1
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
2
|
Wang Q, Yu R, Fu C, Li M, Wang X, Zhu D. The relationship between male and female endogenous reproductive hormones levels and subjective cognitive decline score: A cross-sectional analysis of the Pingyin cohort study. Clin Endocrinol (Oxf) 2024; 101:659-668. [PMID: 38952182 DOI: 10.1111/cen.15104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVE Reproductive hormones might impact disease course in cognitive decline. We examined the association between male and female endogenous reproductive hormones and subjective cognitive decline (SCD) score. DESIGN, PATIENTS AND MEASUREMENTS A cross-sectional study design was used with baseline data from the Pingyin cohort study, involving 1943 participants aged 45-70 years. Oestrogen (E2), testosterone, follicle stimulating hormone (FSH) and luteinizing hormone (LH) were measured in females and E2 and testosterone were measured in males. We categorised hormones into three levels of low, intermediate and high level. The 9-item subjective cognitive decline questionnaire (SCD-Q9) scores were collected to assess the symptoms of SCD. Multivariable logistic regression models were used to estimate odds ratios (ORs) and 95% confidence interval (CI) between categorised hormone levels and SCD status. Multivariable linear regression models were also used. RESULTS Overall, 1943 participants were involved and 1285 (66.1%) were female. The mean age at baseline was 59.1 (standard deviation 7.1) years. Women with high testosterone levels had a higher probability of having SCD compared with those with low testosterone levels (OR 1.43, 95% CI 1.01-2.05). Men with a high level of testosterone (0.59, 0.35-0.98) and high testosterone/E2 ratio (0.55, 0.33-0.90) were related to decreased chances of having SCD. Each one-unit increase of testosterone was linked to reduced SCD score in males [(β: -.029, 95% CI (-0.052, -0.007)]. CONCLUSION There was sex-specific relationship between hormone levels and SCD abnormal. Those with higher testosterone levels in females may increase likelihood of experiencing SCD. Males with higher testosterone levels and higher testosterone/E2 ratio may be associated with reduced likelihood of SCD. The roles of endogenous reproductive hormone levels and their dynamic changes in cognitive function need further investigation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ruihong Yu
- Pingyin Center for Disease Control and Prevention, Jinan, Pingyin, China
| | - Chunying Fu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meiling Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyi Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongshan Zhu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shandong University, Jinan, China
| |
Collapse
|
3
|
Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati A, Liu A, Chen R, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen CJ, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Frolinger T, Zaidi M. Gene-dose-dependent reduction of Fshr expression improves spatial memory deficits in Alzheimer's mice. Mol Psychiatry 2024:10.1038/s41380-024-02824-x. [PMID: 39548323 DOI: 10.1038/s41380-024-02824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of Alzheimer's disease (AD). We have shown recently that FSH directly activates the hippocampal FSH receptors (FSHRs) to drive AD-like pathology and memory loss in mice. To unequivocally establish a role for FSH in memory loss, we depleted the Fshr on a 3xTg background and utilized Morris Water Maze to study deficits in spatial memory. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age. The loss of memory acquisition and retrieval were both rescued in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice-documenting clear gene-dose-dependent prevention of spatial memory loss. Furthermore, at 5 and 8 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the learning and/or retrieval phases, further suggesting that Fshr deletion prevents age-related progression of memory deficits. This prevention was not seen when mice were ovariectomized, except in the 8-month-old 3xTg;Fshr-/- mice. There was also a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels <8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial memory deficits in mice and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of memory loss in post-menopausal women.
Collapse
Affiliation(s)
- Funda Korkmaz
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Sims
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fazilet Sen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Farhath Sultana
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Laurencin
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liam Cullen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anusha Pallapati
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Liu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Chen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satish Rojekar
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgii Pevnev
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Uliana Cheliadinova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darya Vasilyeva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guzel Burganova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Macdonald
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mansi Saxena
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ki Goosens
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Orly Barak
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daria Lizneva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anisa Gumerova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Vitaly Ryu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tony Yuen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Tal Frolinger
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mone Zaidi
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Salcher-Konrad M, Nguyen M, Savović J, Higgins JPT, Naci H. Treatment Effects in Randomized and Nonrandomized Studies of Pharmacological Interventions: A Meta-Analysis. JAMA Netw Open 2024; 7:e2436230. [PMID: 39331390 PMCID: PMC11437387 DOI: 10.1001/jamanetworkopen.2024.36230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/04/2024] [Indexed: 09/28/2024] Open
Abstract
Importance Randomized clinical trials (RCTs) are widely regarded as the methodological benchmark for assessing clinical efficacy and safety of health interventions. There is growing interest in using nonrandomized studies to assess efficacy and safety of new drugs. Objective To determine how treatment effects for the same drug compare when evaluated in nonrandomized vs randomized studies. Data Sources Meta-analyses published between 2009 and 2018 were identified in MEDLINE via PubMed and the Cochrane Database of Systematic Reviews. Data analysis was conducted from October 2019 to July 2024. Study Selection Meta-analyses of pharmacological interventions were eligible for inclusion if both randomized and nonrandomized studies contributed to a single meta-analytic estimate. Data Extraction and Synthesis For this meta-analysis using a meta-epidemiological framework, separate summary effect size estimates were calculated for nonrandomized and randomized studies within each meta-analysis using a random-effects model and then these estimates were compared. The reporting of this study followed the Guidelines for Reporting Meta-Epidemiological Methodology Research and relevant portions of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guideline. Main Outcome and Measures The primary outcome was discrepancies in treatment effects obtained from nonrandomized and randomized studies, as measured by the proportion of meta-analyses where the 2 study types disagreed about the direction or magnitude of effect, disagreed beyond chance about the effect size estimate, and the summary ratio of odds ratios (ROR) obtained from nonrandomized vs randomized studies combined across all meta-analyses. Results A total of 346 meta-analyses with 2746 studies were included. Statistical conclusions about drug benefits and harms were different for 130 of 346 meta-analyses (37.6%) when focusing solely on either nonrandomized or randomized studies. Disagreements were beyond chance for 54 meta-analyses (15.6%). Across all meta-analyses, there was no strong evidence of consistent differences in treatment effects obtained from nonrandomized vs randomized studies (summary ROR, 0.95; 95% credible interval [CrI], 0.89-1.02). Compared with experimental nonrandomized studies, randomized studies produced on average a 19% smaller treatment effect (ROR, 0.81; 95% CrI, 0.68-0.97). There was increased heterogeneity in effect size estimates obtained from nonrandomized compared with randomized studies. Conclusions and Relevance In this meta-analysis of treatment effects of pharmacological interventions obtained from randomized and nonrandomized studies, there was no overall difference in effect size estimates between study types on average, but nonrandomized studies both overestimated and underestimated treatment effects observed in randomized studies and introduced additional uncertainty. These findings suggest that relying on nonrandomized studies as substitutes for RCTs may introduce additional uncertainty about the therapeutic effects of new drugs.
Collapse
Affiliation(s)
- Maximilian Salcher-Konrad
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
- World Health Organization Collaborating Centre for Pharmaceutical Pricing and Reimbursement Policies, Pharmacoeconomics Department, Gesundheit Österreich GmbH (GÖG)/Austrian National Public Health Institute, Vienna, Austria
| | - Mary Nguyen
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
- Department of Family and Community Medicine, University of California, San Francisco
| | - Jelena Savović
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- National Institute for Health and Care Research Applied Research Collaboration West, University Hospitals Bristol and Weston National Health Service Foundation Trust, Bristol, United Kingdom
| | - Julian P. T. Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- National Institute for Health and Care Research Applied Research Collaboration West, University Hospitals Bristol and Weston National Health Service Foundation Trust, Bristol, United Kingdom
| | - Huseyin Naci
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
| |
Collapse
|
5
|
Wang YT, Therriault J, Tissot C, Servaes S, Rahmouni N, Macedo AC, Fernandez-Arias J, Mathotaarachchi SS, Stevenson J, Lussier FZ, Benedet AL, Pascoal TA, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Rosa-Neto P. Hormone therapy is associated with lower Alzheimer's disease tau biomarkers in post-menopausal females -evidence from two independent cohorts. Alzheimers Res Ther 2024; 16:162. [PMID: 39034389 PMCID: PMC11265084 DOI: 10.1186/s13195-024-01509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 06/20/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Females represent approximately 70% of the Alzheimer's disease (AD) cases and the literature has proposed a connection between the decreased estrogen levels during menopause and an increased AD risk. Previous investigations have predominantly focused on assessing how hormone therapy (HT) affects the likelihood of AD development and cognitive deterioration. However, as the research framework has shifted toward a biomarker-defined AD and alterations in specific biomarkers could take place years before cognitive decline becomes discernible, it is crucial to examine how HT influences AD biomarkers. The main goal of this study was to evaluate the impact of HT on AD biomarker-informed pathophysiology in both cognitively unimpaired (CU) and cognitively impaired (CI) post-menopausal females across the aging and AD spectrum. METHODS This cross-sectional study included post-menopausal females without HT history (HT-) and with HT (HT+) at the time of PET imaging assessment from two cohorts: the Translational Biomarkers in Aging and Dementia (TRIAD) cohort, and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants underwent magnetic resonance imaging (MRI), positron emission tomography (PET) and biofluid collection. Voxel-based t-tests were performed to assess the differences in amyloid-β (Aβ) and tau neurofibrillary tangles (NFTs) loads between HT- and HT + females. Linear regression models with interaction terms were also conducted to examine the interactive effects of HT and Aβ-PET on regional tau-PET. RESULTS HT + females demonstrated significantly lower tau-PET standardized uptake value ratio (SUVR) in Braak I-II ROIs (P < 0.05, Hedges' g = 0.73), Braak III-IV ROIs (P < 0.0001, Hedges' g = 0.74) and Braak V-VI ROIs (P < 0.0001, Hedges' g = 0.69) compared to HT- females. HT + females also showed significantly lower CSF p-tau181 (P < 0.001) and plasma p-tau181 (P < 0.0001) concentrations. Additionally, results from multivariate linear regression models indicated that HT interacts with cortical Aβ and is associated with lower regional NFT load. CONCLUSIONS Overall, findings from this observational study suggest that HT is associated with lower tau neuroimaging and fluid biomarkers in postmenopausal females. Due to the close link between tau and cognition, this study highlights the need for large randomized controlled trials designed to systemically study the influences of HT on AD biomarkers and disease progression.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Sulantha S Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada.
- Montreal Neurological Institute, Montreal, QC, Canada.
