicity by reducing LDH, NO release, intracellular ROS accumulation and MMP loss. CONCLUSIONS It was suggested that LO could show neuroprotective effect in AD model in vivo (scopolamine-treated mice) and in vitro (H2O2 induced PC12 cells) via modulating oxidative stress and AChE activity.
Collapse
Affiliation(s)
- Pan Xu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Kezhu Wang
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Cong Lu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Liming Dong
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Li Gao
- Department of Pharmacology and Toxicology Laboratory, Xinjiang Institute of Traditional Uighur Medicine, Urumqi, Xinjiang 830049, China
| | - Ming Yan
- Department of Pharmacology and Toxicology Laboratory, Xinjiang Institute of Traditional Uighur Medicine, Urumqi, Xinjiang 830049, China
| | - Silafu Aibai
- Department of Pharmacology and Toxicology Laboratory, Xinjiang Institute of Traditional Uighur Medicine, Urumqi, Xinjiang 830049, China
| | - Xinmin Liu
- Research Center of Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
14
|
Fedotova J, Soultanov V, Nikitina T, Roschin V, Ordyan N, Hritcu L. Ropren® treatment reverses anxiety-like behavior and monoamines levels in gonadectomized rat model of Alzheimer’s disease. Biomed Pharmacother 2016; 83:1444-1455. [DOI: 10.1016/j.biopha.2016.08.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/19/2016] [Accepted: 08/28/2016] [Indexed: 12/12/2022] Open
|
15
|
Holmes S, Singh M, Su C, Cunningham RL. Effects of Oxidative Stress and Testosterone on Pro-Inflammatory Signaling in a Female Rat Dopaminergic Neuronal Cell Line. Endocrinology 2016; 157:2824-35. [PMID: 27167771 PMCID: PMC4929547 DOI: 10.1210/en.2015-1738] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Parkinson's disease, a progressive neurodegenerative disorder, is associated with oxidative stress and neuroinflammation. These pathological markers can contribute to the loss of dopamine neurons in the midbrain. Interestingly, men have a 2-fold increased incidence for Parkinson's disease than women. Although the mechanisms underlying this sex difference remain elusive, we propose that the primary male sex hormone, testosterone, is involved. Our previous studies show that testosterone, through a putative membrane androgen receptor, can increase oxidative stress-induced neurotoxicity in dopamine neurons. Based on these results, this study examines the role of nuclear factor κ B (NF-κB), cyclooxygenase-2 (COX2), and apoptosis in the deleterious effects of androgens in an oxidative stress environment. We hypothesize, under oxidative stress environment, testosterone via a putative membrane androgen receptor will exacerbate oxidative stress-induced NF-κB/COX2 signaling in N27 dopaminergic neurons, leading to apoptosis. Our data show that testosterone increased the expression of COX2 and apoptosis in dopamine neurons. Inhibiting the NF-κB and COX2 pathway with CAPE and ibuprofen, respectively, blocked testosterone's negative effects on cell viability, indicating that NF-κB/COX2 cascade plays a role in the negative interaction between testosterone and oxidative stress on neuroinflammation. These data further support the role of testosterone mediating the loss of dopamine neurons under oxidative stress conditions, which may be a key mechanism contributing to the increased incidence of Parkinson's disease in men compared with women.
Collapse
Affiliation(s)
- Shaletha Holmes
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Chang Su
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Rebecca L Cunningham
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, Texas 76107
| |
Collapse
|
16
|
Pan W, Han S, Kang L, Li S, Du J, Cui H. Effects of dihydrotestosterone on synaptic plasticity of the hippocampus in mild cognitive impairment male SAMP8 mice. Exp Ther Med 2016; 12:1455-1463. [PMID: 27588067 PMCID: PMC4997989 DOI: 10.3892/etm.2016.3470] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/21/2016] [Indexed: 01/03/2023] Open
Abstract
The current study focused on how dihydrotestosterone (DHT) regulates synaptic plasticity in the hippocampus of mild cognitive impairment male senescence-accelerated mouse prone 8 (SAMP8) mice. Five-month-old SAMP8 mice were divided into the control, castrated and castrated-DHT groups, in which the mice were castrated and treated with physiological doses of DHT for a period of 2 months. To determine the regulatory mechanisms of DHT in the cognitive capacity, the effects of DHT on the morphology of the synapse and the expression of synaptic marker proteins in the hippocampus were investigated using immunohistochemistry, qPCR and western blot analysis. The results showed that the expression of cAMP-response element binding protein (CREB), postsynaptic density protein 95 (PSD95), synaptophysin (SYN) and developmentally regulated brain protein (Drebrin) was reduced in the castrated group compared to the control group. However, DHT promoted the expression of CREB, PSD95, SYN and Drebrin in the hippocampus of the castrated-DHT group. Thus, androgen depletion impaired the synaptic plasticity in the hippocampus of SAMP8 and accelerated the development of Alzheimer's disease (AD)-like neuropathology, suggesting that a similar mechanism may underlie the increased risk for AD in men with low testosterone. In addition, DHT regulated synaptic plasticity in the hippocampus of mild cognitive impairment (MCI) SAMP8 mice and delayed the progression of disease to Alzheimer's dementia. In conclusion, androgen-based hormone therapy is a potentially useful strategy for preventing the progression of MCI in aging men. Androgens enhance synaptic markers (SYN, PSD95, and Drebrin), activate CREB, modulate the fundamental biology of synaptic structure, and lead to the structural changes of plasticity in the hippocampus, all of which result in improved cognitive function.
Collapse
Affiliation(s)
- Wensen Pan
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; Department of Respiration Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shuo Han
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lin Kang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Sha Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Juan Du
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|
17
|
Cognitive-enhancing activities of the polyprenol preparation Ropren® in gonadectomized β-amyloid (25–35) rat model of Alzheimer's disease. Physiol Behav 2016; 157:55-62. [DOI: 10.1016/j.physbeh.2016.01.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/22/2016] [Accepted: 01/24/2016] [Indexed: 11/23/2022]
|
18
|
Soultanov V, Fedotova J, Nikitina T, Roschin V, Ordyan N, Hritcu L. Antidepressant-Like Effect of Ropren® in β-Amyloid-(25–35) Rat Model of Alzheimer’s Disease with Altered Levels of Androgens. Mol Neurobiol 2016; 54:2611-2621. [DOI: 10.1007/s12035-016-9848-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/09/2016] [Indexed: 10/22/2022]
|
19
|
Huo DS, Sun JF, Zhang B, Yan XS, Wang H, Jia JX, Yang ZJ. Protective effects of testosterone on cognitive dysfunction in Alzheimer's disease model rats induced by oligomeric beta amyloid peptide 1-42. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2016; 79:856-863. [PMID: 27599231 DOI: 10.1080/15287394.2016.1193114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cognitive dysfunction is known to be influenced by circulating sex steroidal hormones. The aim of this study was to examine the protective effect and possible protective mechanism of testosterone (T) on cognitive performance in male rats induced by intrahippocampal injections of beta amyloid 1-42 oligomers (Aβ1-42). Treatment with T as evidenced by the Morris water maze (MWM) test significantly shortened escape latency and reduced path length to reach the platform compared to the control (C). During probe trials, the T group displayed a significantly greater percent of time in the target quadrant and improved the number of platform crossings compared with C, flutamide (F), an antiandrogen, and a combined F and T group. Flutamide markedly inhibited the influence of T on cognitive performance. Following Nissl staining, the number of intact pyramidal cells was significantly elevated in the T group, and the effect of T was blocked by F. Immunohistochemisty and Western blot analysis showed that the protein expression level of Aβ 1-42 was markedly decreased and expression levels of synaptophysin (SYN) significantly increased with T, while F inhibited all T-mediated effects. Our data suggest that the influence of T on cognitive performance was mediated via androgen receptors (AR) to remove beta amyloid, which leads to enhanced synaptic plasticity.
