1
|
Lu C, Ren J, Huang S, Wang M, Zhou H, Guo J. S-9-PAHSA Attenuates Aβ Accumulation and Improves Cognitive Deficits by Promoting Mitochondrial Autophagy in 5xFAD Mice. Eur J Neurosci 2025; 61:e70118. [PMID: 40275795 DOI: 10.1111/ejn.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 04/04/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by significant cognitive impairment and predominantly affects the elderly. With no effective cure available, research continues to explore novel therapeutic and preventive strategies. Recently, palmitic acid-hydroxystearic acids (PAHSAs), especially their stereochemistry S-configuration, have shown potential as a dietary supplement with anti-inflammatory and anti-diabetic properties. We previously found that one of the PAHSAs, 9-PAHSA, could improve cognitive impairment in the high-fat-diet mice, however, whether it has an equal effect on AD-like mice remains unclear. Since mitochondrial dysfunction is recognized as a significant pathological feature of AD, with impaired mitophagy leading to the accumulation of dysfunctional mitochondria, thus exacerbating disease progression, in this study, we evaluated the effects of the chiral isomer of 9-PAHSA, S-9-PAHSA, on cognitive dysfunction and mitochondrial dysfunction in 5xFAD mice. Three-month-old mice were treated with S-9-PAHSA 30 mg/kg in their drinking water for 3 months. Behavioral studies were conducted using the Morris Water Maze (MWM) and Y-maze tests, followed by assessments of amyloid-beta (Aβ) plaque deposition, neuronal apoptosis, and mitochondrial function. We found that S-9-PAHSA significantly enhanced spatial learning and memory abilities, reduced amyloid plaque deposition, decreased neuronal apoptosis, and improved mitochondrial homeostasis and autophagy in 5xFAD mice. These findings suggest that S-9-PAHSA holds promise as a supplementary preventive and therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Chenyu Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jiaoqi Ren
- Department of Geriatrics, Huashan Hospital, National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Shanshan Huang
- Department of Geriatrics, Huashan Hospital, National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Meng Wang
- Department of Geriatrics, Huashan Hospital, National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Houguang Zhou
- Department of Geriatrics, Huashan Hospital, National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Jingchun Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Chen S, Hu Z, Zhao M, Sun J, Nie S, Gao X, Huang Y. A comprehensive proteomic analysis reveals novel inflammatory biomarkers in intracranial aneurysms. J Proteomics 2025; 313:105374. [PMID: 39761748 DOI: 10.1016/j.jprot.2025.105374] [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: 12/07/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 02/04/2025]
Abstract
Inflammation is a complex factor in the pathogenesis of intracranial aneurysms (IA), but its specific cellular inflammatory factors remain uncertain. We collected two cohorts and measured the representation of vascular inflammation-related proteins using the Olink CVD II Vascular Inflammation Panel. We subsequently validated our findings using ELISA and RT-qPCR. Our proteomic analysis identified 11 vascular inflammation-related markers that were significantly differentially represented between the IA and control groups. These markers were implicated in leukocyte migration, immune response, triglyceride and lipoprotein metabolism, acute phase response, T cell regulation, and several key biological pathways, including PPAR, HIF-1, cytokine-cytokine interactions, and PI3K-AKT signaling. Further validation with ELISA and RT-qPCR confirmed the differential representation of IL6, PTX3, LPL, and OLR1 between the two groups. Notably, a combination marker incorporating these four factors demonstrated high diagnostic potential for the early detection of IA. Our study has identified a set of informative biomarkers (IL6, PTX3, LPL, and OLR1) that could be valuable for the early diagnosis of IA. Importantly, this is the first report of significantly elevated OLR1 representation in the plasma of IA patients. Further investigation into the role of OLR1 in the pathogenesis of IA is warranted. SIGNIFICANCE: This study significantly advances our understanding of the molecular mechanisms underlying intracranial aneurysm (IA) pathogenesis. By identifying a panel of novel biomarkers, including the previously unreported elevated expression of OLR1 in IA patients, we provide crucial insights into the inflammatory processes involved in aneurysm formation and development. These findings have important clinical implications, as the identified biomarkers could serve as valuable tools for early diagnosis and potentially targeted therapeutic interventions. Furthermore, the study highlights the complex interplay of inflammatory pathways in IA, suggesting that a multi-faceted approach may be necessary for effective management.
Collapse
Affiliation(s)
- Siqi Chen
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China
| | - Ziliang Hu
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China
| | - Mingyue Zhao
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China
| | - Jie Sun
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Sheng Nie
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Xiang Gao
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China
| | - Yi Huang
- Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China.
| |
Collapse
|
3
|
Sultana F, Davis SR, Wolfe RS, McNeil JJ, Islam RM. Associations between blood sex hormones, cognitive decline and incident dementia in community-dwelling older Australian women: a prospective cohort study. Climacteric 2025:1-10. [PMID: 40085743 DOI: 10.1080/13697137.2025.2470458] [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: 09/28/2024] [Revised: 01/21/2025] [Accepted: 02/16/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Whether blood sex hormone concentrations predict cognitive decline and incident dementia in older women is uncertain. The Sex Hormones in Older Women (SHOW) study is a prospective cohort study of Australian women, aged at least 70 years, without cognitive impairment. METHODS Sex hormones were measured by liquid chromatography-tandem mass spectrometry, and comprehensive cognitive testing was performed at baseline and 3 years later. RESULTS Of the 6358 participants who had sex hormones measured, 4444 women (median age at baseline 74 years [Q1-Q3 71.7-77.5]) provided data for cognitive analyses. The findings were limited to a decline in executive function and verbal fluency was positively associated with the highest quartiles of estrone (odds ratio [OR] = 1.21, 95% confidence interval [CI] 1.01-1.45, p = 0.04) and dehydro-epiandrosterone (DHEA) (OR = 1.21, 95% CI 1.01-1.45, p = 0.04), compared with the lowest quartiles. Estrone and DHEA were not associated with any other cognitive decline. Testosterone was not associated with cognitive decline. In an exploratory analysis, cognitive decline was not different in women who had estradiol below the limit of detection (66% of women) compared with women with measurable estradiol. Over a median 4.1 years of follow-up (22,518 person-years), 121 (2.2%) developed dementia; an incident rate of 5.3 per 1000 person-years. There were no associations between any hormone and incident dementia. CONCLUSIONS The finding of a greater likelihood of a decline in executive function and verbal fluency in community-dwelling older women with the highest blood concentrations of DHEA and its metabolite estrone need reaffirmation and their clinical significance should be further investigated. These findings do not support use of estrogen or DHEA therapy to prevent cognitive decline in older women.