- The McGill University Research Centre for Studies in Aging, 6875 LaSalle Boulevard, H4H 1R3, Montreal, QC, Canada.
| |
Collapse
|
6
|
Frolinger T, Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati AR, Liu A, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen C, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Zaidi M. Gene-Dose-Dependent Reduction Fshr Expression Improves Spatial Memory Deficits in Alzheimer's Mice. RESEARCH SQUARE 2024:rs.3.rs-3964789. [PMID: 38463956 PMCID: PMC10925392 DOI: 10.21203/rs.3.rs-3964789/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is a major progressive neurodegenerative disorder of the aging population. High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of AD, and we have shown recently that FSH directly activates the hippocampal Fshr to drive AD-like pathology and memory loss in mice. To establish a role for FSH in memory loss, we used female 3xTg;Fshr+/+, 3xTg;Fshr+/- and 3xTg;Fshr-/- mice that were either left unoperated or underwent sham surgery or ovariectomy at 8 weeks of age. Unoperated and sham-operated 3xTg;Fshr-/- mice were implanted with 17β-estradiol pellets to normalize estradiol levels. Morris Water Maze and Novel Object Recognition behavioral tests were performed to study deficits in spatial and recognition memory, respectively, and to examine the effects of Fshr depletion. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age; both the acquisition and retrieval of the memory were ameliorated in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice- -thus documenting a clear gene-dose-dependent prevention of hippocampal-dependent spatial memory impairment. At 5 and 10 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the acquasition and/or retrieval phases, suggesting that Fshr deletion prevented the progression of spatial memory deficits with age. However, this prevention was not seen when mice were ovariectomized, except in the 10-month-old 3xTg;Fshr-/- mice. In the Novel Object Recognition test performed at 10 months, all groups of mice, except ovariectomized 3xTg;Fshr-/- mice showed a loss of recognition memory. Consistent with the neurobehavioral data, there was a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels < 8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial and recognition memory deficits in mice, and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of cognitive decline in postmenopausal women.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Avi Liu
- Icahn School of Medicine at Mount Sinai
| | | | | | | | | | | | | | | | | | | | | | | | | | - Keqiang Ye
- Shenzhen Institute of Advanced Technology
| | | | | | | |
Collapse
|
7
|
Chen A, Li Q, Huang Y, Li Y, Chuang YN, Hu X, Guo S, Wu Y, Guo Y, Bian J. Feasibility of Identifying Factors Related to Alzheimer's Disease and Related Dementia in Real-World Data. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.10.24302621. [PMID: 38405723 PMCID: PMC10889002 DOI: 10.1101/2024.02.10.24302621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A comprehensive view of factors associated with AD/ADRD will significantly aid in studies to develop new treatments for AD/ADRD and identify high-risk populations and patients for prevention efforts. In our study, we summarized the risk factors for AD/ADRD by reviewing existing meta-analyses and review articles on risk and preventive factors for AD/ADRD. In total, we extracted 477 risk factors in 10 categories from 537 studies. We constructed an interactive knowledge map to disseminate our study results. Most of the risk factors are accessible from structured Electronic Health Records (EHRs), and clinical narratives show promise as information sources. However, evaluating genomic risk factors using RWD remains a challenge, as genetic testing for AD/ADRD is still not a common practice and is poorly documented in both structured and unstructured EHRs. Considering the constantly evolving research on AD/ADRD risk factors, literature mining via NLP methods offers a solution to automatically update our knowledge map.
Collapse
Affiliation(s)
- Aokun Chen
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Qian Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu Huang
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yongqiu Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu-neng Chuang
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Xia Hu
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Serena Guo
- Department of Pharmaceutical Outcomes & Policy, College of Pharmacy, University of Florida, 1225 Center Drive, Gainesville, FL 32610
| | - Yonghui Wu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yi Guo
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| |
Collapse
|
8
|
Gharat R, Dixit G, Khambete M, Prabhu A. Targets, trials and tribulations in Alzheimer therapeutics. Eur J Pharmacol 2024; 962:176230. [PMID: 38042464 DOI: 10.1016/j.ejphar.2023.176230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by abnormal accumulation of extracellular amyloid beta senile plaques and intracellular neurofibrillary tangles in the parts of the brain responsible for cognition. The therapeutic burden for the management of AD relies solely on cholinesterase inhibitors that provide only symptomatic relief. The urgent need for disease-modifying drugs has resulted in intensive research in this domain, which has led to better understanding of the disease pathology and identification of a plethora of new pathological targets. Currently, there are over a hundred and seventy clinical trials exploring disease modification, cognitive enhancement, and reduction of neuro-psychiatric complications. However, the path to developing safe and efficacious AD therapeutics has not been without challenges. Several clinical trials have been terminated in advanced stages due to lack of therapeutic translation or increased incidence of adverse events. This review presents an in-depth look at the various therapeutic targets of AD and the lessons learnt during their clinical assessment. Comprehensive understanding of the implication of modulating various aspects of Alzheimer brain pathology is crucial for development of drugs with potential to halt disease progression in Alzheimer therapeutics.
Collapse
Affiliation(s)
- Ruchita Gharat
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Gargi Dixit
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Mihir Khambete
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
9
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572105. [PMID: 38187564 PMCID: PMC10769303 DOI: 10.1101/2023.12.18.572105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent E2 treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p=1.6×10-51) and upregulation (p=3.8×10-3) of UBE2M across both brain regions, provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p=1.9×10-4; interaction p=3.5×10-2) of LTBR in the PFC, provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step towards understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Kip D. Zimmerman
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Department of Internal Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Larry Wilhelm
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Dongqin Zhu
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Jessica Bodie
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Steven G. Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
10
|
Nerattini M, Jett S, Andy C, Carlton C, Zarate C, Boneu C, Battista M, Pahlajani S, Loeb-Zeitlin S, Havryulik Y, Williams S, Christos P, Fink M, Brinton RD, Mosconi L. Systematic review and meta-analysis of the effects of menopause hormone therapy on risk of Alzheimer's disease and dementia. Front Aging Neurosci 2023; 15:1260427. [PMID: 37937120 PMCID: PMC10625913 DOI: 10.3389/fnagi.2023.1260427] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Despite a large preclinical literature demonstrating neuroprotective effects of estrogen, use of menopausal hormone therapy (HT) for Alzheimer's disease (AD) risk reduction has been controversial. Herein, we conducted a systematic review and meta-analysis of HT effects on AD and dementia risk. Methods Our systematic search yielded 6 RCT reports (21,065 treated and 20,997 placebo participants) and 45 observational reports (768,866 patient cases and 5.5 million controls). We used fixed and random effect meta-analysis to derive pooled relative risk (RR) and 95% confidence intervals (C.I.) from these studies. Results Randomized controlled trials conducted in postmenopausal women ages 65 and older show an increased risk of dementia with HT use compared with placebo [RR = 1.38, 95% C.I. 1.16-1.64, p < 0.001], driven by estrogen-plus-progestogen therapy (EPT) [RR = 1.64, 95% C.I. 1.20-2.25, p = 0.002] and no significant effects of estrogen-only therapy (ET) [RR = 1.19, 95% C.I. 0.92-1.54, p = 0.18]. Conversely, observational studies indicate a reduced risk of AD [RR = 0.78, 95% C.I. 0.64-0.95, p = 0.013] and all-cause dementia [RR = .81, 95% C.I. 0.70-0.94, p = 0.007] with HT use, with protective effects noted with ET [RR = 0.86, 95% C.I. 0.77-0.95, p = 0.002] but not with EPT [RR = 0.910, 95% C.I. 0.775-1.069, p = 0.251]. Stratified analysis of pooled estimates indicates a 32% reduced risk of dementia with midlife ET [RR = 0.685, 95% C.I. 0.513-0.915, p = 0.010] and non-significant reductions with midlife EPT [RR = 0.775, 95% C.I. 0.474-1.266, p = 0.309]. Late-life HT use was associated with increased risk, albeit not significant [EPT: RR = 1.323, 95% C.I. 0.979-1.789, p = 0.069; ET: RR = 1.066, 95% C.I. 0.996-1.140, p = 0.066]. Discussion These findings support renewed research interest in evaluating midlife estrogen therapy for AD risk reduction.
Collapse
Affiliation(s)
- Matilde Nerattini
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Camila Boneu
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Michael Battista
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Susan Loeb-Zeitlin
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States
| | - Yelena Havryulik
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Paul Christos
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Matthew Fink
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Neurology and Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
- Department of Experimental and Clinical Biomedical Sciences, Nuclear Medicine Unit, University of Florence, Florence, Italy
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
11
|
Hao W, Fu C, Dong C, Zhou C, Sun H, Xie Z, Zhu D. Age at menopause and all-cause and cause-specific dementia: a prospective analysis of the UK Biobank cohort. Hum Reprod 2023; 38:1746-1754. [PMID: 37344154 PMCID: PMC10663050 DOI: 10.1093/humrep/dead130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/25/2023] [Indexed: 06/23/2023] Open
Abstract
STUDY QUESTION Are there associations between natural or surgical menopause and incident dementia by age at menopause? SUMMARY ANSWER Compared to age at menopause of 46-50 years, earlier natural menopause (≤40 and 41-45 years) was related to higher risk of all-cause dementia, while a U-shape relationship was observed between age at surgical menopause and risk of dementia. WHAT IS KNOWN ALREADY Menopause marks the end of female reproductive period. Age at menopause reflects the length of exposure to endogenous estrogen. Evidence on the association between age at natural, surgical menopause, and risk of dementia has been inconsistent. STUDY DESIGN, SIZE, DURATION A population-based cohort study involving 160 080 women who participated in the UK Biobank study. PARTICIPANTS/MATERIALS, SETTING, METHODS Women with no dementia at baseline, and had no missing data on key exposure variables and covariates were included. Cox proportional hazards models were used to estimate hazard ratios (HRs) and 95% CIs on the association of categorical menopause age with incident all-cause dementia, Alzheimer's disease (AD) and vascular dementia (VD). Restricted cubic splines were used to model the non-linear relationship between continuous age at natural, surgical menopause, and risk of dementia. In addition, we analyzed the interaction effect of ever-used menopausal hormone therapy (MHT) at baseline, income level, leisure activities, and age at menopause on risk of dementia. MAIN RESULTS AND THE ROLE OF CHANCE Compared to women with age at menopause of 46-50 years, women with earlier natural menopause younger than 40 years (1.36, 1.01-1.83) and 41-45 years (1.19, 1.03-1.39) had a higher risk of all-cause dementia, while late natural menopause >55 years was linked to lower risk of dementia (0.83, 0.71-0.98). Compared to natural menopause, surgical menopause was associated with 10% higher risk of dementia (1.10, 0.98-1.24). A U-shape relationship was observed between surgical menopause and risk of dementia. Women with surgical menopause before age 40 years (1.94, 1.38-2.73) and after age 55 years (1.65, 1.21-2.24) were both linked to increased risk of all-cause dementia. Women with early natural menopause without ever taking MHT at baseline had an increased risk of AD. Also, in each categorized age at the menopause level, higher income level or higher number of leisure activities was linked to a lowers risk of dementia. LIMITATIONS, REASONS FOR CAUTION Menopausal age was based on women's self-report, which might cause recall bias. WIDER IMPLICATION OF THE FINDINGS Women who experienced natural menopause or had surgical menopause at an earlier age need close monitoring and engagement for preventive health measures to delay the development of dementia. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Start-up Foundation for Scientific Research in Shandong University (202099000066), Science Fund Program for Excellent Young Scholars of Shandong Provence (Overseas) (2022HWYQ-030), and the National Natural Science Foundation of China (82273702). There are no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Wenting Hao
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, China
| | - Chunying Fu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Caiyun Dong
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunmiao Zhou
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huizi Sun
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziwei Xie
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongshan Zhu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
12
|
Ali N, Sohail R, Jaffer SR, Siddique S, Kaya B, Atowoju I, Imran A, Wright W, Pamulapati S, Choudhry F, Akbar A, Khawaja UA. The Role of Estrogen Therapy as a Protective Factor for Alzheimer's Disease and Dementia in Postmenopausal Women: A Comprehensive Review of the Literature. Cureus 2023; 15:e43053. [PMID: 37680393 PMCID: PMC10480684 DOI: 10.7759/cureus.43053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
The complete cessation of menstruation for 12 months with associated vasomotor symptoms is termed menopause. Apart from playing a role in reproduction, estrogen significantly affects the central nervous system (CNS). Population-based studies highlighted a substantial difference in the prevalence of dementia between men and women, with Alzheimer-associated dementia being more prevalent in women, indicating that estrogen deficiency might be a risk factor for neurodegenerative diseases. Patients with dementia experience a progressive decline in neurocognitive function, beginning with short-term memory loss that progresses to long-term memory loss and the inability to perform everyday activities, leading ultimately to death. There is currently no cure for dementia, so preventing or slowing the disease's progression is paramount. Accordingly, researchers have widely studied the role of estrogen as a neuroprotective agent. Estrogen prevents dementia by augmenting Hippocampal and prefrontal cortex function, reducing neuroinflammation, preventing degradation of estrogen receptors, decreasing oxidative damage to the brain, and increasing cholinergic and serotonergic function. According to the window phase hypothesis, estrogen's effect on preventing dementia is more pronounced if therapy is started early, during the first five years of menopause. Other studies like The Woman's Health Initiative Memory Study (WHIMS) showed unfavorable effects of estrogen on the brain. This review aims to establish an understanding of the currently available data on estrogen's effect on neurodegeneration, namely, dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- Noor Ali
- Obstetrics and Gynecology, Thumbay University Hospital, Ajman, ARE
- General Physician, Dubai Medical College, DXB, ARE
| | - Rohab Sohail
- Internal Medicine, Quaid-e-Azam Medical College, Bahawalpur, PAK
| | | | - Sadia Siddique
- Gastroenterology, Blackpool Victoria Hospital National Health Services (NHS) Foundation Trust, Blackpool, GBR
| | - Berfin Kaya
- Obstetrics and Gynaecology, Izmir Ataturk Research and Training Hospital, Izmir, TUR
- Obstetrics and Gynaecology, Izmir Kâtip Celebi University, Faculty of Medicine, Izmir, TUR
| | - Inioluwa Atowoju
- Obstetrics and Gynecology, Kharkiv National Medical University, Kharkiv, UKR
| | - Alizay Imran
- Surgery, Windsor University School of Medicine, Chicago, USA
| | - Whitney Wright
- Obstetrics and Gynecology, Texila American University, Georgetown, GUY
| | - Spandana Pamulapati
- Obstetrics and Gynecology, Alluri Sita Rama Raju Academy of Medical Sciences, Eluru, IND
| | - Faiza Choudhry
- Medicine and Surgery, Liaquat University of Medical and Health Sciences, Sindh, PAK
| | - Anum Akbar
- Pediatrics, University of Nebraska Medical Center, Omaha, USA
| | - Uzzam Ahmed Khawaja
- Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Clinical and Translational Research, Dr Ferrer BioPharma, South Miami, USA
| |
Collapse
|
13
|
Xiong J, Zhang Z, Ye K. C/EBPβ/AEP Signaling Drives Alzheimer's Disease Pathogenesis. Neurosci Bull 2023; 39:1173-1185. [PMID: 36735152 PMCID: PMC10313643 DOI: 10.1007/s12264-023-01025-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/02/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia. Almost two-thirds of patients with AD are female. The reason for the higher susceptibility to AD onset in women is unclear. However, hormone changes during the menopausal transition are known to be associated with AD. Most recently, we reported that follicle-stimulating hormone (FSH) promotes AD pathology and enhances cognitive dysfunctions via activating the CCAAT-enhancer-binding protein (C/EBPβ)/asparagine endopeptidase (AEP) pathway. This review summarizes our current understanding of the crucial role of the C/EBPβ/AEP pathway in driving AD pathogenesis by cleaving multiple critical AD players, including APP and Tau, explaining the roles and the mechanisms of FSH in increasing the susceptibility to AD in postmenopausal females. The FSH-C/EBPβ/AEP pathway may serve as a novel therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology (SIAT), Shenzhen, 518034, China.