Collapse
Affiliation(s)
- Dong-Sheng Huo
- a Department of Human Anatomy, Baotou Medical College, Baotou , Inner Mongolia , China
| | - Jian-Fang Sun
- b The First Affiliated Hospital of Baotou Medical College, Baotou , Inner Mongolia , China
| | - Baifeng Zhang
- a Department of Human Anatomy, Baotou Medical College, Baotou , Inner Mongolia , China
| | - Xu-Sheng Yan
- a Department of Human Anatomy, Baotou Medical College, Baotou , Inner Mongolia , China
| | - He Wang
- c School of Health Sciences , University of Newcastle , Newcastle , Australia
| | - Jian-Xin Jia
- a Department of Human Anatomy, Baotou Medical College, Baotou , Inner Mongolia , China
| | - Zhan-Jun Yang
- a Department of Human Anatomy, Baotou Medical College, Baotou , Inner Mongolia , China
| |
Collapse
|
20
|
Jian-xin J, Cheng-li C, Song W, Yan XS, Huo DS, Wang H, Yang ZJ. Effects of Testosterone Treatment on Synaptic Plasticity and Behavior in Senescence Accelerated Mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2015; 78:1311-1320. [PMID: 26529502 DOI: 10.1080/15287394.2015.1085839] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Learning and memory are known to be influenced by circulating sex steroidal hormones and these behavioral processes are diminished in aging. Thus, the aim of this study was to examine the mechanism underlying testosterone-induced effects on cognitive performance in the senescence accelerated mouse P8 (SAMP8) model. Treatment with testosterone (T) as evidenced by the Morris water maze test produced a significantly shorter escape latency and reduced path length to reach the platform compared to the control (C). No significant differences were noted in mean swim speed among all groups. During the probe trials, the T group spent a significantly greater percent of time in the target quadrant and improved the number of platform crossings. Flutamide (F), an antiandrogen, significantly inhibited the effects of T on behavioral and memory performances indicators. Following Nissl staining, the number of intact pyramidal cells was markedly elevated in the treated mice, and this effect was blocked by F. Immunohistochemistry and Western blot analysis showed that the expression levels of NMDAR1, SYN, and p-CREC/CREB protein levels were significantly increased in the T group, while F inhibited the T-mediated effects. Western blot analysis showed that there were no significant differences in the expression levels of SYN, p-CREC/CREB, and NMDAR1 between C, F, and F + T groups. Reverse-transcription polymerase chain reaction (RT-PCR) analysis showed that the mRNA expression levels of NMDAR1 and SYN were significantly increased in T-administered mice, while F inhibited the T-mediated effects. Data suggest that the T-mediated increase in SYN expression levels resulted in improvement in behavioral performances and learning, which may involve stimulation of central nervous system androgen receptors (AR).
Collapse
Affiliation(s)
- Jia Jian-xin
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| | - Cui Cheng-li
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| | - Wei Song
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| | - Xu-sheng Yan
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| | - Dong-sheng Huo
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| | - He Wang
- b School of Health Sciences , University of Newcastle , Newcastle , Australia
| | - Zhan-jun Yang
- a Department of Human Anatomy , Baotou Medical College , Inner , Mongolia , China
| |
Collapse
|
21
|
Cherrier MM, Anderson K, Shofer J, Millard S, Matsumoto AM. Testosterone treatment of men with mild cognitive impairment and low testosterone levels. Am J Alzheimers Dis Other Demen 2015; 30:421-30. [PMID: 25392187 PMCID: PMC10852633 DOI: 10.1177/1533317514556874] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
INTRODUCTION This study investigated the effects of testosterone (T) treatment on cognition, mood, and quality of life in men with mild cognitive impairment (MCI) and low serum T levels. METHODS A total of 351 community-dwelling men were screened, and 37 men evidenced both MCI and low T of whom 27 agreed for further screening. Twenty-two met all the study inclusion/exclusion criteria and enrolled in a 6-month randomized, double-blind, placebo-controlled study. RESULTS Total T levels significantly increased in the T treatment group. No significant changes were observed in measures of cognition, mood, or quality of life other than improvement in 1 objective measure of verbal memory (P < .05) and decreased depression symptoms (P < .02) in the treatment group. CONCLUSIONS Testosterone treatment may modestly improve verbal memory and depression symptoms in men with both MCI and low T.
Collapse
Affiliation(s)
- M M Cherrier
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - K Anderson
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - J Shofer
- Geriatric, Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - S Millard
- Geriatric, Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - A M Matsumoto
- Geriatric, Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
22
|
|
23
|
Blair JA, McGee H, Bhatta S, Palm R, Casadesus G. Hypothalamic-pituitary-gonadal axis involvement in learning and memory and Alzheimer's disease: more than "just" estrogen. Front Endocrinol (Lausanne) 2015; 6:45. [PMID: 25859241 PMCID: PMC4373369 DOI: 10.3389/fendo.2015.00045] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/12/2015] [Indexed: 11/25/2022] Open
Abstract
Accumulating studies affirm the effects of age-related endocrine dysfunction on cognitive decline and increasing risk of neurodegenerative diseases. It is well known that estrogens can be protective for cognitive function, and more recently androgens and luteinizing hormone have also been shown to modulate learning and memory. Understanding the mechanisms underlying hypothalamic-pituitary-gonadal axis-associated cognitive dysfunction is crucial for therapeutic advancement. Here, we emphasize that reproductive hormones are influential in maintaining neuronal health and enhancing signaling cascades that lead to cognitive impairment. We summarize and critically evaluate age-related changes in the endocrine system, their implications in the development of Alzheimer's disease, and the therapeutic potential of endocrine modulation in the prevention of age-related cognitive decline.