Collapse
Affiliation(s)
- Farhana Sultana
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Susan R Davis
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Endocrinology and Diabetes, Alfred Health, Melbourne, VIC, Australia
| | - Rory S Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - John J McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Rakibul M Islam
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
4
|
Perks CM, Barker RM, Alhadrami M, Alkahtani O, Gill E, Grishaw M, Harland AJ, Henley P, Li H, O’Sullivan E, Stone G, Su X, Kehoe PG. Curious Dichotomies of Apolipoprotein E Function in Alzheimer's Disease and Cancer-One Explanatory Mechanism of Inverse Disease Associations? Genes (Basel) 2025; 16:331. [PMID: 40149482 PMCID: PMC11942319 DOI: 10.3390/genes16030331] [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: 02/17/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
An apparent "inverse" relationship exists between two seemingly unconnected conditions, Alzheimer's disease (AD) and cancer, despite sharing similar risk factors, like increased age and obesity. AD is associated with amyloid beta (Aβ) plaques and neurofibrillary tau tangles that cause neural degeneration; cancer, in contrast, is characterized by enhanced cell survival and proliferation. Apolipoprotein E (ApoE) is the main lipoprotein found in the central nervous system and via its high affinity with lipoprotein receptors plays a critical role in cholesterol transport and uptake. ApoE has 3 protein isoforms, ApoE E2, ApoE E3, and ApoE E4, respectively encoded for by 3 allelic variants of APOE (ε2, ε3, and ε4). This review examines the characteristics and function of ApoE described in both AD and cancer to assimilate evidence for its potential contribution to mechanisms that may underly the reported inverse association between the two conditions. Of the genetic risk factors relevant to most cases of AD, the most well-known with the strongest contribution to risk is APOE, specifically the ε4 variant, whereas for cancer risk, APOE has not featured as a significant genetic contributor to risk. However, at the protein level in both conditions, ApoE contributes to disease pathology via affecting lipid physiology and transport. In AD, Aβ-dependent and -independent interactions have been suggested, whereas in cancer, ApoE plays a role in immunoregulation. Understanding the mechanism of action of ApoE in these diametrically opposed diseases may enable differential targeting of therapeutics to provide a beneficial outcome for both.
Collapse
Affiliation(s)
- Claire M. Perks
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Rachel M. Barker
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Mai Alhadrami
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Omar Alkahtani
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Emily Gill
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Mary Grishaw
- Cerebrovascular and Dementia Research Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (M.G.); (P.H.); (E.O.); (G.S.)
| | - Abigail J. Harland
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Peter Henley
- Cerebrovascular and Dementia Research Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (M.G.); (P.H.); (E.O.); (G.S.)
| | - Haonan Li
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Ellie O’Sullivan
- Cerebrovascular and Dementia Research Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (M.G.); (P.H.); (E.O.); (G.S.)
| | - Gideon Stone
- Cerebrovascular and Dementia Research Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (M.G.); (P.H.); (E.O.); (G.S.)
| | - Xiaoyu Su
- Cancer Endocrinology Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.B.); (M.A.); (O.A.); (E.G.); (A.J.H.); (H.L.); (X.S.)
| | - Patrick G. Kehoe
- Cerebrovascular and Dementia Research Group, Bristol Medical School, Learning & Research Building, Level 2, Southmead Hospital, Bristol BS10 5NB, UK; (M.G.); (P.H.); (E.O.); (G.S.)
| |
Collapse
|
5
|
Wood Alexander M, Honer WG, Saloner R, Galea LAM, Bennett DA, Rabin JS, Casaletto KB. The interplay between age at menopause and synaptic integrity on Alzheimer's disease risk in women. SCIENCE ADVANCES 2025; 11:eadt0757. [PMID: 40043118 PMCID: PMC11881898 DOI: 10.1126/sciadv.adt0757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/29/2025] [Indexed: 03/09/2025]
Abstract
Menopause is a major biological transition that may influence women's late-life brain health. Earlier estrogen depletion-via earlier menopause-has been associated with increased risk for Alzheimer's disease (AD). Synaptic dysfunction also incites and exacerbates AD progression. We investigated whether age at menopause and synaptic health together influence AD neuropathology and cognitive trajectories using clinical and autopsy data from 268 female decedents in the Rush Memory and Aging Project. We observed significant interactions between age at menopause and synaptic integrity on cognitive decline and tau tangles, such that earlier menopause strengthened the associations of reduced synaptic integrity with faster cognitive decline and elevated tau. Exploratory analyses showed that these relationships were attenuated in women who took menopausal hormone therapy. These findings suggest that midlife endocrine processes or their sequalae may influence synaptic vulnerability to AD. Interventions addressing both hormonal factors and synaptic health could enhance resilience to dementia in women.
Collapse
Affiliation(s)
- Madeline Wood Alexander
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - William G. Honer
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Liisa A. M. Galea
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, The Centre for Addition and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jennifer S. Rabin
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Division of Neurology, Temerty Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| |
Collapse
|
6
|
Kritzer MF, Adler A, Locklear M. Androgen effects on mesoprefrontal dopamine systems in the adult male brain. Neuroscience 2025; 568:519-534. [PMID: 38977069 DOI: 10.1016/j.neuroscience.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Epidemiological data show that males are more often and/or more severely affected by symptoms of prefrontal cortical dysfunction in schizophrenia, Parkinson's disease and other disorders in which dopamine circuits associated with the prefrontal cortex are dysregulated. This review focuses on research showing that these dopamine circuits are powerfully regulated by androgens. It begins with a brief overview of the sex differences that distinguish prefrontal function in health and prefrontal dysfunction or decline in aging and/or neuropsychiatric disease. This review article then spotlights data from human subjects and animal models that specifically identify androgens as potent modulators of prefrontal cortical operations and of closely related, functionally critical measures of prefrontal dopamine level or tone. Candidate mechanisms by which androgens dynamically control mesoprefrontal dopamine systems and impact prefrontal states of hypo- and hyper-dopaminergia in aging and disease are then considered. This is followed by discussion of a working model that identifies a key locus for androgen modulation of mesoprefrontal dopamine systems as residing within the prefrontal cortex itself. The last sections of this review critically consider the ways in which the organization and regulation of mesoprefrontal dopamine circuits differ in the adult male and female brain, and highlights gaps where more research is needed.
