1
|
Tarrá Marrugo AD. Histopathological changes of nervous tissue in women over 60 years of age with Alzheimer's disease and their relationship with menopause. REVISTA ESPANOLA DE PATOLOGIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ANATOMIA PATOLOGICA Y DE LA SOCIEDAD ESPANOLA DE CITOLOGIA 2025; 58:100800. [PMID: 39889508 DOI: 10.1016/j.patol.2024.100800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/14/2024] [Indexed: 02/03/2025]
Abstract
INTRODUCTION Ageing is a natural and irreversible process that primarily manifests in older age, becoming more common after the age of 60. Currently, a significant increase has been observed in the elderly population, with forecasts indicating that this group will triple in size over the next 50 years. This phenomenon is evident in several countries, including Japan, Mexico, Brazil, and Colombia, where the growing population of older adults is accompanied by an increased risk of neurodegenerative diseases, such as Alzheimer's disease. Studies have shown differences in the onset and progression of the disease between men and women, highlighting menopause and hormonal factors as key determinants in women. An association has been identified between a lower exposure to endogenous oestrogens and a higher risk of dementia in women, linked to the action of the enzyme β-secretase (BACE1), which is involved in the formation of amyloid aggregates associated with Alzheimer's disease. These findings highlight the importance of thoroughly investigating and understanding the impact of ageing and related diseases on the current and future population. OBJECTIVE This study aims to describe the histopathological changes in nervous tissue in women over 60 years of age with Alzheimer's disease and their relationship to menopause. METHODOLOGY A comprehensive search was conducted in databases such as PubMed, ScienceDirect, Frontiers, Scopus, and Springer. RESULTS Two hundred thirteen articles were selected for review and 45 full articles were chosen. CONCLUSIONS Alzheimer's disease is characterised by a progressive loss of cognitive function due to brain lesions, including the accumulation of amyloid-beta plaques and neuronal apoptosis. Hormonal changes during menopause may contribute to the onset of the disease.
Collapse
Affiliation(s)
- Angel David Tarrá Marrugo
- GINUMED, Corporación Universitaria Rafael Núñez, Colombia; Facultad Ciencias de la Salud, Corporación Universitaria Rafael Núñez, Campus Cartagena de Indias, Colombia.
| |
Collapse
|
2
|
Al Amin M, Dehbia Z, Nafady MH, Zehravi M, Kumar KP, Haque MA, Baig MS, Farhana A, Khan SL, Afroz T, Koula D, Tutone M, Nainu F, Ahmad I, Emran TB. Flavonoids and Alzheimer's disease: reviewing the evidence for neuroprotective potential. Mol Cell Biochem 2025; 480:43-73. [PMID: 38568359 DOI: 10.1007/s11010-023-04922-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2025]
Abstract
Neurodegeneration, which manifests as several chronic and incurable diseases, is an age-related condition that affects the central nervous system (CNS) and poses a significant threat to the public's health for the elderly. Recent decades have experienced an alarming increase in the incidence of neurodegenerative disorders (NDDs), a severe public health issue due to the ongoing development of people living in modern civilizations. Alzheimer's disease (AD) is a leading trigger of age-related dementia. Currently, there are no efficient therapeutics to delay, stop, or reverse the disease's course development. Several studies found that dietary bioactive phytochemicals, primarily flavonoids, influence the pathophysiological processes underlying AD. Flavonoids work well as a supplement to manufactured therapies for NDDs. Flavonoids are effective in complementing synthetic approaches to treat NDDs. They are biologically active phytochemicals with promising pharmacological activities, for instance, antiviral, anti-allergic, antiplatelet, anti-inflammatory, antitumor, anti-apoptotic, and antioxidant effects. The production of nitric oxide (NO), tumor necrosis factor (TNF-α), and oxidative stress (OS) are downregulated by flavonoids, which slow the course of AD. Hence, this research turned from preclinical evidence to feasible clinical applications to develop newer therapeutics, focusing on the therapeutic potential of flavonoids against AD.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Zerrouki Dehbia
- Laboratory of Agro-Biotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, 12568, Egypt
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia
| | - Kusuma Pravin Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), Govt. of N.C.T. of Delhi, Pushpvihar, New Delhi, 110017, India
| | - M Akiful Haque
- Department of Pharmaceutical Analysis, School of Pharmacy, Anurag University, Ghatkesar, Hyderabad, 500088, India
| | - Mirza Shahed Baig
- Department of Pharmaceutical Chemistry, Y. B. Chavan College of Pharmacy, Aurangabad, 431001, India
| | - Azmath Farhana
- Department of Pharmacology, School of Pharmacy, Anurag University, Hyderabad, TS, India
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, 413520, Maharashtra, India
| | - Tahmina Afroz
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Doukani Koula
- Department of Biology, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Marco Tutone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, 90123, Italy
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
3
|
Ben Ayed I, Ammar A, Aouichaoui C, Mezghani N, Salem A, Naija S, Ben Amor S, Trabelsi K, Jahrami H, Trabelsi Y, El Massioui F. Does acute aerobic exercise enhance selective attention, working memory, and problem-solving abilities in Alzheimer's patients? A sex-based comparative study. Front Sports Act Living 2024; 6:1383119. [PMID: 38903391 PMCID: PMC11187274 DOI: 10.3389/fspor.2024.1383119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/09/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction The present study aimed to evaluate the effect of acute aerobic exercise on certain cognitive functions known to be affected by Alzheimer's disease (AD), with a particular emphasis on sex differences. Methods A total of 53 patients, with a mean age of 70.54 ± 0.88 years and moderate AD, voluntarily participated in the study. Participants were randomly assigned to two groups: the experimental group (EG), which participated in a 20-min moderate-intensity cycling session (60% of the individual maximum target heart rate recorded at the end of the 6-min walk test); and the control group (CG), which participated in a 20-min reading activity. Cognitive abilities were assessed before and after the physical exercise or reading session using the Stroop test for selective attention, the forward and backward digit span test for working memory, and the Tower of Hanoi task for problem-solving abilities. Results At baseline, both groups had comparable cognitive performance (p > 0.05 in all tests). Regardless of sex, aerobic acute exercise improved attention in the Stroop test (p < 0.001), enhanced memory performance in both forward (p < 0.001) and backward (p < 0.001) conditions, and reduced the time required to solve the problem in the Tower of Hanoi task (p < 0.001). No significant differences were observed in the number of movements. In contrast, the CG did not significantly improve after the reading session for any of the cognitive tasks (p > 0.05). Consequently, the EG recorded greater performance improvements than the CG in most cognitive tasks tested (p < 0.0001) after the intervention session. Discussion These findings demonstrate that, irrespective to sex, a single aerobic exercise session on an ergocycle can improve cognitive function in patients with moderate AD. The results suggest that acute aerobic exercise enhances cognitive function similarly in both female and male patients, indicating promising directions for inclusive therapeutic strategies.
Collapse
Affiliation(s)
- Ines Ben Ayed
- Research Laboratory, Exercise Physiology and Physiopathology: from Integrated to Molecular “Biology, Medicine and Health”, LR19ES09, Faculty of Medicine of Sousse, Sousse University, Sousse, Tunisia
- Laboratory of Human and Artificial Cognition (EA 4004), Psychology UFR, University of Vincennes/Saint-Denis, Saint-Denis, France
- Research Laboratory, Education, Motricity, Sport and Health (EM2S), LR15JS01, High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia
| | - Achraf Ammar
- Department of Training and Movement Science, Institute of Sport Science, Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Laboratory, Molecular Bases of Human Pathology, LR19ES13, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia
- High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia
| | - Chirine Aouichaoui
- Research Laboratory, Exercise Physiology and Physiopathology: from Integrated to Molecular “Biology, Medicine and Health”, LR19ES09, Faculty of Medicine of Sousse, Sousse University, Sousse, Tunisia
- High Institute of Sport and Physical Education of Ksar Saïd, University of Manouba, Cité Nasr, Tunisia
| | - Nourhen Mezghani
- Department of Sport Sciences, College of Education, Taif University, Taif, Saudi Arabia
| | - Atef Salem
- Department of Training and Movement Science, Institute of Sport Science, Johannes Gutenberg-University Mainz, Mainz, Germany
- High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia
| | - Salma Naija
- Neurology Department, University Hospital Sahloul Sousse, Sousse, Tunisia
| | - Sana Ben Amor
- Neurology Department, University Hospital Sahloul Sousse, Sousse, Tunisia
| | - Khaled Trabelsi
- Research Laboratory, Education, Motricity, Sport and Health (EM2S), LR15JS01, High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia
- High Institute of Sport and Physical Education of Sfax, University of Sfax, Sfax, Tunisia
| | - Haitham Jahrami
- College of Medicine and Medical Science, Arabian Gulf University, Manama, Bahrain
| | - Yassine Trabelsi
- Research Laboratory, Exercise Physiology and Physiopathology: from Integrated to Molecular “Biology, Medicine and Health”, LR19ES09, Faculty of Medicine of Sousse, Sousse University, Sousse, Tunisia
| | - Farid El Massioui
- Laboratory of Human and Artificial Cognition (EA 4004), Psychology UFR, University of Vincennes/Saint-Denis, Saint-Denis, France
| |
Collapse
|
4
|
Islam R, Rajan R, Choudhary H, Vrionis F, Hanafy KA. Gender differences in Alzheimer's may be associated with TLR4-LYN expression in damage associated microglia and neuronal phagocytosis. J Cell Physiol 2024; 239:e30916. [PMID: 36409648 PMCID: PMC10199960 DOI: 10.1002/jcp.30916] [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: 07/20/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022]
Abstract
The role of Aβ plaques and neurofibrillary tangles in Alzheimer's disease (AD) pathogenesis have recently come into question due to failure of many pharmaceutical agents targeting these deposits and detection of these misfolded proteins in normal human brains. Therefore, we investigated correlations between microglial activation and toll like receptor 4 (TLR4) and Lck/Yes novel tyrosine (LYN) kinase signaling in an AD mouse model. In this study, we used 5-6-month-old 5XFAD and wild type (WT) male and female mice. Immunohistochemistry (IHC) and flow cytometry (FC) were performed on their brains. Cognitive performance was assessed with the Barnes-Maze. IHC showed more Ab aggregation in microglia of female 5XFAD mice compared to their male counterparts. Increased co-localization of microglial TLR4 and LYN was also observed in AD more than WT and females more than males. IHC also suggests microglial phagocytosis of neurons in AD mice, which is supported by FC data. Our FC data also support the involvement of disease associated microglia (DAMs) in this process based on cytokine secretion. Cognitive assessment by the Barnes maze showed 5XFAD females performed worse than males. In this study, we investigated the relationship between microglial TLR4 and LYN kinase in 5XFAD male and females. Our data reveals a correlation between microglial TLR4 and LYN co-localization and AD pathogenesis, more in females than males. Targeting microglial TLR4 and Lyn in DAMs may offer new therapeutic opportunities in the treatment of AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Robin Rajan
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Hadi Choudhary
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Frank Vrionis
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Khalid A. Hanafy
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| |
Collapse
|
5
|
Tsamou M, Roggen EL. Sex-associated microRNAs potentially implicated in sporadic Alzheimer's disease (sAD). Brain Res 2024; 1829:148791. [PMID: 38307153 DOI: 10.1016/j.brainres.2024.148791] [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: 09/22/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND The onset and pathology of sporadic Alzheimer's disease (sAD) seem to be affected by both sex and genetic mechanisms. Evidence supports that the high prevalence of sAD in women, worldwide, may be attributed to an interplay among aging, sex, and lifestyle, influenced by genetics, metabolic changes, and hormones. Interestingly, epigenetic mechanisms such as microRNAs (miRNAs), known as master regulators of gene expression, may contribute to this observed sexual dimorphism in sAD. OBJECTIVES To investigate the potential impact of sex-associated miRNAs on processes manifesting sAD pathology, as described by the Tau-driven Adverse Outcome Pathway (AOP) leading to memory loss. METHODS Using publicly available human miRNA datasets, sex-biased miRNAs, defined as differentially expressed by sex in tissues possibly affected by sAD pathology, were collected. In addition, sex hormone-related miRNAs were also retrieved from the literature. The compiled sex-biased and sex hormone-related miRNAs were further plugged into the dysregulated processes of the Tau-driven AOP for memory loss. RESULTS Several miRNAs, previously identified as sex-associated, were implicated in dysregulated processes associated with the manifestation of sAD pathology. Importantly, the described pathology processes were not confined to a particular sex. A mechanistic-based approach utilizing miRNAs was adopted in order to elucidate the link between sex and biological processes potentially involved in the development of memory loss. CONCLUSIONS The identification of sex-associated miRNAs involved in the early processes manifesting memory loss may shed light to the complex molecular mechanisms underlying sAD pathogenesis in a sex-specific manner.