Collapse
Affiliation(s)
- Jeffrey A. Blair
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Henry McGee
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Sabina Bhatta
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Russell Palm
- University of Toledo School of Medicine, Toledo, OH, USA
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- *Correspondence: Gemma Casadesus, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH 44242, USA e-mail:
| |
Collapse
|
24
|
Kelly B, Maguire-Herring V, Rose CM, Gore HE, Ferrigno S, Novak MA, Lacreuse A. Short-term testosterone manipulations do not affect cognition or motor function but differentially modulate emotions in young and older male rhesus monkeys. Horm Behav 2014; 66:731-42. [PMID: 25308086 PMCID: PMC4262694 DOI: 10.1016/j.yhbeh.2014.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 07/15/2014] [Accepted: 08/12/2014] [Indexed: 12/28/2022]
Abstract
Human aging is characterized by declines in cognition and fine motor function as well as improved emotional regulation. In men, declining levels of testosterone (T) with age have been implicated in the development of these age-related changes. However, studies examining the effects of T replacement on cognition, emotion and fine motor function in older men have not provided consistent results. Rhesus monkeys (Macaca mulatta) are excellent models for human cognitive aging and may provide novel insights on this issue. We tested 10 aged intact male rhesus monkeys (mean age=19, range 15-25) on a battery of cognitive, motor and emotional tasks at baseline and under low or high T experimental conditions. Their performance was compared to that of 6 young males previously tested in the same paradigm (Lacreuse et al., 2009; Lacreuse et al., 2010). Following a 4-week baseline testing period, monkeys were treated with a gonadotropin releasing hormone agonist (Depot Lupron, 200 μg/kg) to suppress endogenous T and were tested on the task battery under a 4-week high T condition (injection of Lupron+T enanthate, 20 mg/kg, n=8) or 4-week low T condition (injection of Lupron+oil vehicle, n=8) before crossing over to the opposite treatment. The cognitive tasks consisted of the Delayed Non-Matching-to-Sample (DNMS), the Delayed Response (DR), and the Delayed Recognition Span Test (spatial-DRST). The emotional tasks included an object Approach-Avoidance task and a task in which monkeys were played videos of unfamiliar conspecifics in different emotional context (Social Playbacks). The fine motor task was the Lifesaver task that required monkeys to remove a Lifesaver candy from rods of different complexity. T manipulations did not significantly affect visual recognition memory, working memory, reference memory or fine motor function at any age. In the Approach-Avoidance task, older monkeys, but not younger monkeys, spent more time in proximity of novel objects in the high T condition relative to the low T condition. In both age groups, high T increased watching time of threatening social stimuli in the Social Playbacks. These results suggest that T affects some aspects of emotional processing but has no effect on fine motor function or cognition in young or older male macaques. It is possible that the duration of T treatment was not long enough to affect cognition or fine motor function or that T levels were too high to improve these outcomes. An alternative explanation for the discrepancies of our findings with some of the cognitive and emotional effects of T reported in rodents and humans may be the use of a chemical castration, which reduced circulating gonadotropins in addition to T. Further studies will investigate whether the luteinizing hormone LH mediates the effects of T on brain function in male primates.
Collapse
Affiliation(s)
- Brian Kelly
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA; Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA; Behavioral Sciences, Fitchburg State University, Fitchburg MA 01420, USA
| | | | - Christian M Rose
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Heather E Gore
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Stephen Ferrigno
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Melinda A Novak
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA; Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
25
|
Protective effects of testosterone on presynaptic terminals against oligomeric β-amyloid peptide in primary culture of hippocampal neurons. BIOMED RESEARCH INTERNATIONAL 2014; 2014:103906. [PMID: 25045655 PMCID: PMC4086619 DOI: 10.1155/2014/103906] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022]
Abstract
Increasing lines of evidence support that testosterone may have neuroprotective effects. While observational studies reported an association between higher bioavailable testosterone or brain testosterone levels and reduced risk of Alzheimer's disease (AD), there is limited understanding of the underlying neuroprotective mechanisms. Previous studies demonstrated that testosterone could alleviate neurotoxicity induced by β-amyloid (Aβ), but these findings mainly focused on neuronal apoptosis. Since synaptic dysfunction and degeneration are early events during the pathogenesis of AD, we aim to investigate the effects of testosterone on oligomeric Aβ-induced synaptic changes. Our data suggested that exposure of primary cultured hippocampal neurons to oligomeric Aβ could reduce the length of neurites and decrease the expression of presynaptic proteins including synaptophysin, synaptotagmin, and synapsin-1. Aβ also disrupted synaptic vesicle recycling and protein folding machinery. Testosterone preserved the integrity of neurites and the expression of presynaptic proteins. It also attenuated Aβ-induced impairment of synaptic exocytosis. By using letrozole as an aromatase antagonist, we further demonstrated that the effects of testosterone on exocytosis were unlikely to be mediated through the estrogen receptor pathway. Furthermore, we showed that testosterone could attenuate Aβ-induced reduction of HSP70, which suggests a novel mechanism that links testosterone and its protective function on Aβ-induced synaptic damage. Taken together, our data provide further evidence on the beneficial effects of testosterone, which may be useful for future drug development for AD.
Collapse
|
26
|
Testosterone ameliorates streptozotocin-induced memory impairment in male rats. Acta Pharmacol Sin 2014; 35:752-7. [PMID: 24902786 DOI: 10.1038/aps.2014.6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 01/21/2013] [Indexed: 11/09/2022] Open
Abstract
AIM To study the effects of testosterone on streptozotocin (STZ)-induced memory impairment in male rats. METHODS Adult male Wistar rats were intracerebroventricularly (icv) infused with STZ (750 μg) on d 1 and d 3, and a passive avoidance task was assessed 2 weeks after the first injection of STZ. Castration surgery was performed in another group of rats, and the passive avoidance task was assessed 4 weeks after the operation. Testosterone (1 mg·kg(-1)·d(-1), sc), the androgen receptor antagonist flutamide (10 mg·kg(-1)·d(-1), ip), the estrogen receptor antagonist tamoxifen (1 mg·kg(-1)·d(-1), ip) or the aromatase inhibitor letrozole (4 mg·kg(-1)·d(-1), ip) were administered for 6 d after the first injection of STZ. RESULTS STZ administration and castration markedly decreased both STL1 (the short memory) and STL2 (the long memory) in passive avoidance tests. Testosterone replacement almost restored the STL1 and STL2 in castrated rats, and significantly prolonged the STL1 and STL2 in STZ-treated rats. Administration of flutamide, letrozole or tamoxifen significantly impaired the memory in intact rats, and significantly attenuated the testosterone replacement in improving STZ- and castration-induced memory impairment. CONCLUSION Testosterone administration ameliorates STZ- and castration-induced memory impairment in male Wistar rats.