| |
Collapse
|
14
|
Oveisgharan S, Yang J, Yu L, Burba D, Bang W, Tasaki S, Grodstein F, Wang Y, Zhao J, De Jager PL, Schneider JA, Bennett DA. Estrogen Receptor Genes, Cognitive Decline, and Alzheimer Disease. Neurology 2023; 100:e1474-e1487. [PMID: 36697247 PMCID: PMC10104608 DOI: 10.1212/wnl.0000000000206833] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/05/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Lifetime risk of Alzheimer disease (AD) dementia is twofold higher in women compared with men, and low estrogen levels in postmenopause have been suggested as a possible contributor. We examined 3 ER (GPER1, ER2, and ER1) variants in association with AD traits as an indirect method to test the association between estrogen and AD in women. Although the study focus was on women, in a comparison, we separately examined ER molecular variants in men. METHODS Participants were followed for an average of 10 years in one of the 2 longitudinal clinical pathologic studies of aging. Global cognition was assessed using a composite score derived from 19 neuropsychological tests' scores. Postmortem pathologic assessment included examination of 3 AD (amyloid-β and tau tangles determined by immunohistochemistry, and a global AD pathology score derived from diffuse and neurotic plaques and neurofibrillary tangle count) and 8 non-AD pathology indices. ER molecular genomic variants included genotyping and examining ER DNA methylation and RNA expression in brain regions including the dorsolateral prefrontal cortex (DLPFC) that are major players in cognition and often have AD pathology. RESULTS The mean age of women (N = 1711) at baseline was 78.0 (SD = 7.7) years. In women, GPER1 molecular variants had the most consistent associations with AD traits. GPER1 DNA methylation was associated with cognitive decline, tau tangle density, and global AD pathology score. GPER1 RNA expression in DLPFC was related to cognitive decline and tau tangle density. Other associations included associations of ER2 and ER1 sequence variants and DNA methylation with cognition. RNA expressions in DLPFC of genes involved in signaling mechanisms of activated ERs were also associated with cognitive decline and tau tangle density in women. In men (N = 651, average age at baseline: 77.4 [SD = 7.3]), there were less robust associations between ER molecular genomic variants and AD cognitive and pathologic traits. No consistent association was seen between ER molecular genomic variations and non-AD pathologies in either of the sexes. DISCUSSION ER DNA methylation and RNA expression, and to some extent ER polymorphisms, were associated with AD cognitive and pathologic traits in women, and to a lesser extent in men.
Collapse
Affiliation(s)
- Shahram Oveisgharan
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL.
| | - Jingyun Yang
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Lei Yu
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Dominika Burba
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Woojeong Bang
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Shinya Tasaki
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Fran Grodstein
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Yanling Wang
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Jinying Zhao
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Philip Lawrence De Jager
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (S.O., J.Y., L.Y., D.B., W.B., S.T., F.G., Y.W., J.A.S., D.A.B.), Rush University Medical Center, Chicago; Departments of Neurological Sciences (S.O., J.Y., L.Y., S.T., J.A.S., D.A.B.) and Internal Medicine (F.G.), Rush University Medical Center, Chicago, IL; Department of Epidemiology (J.Z.), University of Florida, Gainesville; Center for Translational & Computational Neuroimmunology (P.L.D.J.), Department of Neurology, Columbia University Irving Medical Center, New York; Taub Institute for Research on Alzheimer's Disease and the Aging Brain (P.L.D.J.), Columbia University Irving Medical Center, New York, New York; and Department of Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
15
|
Løkkegaard LE, Thinggaard M, Nygaard M, Hallas J, Osler M, Christensen K. Systemic hormone therapy and dementia: A nested case-control and co-twin control study. Maturitas 2022; 165:113-119. [PMID: 36184115 DOI: 10.1016/j.maturitas.2022.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/12/2022] [Accepted: 04/15/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The effect of systemic hormone therapy (HT) on dementia risk is unclear. Our aim was to investigate the association between HT and dementia. STUDY DESIGN This register-based study consists of a nested case-control study and a co-twin control design, which controls for familial confounding, including shared genetics. MAIN OUTCOME MEASURES Through Danish national registries from 1995 to 2011, we identified: a) 2700 female singletons with incident dementia and 13,492 matched controls; b) 288 female twins with incident dementia and co-twins without dementia. Data on HT and education were retrieved, and analyses were performed using conditional logistic regression and McNemar's χ2-test. HT use decreased dramatically after the Women's Health Initiative study results were published in 2002, and the analyses were stratified accordingly to account for potentially different HT user characteristics. RESULTS The odds ratio (OR) for the association between systemic HT and dementia was 1.05, 95% CI = [0.93-1.19] in singletons and 2.10, 95% CI = [0.99-4.46] in twins. A statistically significant association was found for systemic HT before 2003 in both populations, with an OR of 1.14, 95% CI = [1.01-1.28] in singletons and an OR of 2.20, 95% CI = [1.04-4.65] in twins. CONCLUSION Using Danish nationwide registries and controlling for education and for familial factors in a subsample, systemic HT was found to be associated with increased dementia risk if used before 2003, when HT was more commonly prescribed.
Collapse
Affiliation(s)
- Laura Ekstrøm Løkkegaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark.
| | - Mikael Thinggaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark
| | - Marianne Nygaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark
| | - Jesper Hallas
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense University Hospital, J. B. Winsløws Vej 19, 5000 Odense C, Denmark
| | - Merete Osler
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospitals, Nordre Fasanvej 57, 2000 Frederiksberg, Copenhagen, Denmark
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J. B. Winsløws Vej 9B, 5000 Odense C, Denmark
| |
Collapse
|
16
|
Li J, Wei Z, Wu J, Min K, Li X, Yao Y, Li Y, Zhang N, Shi A, Han J, Qiao C, Yang K. Trends in research related to menopausal hormone therapy from 2000 to 2021: A bibliometric analysis. Front Med (Lausanne) 2022; 9:952487. [DOI: 10.3389/fmed.2022.952487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
We conducted the present bibliometric analysis to explore menopausal hormone therapy (MHT)-related research trends between 2000 and 2021. The Web of Science database was systematically searched from 2000 to 2021 to retrieve MHT-related publications. Visualization mapping and keyword cluster graphs were utilized to illustrate the research topics and hotpots. We included 11,616 MHT-related publications for this bibliometric analysis. The results showed that (1) MHT-related research had a very slow increase in the past 22 years, and the trend fluctuated. Sum of times cited and average citations per item had the same trend: a sharp decline from 2002 to 2003, and a rapid increase from 2003 to 2006, reaching the peak in 2006, then following a downward trend. The average H-index was 57, peaking in 2001; (2) the USA, the League of European Research Universities, and Dr. JoAann Manson from Harvard University contributed the most; (3) Menopause: The Journal of The North American Menopause Society had the most significant number of MHT-related publications; (4) the research hotpots primarily focused on MHT for treating menopausal symptoms and the impact of MHT on women’s health. According to previous studies, MHT was the most effective treatment for managing vasomotor symptoms of menopause, but results from the clinical trials and observational studies regarding MHT adverse events remain inconsistent. Mechanisms are fundamental when clinical studies give conflicting results. Therefore, future studies should focus on adverse events and their mechanisms.