Collapse
Affiliation(s)
- Maria Tsamou
- ToxGenSolutions (TGS), Oxfordlaan 70, 6229EV Maastricht, The Netherlands.
| | - Erwin L Roggen
- ToxGenSolutions (TGS), Oxfordlaan 70, 6229EV Maastricht, The Netherlands
| |
Collapse
|
6
|
Fang W, Qu J, Zhao W, Cao X, Liu J, Han Q, Chen D, Lv W, Xie Y, Sun Y. Monkey multi-organ cell atlas exposed to estrogen. LIFE MEDICINE 2024; 3:lnae012. [PMID: 39872660 PMCID: PMC11749546 DOI: 10.1093/lifemedi/lnae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/21/2024] [Indexed: 01/30/2025]
Abstract
Awareness of estrogen's effects on health is broadening rapidly. The effects of long-term high levels of estrogen on the body involve multiple organs. Here, we used both single-cell chromatin accessibility and RNA sequencing data to analyze the potential effect of estrogen on major organs. The integrated cell map enabled in-depth dissection and comparison of molecular dynamics, cell-type compositions, and cellular heterogeneity across multiple tissues and organs under estrogen stimulation. We also inferred pseudotime cell trajectories and cell-cell communications to uncover key molecular signatures underlying their cellular processes in major organs in response to estrogen. For example, estrogen could induce the differentiation of IFIT3 + neutrophils into S100A9 + neutrophils involved in the function of endosome-to-lysosome transport and the multivesicular body sorting pathway in liver tissues. Furthermore, through integration with human genome-wide association study data, we further identified a subset of risk genes during disease development that were induced by estrogen, such as AKT1 (related to endometrial cancer), CCND1 (related to breast cancer), HSPH1 (related to colorectal cancer), and COVID-19 and asthma-related risk genes. Our work uncovers the impact of estrogen on the major organs, constitutes a useful resource, and reveals the contribution and mechanism of estrogen to related diseases.
Collapse
Affiliation(s)
- Wen Fang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wanjun Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jinran Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Quan Han
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Wen Lv
- Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Yicheng Xie
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
7
|
Is Hormone Replacement Therapy a Risk Factor or a Therapeutic Option for Alzheimer's Disease? Int J Mol Sci 2023; 24:ijms24043205. [PMID: 36834617 PMCID: PMC9964432 DOI: 10.3390/ijms24043205] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accounts for more than half of all dementia cases in the elderly. Interestingly, the clinical manifestations of AD disproportionately affect women, comprising two thirds of all AD cases. Although the underlying mechanisms for these sex differences are not fully elucidated, evidence suggests a link between menopause and a higher risk of developing AD, highlighting the critical role of decreased estrogen levels in AD pathogenesis. The focus of this review is to evaluate clinical and observational studies in women, which have investigated the impact of estrogens on cognition or attempted to answer the prevailing question regarding the use of hormone replacement therapy (HRT) as a preventive or therapeutic option for AD. The articles were retrieved through a systematic review of the databases: OVID, SCOPUS, and PubMed (keywords "memory", "dementia," "cognition," "Alzheimer's disease", "estrogen", "estradiol", "hormone therapy" and "hormone replacement therapy" and by searching reference sections from identified studies and review articles). This review presents the relevant literature available on the topic and discusses the mechanisms, effects, and hypotheses that contribute to the conflicting findings of HRT in the prevention and treatment of age-related cognitive deficits and AD. The literature suggests that estrogens have a clear role in modulating dementia risk, with reliable evidence showing that HRT can have both a beneficial and a deleterious effect. Importantly, recommendation for the use of HRT should consider the age of initiation and baseline characteristics, such as genotype and cardiovascular health, as well as the dosage, formulation, and duration of treatment until the risk factors that modulate the effects of HRT can be more thoroughly investigated or progress in the development of alternative treatments can be made.
Collapse
|
8
|
Koemans EA, Castello JP, Rasing I, Abramson JR, Voigt S, Perosa V, van Harten TW, van Zwet EW, Terwindt GM, Gurol ME, Rosand J, Greenberg SM, van Walderveen MA, Biffi A, Viswanathan A, Wermer MJ. Sex Differences in Onset and Progression of Cerebral Amyloid Angiopathy. Stroke 2023; 54:306-314. [PMID: 36689586 PMCID: PMC9855754 DOI: 10.1161/strokeaha.122.040823] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/02/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Cerebral Amyloid Angiopathy (CAA) disease course is highly variable even in hereditary forms. Sex may be a possible modifying factor. We investigated biological sex differences in clinical disease course and magnetic resonance imaging-markers in sporadic (sCAA) and Dutch-type hereditary CAA (D-CAA). METHODS Patients with D-CAA and sCAA were included from hospital and research databases of the Leiden University Medical Center (2012-2020) and Massachusetts General Hospital (1994-2012). Key outcomes were: sex differences in symptomatic intracerebral hemorrhage (sICH) onset, recurrence and survival (analyzed using Kaplan Meier survival and regression analyses), and sex differences in magnetic resonance imaging-markers in D-CAA (explored using scatterplots), and in sCAA (investigated using regression analysis). RESULTS We included 136 patients with D-CAA (mean age 57 years, 56% women, 64% with previous sICH) and 370 patients with sCAA (mean age 76 years, 51% women, all with previous sICH). Men and women with D-CAA did not differ for sICH onset (median age 54 in men and 56 in women [P=0.13]). Men with D-CAA had a slightly higher number of sICH compared with women (median 2 versus 1; adjusted RR, 1.5 [95% CI, 1.1-1.9]) and a shorter interval between the first and second sICH (median 1.8 years for men and 3.1 years for women, P=0.02). Men with sCAA had their first sICH at an earlier age (median 75 versus 78 years, respectively, P=0.003) and more lobar microbleeds (median 1 versus 0, P=0.022) compared with women with sCAA. No substantial differences were found in the other magnetic resonance imaging markers. Survival after first sICH was comparable between sexes for D-CAA (P=0.12) and sCAA (P=0.23). CONCLUSIONS Men with CAA seem to have an earlier onset (sCAA) and more hemorrhagic disease course (sCAA and D-CAA) compared with women. Future studies are necessary to confirm these findings and determine the underlying role of sex-related factors.
Collapse
Affiliation(s)
- Emma A. Koemans
- Department of Neurology, Leiden University Medical Center, the Netherlands (E.A.K., I.R., S.V., G.M.T., M.J.H.W.)
| | - Juan Pablo Castello
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., J.R., A.B.)
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
- Department of Neurology, University of Miami Miller School of Medicine, FL (J.P.C.)
| | - Ingeborg Rasing
- Department of Neurology, Leiden University Medical Center, the Netherlands (E.A.K., I.R., S.V., G.M.T., M.J.H.W.)
| | - Jessica R. Abramson
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., J.R., A.B.)
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | - Sabine Voigt
- Department of Neurology, Leiden University Medical Center, the Netherlands (E.A.K., I.R., S.V., G.M.T., M.J.H.W.)
- Department of Radiology, Leiden University Medical Center, the Netherlands (S.V., T.W.v.H., M.A.A.v.W.)
| | - Valentina Perosa
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany (V.P.)
| | - Thijs W. van Harten
- Department of Radiology, Leiden University Medical Center, the Netherlands (S.V., T.W.v.H., M.A.A.v.W.)
| | - Erik W. van Zwet
- Department of Biomedical Data Sciences, Leiden University Medical Center, the Netherlands (E.W.v.Z.)
| | - Gisela M. Terwindt
- Department of Neurology, Leiden University Medical Center, the Netherlands (E.A.K., I.R., S.V., G.M.T., M.J.H.W.)
| | - M. Edip Gurol
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | - Jonathan Rosand
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., J.R., A.B.)
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | - Steven M. Greenberg
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | | | - Alessandro Biffi
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., J.R., A.B.)
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | - Anand Viswanathan
- Department of Neurology, J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (J.P.C., J.R.A., V.P., M.E.G., J.R., S.M.G., A.B., A.V.)
| | - Marieke J.H. Wermer
- Department of Neurology, Leiden University Medical Center, the Netherlands (E.A.K., I.R., S.V., G.M.T., M.J.H.W.)
| |
Collapse
|
9
|
Roberta de Souza Mendes Kawamura L, Ferreira Lima Mota I, Santos Vasconcelos A, Renata Mortari M. Challenges in the pharmacological treatment of patients under suspicion of chronic traumatic encephalopathy: A review. Brain Res 2023; 1799:148176. [PMID: 36503890 DOI: 10.1016/j.brainres.2022.148176] [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: 08/12/2022] [Revised: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is caused by progressive neurodegeneration associated with repetitive head impacts. This disease is more common in professionals who practice contact sports, resulting in a concussion and subconcussive trauma. CTE is characterized by the accumulation of hyperphosphorylated tau protein in neurons, astrocytes, and frontotemporal lobe degeneration. Symptoms are usually nonspecific and overlap with other neurodegenerative diseases, such as Alzheimer's disease and frontotemporal dementia, making it difficult to provide drug treatment for patients with this comorbidity. Therefore, the objective of this article is to present an updated review of the pharmacological treatment of chronic traumatic encephalopathy and its challenges.
Collapse
Affiliation(s)
| | - Isabela Ferreira Lima Mota
- Neuropharmacology Laboratory, Institute of Biological Sciences, Department of Physiological Sciences, University of Brasilia, Brazil
| | | | - Márcia Renata Mortari
- Neuropharmacology Laboratory, Institute of Biological Sciences, Department of Physiological Sciences, University of Brasilia, Brazil
| |
Collapse
|
10
|
Kumar R, Fatima F, Yadav G, Singh S, Haldar S, Alexiou A, Ashraf GM. Epigenetic Modifications by Estrogen and Androgen in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:6-17. [PMID: 35232367 DOI: 10.2174/1871527321666220225110501] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
For the development and maintenance of neuron networks in the brain, epigenetic mechanisms are necessary, as indicated by recent findings. This includes some of the high-order brain processes, such as behavior and cognitive functions. Epigenetic mechanisms could influence the pathophysiology or etiology of some neuronal diseases, altering disease susceptibility and therapy responses. Recent studies support epigenetic dysfunctions in neurodegenerative and psychiatric conditions, such as Alzheimer's disease (AD). These dysfunctions in epigenetic mechanisms also play crucial roles in the transgenerational effects of the environment on the brain and subsequently in the inheritance of pathologies. The possible role of gonadal steroids in the etiology and progression of neurodegenerative diseases, including Alzheimer's disease, has become the subject of a growing body of research over the last 20 years. Recent scientific findings suggest that epigenetic changes, driven by estrogen and androgens, play a vital role in brain functioning. Therefore, exploring the role of estrogen and androgen-based epigenetic changes in the brain is critical for the deeper understanding of AD. This review highlights the epigenetic modifications caused by these two gonadal steroids and the possible therapeutic strategies for AD.
Collapse
Affiliation(s)
- Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Faiza Fatima
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Garima Yadav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Simran Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Subhagata Haldar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, Hebersham, 2770 NSW, Australia, and AFNP Med Austria, 1010 Wien, Austria
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Yan QY, Lv JL, Shen XY, Ou-Yang XN, Yang JZ, Nie RF, Lu J, Huang YJ, Wang JY, Shen X. Patchouli alcohol as a selective estrogen receptor β agonist ameliorates AD-like pathology of APP/PS1 model mice. Acta Pharmacol Sin 2022; 43:2226-2241. [PMID: 35091686 PMCID: PMC9433381 DOI: 10.1038/s41401-021-00857-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Clinical evidence shows that postmenpausal women are almost twice as likely to develop Alzheimer's disease (AD) as men of the same age, and estrogen is closely related to the occurrence of AD. Estrogen receptor (ER) α is mainly expressed in the mammary gland and other reproductive organs like uterus while ERβ is largely distributed in the hippocampus and cardiovascular system, suggesting that ERβ selective agonist is a valuable drug against neurodegenerative diseases with low tendency in inducing cancers of breast and other reproductive organs. In this study we identified a natural product patchouli alcohol (PTA) as a selective ERβ agonist which improved the cognitive defects in female APP/PS1 mice, and explore the underlying mechanisms. Six-month-old female APP/PS1 mice were administered PTA (20, 40 mg · kg-1 · d-1, i.g.) for 90 days. We first demonstrated that PTA bound to ERβ with a dissociation constant (KD) of 288.9 ± 35.14 nM in microscale thermophoresis. Then we showed that PTA administration dose-dependently ameliorated cognitive defects evaluated in Morris water maze and Y-maze testes. Furthermore, PTA administration reduced amyloid plaque deposition in the hippocampus by promoting microglial phagocytosis; PTA administration improved synaptic integrity through enhancing BDNF/TrkB/CREB signaling, ameliorated oxidative stress by Catalase level, and regulated Bcl-2 family proteins in the hippocampus. The therapeutic effects of PTA were also observed in vitro: PTA (5, 10, 20 μM) dose-dependently increased phagocytosis of o-FAM-Aβ42 in primary microglia and BV2 cells through enhancing ERβ/TLR4 signaling; PTA treatment ameliorated o-Aβ25-35-induced reduction of synapse-related proteins VAMP2 and PSD95 in primary neurons through enhancing ERβ/BDNF/TrkB/CREB pathways; PTA treatment alleviated o-Aβ25-35-induced oxidative stress in primary neurons through targeting ERβ and increasing Catalase expression. Together, this study has addressed the efficacy of selective ERβ agonist in the amelioration of AD and highlighted the potential of PTA as a drug lead compound against the disease.