Collapse
Affiliation(s)
- Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, United States.
| | - Alexander Adler
- Department of Oncology and Immuno-Oncology, Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, United States
| | | |
Collapse
|
7
|
Yao M, Rosario ER, Soper JC, Pike CJ. Androgens Regulate Tau Phosphorylation Through Phosphatidylinositol 3-Kinase-Protein Kinase B-Glycogen Synthase Kinase 3β Signaling. Neuroscience 2025; 568:503-518. [PMID: 35777535 PMCID: PMC9797620 DOI: 10.1016/j.neuroscience.2022.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 12/31/2022]
Abstract
Age-related testosterone depletion in men is a risk factor for Alzheimer's disease (AD). How testosterone modulates AD risk remains to be fully elucidated, although regulation of tau phosphorylation has been suggested as a contributing protective action. To investigate the relationship between testosterone and tau phosphorylation, we first evaluated the effect of androgen status on tau phosphorylation in 3xTg-AD mice. Depletion of endogenous androgens via gonadectomy resulted in increased tau phosphorylation that was prevented by acute testosterone treatment. Parallel alterations in the phosphorylation of both glycogen synthase kinase 3β (GSK3β) and protein kinase B (Akt) suggest possible components of the underlying signaling pathway. To further explore mechanism, primary cultured neurons were treated with a physiological concentration of testosterone or its active metabolite dihydrotestosterone (DHT). Results showed that testosterone and DHT induced significant decreases in phosphorylated tau and significant increases in phosphorylation of Akt and GSK3β. Pharmacological inhibition of phosphatidylinositol 3-kinase (PI3K) effectively inhibited androgen-induced increases in Akt and GSK3β phosphorylation, and decreases in tau phosphorylation. In addition, androgen receptor (AR) knock-down by small interfering RNA prevented androgen-induced changes in the phosphorylation of Akt, GSK3β and tau, suggesting an AR-dependent mechanism. Additional experiments demonstrated androgen-induced changes in Akt, GSK3β and tau phosphorylation in AR-expressing PC12 cells but not in AR-negative PC12 cells. Together, these results suggest an AR-dependent pathway involving PI3K-Akt-GSK3β signaling through which androgens can reduce tau phosphorylation. These findings identify an additional protective mechanism of androgens that can improve neural health and inhibit development of AD.
Collapse
Affiliation(s)
- Mingzhong Yao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Emily R Rosario
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jenna Carroll Soper
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
8
|
Lemos R, Pérez-Badell Y, De Nisco M, Carpentieri A, Suárez M, Pedatella S. Organic Chimeras Based on Selenosugars, Steroids, and Fullerenes as Potential Inhibitors of the β-amyloid Peptide Aggregation. Chempluschem 2025; 90:e202400404. [PMID: 39235155 DOI: 10.1002/cplu.202400404] [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: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
The aggregation of β-amyloid peptide (Aβ) is associated with neurodegenerative diseases such as Alzheimer's disease (AD). Several therapies aimed at reducing the aggregation of this peptide have emerged as potential strategies for the treatment of AD. This paper describes the design and preparation of new hybrid molecules based on steroids, selenosugars, and [60]fullerene as potential inhibitors of Aβ oligomerization. These moieties were selected based on their antioxidant properties and possible areas of interaction with the Aβ. Cyclopropanations between C60 and malonates bearing different steroid and selenosugar moieties using the Bingel-Hirsch protocol have enabled the synthesis of functionalized molecular hybrids. The obtained derivatives were characterized by physical and spectroscopic techniques. Theoretical calculations for all the selenium compounds were performed using the density functional theory DFT/B3LYP-D3(BJ)/6-311G(2d,p) predicting the most stable conformations of the synthesized derivatives. Relevant geometrical parameters were investigated to relate the stereochemical behavior and the spectroscopic data obtained. The affinity of the compounds for Aβ-peptide was estimated by molecular docking simulation, which predicted an increase in affinity and interactions for Aβ for the hybrids containing the C60 core. In addition, parameters such as lipophilicity, polar surface area, and dipole moment were calculated to predict their potential interaction with membrane cells.
Collapse
Affiliation(s)
- Reinier Lemos
- Laboratorio de Síntesis Orgánica, Facultad de Química, Universidad de la Habana, 10400, La Habana, Cuba
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Yoana Pérez-Badell
- Laboratorio de Química Computacional y Teórica, Facultad de Química, Universidad de La Habana, 10400, La Habana, Cuba
| | - Mauro De Nisco
- Department of Chemical Sciences, University of Basilicata, Viale dell'Ateneo Lucano 10, I-85100, Potenza, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Margarita Suárez
- Laboratorio de Síntesis Orgánica, Facultad de Química, Universidad de la Habana, 10400, La Habana, Cuba
| | - Silvana Pedatella
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| |
Collapse
|
9
|
Ng TKS, Udeh‐Momoh C, Lim M, Gleerup HS, Leifert W, Ajalo C, Ashton N, Zetterberg H, Rissman RA, Winston CN, O’ Bryant S, Jenkins R, Carro E, Orive G, Tamburin S, Olvera‐Rojas M, Solis‐Urra P, Esteban‐Cornejo I, Santos GAAD, Rajan KB, Koh D, Simonsen AH, Slowey PD. Guidelines for the standardization of pre-analytical variables for salivary biomarker studies in Alzheimer's disease research: An updated review and consensus of the Salivary Biomarkers for Dementia Research Working Group. Alzheimers Dement 2025; 21:e14420. [PMID: 39737743 PMCID: PMC11848381 DOI: 10.1002/alz.14420] [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: 07/17/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 01/01/2025]
Abstract
There is a pressing need for accessible biomarkers with high diagnostic accuracy for Alzheimer's disease (AD) diagnosis to facilitate widespread screening, particularly in underserved groups. Saliva is an emerging specimen for measuring AD biomarkers, with distinct contexts of use that could complement blood and cerebrospinal fluid and detect various analytes. An interdisciplinary, international group of AD and related dementias (ADRD) researchers convened and performed a narrative review of published studies on salivary AD biomarkers. We critically appraised the current state of the literature, examining both consistencies and discrepancies in existing pre-analytical variables and methodologies. We discussed how various pre-analytical variables could influence the detection and quantification of salivary biomarkers, showed technologies available to standardize collection procedures, and proposed a standardized pre-analytical protocol to guide future studies on salivary AD biomarker examinations. We identified potential contexts of use, gaps, and priorities and proposed future research directions. HIGHLIGHTS: Given its non-invasive nature, wider accessibility, and cultural acceptability, particularly in low-resourced settings, saliva is a biofluid complementary to blood and CSF. Current salivary AD biomarker studies do not control for many confounding pre-analytical variables during the sampling process, potentially leading to inaccurate salivary biomarker readings and conclusions, contributing to conflicting findings. Reviewing the current literature, including the consistencies and non-consistencies observed in the existing parameters and methodologies, discussing how they can affect salivary AD biomarker detection and quantification. Proposing a standardized salivary pre-analytical protocol, identifying the gaps and prioritizations needed to move this area forward, proposing future directions and potential contexts of use.