Collapse
|
27
|
Panizzon MS, Hauger R, Xian H, Vuoksimaa E, Spoon KM, Mendoza SP, Jacobson KC, Vasilopoulos T, Rana BK, McKenzie R, McCaffery JM, Lyons MJ, Kremen WS, Franz CE. Interaction of APOE genotype and testosterone on episodic memory in middle-aged men. Neurobiol Aging 2013; 35:1778.e1-8. [PMID: 24444806 DOI: 10.1016/j.neurobiolaging.2013.12.025] [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: 08/21/2013] [Revised: 12/19/2013] [Accepted: 12/21/2013] [Indexed: 01/12/2023]
Abstract
Age-related changes in testosterone are believed to be a key component of the processes that contribute to cognitive aging in men. The APOE-ε4 allele may interact with testosterone and moderate the hormone's association with cognition. The goals of the present study were to examine the degree to which free testosterone is associated with episodic memory in a community-based sample of middle-aged men, and examine the potential interaction between free testosterone and the APOE-ε4 allele. Data were used from 717 participants in the Vietnam Era Twin Study of Aging. Average age was 55.4 years (standard deviation = 2.5). Significant positive associations were observed between free testosterone level and verbal episodic memory, as well as a significant interaction between free testosterone and APOE-ε4 status. In ε4 carriers free testosterone was positively associated with verbal episodic memory performance (story recall), whereas no association was observed in ε4 noncarriers. Results support the hypothesis that APOE-ε4 status increases susceptibility to other risk factors, such as low testosterone, which may ultimately contribute to cognitive decline or dementia.
Collapse
Affiliation(s)
- Matthew S Panizzon
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA.
| | - Richard Hauger
- Department of Psychiatry, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Hong Xian
- Department of Biostatistics, St. Louis University, College for Public Health & Social Justice, St. Louis, MO, USA; Research Service, St. Louis Veterans Affairs Medical Center, St. Louis, MO
| | - Eero Vuoksimaa
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Kelly M Spoon
- Computational Science Research Center, San Diego State University, San Diego, CA, USA
| | - Sally P Mendoza
- Department of Psychology, University of California, Davis, CA, USA
| | | | | | - Brinda K Rana
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| | - Ruth McKenzie
- Department of Psychology, Boston University, Boston, MA, USA
| | - Jeanne M McCaffery
- Department of Psychiatry and Human Behavior, The Miriam Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Michael J Lyons
- Department of Psychology, Boston University, Boston, MA, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA; VA San Diego Healthcare System, CA, USA; Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Carol E Franz
- Department of Psychiatry, University of California, San Diego, CA, USA; Twin Research Laboratory, Center for Behavioral Genomics, University of California, San Diego, CA, USA
| |
Collapse
|
28
|
Holmes S, Abbassi B, Su C, Singh M, Cunningham RL. Oxidative stress defines the neuroprotective or neurotoxic properties of androgens in immortalized female rat dopaminergic neuronal cells. Endocrinology 2013; 154:4281-92. [PMID: 23959938 PMCID: PMC3800758 DOI: 10.1210/en.2013-1242] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Males have a higher risk for developing Parkinson's disease and parkinsonism after ischemic stroke than females. Although estrogens have been shown to play a neuroprotective role in Parkinson's disease, there is little information on androgens' actions on dopamine neurons. In this study, we examined the effects of androgens under conditions of oxidative stress to determine whether androgens play a neuroprotective or neurotoxic role in dopamine neuronal function. Mitochondrial function, cell viability, intracellular calcium levels, and mitochondrial calcium influx were examined in response to androgens under both nonoxidative and oxidative stress conditions. Briefly, N27 dopaminergic cells were exposed to the oxidative stressor, hydrogen peroxide, and physiologically relevant levels of testosterone or dihydrotestosterone, applied either before or after oxidative stress exposure. Androgens, alone, increased mitochondrial function via a calcium-dependent mechanism. Androgen pretreatment protected cells from oxidative stress-induced cell death. However, treatment with androgens after the oxidative insult increased cell death, and these effects were, in part, mediated by calcium influx into the mitochondria. Interestingly, the negative effects of androgens were not blocked by either androgen or estrogen receptor antagonists. Instead, a putative membrane-associated androgen receptor was implicated. Overall, our results indicate that androgens are neuroprotective when oxidative stress levels are minimal, but when oxidative stress levels are elevated, androgens exacerbate oxidative stress damage.
Collapse
Affiliation(s)
- Shaletha Holmes
- PhD, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, Texas 76107-2699.
| | | | | | | | | |
Collapse
|
29
|
Jia J, Kang L, Li S, Geng D, Fan P, Wang L, Cui H. Amelioratory effects of testosterone treatment on cognitive performance deficits induced by soluble Aβ1-42 oligomers injected into the hippocampus. Horm Behav 2013; 64:477-86. [PMID: 23954394 DOI: 10.1016/j.yhbeh.2013.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 01/26/2023]
Abstract
This study was undertaken to investigate the protective effects and potential mechanism of testosterone (T) on cognitive performance in adult male rats given bilateral intrahippocampal injections of beta amyloid 1-42 oligomers (Aβ1-42) combined with gonadectomy (Aβ+GDX). A series of experiments were designed to verify the optimal administration time and dose of T and to explore its potential protective mechanisms on spatial ability in Aβ+GDX rats in the Morris water maze test. Aβ1-42 was injected only once two weeks before testing, while T and the androgen receptor (AR) antagonist flutamide (F) were administered daily beginning 2 days before and throughout the 6 days of testing. The Aβ1-42 injection and GDX individually impaired cognitive performance, and the combination of these treatments was additive, leading to even greater impairment. The serum T level peaked at 48 h after administration. T doses ranging from 0.25 to 1.00 mg corresponding to serum T levels of 4.5-21.35 ng/ml improved the spatial ability. Animals administered 0.75 mg of T corresponding to the serum T level of 15.2 ng/ml had the most significantly improved behavioral performances. However, higher T doses of 1.50 and 2.00 mg resulting in serum T levels of 34.8 and 45 ng/ml, respectively, impaired the behavioral performances. F had no effect on the serum T level and spatial ability, but it blocked the activational effect of T. These findings indicate that the effect of T on behavioral performances is partly mediated through ARs.