Collapse
Affiliation(s)
- Qiu-Ying Yan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian-Lu Lv
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xing-Yi Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xing-Nan Ou-Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Juan-Zhen Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui-Fang Nie
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu-Jie Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Ying Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
12
|
Luo M, Zeng Q, Jiang K, Zhao Y, Long Z, Du Y, Wang K, He G. Estrogen deficiency exacerbates learning and memory deficits associated with glucose metabolism disorder in APP/PS1 double transgenic female mice. Genes Dis 2022; 9:1315-1331. [PMID: 35873026 PMCID: PMC9293702 DOI: 10.1016/j.gendis.2021.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 11/27/2022] Open
Abstract
Alterations in glucose metabolism occur in the brain in the early stage of Alzheimer's disease (AD), and menopausal women have more severe metabolic dysfunction and are more prone to dementia than men. Although estrogen deficiency-induced changes in glucose metabolism have been previously studied in animal models, their molecular mechanisms in AD remain elusive. To investigate this issue, double transgenic (APP/PS1) female mice were subjected to bilateral ovariectomy at 3 months of age and were sacrificed 1 week, 1 month and 3 months after surgery to simulate early, middle and late postmenopause, respectively. Our analysis demonstrated that estrogen deficiency exacerbates learning and memory deficits in this mouse model of postmenopause. Estrogen deficiency impairs the function of mitochondria in glucose metabolism. It is possible that the occurrence of AD is associated with the aberrant mitochondrial ERβ-mediated IGF-1/IGF-1R/GSK-3β signaling pathway. In this study, we established a potential mechanism for the increased risk of AD in postmenopausal women and proposed a therapeutic target for AD due to postmenopause.
Collapse
Affiliation(s)
- Min Luo
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China.,Department of Pathology, Suining Municipal Hospital of TCM, Suining, Sichuan 629000, PR China
| | - Qinghua Zeng
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China.,Department of Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| | - Kai Jiang
- Department of Gastroenterology, Suining Central Hospital, Suining, Sichuan 629000, PR China
| | - Yueyang Zhao
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China.,Department of Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| | - Yexiang Du
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China
| | - Kejian Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China.,Department of Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, PR China.,Department of Anatomy, Chongqing Medical University, Chongqing 400016, PR China
| |
Collapse
|
13
|
Pinky PD, Pfitzer JC, Senfeld J, Hong H, Bhattacharya S, Suppiramaniam V, Qureshi I, Reed MN. Recent Insights on Glutamatergic Dysfunction in Alzheimer's Disease and Therapeutic Implications. Neuroscientist 2022:10738584211069897. [PMID: 35073787 DOI: 10.1177/10738584211069897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) poses a critical public health challenge, and there is an urgent need for novel treatment options. Glutamate, the principal excitatory neurotransmitter in the human brain, plays a critical role in mediating cognitive and behavioral functions; and clinical symptoms in AD patients are highly correlated with the loss of glutamatergic synapses. In this review, we highlight how dysregulated glutamatergic mechanisms can underpin cognitive and behavioral impairments and contribute to the progression of AD via complex interactions with neuronal and neural network hyperactivity, Aβ, tau, glial dysfunction, and other disease-associated factors. We focus on the tripartite synapse, where glutamatergic neurotransmission occurs, and evidence elucidating how the tripartite synapse can be pathologically altered in AD. We also discuss promising therapeutic approaches that have the potential to rescue these deficits. These emerging data support the development of novel glutamatergic drug candidates as compelling approaches for treating AD.
Collapse
Affiliation(s)
- Priyanka D Pinky
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jeremiah C Pfitzer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jared Senfeld
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacy, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA.,Center for Neuroscience, Auburn University, Auburn, AL, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA.,Center for Neuroscience, Auburn University, Auburn, AL, USA
| | | | - Miranda N Reed
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA.,Center for Neuroscience, Auburn University, Auburn, AL, USA
| |
Collapse
|
14
|
Schindler LS, Subramaniapillai S, Barth C, van der Meer D, Pedersen ML, Kaufmann T, Maximov II, Linge J, Leinhard OD, Beck D, Gurholt TP, Voldsbekk I, Suri S, Ebmeier KP, Draganski B, Andreassen OA, Westlye LT, de Lange AMG. Associations between abdominal adipose tissue, reproductive span, and brain characteristics in post-menopausal women. Neuroimage Clin 2022; 36:103239. [PMID: 36451350 PMCID: PMC9668664 DOI: 10.1016/j.nicl.2022.103239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
The menopause transition involves changes in oestrogens and adipose tissue distribution, which may influence female brain health post-menopause. Although increased central fat accumulation is linked to risk of cardiometabolic diseases, adipose tissue also serves as the primary biosynthesis site of oestrogens post-menopause. It is unclear whether different types of adipose tissue play diverging roles in female brain health post-menopause, and whether this depends on lifetime oestrogen exposure, which can have lasting effects on the brain and body even after menopause. Using the UK Biobank sample, we investigated associations between brain characteristics and visceral adipose tissue (VAT) and abdominal subcutaneous adipose tissue (ASAT) in 10,251 post-menopausal females, and assessed whether the relationships varied depending on length of reproductive span (age at menarche to age at menopause). To parse the effects of common genetic variation, we computed polygenic scores for reproductive span. The results showed that higher VAT and ASAT were both associated with higher grey and white matter brain age, and greater white matter hyperintensity load. The associations varied positively with reproductive span, indicating more prominent associations between adipose tissue and brain measures in females with a longer reproductive span. The effects were in general small, but could not be fully explained by genetic variation or relevant confounders. Our findings indicate that associations between abdominal adipose tissue and brain health post-menopause may partly depend on individual differences in cumulative oestrogen exposure during reproductive years, emphasising the complexity of neural and endocrine ageing processes in females.
Collapse
Affiliation(s)
- Louise S Schindler
- LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway.
| | - Sivaniya Subramaniapillai
- LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway
| | - Claudia Barth
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Dennis van der Meer
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; School of Mental Health and Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Mads L Pedersen
- Department of Psychology, University of Oslo, Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatry and Psychotherapy, University of Tübingen, Germany
| | - Ivan I Maximov
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Jennifer Linge
- AMRA Medical AB, Linköping, Sweden; Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Olof Dahlqvist Leinhard
- AMRA Medical AB, Linköping, Sweden; Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Dani Beck
- Department of Psychology, University of Oslo, Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tiril P Gurholt
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Irene Voldsbekk
- Department of Psychology, University of Oslo, Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Oxford, UK; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | | | - Bogdan Draganski
- LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Dept. of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Department of Psychology, University of Oslo, Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ann-Marie G de Lange
- LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Tamagno E, Guglielmotto M. Estrogens still represent an attractive therapeutic approach for Alzheimer's disease. Neural Regen Res 2022; 17:93-94. [PMID: 34100436 PMCID: PMC8451543 DOI: 10.4103/1673-5374.314295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Elena Tamagno
- Department of Neuroscience; Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| | - Michela Guglielmotto
- Department of Neuroscience; Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Torino, Italy
| |
Collapse
|
16
|
Liu H, Zhong L, Dai Q, Yang J, Zhang Y, Zhang B, Jiang Y. Zuoguiwan Ameliorates Cognitive Deficits and Neuro-Inflammation in Streptozotocin-Induced Alzheimer's Disease Rats. Neuroimmunomodulation 2022; 29:63-69. [PMID: 34320500 DOI: 10.1159/000516396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Alzheimer's disease is the most popular neurodegenerative disorder with no effective drugs to stop the progression. Zuoguiwan (ZGW), a traditional Chinese herbal medicine, has been applied in many diseases. Our study aimed to detect the function and mechanisms of ZGW in Alzheimer's disease (AD). METHODS The rat models of AD were established by streptozotocin (STZ), and the function of ZGW on cognitive dysfunction was measured with the Morris water maze test. The concentration of pro-inflammatory mediators was accessed by enzyme-linked immunosorbent assay. The relative mRNA expression of ERβ was detected by real-time quantitative PCR. RESULTS The treatment with ZGW could suppress the cognitive impairment by the findings of escape latency and time spent in the target quadrant and the increased concentration of IL-1β, IL-6, and TNF-α induced by STZ. STZ might repress the mRNA levels of ERβ, and ZGW management weakened the declined mRNA expression of ERβ. ZGW might play a protective role in AD rats against the injury of STZ on cognition and neuro-inflammation by improving the mRNA expression of ERβ. CONCLUSION The results indicated that ZGW might be a novel therapeutic strategy to slow the process of AD by modulating ERβ.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
- Postdoctoral Program, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lili Zhong
- Department of Pathology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiaomei Dai
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Yang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuwei Zhang
- Department of Physiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bo Zhang
- Department of Neurobiology, Research Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Jiang
- Department of Biochemistry, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
17
|
Villaseca P, Cisternas P, Inestrosa NC. Menopause and development of Alzheimer's disease: Roles of neural glucose metabolism and Wnt signaling. Front Endocrinol (Lausanne) 2022; 13:1021796. [PMID: 36339406 PMCID: PMC9627150 DOI: 10.3389/fendo.2022.1021796] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/28/2022] [Indexed: 11/26/2022] Open
Abstract
Late onset Alzheimer´s disease (AD) is a neurodegenerative disease with gender differences in its onset and progression, being the prevalence predominant in women and at an earlier age than in men. The pathophysiology of the menopausal condition has been associated to this dementia, playing major roles regarding both endocrine and glucose metabolism changes, amongst other mechanisms. In the current review we address the role of estrogen deficiency in the processes involved in the development of AD, including amyloid precursor protein (APP) processing to form senile plaques, Tau phosphorylation forming neurofibrillary tangles, Wnt signaling and AD neuropathology, the role of glucose brain metabolism, Wnt signaling and glucose transport in the brain, and our research contribution to these topics.
Collapse
Affiliation(s)
- Paulina Villaseca
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Pedro Cisternas
- Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Nibaldo C. Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- Centro de Envejecimiento y Regeneración (CARE UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Nibaldo C. Inestrosa,
| |
Collapse
|
18
|
Zhang S, Fan W, Hu H, Wen L, Gong M, Liu B, Hu J, Li G, Zhang D. Subcortical Volume Changes in Early Menopausal Women and Correlation With Neuropsychological Tests. Front Aging Neurosci 2021; 13:738679. [PMID: 34955807 PMCID: PMC8692945 DOI: 10.3389/fnagi.2021.738679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Background: The aging process and declining estradiol levels are two important factors that cause structural brain alterations. Many prior studies have investigated these two elements and revealed controversial results in menopausal women. Here, a cross-sectional study was designed to individually evaluate estradiol-related structural changes in the brain. Methods: A total of 45 early menopausal women and 54 age-matched premenopausal controls were enrolled and subjected to magnetic resonance imaging (MRI) scans, blood biochemistry tests, and neuropsychological tests. MRI structural images were analyzed using FreeSurfer to detect changes in subcortical and cortical volumes as well as cortical thickness. Finally, structural brain data as well as clinical and neuropsychological data were used for Pearson's correlation analyses to individually determine estradiol-related structural and functional changes in the brains of early menopausal women. Results: Compared with the premenopausal controls, the early menopausal women showed significant subcortical volumetric loss in the left amygdala and right amygdala, higher serum follicle-stimulating hormone (FSH) levels, more recognizable climacteric and depressive symptoms, decreased quality of sleep, and decreased working memory and executive functions. Simultaneously, FSH levels were related to lower working memory accuracy and longer working memory reaction time. Decreased subcortical volume in the bilateral amygdala was also related to lower working memory accuracy and longer executive reaction time in early menopausal women. Conclusion: The data suggest that estradiol deficiency in early menopausal women can lead to subcortical volume and functional brain changes, which may contribute to further understanding the neurobiological role of declined estradiol levels in early menopausal women.