Collapse
Affiliation(s)
- Ted K. S. Ng
- Rush Institute for Healthy AgingDepartment of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
- Edson College of Nursing and Health EducationArizona State UniversityPhoenixArizonaUSA
| | - Chinedu Udeh‐Momoh
- School of Public Health SciencesWake Forest University School of Medicine, North CarolinaMedical Center BoulevardWinston‐SalemNorth CarolinaUSA
- Brain and Mind InstituteAga Khan UniversityNairobiKenya
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska InstitutetStockholmStockholmSweden
- Sheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
| | - Mei‐Ann Lim
- PAPRSB Institute of Health SciencesUniversiti Brunei DarussalamDarussalamBrunei
| | - Helena Sophia Gleerup
- Danish Dementia Research Centre (DDRC)Department of NeurologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Wayne Leifert
- Molecular Diagnostic Solutions GroupHuman Health ProgramCommonwealth Scientific and Industrial Research Organisation (CSIRO) Health and BiosecurityAdelaideSouth AustraliaAustralia
- School of Biological SciencesThe University of AdelaideAdelaideSouth AustraliaAustralia
| | | | - Nicholas Ashton
- Banner Health Foundation and Banner Alzheimer's FoundationPhoenixArizonaUSA
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLMaple HouseLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayScience ParkHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Robert A. Rissman
- Department of Physiology and Neuroscience and Alzheimer's Disease Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Charisse N. Winston
- Department of Physiology and Neuroscience and Alzheimer's Disease Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Sid O’ Bryant
- Family Medicine and Osteopathic Manipulative MedicineInstitute for Healthy AgingInstitute for Translational ResearchTexas College of Osteopathic MedicineUNT Health Science CenterFort WorthTexasUSA
| | | | - Eva Carro
- Neurobiology of Alzheimer's Disease UnitFunctional Unit for Research into Chronic DiseasesInstituto de Salud Carlos IIIMadridSpain
- Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED)ISCIIIFuencarral‐El PardoMadridSpain
| | - Gorka Orive
- NanoBioCel Research GroupSchool of PharmacyUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- Bioaraba, NanoBioCel Research GroupVitoria‐GasteizSpain
- Biomedical Research Networking Centre in BioengineeringBiomaterials and Nanomedicine (CIBER‐BBN)Institute of Health Carlos IIIMadridSpain
- University Institute for Regenerative Medicine and Oral Implantology‐UIRMI (UPV/EHU‐Fundación Eduardo Anitua)Vitoria‐GasteizSpain
| | - Stefano Tamburin
- Department of NeurosciencesBiomedicine, and Movement SciencesUniversity of VeronaVeronaItaly
| | - Marcos Olvera‐Rojas
- Department of Physical Education and SportsFaculty of Sport SciencesSport and Health University Research Institute (iMUDS)University of GranadaGranadaSpain
| | - Patricio Solis‐Urra
- Department of Physical Education and SportsFaculty of Sport SciencesSport and Health University Research Institute (iMUDS)University of GranadaGranadaSpain
- Faculty of Education and Social SciencesUniversity of Andres BelloViña del MarChile
| | - Irene Esteban‐Cornejo
- Department of Physical Education and SportsFaculty of Sport SciencesSport and Health University Research Institute (iMUDS)University of GranadaGranadaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADABeiroGranadaSpain
| | | | - Kumar B. Rajan
- Rush Institute for Healthy AgingDepartment of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | - David Koh
- Saw Swee Hock School of Public HealthNational University of SingaporeSingaporeSingapore
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre (DDRC)Department of NeurologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Paul D. Slowey
- Oasis Diagnostics® CorporationVancouverWashingtonUSA
- Central South UniversityChangshaHunanChina
- RapidDx, Inc. MilwaukeeNew BerlinWisconsinUSA
| | | |
Collapse
|
10
|
Manna PR, Yang S, Manna C, Waters H, Islam MA, Reddy AP, Rawat P, Reddy PH. Steroidogenic acute regulatory protein mediated variations of gender-specific sex neurosteroids in Alzheimer's disease: Relevance to hormonal and neuronal imbalance. Neurosci Biobehav Rev 2025; 169:105969. [PMID: 39631487 DOI: 10.1016/j.neubiorev.2024.105969] [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: 05/13/2024] [Revised: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
The steroidogenic acute regulatory (StAR) protein mediates the rate-liming step in neuro/steroid biosynthesis. Multifaceted and delicate changes during aging, disrupting hormonal and neuronal homeostasis, constitute human senescence, an inevitable phenomenon that attributes to increased morbidity and mortality. Aging, along with progressive decreases in bioactive neurosteroids, is the primary risk factor for Alzheimer's disease (AD), which preferentially impacts two-thirds of women and one-third of men. AD is neuropathologically characterized by the accumulation of extracellular amyloid-β and intracellular phosphorylated Tau containing neurofibrillary tangles, resulting in dementia. Postmortem brains pertaining to gender-specific AD patients exhibit varied suppression of StAR and sex neurosteroid levels compared with age-matched cognitively healthy subjects, in which the attenuation of StAR is inversely correlated with the AD pathological markers. Interestingly, retinoid signaling upregulates StAR-motivated neurosteroid biosynthesis and reinstates various neurodegenerative vulnerabilities that promote AD pathogenesis. This review summarizes current understanding of StAR-driven alterations of sex neurosteroids in gender-specific AD risks and provides biochemical and molecular insights into therapeutic interventions for preventing and/or alleviating dementia for healthy aging.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Hope Waters
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Priyanka Rawat
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
Singh P, Paramanik V. DNA methylation, histone acetylation in the regulation of memory and its modulation during aging. FRONTIERS IN AGING 2025; 5:1480932. [PMID: 39835300 PMCID: PMC11743476 DOI: 10.3389/fragi.2024.1480932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/27/2024] [Indexed: 01/22/2025]
Abstract
Memory formation is associated with constant modifications of neuronal networks and synaptic plasticity gene expression in response to different environmental stimuli and experiences. Dysregulation of synaptic plasticity gene expression affects memory during aging and neurodegenerative diseases. Covalent modifications such as methylation on DNA and acetylation on histones regulate the transcription of synaptic plasticity genes. Changes in these epigenetic marks correlated with alteration of synaptic plasticity gene expression and memory formation during aging. These epigenetic modifications, in turn, are regulated by physiology and metabolism. Steroid hormone estrogen and metabolites such as S-adenosyl methionine and acetyl CoA directly impact DNA and histones' methylation and acetylation levels. Thus, the decline of estrogen levels or imbalance of these metabolites affects gene expression and underlying brain functions. In the present review, we discussed the importance of DNA methylation and histone acetylation on chromatin modifications, regulation of synaptic plasticity gene expression and memory consolidation, and modulation of these epigenetic marks by epigenetic modifiers such as phytochemicals and vitamins. Further, understanding the molecular mechanisms that modulate these epigenetic modifications will help develop recovery approaches.