Collapse
Affiliation(s)
- Jianxin Jia
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | | | |
Collapse
|
30
|
Ferrari R, Dawoodi S, Raju M, Thumma A, Hynan LS, Maasumi SH, Reisch JS, O'Bryant S, Jenkins M, Barber R, Momeni P. Androgen receptor gene and sex-specific Alzheimer's disease. Neurobiol Aging 2013; 34:2077.e19-20. [PMID: 23545426 PMCID: PMC4012749 DOI: 10.1016/j.neurobiolaging.2013.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 02/22/2013] [Indexed: 01/02/2023]
Abstract
Women are at a 2-fold risk of developing late-onset Alzheimer's disease (AD) (onset at 65 years of age or older) compared with men. During perimenopausal years, women undergo hormonal changes that are accompanied by metabolic, cardiovascular, and inflammatory changes. These all together have been suggested as risk factors for late-onset AD. However, not all perimenopausal women develop AD; we hypothesize that certain genetic factors might underlie the increased susceptibility for developing AD in postmenopausal women. We investigated the Androgen Receptor gene (AR) in a clinical cohort of male and female AD patients and normal control subjects by sequencing all coding exons and evaluating the length and distribution of the CAG repeat in exon 1. We could not establish a correlation between the repeat length, sex, and the disease status, nor did we identify possible pathogenic variants. AR is located on the X chromosome; to assess its role in AD, X-inactivation patterns will need to be studied to directly correlate the actual expressed repeat length to a possible sex-specific phenotypic effect.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Texas Tech University, Health Sciences Center, Department of Internal Medicine, Lubbock, TX 79430, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Age-related hormonal decline is gradual and less recognized in men than in women. Symptoms are oftentimes ignored and non-specific. Fatigue, lack of concentration, mood swings, decreased sexual desire, erectile dysfunction, infertility, hair loss, reduced muscle and bone mass, and weight gain are a few of the symptoms of male hypogonadism. This disorder is linked to reduction in quality of life, and poorer health outcomes as it may increase the risk for cardiovascular disease, diabetes mellitus, metabolic syndrome, Alzheimer's disease and premature death. Different modalities of testosterone replacement therapy have evolved over 70 years, and sales continue to grow. Each preparation is differentiated by route of delivery, ease of use, cost and pharmacokinetics. Topical/transdermal testosterone replacement therapy, including patches and gels, are the most modern formulations on the market. These are more expensive treatments, but yield more physiological concentrations of testosterone. Restoration of testosterone levels to the eugonadal range reverses signs and symptoms of hypogonadism, except for infertility, and may alleviate co-morbidities associated with hypogonadism. Patient understanding of and compliance with both treatment and monitoring are of utmost importance to achieve clinical success with maximum benefit and minimum risk. The aim of our review is to summarize the indications, contraindications, benefits and risks of testosterone replacement therapy as they relate to transdermal administration. Further, we compare the various testosterone preparations, focusing on the newest topical/transdermal routes of administration that are currently available.
Collapse
Affiliation(s)
- Katrina A Abadilla
- Johns Hopkins UniversitySinai Hospital Program in Internal Medicine, Baltimore, MD, USA
| | | |
Collapse
|
32
|
Abstract
Astrogliosis is induced by neuronal damage and is also a pathological feature of the major aging-related neurodegenerative disorders. The mechanisms that control the cascade of astrogliosis have not been well established. In a previous study, we identified a novel androgen receptor (AR)-interacting protein, p44/WDR77, that plays a critical role in the proliferation and differentiation of prostate epithelial cells. In the present study, we found that deletion of the p44/WDR77 gene caused premature death with dramatic astrogliosis in mouse brain. We further found that p44/WDR77 is expressed in astrocytes and that loss of p44/WDR77 expression in astrocytes leads to growth arrest and astrogliosis. The astrocyte activation induced by deletion of the p44/WDR77 gene was associated with upregulation of p21(Cip1) expression and NF-κB activation. Silencing p21(Cip1) or NF-κB p65 expression with short hairpin RNA (shRNA) abolished astrocyte activation and rescued the astrocyte growth inhibition induced by deletion of the p44/WDR77 gene. Our results reveal a novel role for p44/WDR77 in the control of astrocyte activation through p21(Cip1) and NF-κB signaling.
Collapse
|
33
|
Palm R, Ayala-Fontanez N, Garcia Y, Lee HG, Smith MA, Casadesus G. Neuroendocrinology-based therapy for Alzheimer's disease. Biofactors 2012; 38:123-32. [PMID: 22438197 DOI: 10.1002/biof.1011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 02/03/2012] [Indexed: 01/11/2023]
Abstract
The nervous system interacts directly with the endocrine system to control a plethora of central nervous system (CNS) functions. Metabolic and reproductive hormones are known to be important in the maintenance of neuronal health and their fluctuations are important for CNS aspects ranging from sleep and appetite regulation to cognitive function. This review will summarize and critically evaluate how age-related changes in sex and metabolic hormones modulate affect cognitive function and the implications of targeting the neuroendocrinological system as a therapeutic strategy in Alzheimer's disease.
Collapse
Affiliation(s)
- Russell Palm
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | | | | | |
Collapse
|
34
|
Neurosteroid biosynthetic pathways changes in prefrontal cortex in Alzheimer's disease. Neurobiol Aging 2011; 32:1964-76. [DOI: 10.1016/j.neurobiolaging.2009.12.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 12/10/2009] [Accepted: 12/17/2009] [Indexed: 12/21/2022]
|
35
|
Genetic influences on hippocampal volume differ as a function of testosterone level in middle-aged men. Neuroimage 2011; 59:1123-31. [PMID: 21983185 DOI: 10.1016/j.neuroimage.2011.09.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 09/18/2011] [Accepted: 09/19/2011] [Indexed: 11/23/2022] Open
Abstract
The hippocampus expresses a large number of androgen receptors; therefore, in men it is potentially vulnerable to the gradual age-related decline of testosterone levels. In the present study we sought to elucidate the nature of the relationship between testosterone and hippocampal volume in a sample of middle-aged male twins (average age 55.8 years). We found no evidence for a correlation between testosterone level and hippocampal volume, as well as no indication of shared genetic influences. However, a significant moderating effect of testosterone on the genetic and environmental determinants of hippocampal volume was observed. Genetic influences on hippocampal volume increased substantially as a function of increasing testosterone level, while environmental influences either decreased or remained stable. These findings provide evidence for an apparent gene-by-hormone interaction on hippocampal volume. To the best of our knowledge, this is the first study to demonstrate that the heritability of a brain structure in adults may be modified by an endogenous biological factor.
Collapse
|
36
|
Thompson CK, Brenowitz EA. Neuroprotective effects of testosterone in a naturally occurring model of neurodegeneration in the adult avian song control system. J Comp Neurol 2011; 518:4760-70. [PMID: 20963827 DOI: 10.1002/cne.22486] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Seasonal regression of the avian song control system, a series of discrete brain nuclei that regulate song learning and production, serves as a useful model for investigating the neuroprotective effects of steroids. In seasonally breeding male songbirds, the song control system regresses rapidly when males are transferred from breeding to nonbreeding physiological conditions. One nucleus in particular, the HVC, regresses in volume by 22% within days of castration and transfer to a nonbreeding photoperiod. This regression is mediated primarily by a 30% decrease in neuron number, a result of a caspase-dependent process of programmed cell death. Here we examine whether testosterone (T) can act locally in the brain to prevent seasonal-like neurodegeneration in HVC. We began to infuse T intracerebrally near HVC on one side of the brain in breeding-condition male white-crowned sparrows 2 days prior to T withdrawal and shifting them to short-day photoperiods. The birds were killed 3 or 7 days later. Local T infusion significantly protected ipsilateral HVC from volume regression and neuron loss. In addition, T infusion significantly reduced the number, density, and number/1,000 neurons of activated caspase-3 cells and cells positive for cleaved PARP, both markers for programmed cell death, in the ipsilateral HVC. T infusion near HVC also prevented regression of ipsilateral efferent targets of HVC neurons, including the volumes of robust nucleus of the arcopallium (RA) and Area X and the soma area and density of RA neurons. Thus T can act locally in the brain to have a neuroprotective effect and act transsynaptically to prevent regression of efferent nuclei.