Collapse
Affiliation(s)
- Si Zhang
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Weijie Fan
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Hao Hu
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Li Wen
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Mingfu Gong
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Bo Liu
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Junhao Hu
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Guanghui Li
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| | - Dong Zhang
- Department of Radiology, XinQiao Hosptial, Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
Human-Induced Pluripotent Stem Cell-Based Models for Studying Sex-Specific Differences in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:57-88. [PMID: 34921676 DOI: 10.1007/5584_2021_683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The prevalence of neurodegenerative diseases is steadily increasing worldwide, and epidemiological studies strongly suggest that many of the diseases are sex-biased. It has long been suggested that biological sex differences are crucial for neurodegenerative diseases; however, how biological sex affects disease initiation, progression, and severity is not well-understood. Sex is a critical biological variable that should be taken into account in basic research, and this review aims to highlight the utility of human-induced pluripotent stem cells (iPSC)-derived models for studying sex-specific differences in neurodegenerative diseases, with advantages and limitations. In vitro systems utilizing species-specific, renewable, and physiologically relevant cell sources can provide powerful platforms for mechanistic studies, toxicity testings, and drug discovery. Matched healthy, patient-derived, and gene-corrected human iPSCs, from both sexes, can be utilized to generate neuronal and glial cell types affected by specific neurodegenerative diseases to study sex-specific differences in two-dimensional (2D) and three-dimensional (3D) human culture systems. Such relatively simple and well-controlled systems can significantly contribute to the elucidation of molecular mechanisms underlying sex-specific differences, which can yield effective, and potentially sex-based strategies, against neurodegenerative diseases.
Collapse
|
20
|
PET imaging of brain aromatase in humans and rhesus monkeys by 11C-labeled cetrozole analogs. Sci Rep 2021; 11:23623. [PMID: 34880350 PMCID: PMC8654920 DOI: 10.1038/s41598-021-03063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/22/2021] [Indexed: 11/08/2022] Open
Abstract
Aromatase is an estrogen synthetic enzyme that plays important roles in brain functions. To quantify aromatase expression in the brain by positron emission tomography (PET), we had previously developed [11C]cetrozole, which showed high specificity and affinity. To develop more efficient PET tracer(s) for aromatase imaging, we synthesized three analogs of cetrozole. We synthesized meta-cetrozole, nitro-cetrozole, and iso-cetrozole, and prepared the corresponding 11C-labeled tracers. The inhibitory activities of these three analogs toward aromatase were evaluated using marmoset placenta, and PET imaging of brain aromatase was performed using the 11C-labeled tracers in monkeys. The most promising analog in the monkey study, iso-cetrozole, was evaluated in the human PET study. The highest to lowest inhibitory activity of the analogs toward aromatase in the microsomal fraction from marmoset placenta was in the following order: iso-cetrozole, nitro-cetrozole, cetrozole, and meta-cetrozole. This order showed good agreement with the order of the binding potential (BP) of each 11C-labeled analog to aromatase in the rhesus monkey brain. A human PET study using [11C]iso-analog showed a similar distribution pattern of binding as that of [11C]cetrozole. The time-activity curves showed that elimination of [11C]iso-cetrozole from brain tissue was faster than that of 11C-cetrozole, indicating more rapid metabolism of [11C]iso-cetrozole. [11C]Cetrozole has preferable metabolic stability for brain aromatase imaging in humans, although [11C]iso-cetrozole might also be useful to measure aromatase level in living human brain because of its high binding potential.
Collapse
|
21
|
Ceccarelli I, Bioletti L, Peparini S, Solomita E, Ricci C, Casini I, Miceli E, Aloisi AM. Estrogens and phytoestrogens in body functions. Neurosci Biobehav Rev 2021; 132:648-663. [PMID: 34890602 DOI: 10.1016/j.neubiorev.2021.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/23/2022]
Abstract
Estrogens are the hormones of reproduction in women as well as of many other important functions in the male and female body. They undergo significant changes in the different phases of life, e.g. during puberty, pregnancy or at menopause/andropause. Phytoestrogens are natural non-steroidal phenolic plant compounds that can mimic the activity of estrogens and their beneficial effects in women and in men. This narrative review summarizes the literature on the physiological role of estrogens and the several potential health benefits of phytoestrogens, with particular attention given to the possible role of phytoestrogens in aging.
Collapse
Affiliation(s)
- Ilaria Ceccarelli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Lucia Bioletti
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Sofia Peparini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Erminia Solomita
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Comasia Ricci
- Department Life Sciences, University of Siena, Siena, Italy
| | - Ilenia Casini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Elisangela Miceli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Anna Maria Aloisi
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
| |
Collapse
|
22
|
Wartchow KM, Rodrigues L, Swierzy I, Buchfelder M, de Souza DO, Gonçalves CA, Kleindienst A. Amyloid-β Processing in Aged S100B Transgenic Mice Is Sex Dependent. Int J Mol Sci 2021; 22:ijms221910823. [PMID: 34639161 PMCID: PMC8509484 DOI: 10.3390/ijms221910823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022] Open
Abstract
(1) Background: Calcium-binding protein S100B is involved in neuroregeneration but has also been associated with neurodegeneration. These contrasting effects may result from concentration or duration of exposure. We investigated the effect of long-term increased S100B levels on amyloid-β processing in one-year-old transgenic (tg) mice with 12 copies of the murine S100B gene with specific consideration of sex and specific brain regions. (2) Methods: S100B and amyloid-β 42 (Aβ42) were quantified in serum, cerebrospinal fluid (CSF), adipose tissue, and different brain regions by ELISA in wild-type (wt) and S100Btg mice (each n = 7 per group). Thioflavin T (ThT) and Aβ immunostaining were performed for visualization of Aβ deposition. (3) Results: S100B in serum, CSF, and brain was significantly increased in S100Btg mice of both sexes. Aβ42 was significantly increased in the hippocampus of male S100Btg mice (p = 0.0075), and the frontal cortex of female S100Btg mice (p = 0.0262). ThT and Aβ immunostaining demonstrated Aβ deposition in different brain regions in S100Btg mice of both sexes and female wt. (4) Conclusion: Our data validate this experimental model for studying the role of S100B in neurodegeneration and indicate that Aβ processing is sex-dependent and brain region-specific, which deserves further investigation of signaling pathways and behavioral responses.
Collapse
Affiliation(s)
- Krista Minéia Wartchow
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil; (K.M.W.); (L.R.); (D.O.d.S.); (C.-A.G.)
- Department of Neurosurgery, Friedrich-Alexander University, 91054 Erlangen, Germany; (I.S.); (M.B.)
| | - Leticia Rodrigues
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil; (K.M.W.); (L.R.); (D.O.d.S.); (C.-A.G.)
- Department of Neurosurgery, Friedrich-Alexander University, 91054 Erlangen, Germany; (I.S.); (M.B.)
| | - Izabela Swierzy
- Department of Neurosurgery, Friedrich-Alexander University, 91054 Erlangen, Germany; (I.S.); (M.B.)
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University, 91054 Erlangen, Germany; (I.S.); (M.B.)
| | - Diogo Onofre de Souza
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil; (K.M.W.); (L.R.); (D.O.d.S.); (C.-A.G.)
| | - Carlos-Alberto Gonçalves
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil; (K.M.W.); (L.R.); (D.O.d.S.); (C.-A.G.)
| | - Andrea Kleindienst
- Department of Neurosurgery, Friedrich-Alexander University, 91054 Erlangen, Germany; (I.S.); (M.B.)
- Correspondence:
| |
Collapse
|
23
|
Guglielmotto M, Manassero G, Vasciaveo V, Venezia M, Tabaton M, Tamagno E. Estrogens Inhibit Amyloid-β-Mediated Paired Helical Filament-Like Conformation of Tau Through Antioxidant Activity and miRNA 218 Regulation in hTau Mice. J Alzheimers Dis 2021; 77:1339-1351. [PMID: 32804095 DOI: 10.3233/jad-200707] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The risk of developing Alzheimer's disease as well as its progression and severity are known to be different in men and women, and cognitive decline is greater in women than in men at the same stage of disease and could be correlated at least in part on estradiol levels. OBJECTIVE In our work we found that biological sex influences the effect of amyloid-β42 (Aβ42) monomers on pathological tau conformational change. METHODS In this study we used transgenic mice expressing the wild-type human tau (hTau) which were subjected to intraventricular (ICV) injections of Aβ peptides in nanomolar concentration. RESULTS We found that Aβ42 produces pathological conformational changes and hyperphosphorylation of tau protein in male or ovariectomized female mice but not in control females. The treatment of ovariectomized females with estradiol replacement protects against the pathological conformation of tau and seems to be mediated by antioxidant activity as well as the ability to modulate the expression of miRNA 218 linked to tau phosphorylation. CONCLUSION Our study indicates that factors as age, reproductive stage, hormone levels, and the interplay with other risk factors should be considered in women, in order to identify the best appropriate therapeutic approach in prevention of cognitive impairment.
Collapse
Affiliation(s)
- Michela Guglielmotto
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Orbassano, Torino, Italy
| | - Giusi Manassero
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Orbassano, Torino, Italy
| | - Valeria Vasciaveo
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Orbassano, Torino, Italy
| | - Marika Venezia
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Orbassano, Torino, Italy
| | - Massimo Tabaton
- Unit of Geriatric Medicine, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
| | - Elena Tamagno
- Department of Neuroscience, University of Torino, Torino, Italy.,Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Orbassano, Torino, Italy
| |
Collapse
|
24
|
Zhao W, Hou Y, Song X, Wang L, Zhang F, Zhang H, Yu H, Zhou Y. Estrogen Deficiency Induces Mitochondrial Damage Prior to Emergence of Cognitive Deficits in a Postmenopausal Mouse Model. Front Aging Neurosci 2021; 13:713819. [PMID: 34335235 PMCID: PMC8319728 DOI: 10.3389/fnagi.2021.713819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Estrogen deficiency contributes to the development of Alzheimer's disease (AD) in menopausal women. In the current study, we examined the impact of estrogen deficiency on mitochondrial function and cognition using a postmenopausal mouse model. Methods: Bilateral ovariectomy was conducted in adult females C57BL/6J. Cognitive function was examined using the Morris water maze (MWM) test at 2 weeks, 1, 2, and 3 months after ovariectomy. Neurodegeneration was assessed using an immunofluorescence assay of microtubule-associated protein 2 (MAP2) in the hippocampus and immunoblotting against postsynaptic density-95 (PSD95). Mitochondrial function in the hippocampus was assessed using immunoblotting for NDUFB8, SDHB, UQCRC2, MTCO1, and ATP5A1. Mitochondrial biogenesis was examined using immunoblotting for PGC-1α, NRF1, and mtTFA. Mitochondrion fission was assessed with immunoblotting for Drp1, whereas mitochondrion fusion was analyzed with immunoblotting for OPA1 and Mfn2. Mitophagy was examined with immunoblotting for PINK1 and LC3B. Mice receiving sham surgery were used as controls. Results: Ovariectomy resulted in significant learning and memory deficits in the MWM test at 3 months, but not at any earlier time points. At 2 weeks after ovariectomy, levels of Drp1 phosphorylated at Ser637 decreased in the hippocampus. At 1 month after ovariectomy, hippocampal levels of NDUFB8, SDHB, PGC-1α, mtTFA, OPA1, and Mfn2 were significantly reduced. At 2 months after ovariectomy, hippocampal levels of MAP2, PSD95, MTCO1, NRF1, and Pink1 were also reduced. At 3 months, levels of LC3B-II were reduced. Conclusions: The cognitive decline associated with estrogen deficiency is preceded by mitochondrial dysfunction, abnormal mitochondrial biogenesis, irregular mitochondrial dynamics, and decreased mitophagy. Thus, mitochondrial damage may contribute to cognitive impairment associated with estrogen deficiency.
Collapse
Affiliation(s)
- Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yue Hou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Fangfang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Hanting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
25
|
Sindi S, Kåreholt I, Ngandu T, Rosenberg A, Kulmala J, Johansson L, Wetterberg H, Skoog J, Sjöberg L, Wang H, Fratiglioni L, Skoog I, Kivipelto M. Sex differences in dementia and response to a lifestyle intervention: Evidence from Nordic population-based studies and a prevention trial. Alzheimers Dement 2021; 17:1166-1178. [PMID: 34255432 PMCID: PMC8361986 DOI: 10.1002/alz.12279] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Evidence on sex differences in the risk for dementia has been mixed. The goal was to assess sex differences in the development of dementia, and in the effects of a lifestyle intervention. METHODS Two strategies were adopted, one using combined data from three large Nordic population-based cohort studies (n = 2289), adopting dementia as outcome, and 2-year multidomain lifestyle intervention (n = 1260), adopting cognitive change as outcome. RESULTS There was higher risk for dementia after age 80 years in women. The positive effects of the lifestyle intervention on cognition did not significantly differ between men and women. Sex-specific analyses suggested that different vascular, lifestyle, and psychosocial risk factors are important for women and men in mid- and late-life. CONCLUSION Women had higher risk for dementia among the oldest individuals. Lifestyle interventions may be effectively implemented among older men and women.