Collapse
|
17
|
Gera S, Kuo TC, Gumerova AA, Korkmaz F, Sant D, DeMambro V, Sudha K, Padilla A, Prevot G, Munitz J, Teunissen A, van Leent MMT, Post TGJM, Fernandes JC, Netto J, Sultana F, Shelly E, Rojekar S, Kumar P, Cullen L, Chatterjee J, Pallapati A, Miyashita S, Kannangara H, Bhongade M, Sengupta P, Ievleva K, Muradova V, Batista R, Robinson C, Macdonald A, Hutchison S, Saxena M, Meseck M, Caminis J, Iqbal J, New MI, Ryu V, Kim SM, Cao JJ, Zaidi N, Fayad ZA, Lizneva D, Rosen CJ, Yuen T, Zaidi M. FSH-blocking therapeutic for osteoporosis. eLife 2022; 11:e78022. [PMID: 36125123 PMCID: PMC9550223 DOI: 10.7554/elife.78022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Pharmacological and genetic studies over the past decade have established the follicle-stimulating hormone (FSH) as an actionable target for diseases affecting millions, namely osteoporosis, obesity, and Alzheimer's disease. Blocking FSH action prevents bone loss, fat gain, and neurodegeneration in mice. We recently developed a first-in-class, humanized, epitope-specific FSH-blocking antibody, MS-Hu6, with a KD of 7.52 nM. Using a Good Laboratory Practice (GLP)-compliant platform, we now report the efficacy of MS-Hu6 in preventing and treating osteoporosis in mice and parameters of acute safety in monkeys. Biodistribution studies using 89Zr-labeled, biotinylated or unconjugated MS-Hu6 in mice and monkeys showed localization to bone and bone marrow. The MS-Hu6 displayed a β phase t½ of 7.5 days (180 hr) in humanized Tg32 mice. We tested 217 variations of excipients using the protein thermal shift assay to generate a final formulation that rendered MS-Hu6 stable in solution upon freeze-thaw and at different temperatures, with minimal aggregation, and without self-, cross-, or hydrophobic interactions or appreciable binding to relevant human antigens. The MS-Hu6 showed the same level of "humanness" as human IgG1 in silico and was non-immunogenic in ELISpot assays for IL-2 and IFN-γ in human peripheral blood mononuclear cell cultures. We conclude that MS-Hu6 is efficacious, durable, and manufacturable, and is therefore poised for future human testing.
Collapse
Affiliation(s)
- Sakshi Gera
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Tan-Chun Kuo
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anisa Azatovna Gumerova
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Damini Sant
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | | | - Karthyayani Sudha
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ashley Padilla
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Geoffrey Prevot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Abraham Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Mandy MT van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Tomas GJM Post
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jessica C Fernandes
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jessica Netto
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Farhath Sultana
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Eleanor Shelly
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Satish Rojekar
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Pushkar Kumar
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jiya Chatterjee
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anusha Pallapati
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Hasni Kannangara
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Megha Bhongade
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Puja Sengupta
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Kseniia Ievleva
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Valeriia Muradova
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Rogerio Batista
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Cemre Robinson
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Susan Hutchison
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Mansi Saxena
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Marcia Meseck
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - John Caminis
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jameel Iqbal
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Maria I New
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jay J Cao
- United States Department of Agriculture, Grand Forks Human Nutrition Research CenterGrand ForksUnited States
| | - Neeha Zaidi
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins UniversityBaltimoreUnited States
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | | | - Tony Yuen
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology and The Mount Sinai Bone Program, Departments of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
18
|
Jett S, Schelbaum E, Jang G, Boneu Yepez C, Dyke JP, Pahlajani S, Diaz Brinton R, Mosconi L. Ovarian steroid hormones: A long overlooked but critical contributor to brain aging and Alzheimer's disease. Front Aging Neurosci 2022; 14:948219. [PMID: 35928995 PMCID: PMC9344010 DOI: 10.3389/fnagi.2022.948219] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/28/2022] [Indexed: 01/19/2023] Open
Abstract
Ovarian hormones, particularly 17β-estradiol, are involved in numerous neurophysiological and neurochemical processes, including those subserving cognitive function. Estradiol plays a key role in the neurobiology of aging, in part due to extensive interconnectivity of the neural and endocrine system. This aspect of aging is fundamental for women's brains as all women experience a drop in circulating estradiol levels in midlife, after menopause. Given the importance of estradiol for brain function, it is not surprising that up to 80% of peri-menopausal and post-menopausal women report neurological symptoms including changes in thermoregulation (vasomotor symptoms), mood, sleep, and cognitive performance. Preclinical evidence for neuroprotective effects of 17β-estradiol also indicate associations between menopause, cognitive aging, and Alzheimer's disease (AD), the most common cause of dementia affecting nearly twice more women than men. Brain imaging studies demonstrated that middle-aged women exhibit increased indicators of AD endophenotype as compared to men of the same age, with onset in perimenopause. Herein, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining cognition in women, with evidence implicating menopause-related declines in 17β-estradiol in cognitive aging and AD risk. We will review research focused on the role of endogenous and exogenous estrogen exposure as a key underlying mechanism to neuropathological aging in women, with a focus on whether brain structure, function and neurochemistry respond to hormone treatment. While still in development, this research area offers a new sex-based perspective on brain aging and risk of AD, while also highlighting an urgent need for better integration between neurology, psychiatry, and women's health practices.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Grace Jang
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Camila Boneu Yepez
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
19
|
Menopausal Hormone Therapy and the Mind: The Role of Hormone Replacement in the Prevention and Treatment of Cognitive Decline, Dementia, and Cognitive Dysfunction of Depression. Harv Rev Psychiatry 2022; 30:215-225. [PMID: 35616610 DOI: 10.1097/hrp.0000000000000339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
LEARNING OBJECTIVES After participating in this activity, learners should be better able to:• Outline the clinical recommendations for menopausal hormone treatment related to cognitive concerns• Debate and discuss the various research pieces on the use of menopausal hormone therapy cognitive decline, dysfunction, and dementia. ABSTRACT Menopause has been associated with subjective cognitive dysfunction and elevated rates of depression. While menopausal hormone therapy (MHT) is Food and Drug Administration-approved for the treatment of vasomotor symptoms related to menopause, a potential role for MHT in treating and preventing cognitive decline, dysfunction, and dementia has remained unclear and a topic of continued interest and debate across decades of research. Increasing numbers of patients are seeking help for subjective cognitive decline, and those with poorer mental health are substantially more likely to perceive themselves to be at high risk of developing dementia; thus, mental health professionals are likely to encounter such patients and may be asked to provide advice concerning MHT, cognition, and indications for MHT use. Here, we synthesize the neurobiological effects of MHT, make recommendations for its use in current clinical practice in the contexts of cognitive dysfunction associated with major depressive disorder, cognitive decline, and Alzheimer's disease, and discuss the frontiers being explored by ongoing research on this topic. We conclude that MHT to improve cognitive functioning has only a few scenarios where it would be recommended and that particular caution may be warranted for carriers of the APOE ε4 allele.
Collapse
|
20
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
21
|
Xiong J, Kang SS, Wang Z, Liu X, Kuo TC, Korkmaz F, Padilla A, Miyashita S, Chan P, Zhang Z, Katsel P, Burgess J, Gumerova A, Ievleva K, Sant D, Yu SP, Muradova V, Frolinger T, Lizneva D, Iqbal J, Goosens KA, Gera S, Rosen CJ, Haroutunian V, Ryu V, Yuen T, Zaidi M, Ye K. FSH blockade improves cognition in mice with Alzheimer's disease. Nature 2022; 603:470-476. [PMID: 35236988 PMCID: PMC9940301 DOI: 10.1038/s41586-022-04463-0] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease has a higher incidence in older women, with a spike in cognitive decline that tracks with visceral adiposity, dysregulated energy homeostasis and bone loss during the menopausal transition1,2. Inhibiting the action of follicle-stimulating hormone (FSH) reduces body fat, enhances thermogenesis, increases bone mass and lowers serum cholesterol in mice3-7. Here we show that FSH acts directly on hippocampal and cortical neurons to accelerate amyloid-β and Tau deposition and impair cognition in mice displaying features of Alzheimer's disease. Blocking FSH action in these mice abrogates the Alzheimer's disease-like phenotype by inhibiting the neuronal C/EBPβ-δ-secretase pathway. These data not only suggest a causal role for rising serum FSH levels in the exaggerated Alzheimer's disease pathophysiology during menopause, but also reveal an opportunity for treating Alzheimer's disease, obesity, osteoporosis and dyslipidaemia with a single FSH-blocking agent.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhihao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Tan-Chun Kuo
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashley Padilla
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pavel Katsel
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jocoll Burgess
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kseniia Ievleva
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Damini Sant
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shan-Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Valeriia Muradova
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tal Frolinger
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jameel Iqbal
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ki A Goosens
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sakshi Gera
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Vahram Haroutunian
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology and Departments of Pharmacological Sciences and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
22
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
23
|
Peters R, Breitner J, James S, Jicha GA, Meyer P, Richards M, Smith AD, Yassine HN, Abner E, Hainsworth AH, Kehoe PG, Beckett N, Weber C, Anderson C, Anstey KJ, Dodge HH. Dementia risk reduction: why haven't the pharmacological risk reduction trials worked? An in-depth exploration of seven established risk factors. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12202. [PMID: 34934803 PMCID: PMC8655351 DOI: 10.1002/trc2.12202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/03/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022]
Abstract
Identifying the leading health and lifestyle factors for the risk of incident dementia and Alzheimer's disease has yet to translate to risk reduction. To understand why, we examined the discrepancies between observational and clinical trial evidence for seven modifiable risk factors: type 2 diabetes, dyslipidemia, hypertension, estrogens, inflammation, omega-3 fatty acids, and hyperhomocysteinemia. Sample heterogeneity and paucity of intervention details (dose, timing, formulation) were common themes. Epidemiological evidence is more mature for some interventions (eg, non-steroidal anti-inflammatory drugs [NSAIDs]) than others. Trial data are promising for anti-hypertensives and B vitamin supplementation. Taken together, these risk factors highlight a future need for more targeted sample selection in clinical trials, a better understanding of interventions, and deeper analysis of existing data.
Collapse
Affiliation(s)
- Ruth Peters
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | - John Breitner
- Douglas Hospital Research Center and McGill UniversityQuebecCanada
| | - Sarah James
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | | | - Pierre‐Francois Meyer
- Center for Studies on the Prevention of Alzheimer's Disease (PREVENT‐AD)VerdunQuebecCanada
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - A. David Smith
- OPTIMADepartment of PharmacologyUniversity of OxfordOxfordUK
| | - Hussein N. Yassine
- Departments of Medicine and NeurologyUniversity of Southern CaliforniaCaliforniaUSA
| | - Erin Abner
- University of KentuckyLexingtonKentuckyUSA
| | - Atticus H. Hainsworth
- Molecular and Clinical Sciences Research InstituteSt GeorgesUniversity of LondonLondonUK
- Department of NeurologySt George's HospitalLondonUK
| | | | | | | | - Craig Anderson
- The George Institute for Global HealthSydneyNew South WalesAustralia
| | - Kaarin J. Anstey
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | | |
Collapse
|
24
|
Zhou HH, Yu Z, Luo L, Xie F, Wang Y, Wan Z. The effect of hormone replacement therapy on cognitive function in healthy postmenopausal women: a meta-analysis of 23 randomized controlled trials. Psychogeriatrics 2021; 21:926-938. [PMID: 34622524 DOI: 10.1111/psyg.12768] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/26/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022]
Abstract
To investigate how hormone replacement therapy (HRT) intervention affects cognitive function in randomized controlled trials of healthy postmenopausal women, the PubMed and Web of Science databases were searched for relevant publications up to 1 May 2020. Random-effects, subgroup analysis, sensitivity analysis and meta-regression analyses were conducted with 23 selected publications. HRT had a significant negative effect on global cognition (standardized mean difference (SMD): -0.04, 95% confidence interval (CI): -0.08 to -0.01). Via subgroup analysis, for those older than 60 years and with more than 6 months' intervention duration, HRT aggravated global cognition (SMD: -0.05, 95% CI: -0.08 to -0.01; SMD: -0.05, 95% CI: -0.08 to -0.01). The results of meta-regression demonstrated no significant association between HRT intervention and global cognition after adjusting for participants' age or intervention duration. In conclusion, HRT had a significant negative effect on global cognition, and this effect might be especially more visible for those aged more than 60 years and with more than 6 months' intervention. Further randomized controlled trials for postmenopausal women with a younger age and short-term HRT exposure are necessary to clarify the effects of HRT on global and domain-specific cognitive functions.