Collapse
Affiliation(s)
- Christopher K Thompson
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington 98195-1525, USA.
| | | |
Collapse
|
37
|
Rosario ER, Chang L, Head EH, Stanczyk FZ, Pike CJ. Brain levels of sex steroid hormones in men and women during normal aging and in Alzheimer's disease. Neurobiol Aging 2011; 32:604-13. [PMID: 19428144 PMCID: PMC2930132 DOI: 10.1016/j.neurobiolaging.2009.04.008] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 03/21/2009] [Accepted: 04/10/2009] [Indexed: 01/12/2023]
Abstract
We examined the relationships between normal aging, Alzheimer's disease (AD), and brain levels of sex steroid hormones in men and women. In postmortem brain tissue from neuropathologically normal, postmenopausal women, we found no age-related changes in brain levels of either androgens or estrogens. In comparing women with and without AD at different ages, brain levels of estrogens and androgens were lower in AD cases aged 80 years and older but not significantly different in the 60-79 year age range. In male brains, we observed that normal aging was associated with significant decreases in androgens but not estrogens. Further, in men aged 60-79 years, brain levels of testosterone but not estrogens were lower in cases with mild neuropathological changes as well as those with advanced AD neuropathology. In male cases over age 80, brain levels hormones did not significantly vary by neuropathological status. To begin investigating the relationships between hormone levels and indices of AD neuropathology, we measured brain levels of soluble β-amyloid (Aβ). In male cases with mild neuropathological changes, we found an inverse relationship between brain levels of testosterone and soluble Aβ. Collectively, these findings demonstrate sex-specific relationships between normal, age-related depletion of androgens and estrogens in men and women, which may be relevant to development of AD.
Collapse
Affiliation(s)
- Emily R. Rosario
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Lilly Chang
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Elizabeth H. Head
- Department of Neurology, University of California Irvine, Irvine, CA 92697
| | - Frank Z. Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
38
|
Thompson CK. Cell death and the song control system: A model for how sex steroid hormones regulate naturally-occurring neurodegeneration. Dev Growth Differ 2011; 53:213-24. [DOI: 10.1111/j.1440-169x.2011.01257.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
The role of oestrogen in the pathogenesis of obesity, type 2 diabetes, breast cancer and prostate disease. Eur J Cancer Prev 2011; 19:256-71. [PMID: 20535861 DOI: 10.1097/cej.0b013e328338f7d2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A detailed review of the literature was performed in a bid to identify the presence of a common link between specific hormone interactions and the increasing prevalence of global disease. The synergistic action of unopposed oestrogen and leptin, compounded by increasing insulin, cortisol and xeno-oestrogen exposure directly initiate, promote and exacerbate obesity, type 2 diabetes, uterine overgrowth, prostatic enlargement, prostate cancer and breast cancer. Furthermore these hormones significantly contribute to the incidence and intensity of anxiety and depression, Alzheimer's disease, heart disease and stroke. This review, in collaboration with hundreds of evidence-based clinical researchers, correlates the significant interactions these hormones exert upon the upregulation of p450 aromatase, oestrogen, leptin and insulin receptor function; the normal status quo of their binding globulins; and how adduct formation alters DNA sequencing to ultimately produce an array of metabolic conditions ranging from menopausal symptoms and obesity to Alzheimer's disease and breast and prostate cancer. It reveals the way that poor diet, increased stress, unopposed endogenous oestrogens, exogenous oestrogens, pesticides, xeno-oestrogens and leptin are associated with increased aromatase activity, and how its products, increased endogenous oestrogen and lowered testosterone, are associated with obesity, type 2 diabetes, Alzheimer's disease and oestrogenic disease. This controversial break-through represents a paradigm shift in medical thinking, which can prevent the raging pandemic of diabetes, obesity and cancer currently sweeping the world, and as such, it will reshape health initiatives, reduce suffering, prevent waste of government expenditure and effectively transform preventative medicine and global health care for decades.
Collapse
|
40
|
Bian C, Zhang D, Guo Q, Cai W, Zhang J. Localization and sex-difference of steroid receptor coactivator-1 immunoreactivities in the brain of adult female and male mice. Steroids 2011; 76:269-79. [PMID: 21145336 DOI: 10.1016/j.steroids.2010.11.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/10/2010] [Accepted: 11/29/2010] [Indexed: 11/24/2022]
Abstract
Females and males are different in brain and behaviors. These differences are mediated by steroids and their nuclear receptors which require coactivators to regulate the transcription of target genes. Studies have shown that these coactivators are critical for modulating steroid hormone action in the brain. Steroid receptor coactivator-1 has been implied in the regulation of reproduction, stress, motor learning, and limited studies have reported the sex-specific difference of SRC-1 mRNA or protein expression in specific brain regions, but the expression and differences of SRC-1 immunoreactivities in adult female and male brain remain unclear. In this study we reported that in both sexes, high levels of SRC-1 immunoreactivities were detected in olfactory bulb, cerebral cortex, hippocampus, Purkinje cells, some limited diencephalon and brainstem nuclei. The immunopositive materials were predominantly detected in cell nucleus, but in some regions they were also detected in the processes or fiber-like structures. In most of the brain regions studied, males possessed significantly higher levels of SRC-1 immunoreactivities than that of females. Higher levels of SRC-1 were detected in some nuclei related to learning and memory, motor regulation and reproduction indicated its potential roles in neurodegeneration and sex-dependent behavior and structure; the region- and sex-specific localization of SRC-1 immunoreactivities in agreement with that of some steroid receptors, indicating this coactivator play important roles in these hormone-reactive regions and cell groups related to reproduction, learning and memory, integration of motor and sense.