Collapse
Affiliation(s)
| | - Vijay Paramanik
- Cellular and Molecular Neurobiology & Drug Targeting Laboratory, Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| |
Collapse
|
12
|
Leisgang Osse AM, Kinney JW, Cummings JL. The Common Alzheimer's Disease Research Ontology (CADRO) for biomarker categorization. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70050. [PMID: 39935614 PMCID: PMC11812129 DOI: 10.1002/trc2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/13/2025]
Abstract
Biomarkers are vital to Alzheimer's disease (AD) drug development and clinical trials, and will have an increasing role in clinical care. In this narrative review, we demonstrate the use of the National Institutes on Aging/Alzheimer's Association (NIA/AA) Common Alzheimer's Disease Research Ontology (CADRO) system for the categorization of biomarkers based on the primary mechanism on which they report. We show that biomarkers are available (in various levels of validation) for all CADRO processes. Application of the CADRO system demonstrates gaps in the field where novel biomarkers are needed for specific aspects of the disease, and assays to detect and measure biological changes, in individuals with symptomatic or preclinical AD. We demonstrate the CADRO system as a means of categorizing established and candidate AD biomarkers, showing the feasibility and practicality of the system. CADRO can assist with biomarker selection for AD clinical trials and drug development, and may eventually be applied to implementing biomarkers in patient care. Highlights The Common Alzheimer's Disease Research Ontology (CADRO) system can be used to categorize biomarkers for drug development.We demonstrate the use of CADRO with Alzheimer's disease (AD) biomarkers.We identified AD biomarkers in each of the CADRO categories.CADRO can be incorporated into current AD drug development and clinical trial systems.
Collapse
Affiliation(s)
- Amanda M. Leisgang Osse
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Jefferson W. Kinney
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Jeffrey L. Cummings
- Department of Brain Health, Kirk Kerkorian School of MedicineUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
13
|
Almutairi JA, Kidd EJ. Biological Sex Disparities in Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:79-104. [PMID: 39485650 DOI: 10.1007/7854_2024_545] [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] [Indexed: 11/03/2024]
Abstract
Alzheimer's disease is a highly complex and multifactorial neurodegenerative disorder, with age being the most significant risk factor. The incidence of Alzheimer's disease doubles every 5 years after the age of 65. Consequently, one of the major challenges in Alzheimer's disease research is understanding how the brain changes with age. Gaining insights into these changes could help identify individuals who are more prone to developing Alzheimer's disease as they age. Over the past 25 years, studies on brain aging have examined thousands of human brains to explore the neuronal basis of age-related cognitive decline. However, most of these studies have focused on adults over 60, often neglecting the critical menopause transition period. During menopause, women experience a substantial decline in ovarian sex hormone production, with a decrease of about 90% in estrogen levels. Estrogen is known for its neuroprotective effects, and its significant loss during menopause affects various biological systems, including the brain. Importantly, despite known differences in dementia risk between sexes, the impact of biological sex and sex hormones on brain aging and the development of Alzheimer's disease remains underexplored.
Collapse
Affiliation(s)
- Jawza A Almutairi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Emma J Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
14
|
Devraj K, Kulkarni O, Liebner S. Regulation of the blood-brain barrier function by peripheral cues in health and disease. Metab Brain Dis 2024; 40:61. [PMID: 39671124 PMCID: PMC11645320 DOI: 10.1007/s11011-024-01468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 12/14/2024]
Abstract
The blood-brain barrier (BBB) is formed by microvascular endothelial cells which are ensembled with pericytes, astrocytes, microglia and neurons in the neurovascular unit (NVU) that is crucial for neuronal function. Given that the NVU and the BBB are highly dynamic and regulated structures, their integrity is continuously challenged by intrinsic and extrinsic factors. Herein, factors from peripheral organs such as gonadal and adrenal hormones may influence vascular function also in CNS endothelial cells in a sex- and age-dependent manner. The communication between the periphery and the CNS likely takes place in specific areas of the brain among which the circumventricular organs have a central position due to their neurosensory or neurosecretory function, owing to physiologically leaky blood vessels. In acute and chronic pathological conditions like liver, kidney, pulmonary disease, toxins and metabolites are generated that reach the brain via the circulation and may directly or indirectly affect BBB functionality via the activation of the immunes system. For example, chronic kidney disease (CKD) currently affects more than 840 million people worldwide and is likely to increase along with western world comorbidities of the cardio-vascular system in continuously ageing societies. Toxins leading to the uremic syndrome, may further lead to neurological complications such as cognitive impairment and uremic encephalopathy. Here we summarize the effects of hormones, toxins and inflammatory reactions on the brain vasculature, highlighting the urgent demand for mechanistically exploring the communication between the periphery and the CNS, focusing on the BBB as a last line of defense for brain protection.