Collapse
Affiliation(s)
- Huan-Huan Zhou
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Zengli Yu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Lan Luo
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Fangfei Xie
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yun Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhongxiao Wan
- School of Public Health, Medical College of Soochow University, Suzhou, China.,College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Zhang GQ, Chen JL, Luo Y, Mathur MB, Anagnostis P, Nurmatov U, Talibov M, Zhang J, Hawrylowicz CM, Lumsden MA, Critchley H, Sheikh A, Lundbäck B, Lässer C, Kankaanranta H, Lee SH, Nwaru BI. Menopausal hormone therapy and women's health: An umbrella review. PLoS Med 2021; 18:e1003731. [PMID: 34339416 PMCID: PMC8366967 DOI: 10.1371/journal.pmed.1003731] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 08/16/2021] [Accepted: 07/12/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND There remains uncertainty about the impact of menopausal hormone therapy (MHT) on women's health. A systematic, comprehensive assessment of the effects on multiple outcomes is lacking. We conducted an umbrella review to comprehensively summarize evidence on the benefits and harms of MHT across diverse health outcomes. METHODS AND FINDINGS We searched MEDLINE, EMBASE, and 10 other databases from inception to November 26, 2017, updated on December 17, 2020, to identify systematic reviews or meta-analyses of randomized controlled trials (RCTs) and observational studies investigating effects of MHT, including estrogen-alone therapy (ET) and estrogen plus progestin therapy (EPT), in perimenopausal or postmenopausal women in all countries and settings. All health outcomes in previous systematic reviews were included, including menopausal symptoms, surrogate endpoints, biomarkers, various morbidity outcomes, and mortality. Two investigators independently extracted data and assessed methodological quality of systematic reviews using the updated 16-item AMSTAR 2 instrument. Random-effects robust variance estimation was used to combine effect estimates, and 95% prediction intervals (PIs) were calculated whenever possible. We used the term MHT to encompass ET and EPT, and results are presented for MHT for each outcome, unless otherwise indicated. Sixty systematic reviews were included, involving 102 meta-analyses of RCTs and 38 of observational studies, with 102 unique outcomes. The overall quality of included systematic reviews was moderate to poor. In meta-analyses of RCTs, MHT was beneficial for vasomotor symptoms (frequency: 9 trials, 1,104 women, risk ratio [RR] 0.43, 95% CI 0.33 to 0.57, p < 0.001; severity: 7 trials, 503 women, RR 0.29, 95% CI 0.17 to 0.50, p = 0.002) and all fracture (30 trials, 43,188 women, RR 0.72, 95% CI 0.62 to 0.84, p = 0.002, 95% PI 0.58 to 0.87), as well as vaginal atrophy (intravaginal ET), sexual function, vertebral and nonvertebral fracture, diabetes mellitus, cardiovascular mortality (ET), and colorectal cancer (EPT), but harmful for stroke (17 trials, 37,272 women, RR 1.17, 95% CI 1.05 to 1.29, p = 0.027) and venous thromboembolism (23 trials, 42,292 women, RR 1.60, 95% CI 0.99 to 2.58, p = 0.052, 95% PI 1.03 to 2.99), as well as cardiovascular disease incidence and recurrence, cerebrovascular disease, nonfatal stroke, deep vein thrombosis, gallbladder disease requiring surgery, and lung cancer mortality (EPT). In meta-analyses of observational studies, MHT was associated with decreased risks of cataract, glioma, and esophageal, gastric, and colorectal cancer, but increased risks of pulmonary embolism, cholelithiasis, asthma, meningioma, and thyroid, breast, and ovarian cancer. ET and EPT had opposite effects for endometrial cancer, endometrial hyperplasia, and Alzheimer disease. The major limitations include the inability to address the varying effects of MHT by type, dose, formulation, duration of use, route of administration, and age of initiation and to take into account the quality of individual studies included in the systematic reviews. The study protocol is publicly available on PROSPERO (CRD42017083412). CONCLUSIONS MHT has a complex balance of benefits and harms on multiple health outcomes. Some effects differ qualitatively between ET and EPT. The quality of available evidence is only moderate to poor.
Collapse
Affiliation(s)
- Guo-Qiang Zhang
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jin-Liang Chen
- Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Luo
- Department of Gastroenterology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Maya B. Mathur
- Quantitative Sciences Unit, Stanford University, Palo Alto, California, United States of America
| | - Panagiotis Anagnostis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ulugbek Nurmatov
- Division of Population Medicine, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Madar Talibov
- UMR1086 INSERM–Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers (ANTICIPE), Centre de Lutte Contre le Cancer François Baclesse, Caen, France
| | - Jing Zhang
- Department of Intensive Care Unit, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Catherine M. Hawrylowicz
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Mary Ann Lumsden
- Section of Reproductive and Maternal Medicine, Glasgow Royal Infirmary, School of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Hilary Critchley
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Aziz Sheikh
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Bo Lundbäck
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hannu Kankaanranta
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Siew Hwa Lee
- School of Nursing, Midwifery and Paramedic Practice, Robert Gordon University, Aberdeen, United Kingdom
| | - Bright I. Nwaru
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Pourhadi N, Mørch LS, Holm EA, Torp-Pedersen CT, Meaidi A. Vaginal estrogen and association with dementia: A nationwide population-based study. Alzheimers Dement 2021; 18:625-634. [PMID: 34322991 DOI: 10.1002/alz.12417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/13/2021] [Accepted: 06/09/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Use of systemic hormone therapy has been positively associated with development of dementia. Little is known about the dose-dependent effect of vaginal estradiol on dementia risk. METHODS We assessed associations between cumulative dose of vaginal estradiol tablets and dementia in a case-control study nested in a nationwide Danish cohort of women aged 50 to 60 years at study initiation, who did not use systemic hormone therapy. Each case was age-matched to 10 female controls. RESULTS A total of 4574 dementia cases were matched to 45,740 controls. Cumulative use of vaginal estradiol tablets was not associated with all-cause dementia; adjusted hazard ratio 1.02 (95% confidence interval [CI] 0.89-1.18) for low dose (< 750 mcg), 1.07 (0.94-1.21) for medium dose (750-2000 mcg), and 0.93 (0.84-1.03) for high dose (> 2000 mcg). Similarly, Alzheimer's disease (AD) only was not associated with vaginal estradiol. DISCUSSION Exposure to vaginal estradiol tablets was not associated with all-cause dementia or AD only.
Collapse
Affiliation(s)
- Nelsan Pourhadi
- Memory Clinic, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Lina S Mørch
- Cancer Surveillance and Pharmacoepidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Ellen A Holm
- Department of Medicine, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian T Torp-Pedersen
- Department of Cardiology, Nordsjaellands Hospital, Hillerød, Denmark.,Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Amani Meaidi
- Department of Gynaecology, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
27
|
Kim YJ, Soto M, Branigan GL, Rodgers K, Brinton RD. Association between menopausal hormone therapy and risk of neurodegenerative diseases: Implications for precision hormone therapy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12174. [PMID: 34027024 PMCID: PMC8118114 DOI: 10.1002/trc2.12174] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The impact of menopausal hormone therapy (HT) on age-associated Alzheimer's and neurodegenerative diseases (NDDs) remains unresolved. To determine the effect of HT, formulation, type, and duration on risk of NDDs, a retrospective analysis was performed using a 10-year Humana claims dataset. METHODS Study population included women aged 45 years or older with or without claim records of HT medications. Patients diagnosed with NDDs including Alzheimer's disease (AD), Parkinson's disease (PD), dementia, multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS) were identified. Relative risk (RR) ratios and 95% confidence intervals (CI) for combined NDDs, or AD, PD, dementia, MS, and ALS were determined. Cumulative hazard ratios were determined to investigate the association between HT and NDDs at different age groups. RESULTS In 379,352 women with or without claim records of HT, use of HT was associated with significantly reduced risk for combined NDDs (RR 0.42, 95% CI 0.40-0.43, P < 0.001). Average follow-up time was 5.1 [2.3] years. Formulations containing natural steroids 17β-estradiol and/or progesterone were associated with greater reduction in NDD risk. Oral- HT users showed significantly reduced RRs (0.42, 0.41-0.44, P < 0.001) for combined NDDs compared to non-HT users. The RRs for transdermal-HT users were significantly decreased for all-cause dementia (0.73, 0.60-0.88, P = 0.001) and MS (0.55, 0.36-0.84, P = 0.005). Greatest reduction in risk of NDD, AD, and dementia emerged in patients aged 65 years or older. Further, the protective effect of long-term therapy (>1 year) on combined NDDs, AD, PD, and dementia was greater compared to short-term therapy (≤1 year). DISCUSSION HT was associated with reduced risk of all NDDs including AD and dementia, with greater duration of therapy and natural steroid formulations associated with greater efficacy. These findings advance precision HT to prevent NDDs including AD.
Collapse
Affiliation(s)
- Yu Jin Kim
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of MedicineMD‐PhD Training ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of NeurologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
28
|
Li N, Xu X, Mao S, Jiang Y, Hu Y, Xing R, Chen Y, Ye J, Ling L, Zeng X, Han G. Association of dyslipidaemia with Alzheimer's disease in a cohort of postmenopausal women. J Int Med Res 2021; 48:300060520926020. [PMID: 32567449 PMCID: PMC7309399 DOI: 10.1177/0300060520926020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective To evaluate the association between dyslipidaemia and Alzheimer’s disease (AD) in a cohort of postmenopausal women. Methods This retrospective study analysed data from postmenopausal women with early AD (group AD) and a cohort of healthy age- and sex-matched control subjects (group NC) that were considered to be within standard limits according to a neuropsychological assessment between March 2010 and March 2019. The primary endpoints were body mass index and lipid-related laboratory parameters, including leptin, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), total cholesterol, adiponectin, triglycerides, apolipoprotein A1, apolipoprotein B and apolipoprotein E4, which were evaluated using multivariate binary logistic analysis. Results The study enrolled 200 postmenopausal women with early AD (mean ± SD age 69.34 ± 6.25 years) and 180 control subjects (mean ± SD age 67.48 ± 7.42 years). Lower HDL-C and higher LDL-C were risk factors for AD. A multivariate binary logistic regression model demonstrated that lower HDL-C and higher LDL-C were the only variables associated with the development of AD (odds ratio [OR] 21.14, 95% confidence interval [CI] 2.47, 4.13; OR 36.35, 95% CI 1.24, 3.38; respectively). Conclusion Both low HDL-C and high LDL-C were associated with the occurrence of AD in a cohort of postmenopausal women.
Collapse
Affiliation(s)
- Ning Li
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Xiaoying Xu
- Department of Ophthalmology, Baoding First Central Hospital, Baoding, Hebei Province, China
| | - Shuai Mao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Ye Jiang
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Yadong Hu
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Ruowei Xing
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Yajing Chen
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Junxing Ye
- Department of Orthopaedics, The Third People’s Hospital of Wuxi and The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Li Ling
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Xianshang Zeng
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Xianshang Zeng, Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Yuexiu District, Guangzhou 510080, Guangdong Province, China. Guowei Han, Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan 2nd Road, Yuexiu District, Guangzhou 510080, Guangdong Province, China.
| | - Guowei Han
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
29
|
Bacon ER, Brinton RD. Epigenetics of the developing and aging brain: Mechanisms that regulate onset and outcomes of brain reorganization. Neurosci Biobehav Rev 2021; 125:503-516. [PMID: 33657435 DOI: 10.1016/j.neubiorev.2021.02.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Brain development is a life-long process that encompasses several critical periods of transition, during which significant cognitive changes occur. Embryonic development, puberty, and reproductive senescence are all periods of transition that are hypersensitive to environmental factors. Rather than isolated episodes, each transition builds upon the last and is influenced by consequential changes that occur in the transition before it. Epigenetic marks, such as DNA methylation and histone modifications, provide mechanisms by which early events can influence development, cognition, and health outcomes. For example, parental environment influences imprinting patterns in gamete cells, which ultimately impacts gene expression in the embryo which may result in hypersensitivity to poor maternal nutrition during pregnancy, raising the risks for cognitive impairment later in life. This review explores how epigenetics induce and regulate critical periods, and also discusses how early environmental interactions prime a system towards a particular health outcome and influence susceptibility to disease or cognitive impairment throughout life.