Collapse
Affiliation(s)
- Shireen Sindi
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Ageing Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
| | - Ingemar Kåreholt
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Institute of GerontologySchool of Health and WelfareAging Research Network – Jönköping (ARN‐J)Jönköping UniversityJönköpingSweden
| | - Tiia Ngandu
- Public Health Promotion UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Anna Rosenberg
- Institute of Public Health and Clinical Nutrition and Institute of Clinical MedicineNeurologyUniversity of Eastern FinlandKuopioFinland
| | - Jenni Kulmala
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Public Health Promotion UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Lena Johansson
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Hanna Wetterberg
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Johan Skoog
- Department of PsychologyCenter for Health and Ageing (AGECAP)University of GothenburgGothenburgSweden
| | - Linnea Sjöberg
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
| | - Hui‐Xin Wang
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Stress Research InstituteStockholom UniversityStockholmSweden
| | - Laura Fratiglioni
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Stockholm Gerontology Research CenterStockholmSweden
| | - Ingmar Skoog
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Miia Kivipelto
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Ageing Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
- Institute of Public Health and Clinical Nutrition and Institute of Clinical MedicineNeurologyUniversity of Eastern FinlandKuopioFinland
- Theme AgingKarolinska University HospitalStockholmSweden
| |
Collapse
|
26
|
Han M, Chang J, Choi S, Cho Y, Lee G, Park SM. Association of tibolone and dementia risk: a cohort study using Korean claims data. Gynecol Endocrinol 2021; 37:567-571. [PMID: 33150798 DOI: 10.1080/09513590.2020.1843624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Few studies have examined whether tibolone (TIB), a type of hormone replacement therapy widely used in Asia and Europe, affects dementia risk in postmenopausal women. Our study aims to investigate the association of TIB and dementia risk in Korean women aged 50-80 years. METHODS A population-based longitudinal study was conducted using the Korean National Health Insurance Service claims database merged with national health examination data from 2002 to 2015. Among 13,110 participants, exposure to TIB was determined using the standardized defined daily dose (DDD) system from 2003 to 2007. Starting from 2007, participants were followed up for overall dementia, Alzheimer's disease (AD) and vascular dementia (VD) until 2015. Cox proportional hazards regression was used to determine the adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs) of dementia according to TIB use. RESULTS TIB use was not significantly associated with the risk of total dementia (aHR = 1.040; 95% CI = 0.734-1.472; p = .827), AD (aHR = 0.949; 95% CI = 0.652-1.381; p = .785) and VD (aHR = 1.245; 95% CI = 0.631-2.457; p = .528). CONCLUSIONS Our results suggest that TIB use does not have a significant association with dementia risk. Further randomized controlled trials are necessary to elucidate the role of exogenous hormones in the development of dementia.
Collapse
Affiliation(s)
- Minjung Han
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jooyoung Chang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Seulggie Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Yoosun Cho
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Gyeongsil Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Min Park
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| |
Collapse
|
27
|
Lan Z, Meng Z, Lian B, Liu M, Sun T, Sun H, Liu Z, Hu Z, Guo Q, Zhang J. Hippocampal Aromatase Knockdown Aggravates Ovariectomy-Induced Spatial Memory Impairment, Aβ Accumulation and Neural Plasticity Deficiency in Adult Female Mice. Neurochem Res 2021; 46:1188-1202. [PMID: 33559105 DOI: 10.1007/s11064-021-03258-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 12/23/2022]
Abstract
Ovarian estrogens (mainly 17β estradiol, E2) have been involved in the regulation of the structure of hippocampus, the center of spatial memory. In recent years, high levels of aromatase (AROM), the estrogen synthase, has been localized in hippocampus; and this hippocampus-derived E2 seems to be functional in synaptic plasticity and spatial memory as ovarian E2 does. However, the contribution of ovarian E2 and hippocampal E2 to spatial memory and neural plasticity remains unclear. In this study, AROM-specific RNA interference AAVs (shAROM) were constructed and injected into the hippocampus of control or ovariectomized (OVX) mice. Four weeks later the spatial learning and memory behavior was examined with Morris water maze, the expression of hippocampal Aβ related proteins, selected synaptic proteins and CA1 synapse density, actin polymerization related proteins and CA1 spine density were also examined. The results showed that while OVX and hippocampal shAROM contributed similarly to most of the parameters examined, shAROM induced more increase in BACE1 (amyloidogenic β-secretase), more decrease in neprilysin (Aβ remover) and Profilin-1 (actin polymerization inducer). More importantly, combined OVX and shAROM treatment displayed most significant impairment of spatial learning and memory as well as decrease in synaptic plasticity compared to OVX or shAROM alone. In conclusion, the above results clearly demonstrated the crucial role of hippocampal E2 in the regulation of the structure and function of hippocampus besides ovarian E2, indicating that hippocampal E2 content should also be taken into consideration during estrogenic replacement.
Collapse
Affiliation(s)
- Zhen Lan
- Department of Neurobiology, Army Medical University, Chongqing, China
| | - Zhaoyou Meng
- Department of Neurobiology, Army Medical University, Chongqing, China
| | - Biyao Lian
- Department of Neurobiology, Army Medical University, Chongqing, China
- Department of Pediatrics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mengying Liu
- Department of Neurobiology, Army Medical University, Chongqing, China
- The 305 Hospital of PLA, Beijing, China
| | - Tao Sun
- Department of Neurobiology, Army Medical University, Chongqing, China
- The 63650 Hospital of PLA, Malan, China
| | - Huan Sun
- Department of Neurobiology, Army Medical University, Chongqing, China
- Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhi Liu
- Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Zhenxin Hu
- Battalion One of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Qiang Guo
- Department of Basic Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing, China.
| |
Collapse
|
28
|
Dean B, Gogos A. The impact of ovariectomy and chronic estrogen treatment on gene expression in the rat cortex: Implications for psychiatric disorders. Psychoneuroendocrinology 2021; 127:105192. [PMID: 33730612 DOI: 10.1016/j.psyneuen.2021.105192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023]
Abstract
Estrogens, via estrogen-mediated changes in CNS function, have been suggested to be beneficial in the treatment of several psychiatric disorders. Few studies have used transcriptomic technologies to determine the effect of estrogen on gene expression in the CNS. Thus, we aimed to examine the impact of ovariectomy (the removal of all ovarian hormones) and estrogen replacement on rat frontal cortical gene expression. We used the Agilent SurePrint G3 Gene Expression Rat Array to measure levels of RNA in intact (cycling) female rats and in ovariectomized rats that were, or were not, given 17β-estradiol in implants for 4 weeks. Compared to untreated ovariectomized rats, intact rats (effect of ovarian hormones; comparison 1) and rats receiving 17β-estradiol replacement (estrogen-specific effects; comparison 2) showed significant changes in cortical gene expression (58 and 36 genes, respectively). These changes in gene expression would be expected to affect pathways that regulate neurotransmitters, glutathione and sphingolipids; pathways known to be implicated in the pathophysiologies of psychiatric disorders. When we compared the levels of gene expression in the two comparisons that had a significance of p < 0.01 independent of magnitude of change, there was a strong correlation between fold changes in gene expression for 127 genes. We posit that this correlation is due to the level of expression of these genes being strongly influenced by both cycling and replacement estrogen. Further exploration of ovarian hormone- and estrogen-sensitive gene expression may provide new insight into the aetiology of aspects of psychiatric disorders that show sex differences.
Collapse
Affiliation(s)
- Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
29
|
Abstract
OBJECTIVE The impact of sex hormones milieu on women's cognitive performance at different reproductive stages has caused increased caution. Our research aims to explore whether parity is negatively correlated with cognitive function. METHODS There were 1,093 postmenopausal participants recruited from the Health and Nutrition Examination Survey dataset. Cognitive functioning was evaluated by digit symbol substitution test (DSST). We performed log transformation to normalize the distributions of the DSST values. RESULTS Participants were categorized into tertile groups based on the number of pregnancies. Using the zero to one pregnancy group as the reference, there was a reduced DSST scores with β values of -0.13 (95% confidence interval [CI] -0.23 to -0.03; P = 0.008) in the ≥5 pregnancies group after adjusting for socioeconomic, medical disease, lifestyle, and reproductive components. Moreover, women who had their last pregnancy after 28 years old and education less than 12 years also was correlate with cognitive malfunction after adjusting relevant covariates (both P < 0.001). CONCLUSIONS Women with at least five pregnancies had poorer cognitive performance. Last pregnancy after 28 years old and education less than 12 years also was associated with poorer DSST scores. VIDEO SUMMARY:: http://links.lww.com/MENO/A634.
Collapse
|
30
|
Pan Y, Zhang Y, Liu N, Lu W, Yang J, Li Y, Liu Z, Wei Y, Lou Y, Kong J. Vitamin D Attenuates Alzheimer-like Pathology Induced by Okadaic Acid. ACS Chem Neurosci 2021; 12:1343-1350. [PMID: 33818056 DOI: 10.1021/acschemneuro.0c00812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many elderly individuals suffer from Alzheimer's disease (AD), which causes a growing concern. We investigated the mechanism underlying the effects of vitamin D (VD) as a prophylactic treatment. A mouse model of okadaic-acid-induced AD-like pathology was used in vivo and in vitro. Morris water maze and field trials were used to assess cognitive function. The expression levels of VDR, MTHFR, LCMT-1, PP2A, p-TAU (Thr396), and T-TAU and the methylation level of PP2A were measured by Western blotting, and a reversal of the increase in the levels of these proteins in an AD cell model was observed. We used MTHFR-knockdown SH-SY5Y cells to further test the effects of VD, treated these cells with cycloheximide and MG132, and used RT-PCR to explore the mechanism underlying MTHFR targeting. We found that the effects of VD on AD were impaired by MTHFR knockdown through a pretranscriptional mechanism. In addition, VD attenuated AD-induced cognitive impairment and significantly suppressed the expression of TAU. Our findings indicated that VD treatment alleviated TAU accumulation and rescued methylated PP2A by increasing the expression of LCMT-1 and MTHFR.
Collapse
Affiliation(s)
- Yiming Pan
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yalin Zhang
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ning Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Wanyi Lu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jingxin Yang
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ye Li
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zuwang Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yinghong Wei
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yan Lou
- Department of Computer Science, China Medical University, Shenyang 110013, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
31
|
Kauffman RP, Young C, Castracane VD. Perils of prolonged ovarian suppression and hypoestrogenism in the treatment of breast cancer: Is the risk of treatment worse than the risk of recurrence? Mol Cell Endocrinol 2021; 525:111181. [PMID: 33529690 DOI: 10.1016/j.mce.2021.111181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal breast cancer is usually estrogen receptor positive, and hence, prolonged ovarian suppression by medical or surgical means to prevent recurrence has become standard of management to improve disease-free survival. Ten-year adjuvant tamoxifen therapy is associated with 3.5% fewer recurrences compared to five years. The SOFT trial demonstrated small but statistically significant incremental improvements in long-term disease-free survival by the addition of gonadotropin-releasing hormone analog treatment (triptorelin) to an aromatase inhibitor (exemestane). Profound hypoestrogenism in the premenopausal age group may not be well tolerated due to a host of bothersome side effects (primarily vasomotor symptoms, musculoskeletal complaints, genitourinary syndrome of menopause, and mood disorders). Prolonged hypoestrogenism in younger women is associated with premature development of cardiovascular disease, bone loss, cognitive decline, and all-cause mortality. This paper explores multi-system consequences of prolonged hypoestrogenism in premenopausal women derived from studies of women with and without breast cancer. Pretreatment counseling in estrogen receptor positive breast cancer should emphasize the benefit of prolonged estrogen suppression on breast cancer recurrence and established risks of lifelong hypoestrogenism on quality of life and all-cause mortality. Future genomic research may help identify the best candidates for extended ovarian suppression to avoid treating many women when only a minority benefit.