Collapse
Affiliation(s)
- Kavi Devraj
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India.
| | - Onkar Kulkarni
- Metabolic Disorders and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Partner Site Frankfurt, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
15
|
Yu L, Chen X, Bai X, Fang J, Sui M. Microbiota Alters and Its Correlation with Molecular Regulation Underlying Depression in PCOS Patients. Mol Neurobiol 2024; 61:9977-9992. [PMID: 37995075 DOI: 10.1007/s12035-023-03744-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/27/2023] [Indexed: 11/24/2023]
Abstract
Depression is one of the complications in patients with polycystic ovary syndrome (PCOS) that leads to considerable mental health. Accumulating evidence suggests that human gut microbiomes are associated with the progression of PCOS and depression. However, whether microbiota influences depression development in PCOS patients is still uncharacterized. In this study, we employed metagenomic sequencing and transcriptome sequencing (RNA-seq) to profile the composition of the fecal microbiota and gene expression of peripheral blood mononuclear cells in depressed women with PCOS (PCOS-DP, n = 27) in comparison to mentally healthy women with PCOS (PCOS, n = 18) and compared with healthy control (HC, n = 27) and patients with major depressive disorder (MDD, n = 29). Gut microbiota assessment revealed distinct patterns in the relative abundance in the PCOS-DP compared to HC, MDD, and PCOS groups. Several gut microbes exhibited uniquely and significantly higher abundance in the PCOS-DP compared to PCOS patients, inducing EC Ruminococcus torques, Coprococcus comes, Megasphaera elsdenii, Acidaminococcus intestini, and Barnesiella viscericola. Bacteroides eggerthii was a potential gut microbial biomarker for the PCOS-DP. RNA-seq profiling identified that 35 and 37 genes were significantly elevated and downregulated in the PCOS-DP, respectively. The enhanced differential expressed genes (DEGs) in the PCOS-DP were enriched in pathways involved in signal transduction and endocrine and metabolic diseases, whereas several lipid metabolism pathways were downregulated. Intriguingly, genes correlated with the gut microbiota were found to be significantly enriched in pathways of neurodegenerative diseases and the immune system, suggesting that changes in the microbiota may have a systemic impact on the expression of neurodegenerative diseases and immune genes. Gut microbe-related DEGs of CREB3L3 and CCDC173 were possible molecular biomarkers and therapeutic targets of women with PCOS-DP. Our multi-omics data indicate shifts in the gut microbiome and host gene regulation in PCOS patients with depression, which is of possible etiological and diagnostic importance.
Collapse
Affiliation(s)
- Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Xiaoyu Chen
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xuefeng Bai
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Jingping Fang
- College of Life Science, Fujian Normal University, Fuzhou, 350117, China
| | - Ming Sui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| |
Collapse
|
16
|
Schweitzer N, Son SJ, Thurston RC, Li J, Chen CL, Aizenstein H, Yang S, Iordanova B, Hong CH, Roh HW, Cho YH, Hong S, Nam YJ, Lee DY, Park B, Kim NR, Choi JW, Cheong J, Seo SW, An YS, Moon SY, Han SJ, Wu M. Sex-specific risk factors and clinical dementia outcomes for white matter hyperintensities in a large South Korean cohort. Alzheimers Res Ther 2024; 16:243. [PMID: 39482724 PMCID: PMC11529246 DOI: 10.1186/s13195-024-01598-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE White matter hyperintensities (WMH) on brain MRI images are the most common feature of cerebral small vessel disease (CSVD). Studies have yielded divergent findings on the modifiable risk factors for WMH and WMH's impact on cognitive decline. Mounting evidence suggests sex differences in WMH burden and subsequent effects on cognition. Thus, we aimed to identify sex-specific modifiable risk factors for WMH. We then explored whether there were sex-specific associations of WMH to longitudinal clinical dementia outcomes. METHODS Participants aged 49-89 years were recruited at memory clinics and underwent a T2-weighted fluid-attenuated inversion recovery (FLAIR) 3T MRI scan to measure WMH volume. Participants were then recruited for two additional follow-up visits, 1-2 years apart, where clinical dementia rating sum of boxes (CDR-SB) scores were measured. We first explored which known modifiable risk factors for WMH were significant when tested for a sex-interaction effect. We additionally tested which risk factors were significant when stratified by sex. We then tested to see whether WMH is longitudinally associated with clinical dementia that is sex-specific. RESULTS The study utilized data from 713 participants (241 males, 472 females) with a mean age of 72.3 years and 72.8 years for males and females, respectively. 57.3% and 59.5% of participants were diagnosed with mild cognitive impairment (MCI) for males and females, respectively. 40.7% and 39.4% were diagnosed with dementia for males and females, respectively. Of the 713 participants, 181 participants had CDR-SB scores available for three longitudinal time points. Compared to males, females showed stronger association of age to WMH volume. Type 2 Diabetes was associated with greater WMH burden in females but not males. Finally, baseline WMH burden was associated with worse clinical dementia outcomes longitudinally in females but not in males. DISCUSSION Older females have an accelerated increase in cerebrovascular burden as they age, and subsequently are more vulnerable to clinical dementia decline due to CSVD. Additionally, females are more susceptible to the cerebrovascular consequences of diabetes. These findings emphasize the importance of considering sex when examining the consequences of CSVD. Future research should explore the underlying mechanisms driving these sex differences and personalized prevention and treatment strategies. CLINICAL TRIAL REGISTRATION The BICWALZS is registered in the Korean National Clinical Trial Registry (Clinical Research Information Service; identifier, KCT0003391). Registration Date 2018/12/14.