Collapse
Affiliation(s)
- Eliza R Bacon
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; The Center for Precision Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; Center for Innovation in Brain Science, School of Medicine, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
30
|
Anstey KJ, Ee N, Eramudugolla R, Jagger C, Peters R. A Systematic Review of Meta-Analyses that Evaluate Risk Factors for Dementia to Evaluate the Quantity, Quality, and Global Representativeness of Evidence. J Alzheimers Dis 2020; 70:S165-S186. [PMID: 31306123 PMCID: PMC6700718 DOI: 10.3233/jad-190181] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: The translation of evidence on dementia risk factors into clinical advice requires careful evaluation of the methodology and scope of data from which risk estimates are obtained. Objective: To evaluate the quantity, quality, and representativeness of evidence, we conducted a review of reviews of risk factors for Alzheimer’s disease (AD), Vascular dementia (VaD), and Any Dementia. Methods: PubMed, Cochrane library, and the Global Index Medicus were searched to identify meta-analyses of observational studies of risk factors for AD, VaD, and Any Dementia. PROSPERO CRD42017053920. Results: Meta-analysis data were available for 34 risk factors for AD, 26 risk factors for Any Dementia and eight for VaD. Quality of evidence varied greatly in terms of the number of contributing studies, whether data on midlife exposure was available, and consistency of measures. The most evidence was available for cardiovascular risk factors. The most geographically representative evidence (five of six global regions) was available for alcohol, physical activity, diabetes, high midlife BMI, antihypertensives, and motor function. Evidence from Australia/Oceana or Africa was limited. With the exception of diabetes, meta-analysis data were unavailable from Latin America/Caribbean. Midlife specific data were only available for cholesterol and arthritis. Conclusion: There is a lack of midlife specific data, limited data on VaD, and a lack of geographical representation for many risk factors for dementia. The quality, quantity, and representativeness of evidence needs to be considered before recommendations are made about the relevance of risk factors in mid- or late-life or for dementia subtypes.
Collapse
Affiliation(s)
- Kaarin J Anstey
- School of Psychology, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
| | - Nicole Ee
- School of Psychology, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
| | | | - Carol Jagger
- Newcastle Institute for Ageing, Newcastle University, Newcastle, UK
| | - Ruth Peters
- School of Psychology, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia
| |
Collapse
|
31
|
Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: Sensitivity, Specificity and Potential for Clinical Use. J Pers Med 2020; 10:jpm10030116. [PMID: 32911755 PMCID: PMC7565390 DOI: 10.3390/jpm10030116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer's disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.
Collapse
|
32
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
33
|
Song YJ, Li SR, Li XW, Chen X, Wei ZX, Liu QS, Cheng Y. The Effect of Estrogen Replacement Therapy on Alzheimer's Disease and Parkinson's Disease in Postmenopausal Women: A Meta-Analysis. Front Neurosci 2020; 14:157. [PMID: 32210745 PMCID: PMC7076111 DOI: 10.3389/fnins.2020.00157] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Estrogen replacement therapy (ERT) is a common treatment method for menopausal syndrome; however, its therapeutic value for the treatment of neurological diseases is still unclear. Epidemiological studies were performed, and the effect of postmenopausal ERT on treating neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), was summarized through a meta-analysis. Methods: Twenty-one articles were selected using a systematic searching of the contents listed on PubMed and Web of Science before June 1, 2019. Epidemiological studies were extracted, and relevant research data were obtained from the original articles based on the predefined inclusion criteria and data screening principles. The Comprehensive Meta-Analysis Version 2 software was used to pool effective size, test heterogeneity, conduct meta-regression and subgroup analysis, and to calculate publication bias. Results: Our results showed that ERT significantly decreased the risk of onset and/or development of AD [odds ratio (OR): 0.672; 95% CI: 0.581–0.779; P < 0.001] and PD (OR: 0.470; 95% CI: 0.368–0.600; P < 0.001) compared with the control group. A subgroup and meta-regression analysis showed that study design and measure of effect were the source of heterogeneity. Age, sample size, hormone therapy ascertainment, duration of the treatment, or route of administration did not play a significant role in affecting the outcome of the meta-analysis. Conclusion: We presented evidence here to support the use of estrogen therapy for the treatment of AD and PD.
Collapse
Affiliation(s)
- Yu-Jia Song
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Shu-Ran Li
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiao-Wan Li
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Xi Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Ze-Xu Wei
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Qing-Shan Liu
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| |
Collapse
|
34
|
Frame G, Bretland KA, Dengler-Crish CM. Mechanistic complexities of bone loss in Alzheimer's disease: a review. Connect Tissue Res 2020; 61:4-18. [PMID: 31184223 DOI: 10.1080/03008207.2019.1624734] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: Alzheimer's disease (AD), the primary cause of dementia in the elderly, is one of the leading age-related neurodegenerative diseases worldwide. While AD is notorious for destroying memory and cognition, dementia patients also experience greater incidence of bone loss and skeletal fracture than age-matched neurotypical individuals, greatly impacting their quality of life. Despite the significance of this comorbidity, there is no solid understanding of the mechanisms driving early bone loss in AD. Here, we review studies that have evaluated many of the obvious risk factors shared by dementia and osteoporosis, and illuminate emerging work investigating covert pathophysiological mechanisms shared between the disorders that may have potential as new risk biomarkers or therapeutic targets in AD.Conclusions: Skeletal deficits emerge very early in clinical Alzheimer's progression, and cannot be explained by coincident factors such as aging, female sex, mobility status, falls, or genetics. While research in this area is still in its infancy, studies implicate several potential mechanisms in disrupting skeletal homeostasis that include direct effects of amyloid-beta pathology on bone cells, neurofibrillary tau-induced damage to neural centers regulating skeletal remodeling, and/or systemic Wnt/Beta-catenin signaling deficits. Data from an increasing number of studies substantiate a role for the newly discovered "exercise hormone" irisin and its protein precursor FNDC5 in bone loss and AD-associated neurodegeneration. We conclude that the current status of research on bone loss in AD is insufficient and merits critical attention because this work could uncover novel diagnostic and therapeutic opportunities desperately needed to address AD.
Collapse
Affiliation(s)
- Gabrielle Frame
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.,Biomedical Sciences Program, Kent State University, Kent, OH, USA
| | - Katie A Bretland
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.,Integrated Pharmaceutical Medicine Program, Northeast Ohio Medical University, Rootstown, OH, USA
| | | |
Collapse
|
35
|
Gava G, Orsili I, Alvisi S, Mancini I, Seracchioli R, Meriggiola MC. Cognition, Mood and Sleep in Menopausal Transition: The Role of Menopause Hormone Therapy. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E668. [PMID: 31581598 PMCID: PMC6843314 DOI: 10.3390/medicina55100668] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022]
Abstract
During the menopausal transition, which begins four to six years before cessation of menses, middle-aged women experience a progressive change in ovarian activity and a physiologic deterioration of hypothalamic-pituitary-ovarian axis function associated with fluctuating hormone levels. During this transition, women can suffer symptoms related to menopause (such as hot flushes, sleep disturbance, mood changes, memory complaints and vaginal dryness). Neurological symptoms such as sleep disturbance, "brain fog" and mood changes are a major complaint of women transitioning menopause, with a significant impact on their quality of life, productivity and physical health. In this paper, we consider the associations between menopausal stage and/or hormone levels and sleep problems, mood and reduced cognitive performance. The role of estrogen and menopause hormone therapy (MHT) in cognitive function, sleep and mood are also discussed.
Collapse
Affiliation(s)
- Giulia Gava
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy.
| | - Isabella Orsili
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Stefania Alvisi
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Ilaria Mancini
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Renato Seracchioli
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Maria Cristina Meriggiola
- Gynecology and Physiopathology of Human Reproduction, S. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
36
|
Savolainen-Peltonen H, Rahkola-Soisalo P, Hoti F, Vattulainen P, Gissler M, Ylikorkala O, Mikkola TS. Use of postmenopausal hormone therapy and risk of Alzheimer's disease in Finland: nationwide case-control study. BMJ 2019; 364:l665. [PMID: 30842086 PMCID: PMC6402043 DOI: 10.1136/bmj.l665] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To compare the use of hormone therapy between Finnish postmenopausal women with and without a diagnosis for Alzheimer's disease. DESIGN Nationwide case-control study. SETTING Finnish national population and drug register, between 1999 and 2013. PARTICIPANTS All postmenopausal women (n=84 739) in Finland who, between 1999 and 2013, received a diagnosis of Alzheimer's disease from a neurologist or geriatrician, and who were identified from a national drug register. Control women without a diagnosis (n=84 739), matched by age and hospital district, were traced from the Finnish national population register. INTERVENTIONS Data on hormone therapy use were obtained from the Finnish national drug reimbursement register. MAIN OUTCOME MEASURES Odds ratios and 95% confidence intervals for Alzheimer's disease, calculated with conditional logistic regression analysis. RESULTS In 83 688 (98.8%) women, a diagnosis for Alzheimer's disease was made at the age of 60 years or older, and 47 239 (55.7%) women had been over 80 years of age at diagnosis. Use of systemic hormone therapy was associated with a 9-17% increased risk of Alzheimer's disease. The risk of the disease did not differ significantly between users of estradiol only (odds ratio 1.09, 95% confidence interval 1.05 to 1.14) and those of oestrogen-progestogen (1.17, 1.13 to 1.21). The risk increases in users of oestrogen-progestogen therapy were not related to different progestogens (norethisterone acetate, medroxyprogesterone acetate, or other progestogens); but in women younger than 60 at hormone therapy initiation, these risk increases were associated with hormone therapy exposure over 10 years. Furthermore, the age at initiation of systemic hormone therapy was not a decisive determinant for the increase in risk of Alzheimer's disease. The exclusive use of vaginal estradiol did not affect the risk of the disease (0.99, 0.96 to 1.01). CONCLUSIONS Long term use of systemic hormone therapy might be accompanied with an overall increased risk of Alzheimer's disease, which is not related to the type of progestogen or the age at initiation of systemic hormone therapy. By contrast, use of vaginal estradiol shows no such risk. Even though the absolute risk increase for Alzheimer's disease is small, our data should be implemented into information for present and future users of hormone therapy.