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Shapingba, Chongqing, China
| | | | | | | | | |
Collapse
|
41
|
Caspase inhibitor infusion protects an avian song control circuit from seasonal-like neurodegeneration. J Neurosci 2008; 28:7130-6. [PMID: 18614682 DOI: 10.1523/jneurosci.0663-08.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Sex steroids such as androgens and estrogens have trophic effects on the brain and can ameliorate neurodegeneration, and the withdrawal of circulating steroids induces neurodegeneration in several hormone-sensitive brain areas. Very little is known about the underlying molecular mechanisms that mediate neuronal regression caused by hormone-withdrawal, however. Here we show that reduction of programmed cell death by local infusion of caspase inhibitors rescues a telencephalic nucleus in the adult avian song control system from neurodegeneration that is induced by hormone withdrawal. This treatment also has trans-synaptic effects that provide some protection of an efferent target region. We found that unilateral infusion of caspase inhibitors in vivo in adult white-crowned sparrows rescued neurons within the hormone-sensitive song nucleus HVC (used as a proper name) from programmed cell death for as long as seven days after withdrawal of testosterone and a shift to short-day photoperiod and that the activation of caspase-3 was reduced by 59% on average in the ipsilateral HVC compared with the unmanipulated contralateral HVC. Caspase inhibitor infusion near HVC was sufficient to preserve neuron size ipsilaterally in a downstream nucleus, the robust nucleus of the arcopallium. This is the first report that sustained local application of caspase inhibitors can protect a telencephalic brain area from neurodegeneration in vivo and that a degenerating neural circuit rescued with caspase inhibitors produces sufficient trophic support to protect attributes of a downstream target that would otherwise degenerate. These results strengthen the case for the possible therapeutic use of caspase inhibitors under certain neurodegenerative conditions.
Collapse
|
42
|
Meitzen J, Thompson CK. Seasonal-like growth and regression of the avian song control system: neural and behavioral plasticity in adult male Gambel's white-crowned sparrows. Gen Comp Endocrinol 2008; 157:259-65. [PMID: 18457836 PMCID: PMC2518090 DOI: 10.1016/j.ygcen.2008.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/12/2008] [Accepted: 03/17/2008] [Indexed: 12/16/2022]
Abstract
Birdsong is regulated by a series of discrete brain nuclei known as the song control system. In seasonally-breeding male songbirds, seasonal changes in steroid sex hormones regulate the structure and electrophysiology of song control system neurons, resulting in dramatic changes in singing behavior. Male songbirds can be brought into the laboratory, where circulating levels of steroid hormone and photoperiod can be abruptly manipulated, providing controlled conditions under which rapid "seasonal-like" changes in behavior and morphology can be carefully studied. In this mini-review, we discuss the steroidal and cellular mechanisms underlying seasonal-like growth and regression of the song control system in adult male Gambel's white-crowned sparrows (Zonotrichia leucophrys gambelii), and its impact on song behavior. Specifically, we discuss recent advances concerning: (1) the role of androgen and estrogen receptors in inducing seasonal-like growth of the song control system; (2) how photoperiod modulates the time course of testosterone-induced growth of the song control system; (3) how bilateral intracerebral infusion of androgen and estrogen receptor antagonists near the song control nucleus HVC prevents seasonal-like increases in song stereotypy but not song rate; and (4) the steroidal and cellular mechanisms that mediate rapid regression of the song control system. Throughout this mini-review we compare data collected from white-crowned sparrows to that from other songbird species. We conclude by outlining avenues of future research.
Collapse
Affiliation(s)
- John Meitzen
- Graduate Program in Neurobiology and Behavior, University of Washington, Box 356515, Seattle, WA 98195-6515, USA.
| | | |
Collapse
|
43
|
Nguyen TVV, Galvan V, Huang W, Banwait S, Tang H, Zhang J, Bredesen DE. Signal transduction in Alzheimer disease: p21-activated kinase signaling requires C-terminal cleavage of APP at Asp664. J Neurochem 2007; 104:1065-80. [PMID: 17986220 DOI: 10.1111/j.1471-4159.2007.05031.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The deficits in Alzheimer disease (AD) stem at least partly from neurotoxic beta-amyloid peptides generated from the amyloid precursor protein (APP). APP may also be cleaved intracellularly at Asp664 to yield a second neurotoxic peptide, C31. Previously, we showed that cleavage of APP at the C-terminus is required for the impairments seen in APP transgenic mice, by comparing elements of the disease in animals modeling AD, with (platelet-derived growth factor B-chain promoter-driven APP transgenic mice; PDAPP) versus without (PDAPP D664A) a functional Asp664 caspase cleavage site. However, the signaling mechanism(s) by which Asp664 contributes to these deficits remains to be elucidated. In this study, we identify a kinase protein, recently shown to bind APP at the C-terminus and to contribute to AD, whose activity is modified in PDAPP mice, but normalized in PDAPP D664A mice. Specifically, we observed a significant increase in nuclear p21-activated kinase (isoforms 1, 2, and or 3; PAK-1/2/3) activation in hippocampus of 3 month old PDAPP mice compared with non-transgenic littermates, an effect completely prevented in PDAPP D664A mice. In contrast, 13 month old PDAPP mice displayed a significant decrease in PAK-1/2/3 activity, which was once again absent in PDAPP D664A mice. Similarly, in hippocampus of early and severe AD subjects, there was a progressive and subcellular-specific reduction in active PAK-1/2/3 compared with normal controls. Interestingly, total PAK-1/2/3 protein was increased in early AD subjects, but declined in moderate AD and declined further, to significantly below that of control levels, in severe AD. These findings are compatible with previous suggestions that PAK may be involved in the pathophysiology of AD, and demonstrate that both early activation and late inactivation in the murine AD model require the cleavage of APP at Asp664.
Collapse
|
44
|
Thompson CK, Bentley GE, Brenowitz EA. Rapid seasonal-like regression of the adult avian song control system. Proc Natl Acad Sci U S A 2007; 104:15520-5. [PMID: 17875989 PMCID: PMC2000488 DOI: 10.1073/pnas.0707239104] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We analyzed how rapidly avian song control nuclei regress after testosterone (T) withdrawal. Regression of neuronal attributes resulting from T withdrawal has been observed in several animal models. The time course over which regression occurs is not known, however. To address this issue, we castrated adult male white-crowned sparrows and rapidly shifted them to short-day photoperiods after being held under breeding conditions (long-day photoperiod and systemic T exposure) for 3 weeks. We found that the volume of one song nucleus, HVC, regressed 22% within 12 h after T withdrawal. Changes in HVC neuron density after T withdrawal were dynamic; density increased at 12 h and then decreased by 4 days. HVC neuron number was reduced by 26% by 4 days. The volumes of Area X and the robust nucleus of the arcopallium (RA) were significantly regressed by 7 and 20 days, respectively. RA somatic area and neuronal spacing were significantly reduced by 2 days. The rapidity of HVC regression is unprecedented among vertebrate models of hormone-sensitive neural circuits. These results reveal that the rapid regression of the song control system provides a model for the important role sex steroid hormones play in mediating adult neural plasticity and in neuroprotection.