Collapse
Affiliation(s)
- Noah Schweitzer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Jinghang Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chang-Le Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Howard Aizenstein
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Shaolin Yang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Bistra Iordanova
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yong Hyuk Cho
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sunhwa Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - You Jin Nam
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Bumhee Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
- Office of Biostatistics, Medical Research Collaborating Centre, Ajou Research Institute for Innovative Medicine, Ajou University Medical Centre, Suwon, Republic of Korea
| | - Na-Rae Kim
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jin Wook Choi
- Department of Radiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jaeyoun Cheong
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Republic of Korea
- Human Genome Research and Bio-Resource Centre, Ajou University Medical Centre, Suwon, Republic of Korea
| | - Sang Woon Seo
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, Republic of Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
17
|
Dobson AJ, XU Z, Wilson LF, Chung HF, Sandin S, Van der Schouw YT, Demakakos P, Weiderpass E, Mishra GD. Menopause age and type and dementia risk: a pooled analysis of 233 802 women. Age Ageing 2024; 53:afae254. [PMID: 39562342 PMCID: PMC11576136 DOI: 10.1093/ageing/afae254] [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: 06/19/2024] [Indexed: 11/21/2024] Open
Abstract
OBJECTIVES It is not clear whether the association between younger age at menopause and increased risk of dementia is modified by type of menopause. We examined the association of age at menopause or hysterectomy with dementia risk in three groups of women: those with natural menopause, premenopausal bilateral oophorectomy (surgical menopause) or premenopausal hysterectomy (without bilateral oophorectomy). STUDY DESIGN Individual-level data from 233 802 women in five prospective cohort studies (from four countries) were harmonized and pooled. Cox proportional hazards models were used to assess the associations of age at natural menopause, surgical menopause or premenopausal hysterectomy, with age at dementia, death (where available) or end of follow-up, whichever came first. RESULTS The study followed women to the median age of 72 years (quartiles 67, 76 years). The median follow-up time was 13 years, with 3262 dementia cases during this period. Compared with women with menopause at 50-52 years, women with menopause <40 years had a higher risk of dementia (adjusted hazard ratio (aHR): 1.47, 95% confidence interval (CI): 1.39, 1.56). This level of risk was comparable to that of current smoking and stroke, which are well-established risk factors for dementia. Increased risk of dementia associated with surgical menopause or premenopausal hysterectomy (compared to natural menopause) was not apparent after adjustment for age at menopause (aHR 0.99, 95% CI: 0.93, 1.04 and aHR 0.97, 95% CI: 0.95, 1.00, respectively). CONCLUSION Women who experience menopause before the age of 40 years have a higher risk of dementia irrespective of type of menopause.
Collapse
Affiliation(s)
- Annette J Dobson
- The University of Queensland School of Public Health, NHMRC Centre for Research Excellence on Women and Non-communicable Diseases (CRE WaND), Brisbane, Australia
| | - Zhiwei XU
- The University of Queensland School of Public Health, NHMRC Centre for Research Excellence on Women and Non-communicable Diseases (CRE WaND), Brisbane, Australia
| | - Louise F Wilson
- The University of Queensland School of Public Health, NHMRC Centre for Research Excellence on Women and Non-communicable Diseases (CRE WaND), Brisbane, Australia
| | - Hsin-Fang Chung
- The University of Queensland School of Public Health, NHMRC Centre for Research Excellence on Women and Non-communicable Diseases (CRE WaND), Brisbane, Australia
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Yvonne T Van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organisation, Lyon, France
| | - Gita D Mishra
- The University of Queensland School of Public Health, NHMRC Centre for Research Excellence on Women and Non-communicable Diseases (CRE WaND), Brisbane, Australia
| |
Collapse
|
18
|
Yao CY, Chung CH, Chien WC, Li ST, Lee ST, Huang CC, Yang CC, Tzeng NS. Ectopic pregnancy, its potential links to dementia risk and interactions with depression: insights from a nationwide cohort study. Front Psychiatry 2024; 15:1410685. [PMID: 39279812 PMCID: PMC11392761 DOI: 10.3389/fpsyt.2024.1410685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024] Open
Abstract
Background Dementia poses a growing global mental health impact, with variations in prevalence by gender, possibly influenced by reproductive factors. Ectopic pregnancy (EP), known for its association with cardiovascular diseases and depression, which are also predictors of dementia, prompted an exploration of their interplay. Methods Using Taiwan's National Health Insurance Research Database, this nationwide cohort study examined 53,096 individuals to investigate the link between EP and dementia. Covariates included age, insured premiums, comorbidity by Charlson Comorbidity Index revised by excluding dementia, level of care, and residence. Surgical approaches, number of EP episodes, and dementia subtypes were considered in outcomes analysis using Cox regression. Results Among 13,274 women diagnosed with EP, 791 developed dementia over a 15-year follow-up, particularly vascular dementia. Adjusting for the covariates, the adjusted sub-distribution Hazard Ratio (asHR) with competing risks was 1.644 (95% CI, 1.394-2.053; p < 0.001). For patients with more than one episode, it was even higher (asHR=1.670 [95% CI, 1.419-2.092; p < 0.001]). Post-ectopic depression, prevalent in 62.2% within four weeks, was associated with a greater dementia risk compared to those without (asHR=1.702 [95% CI, 1.444-2.125; p<0.001] vs. asHR=1.551 [95%CI, 1.310-1.937; p<0.001]). Antidepressant treatments showed a partial protective effect, reducing the increased risk by 14.7%. Conclusion An EP history is linked to an earlier onset and a higher risk of overall dementia, VaD in particular, in a dose dependent manner, regardless of surgical intervention and stroke. Post-ectopic depression exacerbates dementia risk, while antidepressants offer partial protection. These findings underscore the potential benefit of screening and treating depression in women following EPs.
Collapse
Affiliation(s)
- Chia-Yi Yao
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Sung-Tao Li
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Siou-Ting Lee
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chih-Chung Huang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chuan-Chi Yang
- Department of Psychiatry, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu, Taiwan
- Department of Psychiatry, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Nian-Sheng Tzeng
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Counseling Center, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
19
|
Del Río JP, Tapia V, Soto H, Vigil P. Neuroactive hormones and personal growth: associations in Chilean adolescents (ages 12-25) with ovulatory dysfunction. Front Psychol 2024; 15:1433437. [PMID: 39233885 PMCID: PMC11371675 DOI: 10.3389/fpsyg.2024.1433437] [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: 05/15/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Hormones produced by the hypothalamic-pituitary-adrenal-gonadal (HPAG) axis are crucial for modulating central nervous system (CNS) function and development throughout a person's life. Disruptions in HPAG function can impact psychological development, particularly during adolescence-a period marked by psychological growth and the maturation of the HPAG axis. An early indicator of HPAG alterations is ovulatory dysfunction (OD), a common condition among adolescents. Methods This study explored the associations between neuroactive hormones and personal growth in adolescents with OD. Female participants aged 12-25 years with OD were recruited, and assessments were conducted to profile their basic hormonal levels and various dimensions of individual development, including self-concept clarity, sense of coherence, self-esteem, perfectionism, self-control, and mood states. Results Adolescents with OD (n = 117) had lower self-concept clarity and self-esteem compared to reference data. A significant portion of the sample displayed elevated levels of tension (71.25%), confusion (62.5%), fatigue (58.22%), and depression (52.6%). Self-esteem scores were negatively correlated with DHEAS (r = -0.224; p = 0.026) and glucose (r = -0.249; p = 0.010). Higher levels of free testosterone were associated with increased depression scores (coef = 0.2398; p = 0.002), whereas higher estradiol levels were linked to lower aggressiveness scores (coef = -0.0648; p = 0.001). Discussion These findings indicate that hormonal imbalances in adolescents with OD could affect personal growth. Further research is needed to establish causal relationships between the variables considered.