Collapse
Affiliation(s)
- Robert P Kauffman
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA.
| | - Christina Young
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| | - V Daniel Castracane
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| |
Collapse
|
32
|
Means JC, Lopez AA, Koulen P. Estrogen Protects Optic Nerve Head Astrocytes Against Oxidative Stress by Preventing Caspase-3 Activation, Tau Dephosphorylation at Ser 422 and the Formation of Tau Protein Aggregates. Cell Mol Neurobiol 2021; 41:449-458. [PMID: 32385548 PMCID: PMC7648721 DOI: 10.1007/s10571-020-00859-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
Glaucoma is a neurodegenerative disorder that leads to the slow degeneration of retinal ganglion cells, and results in damage to the optic nerve and concomitant vision loss. As in other disorders affecting the viability of central nervous system neurons, neurons affected by glaucoma do not have the ability to regenerate after injury. Recent studies indicate a critical role for optic nerve head astrocytes (ONHAs) in this process of retinal ganglion cell degeneration. Cleavage of tau, a microtubule stabilizing protein and constituent of neurofibrillary tangles (NFT), plays a major part in the mechanisms that lead to toxicity in CNS neurons and astrocytes. Here, we tested the hypothesis that estrogen, a pleiotropic neuro- and cytoprotectant with high efficacy in the CNS, prevents tau cleavage, and hence, protects ONHAs against cell damage caused by oxidative stress. Our results indicate that estrogen prevents caspase-3 mediated tau cleavage, and thereby decreases the levels of the resulting form of proteolytically cleaved tau protein, which leads to a decrease in NFT formation, which requires proteolytically cleaved tau protein. Overall, our data propose that by stopping the reduction of estrogen levels involved with aging the sensitivity of the optic nerve to glaucomatous damage might be reduced. Furthermore, our data suggest that therapeutic use of estrogen may be beneficial in slowing or preventing the onset or severity of neurodegenerative diseases such as glaucoma and potentially also other degenerative diseases of the CNS through direct control of posttranslational modifications of tau protein.
Collapse
Affiliation(s)
- John C Means
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA
| | - Adam A Lopez
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA
| | - Peter Koulen
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA.
- Department of Biomedical Sciences, School of Medicine, University of Missouri - Kansas City, 2411 Holmes St., Kansas City, MO, USA.
| |
Collapse
|
33
|
Noori T, Dehpour AR, Sureda A, Sobarzo-Sanchez E, Shirooie S. Role of natural products for the treatment of Alzheimer's disease. Eur J Pharmacol 2021; 898:173974. [PMID: 33652057 DOI: 10.1016/j.ejphar.2021.173974] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Negative psychological and physiological consequences of neurodegenerative disorders represent a high social and health cost. Among the neurodegenerative disorders Alzheimer's disease (AD) is recognized as a leading neurodegenerative condition and a primary cause of dementia in the elderlys. AD is considered as neurodegenerative disorder that progressively impairs cognitive function and memory. According to current epidemiological data, about 50 milLion people worldwide are suffering from AD. The primary symptoms of AD are almost inappreciable and usually comprise forgetfulness of recent events. Numerous processes are involved in the development of AD, for example oxidative stress (OS) mainly due to mitochondrial dysfunction, intracellular the accumulation of hyperphosphorylated tau (τ) proteins in the form of neurofibrillary tangles, excessive the accumulation of extracellular plaques of beta-amyloid (Aβ), genetic and environmental factors. Running treatments only attenuate symptoms and temporarily reduce the rate of cognitive progression associated with AD. This means that most treatments focus only on controlLing symptoms, particularly in the initial stages of the disease. In the past, the first choice of treatment was based on natural ingredients. In this sense, diverse natural products (NPs) are capable to decrease the symptoms and alleviate the development of several diseases including AD attracting the attention of the scientific community and the pharmaceutical industry. Specifically, numerous NPs including flavonoids, gingerols, tannins, anthocyanins, triterpenes and alkaloids have been shown anti-inflammatory, antioxidant, anti-amyloidogenic, and anti-choLinesterase properties. This review provide a summary of the pathogenesis and the therapeutic goals of AD. It also discusses the available data on various plants and isolated natural compounds used to prevent and diminish the symptoms of AD.
Collapse
Affiliation(s)
- Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, TUMS, Tehran, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX), University Research Institute of Health Sciences (IUNICS), and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile; Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Spain
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
34
|
Predicting the Potency of Anti-Alzheimer’s Drug Combinations Using Machine Learning. Processes (Basel) 2021. [DOI: 10.3390/pr9020264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Clinical trials of single drugs intended to slow the progression of Alzheimer’s Disease (AD) have been notoriously unsuccessful. Combinations of repurposed drugs could provide effective treatments for AD. The challenge is to identify potentially effective combinations. To meet this challenge, machine learning (ML) was used to extract the knowledge from two leading AD databases, and then “the machine” predicted which combinations of the drugs in common between the two databases would be the most effective as treatments for AD. Specifically, three-layered artificial neural networks (ANNs) with compound, gated units in their internal layer were trained using ML to predict the cognitive scores of participants, separately in either database, given other data fields including age, demographic variables, comorbidities, and drugs taken. The predictions from the separately trained ANNs were statistically highly significantly correlated. The best drug combinations, jointly determined from both sets of predictions, were high in nonsteroidal anti-inflammatory drugs; anticoagulant, lipid-lowering, and antihypertensive drugs; and female hormones. The results suggest that the neurodegenerative processes that underlie AD and other dementias could be effectively treated using a combination of repurposed drugs. Predicted drug combinations could be evaluated in clinical trials.
Collapse
|
35
|
Shen Y, Zhang B, Pang X, Yang R, Chen M, Zhao J, Wang J, Wang Z, Yu Z, Wang Y, Li L, Liu A, Du G. Network Pharmacology-Based Analysis of Xiao-Xu-Ming Decoction on the Treatment of Alzheimer's Disease. Front Pharmacol 2021; 11:595254. [PMID: 33390981 PMCID: PMC7774966 DOI: 10.3389/fphar.2020.595254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/21/2020] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) has become a worldwide disease that is harmful to human health and brings a heavy economic burden to healthcare system. Xiao-Xu-Ming Decoction (XXMD) has been widely used to treat stroke and other neurological diseases for more than 1000 years in China. However, the synergistic mechanism of the constituents in XXMD for the potential treatment of AD is still unclear. Therefore, the present study aimed to predict the potential targets and uncover the material basis of XXMD for the potential treatment of AD. A network pharmacology-based method, which combined data collection, drug-likeness filtering and absorption, distribution, metabolism, excretion and toxicity (ADME/T) properties filtering, target prediction and network analysis, was used to decipher the effect and potential targets of XXMD for the treatment of AD. Then, the acetylcholinesterase (AChE) inhibitory assay was used to screen the potential active constituents in XXMD for the treatment of AD, and the molecular docking was furtherly used to identify the binding ability of active constituents with AD-related target of AChE. Finally, three in vitro cell models were applied to evaluate the neuroprotective effects of potential lead compounds in XXMD. Through the China Natural Products Database, Traditional Chinese Medicine Systems Pharmacology (TCMSP) Database, Traditional Chinese Medicine (TCM)-Database @Taiwan and literature, a total of 1481 compounds in XXMD were finally collected. After ADME/T properties filtering, 908 compounds were used for the further study. Based on the prediction data, the constituents in XXMD formula could interact with 41 AD-related targets. Among them, cyclooxygenase-2 (COX-2), estrogen receptor α (ERα) and AChE were the major targets. The constituents in XXMD were found to have the potential to treat AD through multiple AD-related targets. 62 constituents in it were found to interact with more than or equal to 10 AD-related targets. The prediction results were further validated by in vitro biology experiment, resulting in several potential anti-AD multitarget-directed ligands (MTDLs), including two AChE inhibitors with the IC50 values ranging from 4.83 to 10.22 μM. Moreover, fanchinoline was furtherly found to prevent SH-SY5Y cells from the cytotoxicities induced by sodium nitroprusside, sodium dithionate and potassium chloride. In conclusion, XXMD was found to have the potential to treat AD by targeting multiple AD-related targets and canonical pathways. Fangchinoline and dauricine might be the potential lead compounds in XXMD for the treatment of AD.
Collapse
Affiliation(s)
- Yanjia Shen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyue Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaocong Pang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ran Yang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao Chen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaying Zhao
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziru Yu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehua Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ailin Liu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanhua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
36
|
Subramaniapillai S, Almey A, Natasha Rajah M, Einstein G. Sex and gender differences in cognitive and brain reserve: Implications for Alzheimer's disease in women. Front Neuroendocrinol 2021; 60:100879. [PMID: 33137359 DOI: 10.1016/j.yfrne.2020.100879] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022]
Abstract
Women represent ⅔ of the cases of Alzheimer's disease (AD). Current research has focused on differential risks to explain higher rates of AD in women. However, factors that reduce risk for AD, like cognitive/brain reserve, are less well explored. We asked: what is known about sex and gender differences in how reserve mitigates risk for AD? We conducted a narrative review of the literature, with keywords: "sex/gender differences", "cognitive/brain reserve", "Alzheimer's Disease", and the following cognitive reserve contributors: "education", "IQ", "occupation", "cognitive stimulation", "bilingualism", "socioeconomic status", "physical activity", "social support". Sixteen papers disaggregated their data by sex. Those papers observed sex and gender differences in reserve contributors. There is also evidence that greater reserve may be more beneficial in lowering AD risk in women, although more research is needed. We discuss how traditional reserve contributors are gendered and may not capture factors that support cognition in aging women.
Collapse
Affiliation(s)
- Sivaniya Subramaniapillai
- Department of Psychology, McGill University, 2001 Avenue McGill College, Montréal, QC H3A 1G1, Canada; Brain Imaging Centre, Douglas Institute Research Centre, 6875 LaSalle Blvd Verdun, Montréal, QC H4H 1R3, Canada.
| | - Anne Almey
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada
| | - M Natasha Rajah
- Brain Imaging Centre, Douglas Institute Research Centre, 6875 LaSalle Blvd Verdun, Montréal, QC H4H 1R3, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, 1033 Avenue des Pins, Montréal, QC H3A 1A1, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada; Rotman Research Institute, Baycrest Hospital, 3560 Bathurst St, Toronto, ON M6A 2E1, Canada; Tema Genus, Linköping University, TEMA-huset, Entrance 37, Room E433, Campus Valla, Linköping, Sweden
| |
Collapse
|
37
|
Boyle CP, Raji CA, Erickson KI, Lopez OL, Becker JT, Gach HM, Kuller LH, Longstreth W, Carmichael OT, Riedel BC, Thompson PM. Estrogen, brain structure, and cognition in postmenopausal women. Hum Brain Mapp 2021; 42:24-35. [PMID: 32910516 PMCID: PMC7721237 DOI: 10.1002/hbm.25200] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/28/2020] [Accepted: 08/02/2020] [Indexed: 12/17/2022] Open
Abstract
Declining estrogen levels before, during, and after menopause can affect memory and risk for Alzheimer's disease. Undesirable side effects of hormone variations emphasize a role for hormone therapy (HT) where possible benefits include a delay in the onset of dementia-yet findings are inconsistent. Effects of HT may be mediated by estrogen receptors found throughout the brain. Effects may also depend on lifestyle factors, timing of use, and genetic risk. We studied the impact of self-reported HT use on brain volume in 562 elderly women (71-94 years) with mixed cognitive status while adjusting for aforementioned factors. Covariate-adjusted voxelwise linear regression analyses using a model with 16 predictors showed HT use as positively associated with regional brain volumes, regardless of cognitive status. Examinations of other factors related to menopause, oophorectomy and hysterectomy status independently yielded positive effects on brain volume when added to our model. One interaction term, HTxBMI, out of several examined, revealed significant negative association with overall brain volume, suggesting a greater reduction in brain volume than BMI alone. Our main findings relating HT to regional brain volume were as hypothesized, but some exploratory analyses were not in line with existing hypotheses. Studies suggest lower levels of estrogen resulting from oophorectomy and hysterectomy affect brain volume negatively, and the addition of HT modifies the relation between BMI and brain volume positively. Effects of HT may depend on the age range assessed, motivating studies with a wider age range as well as a randomized design.