Collapse
Affiliation(s)
- Hanna Savolainen-Peltonen
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
| | - Päivi Rahkola-Soisalo
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
| | | | | | - Mika Gissler
- National Institute for Health and Welfare, Helsinki, Finland
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Family Medicine, Huddinge, Sweden
- University of Turku, Research Centre for Child Psychiatry, Turku, Finland
| | - Olavi Ylikorkala
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
| | - Tomi S Mikkola
- University of Helsinki and Helsinki University Hospital, Obstetrics and Gynecology, Haartmaninkatu 2, PO Box 140, FIN-00029 HUS, 00029 Helsinki, Finland
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
| |
Collapse
|
37
|
Grossberg GT, Tong G, Burke AD, Tariot PN. Present Algorithms and Future Treatments for Alzheimer's Disease. J Alzheimers Dis 2019; 67:1157-1171. [PMID: 30741683 PMCID: PMC6484274 DOI: 10.3233/jad-180903] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2018] [Indexed: 12/13/2022]
Abstract
An estimated 47 million people live with Alzheimer's disease (AD) and other forms of dementia worldwide. Although no disease-modifying treatments are currently available for AD, earlier diagnosis and proper management of the disease could have considerable impact on patient and caregiver quality of life and functioning. Drugs currently approved for AD treat the cognitive, behavioral, and functional symptoms of the disease and consist of three cholinesterase inhibitors (ChEIs) and the N-methyl-D-aspartate receptor antagonist memantine. Treatment of patients with mild to moderate AD is generally initiated with a ChEI. Patients who show progression of symptoms while on ChEI monotherapy may be switched to another ChEI and/or memantine can be added to the treatment regimen. In recent years, putative disease-modifying therapies have emerged that aim to slow the progression of AD instead of only addressing its symptoms. However, many therapies have failed in clinical trials in patients with established AD, suggesting that, once developed, disease-modifying agents may need to be deployed earlier in the course of illness. The goal of this narrative literature review is to discuss present treatment algorithms and potential future therapies in AD.
Collapse
|
38
|
Karahan H, Lüle S, Kelicen-Uğur P. Aromatase/Seladin-1 Interactions in Human Neuronal Cell Culture, the Hippocampus of Healthy Rats and Transgenic Alzheimer’s Disease Mice. Pharmacology 2018; 102:42-52. [DOI: 10.1159/000488765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/26/2018] [Indexed: 01/15/2023]
Abstract
Background/Aims: Decreasing levels of aromatase and seladin-1 could be one of the molecular mechanisms of Alzheimer’s disease (AD). Aromatase is an enzyme that catalyzes estrogen biosynthesis from androgen precursors, and seladin-1 is an enzyme that converts desmosterol to cholesterol, which is the precursor of all hormones. Verifying the potential relationship between these proteins and accordingly determining new therapeutic targets constitute the aims of this study. Methods: Changes in protein levels were compared in vitro in aromatase and seladin-1 inhibitor-administered human neuroblastoma (SH-SY5Y) cells in vivo in intracerebroventricular (icv) aromatase or seladin-1 inhibitor-administered rats, as well as in transgenic AD mice in which the genes encoding these proteins were knocked out. Results and Conclusions: In the cell cultures, we observed that seladin-1 protein levels increased after aromatase enzyme inhibition. The hippocampal aromatase protein levels decreased following chronic seladin-1 inhibition in icv inhibitor-administered rats; however, the aromatase levels in the dentate gyrus of seladin-1 knockout (SelKO) AD male mice increased. These findings indicate a partial relationship between these proteins and their roles in AD pathology.
Collapse
|
39
|
Egashira N, Akiyoshi Y, Iba H, Arai T, Hatip-Al-Khatib I, Mishima K, Iwasaki K. Tokishakuyakusan ameliorates spatial memory deficits induced by ovariectomy combined with β-amyloid in rats. J Pharmacol Sci 2018; 136:149-154. [DOI: 10.1016/j.jphs.2018.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/20/2018] [Accepted: 01/26/2018] [Indexed: 12/18/2022] Open
|
40
|
Bulboacă AE, Bolboacă SD, Bulboacă AC, Prodan CI. Association between low thyroid-stimulating hormone, posterior cortical atrophy and nitro-oxidative stress in elderly patients with cognitive dysfunction. Arch Med Sci 2017; 13:1160-1167. [PMID: 28883858 PMCID: PMC5575209 DOI: 10.5114/aoms.2016.60129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 04/22/2016] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Cortical atrophy is known to be a valuable sign of cognitive decline. The purpose of this study was to assess the association between low thyroid-stimulating hormone (TSH), posterior cortical atrophy (Koedam score - KS) and nitro-oxidative stress in elderly patients. MATERIAL AND METHODS A study (SG) and a control group (CG), each subdivided by gender, were investigated. Subjects older than 59 years with low serum TSH level and with mild cognitive impairment were included in the SG. The CG was formed by subjects free of significant cortical atrophy and free or thyroid dysfunction. Demographic and clinical characteristics of the patients (Mini Mental State Examination, MMSE), Koedam score on cranial magnetic resonance imaging, and blood parameters (TSH, FT4, and nitric oxide - NOx) were assessed. RESULTS Subjects in the study group had fewer years of education above the 8th grade compared with the control group (p < 0.0001). A significantly higher percentage of subjects in the study group had a Koedam score of 2 or 3 compared with controls, who had in the majority of cases a Koedam score of zero (p < 0.02). Significantly higher NOx levels were observed when study groups of both genders were compared with corresponding controls (p < 0.001). No significant differences were observed with regard to FT4 (p > 0.70). Nitric oxide was found to be significantly associated with TSH (p < 0.03) and KS (p < 0.002) when the whole study group was considered as well as when just the non-smoker study group was investigated. CONCLUSIONS Our study revealed an association between subclinical thyroid hypofunction, nitro-oxidative stress, and posterior cortical atrophy as an early stage of global atrophy.
Collapse
Affiliation(s)
- Adriana E. Bulboacă
- Department of Pathophysiology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sorana D. Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Angelo C. Bulboacă
- Department of Neurology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Călin I. Prodan
- Department of Neurology, The University of Oklahoma Health Sciences Center & VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
41
|
Risk factors associated with the onset and progression of Alzheimer’s disease: A systematic review of the evidence. Neurotoxicology 2017; 61:143-187. [DOI: 10.1016/j.neuro.2017.03.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/22/2017] [Indexed: 12/25/2022]
|
42
|
Matsushita H, Minami A, Kanazawa H, Suzuki T, Subhadhirasakul S, Watanabe K, Wakatsuki A. Long-term supplementation with young coconut juice does not prevent bone loss but rather alleviates body weight gain in ovariectomized rats. Biomed Rep 2017; 6:585-591. [PMID: 28529739 DOI: 10.3892/br.2017.883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/22/2017] [Indexed: 12/24/2022] Open
Abstract
Young coconut (Cocos nucifera Linn.) juice (YCJ) has traditionally been consumed to alleviate symptoms associated with the menopause. Recently, the authors demonstrated that short-term (6-week) YCJ supplementation to ovariectomized rats resulted in increased bone mass and bone formation parameter, suggesting that YCJ consumption has a positive effect on bone metabolism and may represent an intervention to help slow the bone loss during menopause transition. The present study sought to determine how long-term (12-week) YCJ supplementation affects bone metabolism in ovariectomized rats, to investigate whether such supplementation may be helpful to in osteoporosis treatment. Ten-week-old female Wistar rats were subjected to either a sham operation (Sham) or bilateral ovariectomy (Ovx). The Ovx+YCJ group received 5X-concentrated YCJ at a dose of 15 ml/kg/day for 12 weeks. Rats in the Ovx group had significantly lower femur bone mineral density than those in the Sham group. YCJ supplementation did not significantly affect this difference. However, YCJ prevented the increase in bone area of the mid third of the femur, a site high in cortical bone, and body weight gain observed following Ovx. Our findings indicate that long-term YCJ intake does not alter bone loss, but rather alleviates body weight gain following menopause.
Collapse
Affiliation(s)
- Hiroshi Matsushita
- Department of Obstetrics and Gynecology, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akira Minami
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Hiroaki Kanazawa
- Department of Functional Anatomy, School of Nursing, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Takashi Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Sanan Subhadhirasakul
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Hat-Yai, Songkhla 90110, Thailand
| | - Kazushi Watanabe
- Department of Obstetrics and Gynecology, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akihiko Wakatsuki
- Department of Obstetrics and Gynecology, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
43
|
Bellou V, Belbasis L, Tzoulaki I, Middleton LT, Ioannidis JPA, Evangelou E. Systematic evaluation of the associations between environmental risk factors and dementia: An umbrella review of systematic reviews and meta-analyses. Alzheimers Dement 2017; 13:406-418. [PMID: 27599208 DOI: 10.1016/j.jalz.2016.07.152] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/05/2016] [Accepted: 07/22/2016] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Dementia is a heterogeneous neurodegenerative disease, whose etiology results from a complex interplay between environmental and genetic factors. METHODS We searched PubMed to identify meta-analyses of observational studies that examined associations between nongenetic factors and dementia. We estimated the summary effect size using random-effects and fixed-effects model, the 95% CI, and the 95% prediction interval. We assessed the between-study heterogeneity (I-square), evidence of small-study effects, and excess significance. RESULTS A total of 76 unique associations were examined. By applying standardized criteria, seven associations presented convincing evidence. These associations pertained to benzodiazepines use, depression at any age, late-life depression, and frequency of social contacts for all types of dementia; late-life depression for Alzheimer's disease; and type 2 diabetes mellitus for vascular dementia and Alzheimer's disease. DISCUSSION Several risk factors present substantial evidence for association with dementia and should be assessed as potential targets for interventions, but these associations may not necessarily be causal.
Collapse
Affiliation(s)
- Vanesa Bellou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Lazaros Belbasis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ioanna Tzoulaki
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Lefkos T Middleton
- Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, UK
| | - John P A Ioannidis
- Department of Medicine, Stanford Prevention Research Center, Stanford, CA, USA; Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA, USA; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA; Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, USA
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
44
|
Mo MS, Huang W, Sun CC, Zhang LM, Cen L, Xiao YS, Li GF, Yang XL, Qu SG, Xu PY. Association Analysis of Proteasome Subunits and Transporter Associated with Antigen Processing on Chinese Patients with Parkinson's Disease. Chin Med J (Engl) 2017; 129:1053-8. [PMID: 27098790 PMCID: PMC4852672 DOI: 10.4103/0366-6999.180513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Proteasome subunits (PSMB) and transporter associated with antigen processing (TAP) loci are located in the human leukocyte antigen (HLA) Class II region play important roles in immune response and protein degradation in neurodegenerative diseases. This study aimed to explore the association between single nucleotide polymorphisms (SNPs) of PSMB and TAP and Parkinson's disease (PD). Methods: A case–control study was conducted by genotyping SNPs in PSMB8, PSMB9, TAP1, and TAP2 genes in the Chinese population. Subjects included 542 sporadic patients with PD and 674 healthy controls. Nine identified SNPs in PSMB8, PSMB9, TAP1, and TAP2 were genotyped through SNaPshot testing. Results: The stratified analysis of rs17587 was specially performed on gender. Data revealed that female patients carry a higher frequency of rs17587-G/G versus (A/A + G/A) compared with controls. But there was no significant difference with respect to the genotypic frequencies of the SNPs in PSMB8, TAP1, and TAP2 loci in PD patients. Conclusion: Chinese females carrying the rs17587-G/G genotype in PSMB9 may increase a higher risk for PD, but no linkage was found between other SNPs in HLA Class II region and PD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080; Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
45
|
Abstract
This report describes the public health impact of Alzheimer's disease, including incidence and prevalence, mortality rates, costs of care, and the overall impact on caregivers and society. It also examines in detail the financial impact of Alzheimer's on families, including annual costs to families and the difficult decisions families must often make to pay those costs. An estimated 5.4 million Americans have Alzheimer's disease. By mid-century, the number of people living with Alzheimer's disease in the United States is projected to grow to 13.8 million, fueled in large part by the aging baby boom generation. Today, someone in the country develops Alzheimer's disease every 66 seconds. By 2050, one new case of Alzheimer's is expected to develop every 33 seconds, resulting in nearly 1 million new cases per year. In 2013, official death certificates recorded 84,767 deaths from Alzheimer's disease, making it the sixth leading cause of death in the United States and the fifth leading cause of death in Americans age ≥ 65 years. Between 2000 and 2013, deaths resulting from stroke, heart disease, and prostate cancer decreased 23%, 14%, and 11%, respectively, whereas deaths from Alzheimer's disease increased 71%. The actual number of deaths to which Alzheimer's disease contributes is likely much larger than the number of deaths from Alzheimer's disease recorded on death certificates. In 2016, an estimated 700,000 Americans age ≥ 65 years will die with Alzheimer's disease, and many of them will die because of the complications caused by Alzheimer's disease. In 2015, more than 15 million family members and other unpaid caregivers provided an estimated 18.1 billion hours of care to people with Alzheimer's and other dementias, a contribution valued at more than $221 billion. Average per-person Medicare payments for services to beneficiaries age ≥ 65 years with Alzheimer's disease and other dementias are more than two and a half times as great as payments for all beneficiaries without these conditions, and Medicaid payments are 19 times as great. Total payments in 2016 for health care, long-term care and hospice services for people age ≥ 65 years with dementia are estimated to be $236 billion. The costs of Alzheimer's care may place a substantial financial burden on families, who often have to take money out of their retirement savings, cut back on buying food, and reduce their own trips to the doctor. In addition, many family members incorrectly believe that Medicare pays for nursing home care and other types of long-term care. Such findings highlight the need for solutions to prevent dementia-related costs from jeopardizing the health and financial security of the families of people with Alzheimer's and other dementias.