Collapse
Affiliation(s)
- Christopher K Thompson
- Graduate Program in Neurobiology and Behavior, University of Washington, Box 351525, Seattle, WA 98195-1525, USA.
| | | | | |
Collapse
|
45
|
Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D. Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci 2007; 25:3039-46. [PMID: 17561817 DOI: 10.1111/j.1460-9568.2007.05563.x] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous studies have shown that the neuroprotective hormone, testosterone, administered immediately after neural injury, reduces reactive astrogliosis. In this study we have assessed the effect of early and late therapy with testosterone or its metabolites, oestradiol and dihydrotestosterone, on reactive astroglia and reactive microglia after a stab wound brain injury in orchidectomized Wistar rats. Animals received daily s.c. injections of testosterone, oestradiol or dihydrotestosterone on days 0-2 or on days 5-7 after injury. The number of vimentin immunoreactive astrocytes and the volume fraction of major histocompatibility complex-II (MHC-II) immunoreactive microglia were estimated in the hippocampus in the lateral border of the wound. Both early and delayed administration of testosterone or oestradiol, but not dihydrotestosterone, resulted in a significant decrease in the number of vimentin-immunoreactive astrocytes. The volume fraction of MHC-II immunoreactive microglia was significantly decreased in the animals that received testosterone or oestradiol in both early and delayed treatments and in animals that received early dihydrotestosterone administration. Thus, both early and delayed administration of testosterone reduces reactive astroglia and reactive microglia and these effects may be at least in part mediated by oestradiol, while dihydrotestosterone may mediate part of the early effects of testosterone on reactive microglia. In conclusion, testosterone controls reactive gliosis and its metabolites, oestradiol and dihydrotestosterone, may be involved in this hormonal effect. The regulation of gliosis may be part of the neuroprotective mechanism of testosterone.
Collapse
|
46
|
Rosario ER, Pike CJ. Androgen regulation of beta-amyloid protein and the risk of Alzheimer's disease. ACTA ACUST UNITED AC 2007; 57:444-53. [PMID: 17658612 PMCID: PMC2390933 DOI: 10.1016/j.brainresrev.2007.04.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/25/2007] [Accepted: 04/27/2007] [Indexed: 01/08/2023]
Abstract
Advancing age is the most significant risk factor for the development of Alzheimer's disease (AD), however the age-related changes that underlie this effect remain unclear. In men, one normal consequence of aging is a robust decline in circulating and brain levels of the sex steroid hormone testosterone. Testosterone depletion leads to functional impairments and increased risk of disease in androgen-responsive tissues throughout the body, including brain. In this review we discuss the relationship between age-related testosterone depletion and the development of AD. Specifically, we focus on androgen regulation of beta-amyloid protein (Abeta), the accumulation of which is a key initiating factor in AD pathogenesis. Emerging data suggest that the regulatory actions of androgens on both Abeta and the development of AD support consideration of androgen therapy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Emily R Rosario
- Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | | |
Collapse
|
47
|
Nguyen TVV, Yao M, Pike CJ. Flutamide and cyproterone acetate exert agonist effects: induction of androgen receptor-dependent neuroprotection. Endocrinology 2007; 148:2936-43. [PMID: 17347309 DOI: 10.1210/en.2006-1469] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Androgens can exert profound effects on the organization, development, and function of the nervous system through activation of androgen receptors (ARs). Nonsteroidal and steroidal antiandrogens antagonize AR-mediated, classic genomic actions of androgens. However, emerging studies in nonneuronal cells indicate that antiandrogens can act as partial agonists for the AR. Here we investigated the effects of the antiandrogens flutamide and cyproterone acetate on neuroprotection induced by dihydrotestosterone (DHT). We observed that, although flutamide and cyproterone acetate blocked androgen-induced gene expression, they failed to inhibit DHT protection against apoptotic insults in cultured hippocampal neurons. Interestingly, flutamide and cyproterone acetate alone, like DHT, significantly reduced apoptosis. Furthermore, the protective actions of flutamide and cyproterone acetate were observed specifically in AR-expressing cell lines, suggesting a role for AR in the agonist effects of antiandrogens. Our results indicate that, in contrast to the classic antiandrogen properties of flutamide and cyproterone acetate, these AR modulators display agonist activities at the level of neuroprotection. These findings provide new insight into the agonist vs. antagonist properties of antiandrogens, information that will be crucial to understanding the neural implications of clinically used AR-modulating drugs.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- University of Southern California, Davis School of Gerontology, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | | | | |
Collapse
|
48
|
Rosario ER, Carroll JC, Oddo S, LaFerla FM, Pike CJ. Androgens regulate the development of neuropathology in a triple transgenic mouse model of Alzheimer's disease. J Neurosci 2007; 26:13384-9. [PMID: 17182789 PMCID: PMC6674990 DOI: 10.1523/jneurosci.2514-06.2006] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Normal age-related testosterone depletion in men is a recently identified risk factor for Alzheimer's disease (AD), but how androgen loss affects the development of AD is unclear. To investigate the relationship between androgen depletion and AD, we compared how androgen status affects the progression of neuropathology in the triple transgenic mouse model of AD (3xTg-AD). Adult male 3xTg-AD mice were sham gonadectomized (GDX) or GDX to deplete endogenous androgens and then exposed for 4 months to either the androgen dihydrotestosterone (DHT) or to placebo. In comparison to gonadally intact 3xTg-AD mice, GDX mice exhibited robust increases in the accumulation of beta-amyloid (Abeta), the protein implicated as the primary causal factor in AD pathogenesis, in both hippocampus and amygdala. In parallel to elevated levels of Abeta, GDX mice exhibited significantly impaired spontaneous alternation behavior, indicating deficits in hippocampal function. Importantly, DHT treatment of GDX 3xTg-AD mice attenuated both Abeta accumulation and behavioral deficits. These data demonstrate that androgen depletion accelerates the development of AD-like neuropathology, suggesting that a similar mechanism may underlie the increased risk for AD in men with low testosterone. In addition, our finding that DHT protects against acceleration of AD-like neuropathology predicts that androgen-based hormone therapy may be a useful strategy for the prevention and treatment of AD in aging men.
Collapse
Affiliation(s)
| | | | - Salvatore Oddo
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California 92697
| | - Frank M. LaFerla
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California 92697
| | - Christian J. Pike
- Neuroscience Graduate Program and
- Davis School of Gerontology, University of Southern California, Los Angeles, California 90089, and
| |
Collapse
|
49
|
Abstract
Many treatment options are now available for pets with age-related cognitive dysfunction, including drugs, nutritional supplements, and diets. This article describes the theory, evidence of efficacy, and potential neuroprotective effects of products used to treat cognitive dysfunction. Cognitive dysfunction is a diagnosis of exclusion, because many painful conditions and health problems may present with similar signs. Practitioners must, therefore, actively question owners of senior pets in order to diagnose cognitive dysfunction and to assess the pet’s general health and well-being.
Collapse
Affiliation(s)
- Gary Landsberg
- Doncaster Animal Clinic, 99 Henderson Avenue, Thornhill, Ontario, L3T 2K9, Canada
| |
Collapse
|