Collapse
Affiliation(s)
- Christina P. Boyle
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Cyrus A. Raji
- Mallinckrodt Institute of RadiologyWashington UniversitySt. LouisMissouriUSA
| | - Kirk I. Erickson
- Department of PsychologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Oscar L. Lopez
- Department of NeurologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - James T. Becker
- Department of PsychologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
- Department of PsychiatryUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - H. Michael Gach
- Departments of Radiation Oncology, Radiology, and Biomedical EngineeringWashington UniversitySt. LouisMissouriUSA
| | - Lewis H. Kuller
- Department of EpidemiologyUniversity of Pittsburgh, Graduate School of Public HealthPittsburghPennsylvaniaUSA
| | - William Longstreth
- Departments of Neurology and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | | | - Brandalyn C. Riedel
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
| |
Collapse
|
38
|
Di Zhang D, Wang YG, Liu CY, Wang ZH, Wang YF. Aminoguanidine ameliorates ovariectomy-induced neuronal deficits in rats by inhibiting AGE-mediated Aβ production. Acta Neurobiol Exp (Wars) 2021; 81:10-20. [PMID: 33949165 DOI: 10.21307/ane-2021-002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 10/08/2020] [Indexed: 11/11/2022]
Abstract
Advanced glycation end products (AGEs) have been reported to cause neurodegeneration, senile plaque formation and spatial learning and memory deficits. There is much evidence describing the beneficial effects of aminoguanidine (AG) on the central nervous system; AG is able to inhibit the receptor for AGEs and beta-amyloid (Aβ) deposition in the brain, thus preventing cognitive decline and neurodegeneration. In this study, we investigated whether AG protects against ovariectomy-induced neuronal deficits and Aβ deposition in rats. Animals in the ovariectomy group (OVX) group, and those in the OVX+AG group were treated with AG (100 mg/kg/day) for 8 weeks. Learning and memory were evaluated using the electric Y maze. AGE and Aβ1-40 biochemical assessments were performed using enzyme-linked immunosorbent assay (ELISA) kits. Furthermore, evaluations of brain amyloid precursor protein 695 (APP695) mRNA expression by RT-PCR and AGE expression by immunohistochemistry were carried out. Ovariectomized rats exhibited memory impairment and Aβ production disorder with upregulated APP695 mRNA and AGE expression levels. AG pretreatment relieved the ovariectomy-induced learning and memory disorder and significantly ameliorated the Aβ production disturbance and AGE generation. Additionally, pathological changes in morphology were also significantly recovered. Our data reveal that AG plays a potentially neuroprotective role against ovariectomy-induced learning and cognitive impairment and Aβ production disorder. Advanced glycation end products (AGEs) have been reported to cause neurodegeneration, senile plaque formation and spatial learning and memory deficits. There is much evidence describing the beneficial effects of aminoguanidine (AG) on the central nervous system; AG is able to inhibit the receptor for AGEs and beta-amyloid (Aβ) deposition in the brain, thus preventing cognitive decline and neurodegeneration. In this study, we investigated whether AG protects against ovariectomy-induced neuronal deficits and Aβ deposition in rats. Animals in the ovariectomy group (OVX) group, and those in the OVX+AG group were treated with AG (100 mg/kg/day) for 8 weeks. Learning and memory were evaluated using the electric Y maze. AGE and Aβ1-40 biochemical assessments were performed using enzyme-linked immunosorbent assay (ELISA) kits. Furthermore, evaluations of brain amyloid precursor protein 695 (APP695) mRNA expression by RT-PCR and AGE expression by immunohistochemistry were carried out. Ovariectomized rats exhibited memory impairment and Aβ production disorder with upregulated APP695 mRNA and AGE expression levels. AG pretreatment relieved the ovariectomy-induced learning and memory disorder and significantly ameliorated the Aβ production disturbance and AGE generation. Additionally, pathological changes in morphology were also significantly recovered. Our data reveal that AG plays a potentially neuroprotective role against ovariectomy-induced learning and cognitive impairment and Aβ production disorder.
Collapse
Affiliation(s)
- Dan Di Zhang
- Department of Geriatric Medicine , Beijing Luhe Hospital Affiliated to Capital Medical University , Beijing , China
| | - Yan Gang Wang
- Department of Gastroenterology , Hebei Provincial Hospital of Traditional Chinese Medicine , Hebei , China
| | - Chun Yan Liu
- Department of Rheumatology , The Third Hospital of Hebei Medical University , Hebei , China
| | - Ze Hou Wang
- Department of Chinese Medicine , Beijing University of Chinese Medicine , Beijing , China
| | - Yue Fen Wang
- Department of Nephropathy , Beijing Hospital of Traditional Chinese Medicine , Capital Medical University , Beijing , China
| |
Collapse
|
39
|
Cáceres A, González JR. Female-specific risk of Alzheimer's disease is associated with tau phosphorylation processes: A transcriptome-wide interaction analysis. Neurobiol Aging 2020; 96:104-108. [PMID: 32977080 DOI: 10.1016/j.neurobiolaging.2020.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 01/09/2023]
Abstract
The levels of tau phosphorylation differ between sexes in Alzheimer's disease (AD). Transcriptome-wide associations of sex by disease interaction could indicate whether specific genes underlie sex differences in tau pathology; however, no such study has been reported yet. We report the first analysis of the effect of the interaction between disease status and sex on differential gene expression, meta-analyzing transcriptomic data from the 3 largest publicly available case-control studies (N = 785) in the brain to date. A total of 128 genes, significantly associated with sex-AD interactions, were enriched in phosphoproteins (false discovery rate (FDR) = 0.001). High and consistent associations were found for the overexpressions of NCL (FDR = 0.002), whose phosphorylated protein generates an epitope against neurofibrillary tangles and KIF2A (FDR = 0.005), a microtubule-associated motor protein gene. Transcriptome-wide interaction analyses suggest sex-modulated tau phosphorylation, at sites like Thr231, Ser199, or Ser202 that could increase the risk of women to AD and indicate sex-specific strategies for intervention and prevention.
Collapse
Affiliation(s)
- Alejandro Cáceres
- Division of Noncommunicable Diseases, Instituto de Salud Global de Barcelona (ISGlobal), Barcelona, Spain; Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| | - Juan R González
- Division of Noncommunicable Diseases, Instituto de Salud Global de Barcelona (ISGlobal), Barcelona, Spain; Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Department of Mathematics, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Holingue C, Budavari AC, Rodriguez KM, Zisman CR, Windheim G, Fallin MD. Sex Differences in the Gut-Brain Axis: Implications for Mental Health. Curr Psychiatry Rep 2020; 22:83. [PMID: 33216233 PMCID: PMC7717677 DOI: 10.1007/s11920-020-01202-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this article is to highlight how sex differences in the gut-brain axis may contribute to the discrepancies in incidence of neurodevelopmental, psychiatric, and neurodegenerative disorders between females and males. We focus on autism spectrum disorder, psychotic disorders, stress and anxiety disorders, depression, Alzheimer's disease, and Parkinson's disease and additionally discuss the comorbidity between inflammatory bowel disorder and mental health disorders. RECENT FINDINGS Human and animal studies show that sex may modify the relationship between the gut or immune system and brain and behavior. Sex also appears to modify the effect of microbial treatments such as probiotics and antibiotics on brain and behavior. There is emerging evidence that assessing the role of sex in the gut-brain axis may help elucidate the etiology of and identify effective treatments for neurodevelopmental, psychiatric, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, USA.
- , Baltimore, USA.
| | - Alexa Curhan Budavari
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Katrina M Rodriguez
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Corina R Zisman
- Section on the Neurobiology of Fear and Anxiety, National Institute of Mental Health, Bethesda, MD, USA
| | - Grace Windheim
- Public Health Studies, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| |
Collapse
|
41
|
Agca C, Klakotskaia D, Stopa EG, Schachtman TR, Agca Y. Ovariectomy Influences Cognition and Markers of Alzheimer's Disease. J Alzheimers Dis 2020; 73:529-541. [PMID: 31796679 DOI: 10.3233/jad-190935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the most devastating and costly diseases, and prevalence of AD increases with age. Furthermore, females are twice as likely to suffer from AD compared to males. The cessation of reproductive steroid hormone production during menopause is hypothesized to cause this difference. Two rodent AD models, APP21 and APP+PS1, and wild type (WT) rats underwent an ovariectomy or sham surgery. Changes in learning and memory, brain histology, amyloid-β (Aβ) deposition, levels of mRNAs involved in Aβ production and clearance, and synaptic and cognitive function were determined. Barnes maze results showed that regardless of ovariectomy status, APP+PS1 rats learned slower and had poor memory retention. Ovariectomy caused learning impairment only in the APP21 rats. High levels of Aβ42 and very low levels of Aβ40 were observed in the brain cortices of APP+PS1 rats indicating limited endogenous PS1. The APP+PS1 rats had 43-fold greater formic acid soluble Aβ42 than Aβ40 at 17 months. Furthermore, levels of formic acid soluble Aβ42 increased 57-fold in ovariectomized APP+PS1 rats between 12 and 17 months of age. The mRNA encoding Grin1 significantly decreased due to ovariectomy whereas levels of Bace1, Chat, and Prkcb all decreased with age. The expression levels of mRNAs involved in Aβ degradation and AβPP cleavage (Neprilysin, Ide, Adam9, and Psenen) were found to be highly correlated with each other as well as hippocampal Aβ deposition. Taken together, these results indicate that both ovariectomy and genotype influence AD markers in a complex manner.
Collapse
Affiliation(s)
- Cansu Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Edward G Stopa
- Departments of Pathology and Neurosurgery, Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
42
|
Um YH, Lim HK. Orexin and Alzheimer's Disease: A New Perspective. Psychiatry Investig 2020; 17:621-626. [PMID: 32517419 PMCID: PMC7385219 DOI: 10.30773/pi.2020.0136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Orexin's role in human cognition has recently been emphasized and emerging evidences indicate its close relationship with Alzheimer's disease (AD). This review aimed to demonstrate recent research on the relationship between orexin and AD. Orexin's role in stress regulation and memory is discussed, with significant findings related to sexual disparities in stress response, with potential clinical implications pertaining to AD pathology. There are controversies regarding the orexin levels in AD patients, but the role of orexin in the trajectory of AD is still emphasized in recent literatures. Orexin is also accentuated in the context of tau pathology, and orexin as a potential therapeutic target for AD is frequently discussed. Future directions with regard to the relationship between orexin and AD are suggested: 1) consideration for AD trajectory in the measurement of orexin levels, 2) the need for objective measure such as polysomnography and actigraphy, 3) the need for close observation of cognitive profiles of orexin-deficient narcolepsy patients, 4) the need for validation studies by neuroimaging 5) the need for taking account sexual disparities in orexinergic activiation, and 6) consideration for orexin's role as a stress regulator. The aforementioned new perspectives could help unravel the relationship between orexin and AD.
Collapse
Affiliation(s)
- Yoo Hyun Um
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
43
|
Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer's Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor α. Cells 2020; 9:E1215. [PMID: 32422896 PMCID: PMC7290654 DOI: 10.3390/cells9051215] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Mutations in genes encoding proteins involved in amyloid-β peptide (Aβ) production are responsible for inherited AD cases. The amyloid cascade hypothesis was proposed to explain the pathogeny. Despite the fact that Aβ is considered as the main culprit of the pathology, most clinical trials focusing on Aβ failed and suggested that earlier interventions are needed to influence the course of AD. Therefore, identifying risk factors that predispose to AD is crucial. Among them, the epsilon 4 allele of the apolipoprotein E gene that encodes the major brain lipid carrier and metabolic disorders such as obesity and type 2 diabetes were identified as AD risk factors, suggesting that abnormal lipid metabolism could influence the progression of the disease. Among lipids, fatty acids (FAs) play a fundamental role in proper brain function, including memory. Peroxisome proliferator-activated receptor α (PPARα) is a master metabolic regulator that regulates the catabolism of FA. Several studies report an essential role of PPARα in neuronal function governing synaptic plasticity and cognition. In this review, we explore the implication of lipid metabolism in AD, with a special focus on PPARα and its potential role in AD therapy.
Collapse
Affiliation(s)
- Francisco Sáez-Orellana
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Jean-Noël Octave
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Nathalie Pierrot
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| |
Collapse
|
44
|
Christensen A, Liu J, Pike CJ. Aging Reduces Estradiol Protection Against Neural but Not Metabolic Effects of Obesity in Female 3xTg-AD Mice. Front Aging Neurosci 2020; 12:113. [PMID: 32431604 PMCID: PMC7214793 DOI: 10.3389/fnagi.2020.00113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vulnerability to Alzheimer's disease (AD) is increased by several risk factors, including midlife obesity, female sex, and the depletion of estrogens in women as a consequence of menopause. Conversely, estrogen-based hormone therapies have been linked with protection from age-related increases in adiposity and dementia risk, although treatment efficacy appears to be affected by the age of initiation. Potential interactions between obesity, AD, aging, and estrogen treatment are likely to have significant impact on optimizing the use of hormone therapies in postmenopausal women. In the current study, we compared how treatment with the primary estrogen, 17β-estradiol (E2), affects levels of AD-like neuropathology, behavioral impairment, and other neural and systemic effects of preexisting diet-induced obesity in female 3xTg-AD mice. Importantly, experiments were conducted at chronological ages associated with both the early and late stages of reproductive senescence. We observed that E2 treatment was generally associated with significantly improved metabolic outcomes, including reductions in body weight, adiposity, and leptin, across both age groups. Conversely, neural benefits of E2 in obese mice, including decreased β-amyloid burden, improved behavioral performance, and reduced microglial activation, were observed only in the early aging group. These results are consistent with the perspective that neural benefits of estrogen-based therapies require initiation of treatment during early rather than later phases of reproductive aging. Further, the discordance between E2 protection against systemic versus neural effects of obesity across age groups suggests that pathways other than general metabolic function, perhaps including reduced microglial activation, contribute to the mechanism(s) of the observed E2 actions. These findings reinforce the potential systemic and neural benefits of estrogen therapies against obesity, while also highlighting the critical role of aging as a mediator of estrogens' protective actions.