Collapse
|
46
|
Preciados M, Yoo C, Roy D. Estrogenic Endocrine Disrupting Chemicals Influencing NRF1 Regulated Gene Networks in the Development of Complex Human Brain Diseases. Int J Mol Sci 2016; 17:E2086. [PMID: 27983596 PMCID: PMC5187886 DOI: 10.3390/ijms17122086] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
During the development of an individual from a single cell to prenatal stages to adolescence to adulthood and through the complete life span, humans are exposed to countless environmental and stochastic factors, including estrogenic endocrine disrupting chemicals. Brain cells and neural circuits are likely to be influenced by estrogenic endocrine disruptors (EEDs) because they strongly dependent on estrogens. In this review, we discuss both environmental, epidemiological, and experimental evidence on brain health with exposure to oral contraceptives, hormonal therapy, and EEDs such as bisphenol-A (BPA), polychlorinated biphenyls (PCBs), phthalates, and metalloestrogens, such as, arsenic, cadmium, and manganese. Also we discuss the brain health effects associated from exposure to EEDs including the promotion of neurodegeneration, protection against neurodegeneration, and involvement in various neurological deficits; changes in rearing behavior, locomotion, anxiety, learning difficulties, memory issues, and neuronal abnormalities. The effects of EEDs on the brain are varied during the entire life span and far-reaching with many different mechanisms. To understand endocrine disrupting chemicals mechanisms, we use bioinformatics, molecular, and epidemiologic approaches. Through those approaches, we learn how the effects of EEDs on the brain go beyond known mechanism to disrupt the circulatory and neural estrogen function and estrogen-mediated signaling. Effects on EEDs-modified estrogen and nuclear respiratory factor 1 (NRF1) signaling genes with exposure to natural estrogen, pharmacological estrogen-ethinyl estradiol, PCBs, phthalates, BPA, and metalloestrogens are presented here. Bioinformatics analysis of gene-EEDs interactions and brain disease associations identified hundreds of genes that were altered by exposure to estrogen, phthalate, PCBs, BPA or metalloestrogens. Many genes modified by EEDs are common targets of both 17 β-estradiol (E2) and NRF1. Some of these genes are involved with brain diseases, such as Alzheimer's Disease (AD), Parkinson's Disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, Autism Spectrum Disorder, and Brain Neoplasms. For example, the search of enriched pathways showed that top ten E2 interacting genes in AD-APOE, APP, ATP5A1, CALM1, CASP3, GSK3B, IL1B, MAPT, PSEN2 and TNF-underlie the enrichment of the Kyoto Encyclopedia of Genes and Genomes (KEGG) AD pathway. With AD, the six E2-responsive genes are NRF1 target genes: APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1. These genes are also responsive to the following EEDs: ethinyl estradiol (APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1), BPA (APBB2, EIF2S1, ENO1, MAPT, and PAXIP1), dibutyl phthalate (DPYSL2, EIF2S1, and ENO1), diethylhexyl phthalate (DPYSL2 and MAPT). To validate findings from Comparative Toxicogenomics Database (CTD) curated data, we used Bayesian network (BN) analysis on microarray data of AD patients. We observed that both gender and NRF1 were associated with AD. The female NRF1 gene network is completely different from male human AD patients. AD-associated NRF1 target genes-APLP1, APP, GRIN1, GRIN2B, MAPT, PSEN2, PEN2, and IDE-are also regulated by E2. NRF1 regulates targets genes with diverse functions, including cell growth, apoptosis/autophagy, mitochondrial biogenesis, genomic instability, neurogenesis, neuroplasticity, synaptogenesis, and senescence. By activating or repressing the genes involved in cell proliferation, growth suppression, DNA damage/repair, apoptosis/autophagy, angiogenesis, estrogen signaling, neurogenesis, synaptogenesis, and senescence, and inducing a wide range of DNA damage, genomic instability and DNA methylation and transcriptional repression, NRF1 may act as a major regulator of EEDs-induced brain health deficits. In summary, estrogenic endocrine disrupting chemicals-modified genes in brain health deficits are part of both estrogen and NRF1 signaling pathways. Our findings suggest that in addition to estrogen signaling, EEDs influencing NRF1 regulated communities of genes across genomic and epigenomic multiple networks may contribute in the development of complex chronic human brain health disorders.
Collapse
Affiliation(s)
- Mark Preciados
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| | - Changwon Yoo
- Department of Biostatistics, Florida International University, Miami, FL 33199, USA.
| | - Deodutta Roy
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
47
|
Depypere H, Vierin A, Weyers S, Sieben A. Alzheimer’s disease, apolipoprotein E and hormone replacement therapy. Maturitas 2016; 94:98-105. [DOI: 10.1016/j.maturitas.2016.09.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 08/05/2016] [Accepted: 09/13/2016] [Indexed: 01/27/2023]
|
48
|
Messina M. Soy and Health Update: Evaluation of the Clinical and Epidemiologic Literature. Nutrients 2016; 8:E754. [PMID: 27886135 PMCID: PMC5188409 DOI: 10.3390/nu8120754] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 01/10/2023] Open
Abstract
Soyfoods have long been recognized as sources of high-quality protein and healthful fat, but over the past 25 years these foods have been rigorously investigated for their role in chronic disease prevention and treatment. There is evidence, for example, that they reduce risk of coronary heart disease and breast and prostate cancer. In addition, soy alleviates hot flashes and may favorably affect renal function, alleviate depressive symptoms and improve skin health. Much of the focus on soyfoods is because they are uniquely-rich sources of isoflavones. Isoflavones are classified as both phytoestrogens and selective estrogen receptor modulators. Despite the many proposed benefits, the presence of isoflavones has led to concerns that soy may exert untoward effects in some individuals. However, these concerns are based primarily on animal studies, whereas the human research supports the safety and benefits of soyfoods. In support of safety is the recent conclusion of the European Food Safety Authority that isoflavones do not adversely affect the breast, thyroid or uterus of postmenopausal women. This review covers each of the major research areas involving soy focusing primarily on the clinical and epidemiologic research. Background information on Asian soy intake, isoflavones, and nutrient content is also provided.
Collapse
Affiliation(s)
- Mark Messina
- Nutrition Matters, Inc., 26 Spadina Parkway, Pittsfield, MA 01201, USA.
| |
Collapse
|
49
|
Georgakis MK, Kalogirou EI, Diamantaras AA, Daskalopoulou SS, Munro CA, Lyketsos CG, Skalkidou A, Petridou ET. Age at menopause and duration of reproductive period in association with dementia and cognitive function: A systematic review and meta-analysis. Psychoneuroendocrinology 2016; 73:224-243. [PMID: 27543884 DOI: 10.1016/j.psyneuen.2016.08.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 07/27/2016] [Accepted: 08/02/2016] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The preponderance of dementia among postmenopausal women compared with same-age men and the female sex hormones neuroprotective properties support a tentative role of their deficiency in the dementia pathogenesis. METHODS Pairs of independent reviewers screened 12,323 publications derived from a search strategy for MEDLINE to identify articles investigating the association of age at menopause/reproductive period with (i) dementia and (ii) cognitive function; a snowball of eligible articles and reviews was conducted and authors were contacted for additional information. Random-effect models were used for the meta-analysis. RESULTS Age at menopause (13 studies; 19,449 participants) and reproductive period (4 studies; 9916 participants) in the highest categories were not associated with odds of dementia (effect size [ES]: 0.97 [0.78-1.21]) and Alzheimer's disease (ES: 1.06 [0.71-1.58]). Significant heterogeneity was however noted in both analyses (I2: 63.3%, p=0.003 and I2: 72.6%, p=0.01, respectively). Subgroup analyses by outcome assessment, study design, level of adjustment and study quality did not materially change the findings. In 9/13 studies assessing cognitive function, advanced age at menopause/longer reproductive period was significantly associated with better cognitive performance/lower decline. Due to statistical differences, no meta-analysis was possible for cognitive function. CONCLUSIONS Existing evidence does not support an association between indices of prolonged exposure to female hormones and lower dementia risk. There are indications, however, for better cognitive performance and delayed cognitive decline, supporting a link between female hormone deficiency and cognitive aging. Current literature limitations, indicated by the heterogeneous study-set, point towards research priorities in this clinically relevant area.
Collapse
Affiliation(s)
- Marios K Georgakis
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| | - Eleni I Kalogirou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| | - Andreas-Antonios Diamantaras
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece; Program Medical Neurosciences, Charité-Universitätsmedizin, Berlin, 10117, Germany.
| | - Stella S Daskalopoulou
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, H3G 1A4, Canada.
| | - Cynthia A Munro
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Constantine G Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Alkistis Skalkidou
- Department of Women's and Children's Health, Uppsala University, Uppsala, 751 85, Sweden.
| | - Eleni Th Petridou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| |
Collapse
|
50
|
Bolton JL. Menopausal Hormone Therapy, Age, and Chronic Diseases: Perspectives on Statistical Trends. Chem Res Toxicol 2016; 29:1583-1590. [PMID: 27636306 PMCID: PMC5069683 DOI: 10.1021/acs.chemrestox.6b00272] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Indexed: 02/07/2023]
Abstract
The release of the Women's Health Initiative (WHI) study in 2002 was a shock to the medical community. Hormone therapy (HT) had generally been considered to be highly beneficial for postmenopausal women since it was the gold standard for relief of menopausal symptoms (hot flashes, night sweats, vaginal atrophy) and it was thought to protect women from osteoporosis, heart disease, and cognitive decline and to generally improve quality of life. However, WHI showed a statistically significant increase in a number of disease states, including breast cancer, cardiovascular disease, and stroke. One problem with the WHI study was that the average age of women in the study was 63, which is considerably older than the age at which most women enter menopause (about 51). The timing hypothesis attempts to rationalize the effect of age on response to HT and risk of various diseases. The data suggests that younger women (50-60) may be protected from heart disease with only a slight increase in breast cancer risk. In contrast, older women (>65) are more susceptible to breast cancer and heart disease and should avoid HT. This Perspective on Statistical Trends evaluates the current data on HT and risk for chronic diseases as a function of age.
Collapse
Affiliation(s)
- Judy L. Bolton
- Department of Medicinal Chemistry
and Pharmacognosy (M/C 781) College of Pharmacy, University of Illinois at Chicago, 833 S. Wood St., Chicago, Illinois 60612-7231, United States
| |
Collapse
|