Collapse
Affiliation(s)
| | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
45
|
Chen YC, Oyang YJ, Lin TY, Sun WZ. Risk assessment of dementia after hysterectomy: Analysis of 14-year data from the National Health Insurance Research Database in Taiwan. J Chin Med Assoc 2020; 83:394-399. [PMID: 32149891 DOI: 10.1097/jcma.0000000000000286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND Anesthesia and surgery may increase the risk of dementia in the elderly, but the higher prevalence of dementia in women and other evidence suggest that dementia risk increases in younger women undergoing hysterectomy. In this study, we assessed the risk of dementia after hysterectomy. METHODS Hysterectomies registered in the National Health Insurance Research Database from 2000 to 2013 were evaluated using a retrospective generational research method. Multivariate Cox regression analysis was used to assess the effect of age at surgery, anesthesia method, and surgery type on the hazard ratio (HR) for the development of dementia. RESULTS Among 280 308 patients who underwent hysterectomy, 4753 (1.7%) developed dementia. Age at surgery and anesthesia method were associated with the occurrence of dementia, independent of surgery type. Among patients 30-49 years of age, general anesthesia (GA) was associated with a higher risk of dementia than spinal anesthesia (SA). The HR for GA was 2.678 (95% confidence interval [CI] = 1.269-5.650) and the risk of dementia increased by 7.4% for every 1-year increase in age (HR = 1.074; 95% CI = 1.048-1.101). In patients >50 years of age, the HR for GA was 1.206 (95% CI = 1.057-1.376), and the risk of dementia increased by 13.0% for every 1-year increase in age (HR = 1.130; 95% CI = 1.126-1.134). CONCLUSION The risk of dementia in women who underwent hysterectomy was significantly affected by older age at surgery, and the risk might not increase linearly with age, but show instead an S-curve with exponential increase at about 50 years of age. Although less significant, GA was associated with higher risk than SA, and the effect of the anesthesia method was greater in patients <50 years of age. In contrast, the surgical procedure used was not associated to the risk of dementia.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Yen-Jen Oyang
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering & Computer Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Yun Lin
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Wei-Zen Sun
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering & Computer Science, National Taiwan University, Taipei, Taiwan, ROC
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
46
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
47
|
Branigan GL, Soto M, Neumayer L, Rodgers K, Brinton RD. Association Between Hormone-Modulating Breast Cancer Therapies and Incidence of Neurodegenerative Outcomes for Women With Breast Cancer. JAMA Netw Open 2020; 3:e201541. [PMID: 32207833 PMCID: PMC7093781 DOI: 10.1001/jamanetworkopen.2020.1541] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/26/2020] [Indexed: 12/17/2022] Open
Abstract
Importance The association between exposure to hormone-modulating therapy (HMT) as breast cancer treatment and neurodegenerative disease (NDD) is unclear. Objective To determine whether HMT exposure is associated with the risk of NDD in women with breast cancer. Design, Setting, and Participants This retrospective cohort study used the Humana claims data set from January 1, 2007, to March 31, 2017. The Humana data set contains claims from private-payer and Medicare insurance data sets from across the United States with a population primarily residing in the Southeast. Patient claims records were surveyed for a diagnosis of NDD starting 1 year after breast cancer diagnosis for the duration of enrollment in the claims database. Participants were 57 843 women aged 45 years or older with a diagnosis of breast cancer. Patients were required to be actively enrolled in Humana claims records for 6 months prior to and at least 3 years after the diagnosis of breast cancer. The analyses were conducted between January 1 and 15, 2020. Exposure Hormone-modulating therapy (selective estrogen receptor modulators, estrogen receptor antagonists, and aromatase inhibitors). Main Outcomes and Measures Patients receiving HMT for breast cancer treatment were identified. Survival analysis was used to determine the association between HMT exposure and diagnosis of NDD. A propensity score approach was used to minimize measured and unmeasured selection bias. Results Of the 326 485 women with breast cancer in the Humana data set between 2007 and 2017, 57 843 met the study criteria. Of these, 18 126 (31.3%; mean [SD] age, 76.2 [7.0] years) received HMT, whereas 39 717 (68.7%; mean [SD] age, 76.8 [7.0] years) did not receive HMT. Mean (SD) follow-up was 5.5 (1.8) years. In the propensity score-matched population, exposure to HMT was associated with a decrease in the number of women who received a diagnosis of NDD (2229 of 17 878 [12.5%] vs 2559 of 17 878 [14.3%]; relative risk, 0.89; 95% CI, 0.84-0.93; P < .001), Alzheimer disease (877 of 17 878 [4.9%] vs 1068 of 17 878 [6.0%]; relative risk, 0.82; 95% CI, 0.75-0.90; P < .001), and dementia (1862 of 17 878 [10.4%] vs 2116 of 17 878 [11.8%]; relative risk, 0.88; 95% CI, 0.83-0.93; P < .001). The number needed to treat was 62.51 for all NDDs, 93.61 for Alzheimer disease, and 69.56 for dementia. Conclusions and Relevance Among patients with breast cancer, tamoxifen and steroidal aromatase inhibitors were associated with a decrease in the number who received a diagnosis of NDD, specifically Alzheimer disease and dementia.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
- MD-PhD Training Program, University of Arizona College of Medicine, Tucson
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
| | - Leigh Neumayer
- Department of Surgery, University of Arizona College of Medicine, Tucson
- Department of Obstetrics and Gynecology, University of Arizona College of Medicine, Tucson
| | - Kathleen Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
- Department of Neurology, University of Arizona College of Medicine, Tucson
| |
Collapse
|
48
|
Yang L, Zhou R, Tong Y, Chen P, Shen Y, Miao S, Liu X. Neuroprotection by dihydrotestosterone in LPS-induced neuroinflammation. Neurobiol Dis 2020; 140:104814. [PMID: 32087283 DOI: 10.1016/j.nbd.2020.104814] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/16/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Microglia-induced neuroinflammation plays a vital role in the etiology and progression of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and multiple sclerosis. The neuroprotective role of androgens, including testosterone and its metabolite dihydrotestosterone (DHT), has been increasingly demonstrated in these diseases, but few studies investigated the effects of androgen on neuroinflammation. This study investigated the role of DHT in lipopolysaccharide (LPS)-induced neuroinflammation, neuronal damage and behavioral dysfunction, as well as underlying mechanisms. We showed that DHT inhibited LPS-induced release of proinflammatory factors, including TNF-α, IL-1β, IL-6; iNOS, COX-2, NO, and PGE2 in BV2 cells and primary microglia by suppressing the TLR4-mediated NF-κB and MAPK p38 signaling pathways, thus protecting SH-SY5Y neurons from inflammatory damage induced by activated microglia. In an LPS-induced neuroinflammation mouse model, endogenous DHT depletion by castration exacerbated inflammatory responses by upregulating the levels of TNF-α, IL-1β, IL-6, iNOS, and COX-2 in the serum and brain by increasing the LR4-mediated NF-κB and MAPK pathway activation, but these effects were restored by exogenous DHT supplementation. Moreover, DHT also regulated the mRNA levels of the anti-inflammatory cytokines IL-10 and IL-13 in the brain. In addition, DHT modulated the expression of Aβ, the apoptotic proteins caspase-3, Bcl-2, and Bax, and synaptophysin, as well as neuronal damage in LPS-treated mouse brains. Further behavioral tests revealed that DHT ameliorated LPS-induced spatial and learning impairment and motor incoordination, and partly improved the locomotor activity in LPS-injected mice. Therefore, this study suggests that DHT exerts anti-neuroinflammatory and neuroprotective effects; thus, androgen replacement therapy is a potential therapeutic strategy for improving cognitive and behavioral function in neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Lei Yang
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Renyuan Zhou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin Medical University 300070 Tianjin, China; Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Yu Tong
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Pengfei Chen
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Yu Shen
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Shuai Miao
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai 200040, China.
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin Medical University 300070 Tianjin, China.
| |
Collapse
|
49
|
Avila JA, Kiprowska M, Jean-Louis T, Rockwell P, Figueiredo-Pereira ME, Serrano PA. PACAP27 mitigates an age-dependent hippocampal vulnerability to PGJ2-induced spatial learning deficits and neuroinflammation in mice. Brain Behav 2020; 10:e01465. [PMID: 31769222 PMCID: PMC6955932 DOI: 10.1002/brb3.1465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/20/2019] [Accepted: 10/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Inflammation in the brain is mediated by the cyclooxygenase pathway, which leads to the production of prostaglandins. Prostaglandin (PG) D2, the most abundant PG in the brain, increases under pathological conditions and is spontaneously metabolized to PGJ2. PGJ2 is highly neurotoxic, with the potential to transition neuroinflammation into a chronic state and contribute to neurodegeneration as seen in many neurological diseases. Conversely, PACAP27 is a lipophilic peptide that raises intracellular cAMP and is an anti-inflammatory agent. The aim of our study was to investigate the therapeutic potential of PACAP27 to counter the behavioral and neurotoxic effects of PGJ2 observed in aged subjects. METHODS PGJ2 was injected bilaterally into the hippocampal CA1 region of 53-week-old and 12-week-old C57BL/6N male mice, once per week over 3 weeks (three total infusions) and included co-infusions of PACAP27 within respective treatment groups. Our behavioral assessments looked at spatial learning and memory performance on the 8-arm radial maze, followed by histological analyses of fixed hippocampal tissue using Fluoro-Jade C and fluorescent immunohistochemistry focused on IBA-1 microglia. RESULTS Aged mice treated with PGJ2 exhibited spatial learning and long-term memory deficits, as well as neurodegeneration in CA3 pyramidal neurons. Aged mice that received co-infusions of PACAP27 exhibited remediated learning and memory performance and decreased neurodegeneration in CA3 pyramidal neurons. Moreover, microglial activation in the CA3 region was also reduced in aged mice cotreated with PACAP27. CONCLUSIONS Our data show that PGJ2 can produce a retrograde spread of damage not observed in PGJ2-treated young mice, leading to age-dependent neurodegeneration of hippocampal neurons producing learning and memory deficits. PACAP27 can remediate the behavioral and neurodegenerative effects that PGJ2 produces in aged subjects. Targeting specific neurotoxic prostaglandins, such as PGJ2, offers great promise as a new therapeutic strategy downstream of cyclooxygenases, to combat the neuronal deficits induced by chronic inflammation.
Collapse
Affiliation(s)
- Jorge A Avila
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| | - Magdalena Kiprowska
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Teneka Jean-Louis
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Patricia Rockwell
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Maria E Figueiredo-Pereira
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Peter A Serrano
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| |
Collapse
|
50
|
Zhou C, Wu Q, Wang Z, Wang Q, Liang Y, Liu S. The Effect of Hormone Replacement Therapy on Cognitive Function in Female Patients With Alzheimer's Disease: A Meta-Analysis. Am J Alzheimers Dis Other Demen 2020; 35:1533317520938585. [PMID: 32677442 PMCID: PMC10624041 DOI: 10.1177/1533317520938585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Previous studies have indicated that estrogen may delay disease progression and minimize the cognitive decline in patients with Alzheimer's disease (AD). However, the evidence for an estrogen deficiency in women with dementia and cognitive dysfunction is inconsistent. In the present review, a fixed effect meta-analysis revealed that the hormone replacement therapy (HRT) group exhibited significant improvements in Alzheimer Disease Assessment Scale-Cognitive subscale scores relative to those observed in the placebo group, suggesting that HRT is feasible for treating cognitive decline in patients with AD. However, no significant differences in Mini-Mental State Examination and Clinical Dementia Rating scale scores were observed between the 2 groups. The results of our systematic review indicate that HRT can improve cognitive function in female patients with AD. Due to limitations in sample size and the available literature, further multicenter trials with larger sample sizes are required to support these findings.
Collapse
Affiliation(s)
- ChengCheng Zhou
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- ChengCheng Zhou and Qingguang Wu are co-authors and contributed equally to this article
| | - Qingguang Wu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- ChengCheng Zhou and Qingguang Wu are co-authors and contributed equally to this article
| | - Zongwei Wang
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youya Liang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Sijun Liu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|