Collapse
Affiliation(s)
- Juan Pablo Del Río
- Departamento de Psiquiatría y Salud Mental de la Infancia y de la Adolescencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Millennium Nucleus to Improve the Mental Health of Adolescents and Youths Imhay, Santiago, Chile
- Reproductive Health Research Institute, Santiago, Chile
| | - Valeska Tapia
- Reproductive Health Research Institute, Santiago, Chile
| | - Hugo Soto
- Reproductive Health Research Institute, Santiago, Chile
| | - Pilar Vigil
- Reproductive Health Research Institute, Santiago, Chile
| |
Collapse
|
20
|
Tang S, Xiao Z, Lin F, Liang X, Ma X, Wu J, Zhou X, Zhao Q, Gao J, Xiao Q, Ding D. Joint effect of testosterone and neurofilament light chain on cognitive decline in men: The Shanghai Aging Study. Alzheimers Dement 2024; 20:5290-5298. [PMID: 38837321 PMCID: PMC11350006 DOI: 10.1002/alz.13889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION The association of testosterone and cognitive decline is inconclusive, and its joint effect with neurofilaments light chain (NfL) remains largely unknown. METHODS A total of 581 non-demented older men in the Shanghai Aging Study were included. Blood total testosterone (TT), free testosterone (FT), and NfL were measured at baseline. The relationships between TT, FT, TT/FT-NfL, and cognitive decline were explored by Cox regression models. RESULTS During a median follow-up of 6.7 years, there was an inverse association between TT/FT and cognitive decline (TT, trend p = 0.004, Q1 vs Q4, hazard ratio [HR] = 4.39, 95% confidence interval [CI] = 1.60 to 12.04; FT, trend p = 0.002, Q1 vs Q4, HR = 5.29, 95% CI = 1.50 to 16.89). Compared to participants with high TT/FT-low NfL, those with low TT/FT-high NfL had significantly higher risks of cognitive decline (TT, HR = 5.10, 95% CI = 1.11 to 23.40; FT, HR = 6.14, 95% CI = 1.34 to 28.06). DISCUSSION Our findings suggest that the combination of testosterone and neurodegenerative markers may provide reliable predictive insights into future cognitive decline. HIGHLIGHTS Testosterone is inversely associated with cognitive decline in older men. There is a joint effect of testosterone and NfL on cognitive decline. Sex hormone and neurodegeneration may synergistically contribute to cognitive deterioration.
Collapse
Affiliation(s)
- Shuning Tang
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
| | - Zhenxu Xiao
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Fangting Lin
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
| | - Xiaoniu Liang
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaoxi Ma
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Jie Wu
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaowen Zhou
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Qianhua Zhao
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
- MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Junling Gao
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
- Health Communication InstituteFudan UniversityShanghaiChina
| | - Qianyi Xiao
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
- Health Communication InstituteFudan UniversityShanghaiChina
| | - Ding Ding
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
21
|
Liang C, Dobson AJ, Chung HF, van der Schouw YT, Sandin S, Weiderpass E, Mishra GD. Association of infertility and recurrent pregnancy loss with the risk of dementia. Eur J Epidemiol 2024; 39:785-793. [PMID: 38888679 PMCID: PMC11343804 DOI: 10.1007/s10654-024-01135-3] [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: 02/23/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Emerging evidence has shown the association between female reproductive histories (e.g., menarche age, parity, premature and early menopause) and the risk of dementia. However, little attention has been given to infertility and pregnancy loss. To examine the associations of infertility, recurrent miscarriages, and stillbirth with the risk of dementia, this study used data from four cohorts in the International Collaboration for a Life Course Approach to Reproductive Health and Chronic Disease Events. Women with data on at least one of the reproductive exposures of interest, dementia, and all covariates were included. Histories of infertility, miscarriage, and stillbirth were self-reported. Dementia (including Alzheimer's disease) was identified through surveys, aged care, pharmaceutical, hospital, and death registry data. Cause-specific Cox regression models were used to estimate the hazard ratios of dementia, accounting for well-established risk factors of dementia, study variability, and within-study correlation. Overall, 291,055 women were included at a median (interquartile range) age of 55.0 (47.0-62.0) at baseline. During the median (interquartile range) follow-up period of 13.0 (12.0-14.0) years, 3334 (1.2%) women developed dementia. Compared to women without stillbirth, a history of recurrent stillbirths (≥ 2) was associated with 64% higher risk of dementia (adjusted hazard ratio = 1.64, 95% confidence interval: 1.46-1.85). Compared to women without miscarriage, women with recurrent miscarriages (≥ 3) were at 22% higher risk of dementia (adjusted hazard ratio = 1.22, 95% confidence interval: 1.19-1.25). These findings suggest that recurrent stillbirths is a risk factor for dementia and may need to be considered in risk assessment of dementia in women.
Collapse
Affiliation(s)
- Chen Liang
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Annette J Dobson
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia.
| | - Hsin-Fang Chung
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Gita D Mishra
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| |
Collapse
|
22
|
Schweitzer N, Son SJ, Thurston RC, Li J, Chen CL, Aizenstein H, Yang S, Iordanova B, Hong CH, Roh HW, Cho YH, Hong S, Nam YJ, Lee DY, Park B, Kim NR, Choi JW, Cheong J, Seo SW, An YS, Moon SY, Han SJ, Wu M. Sex-Specific Risk Factors and Clinical Dementia Outcomes for White Matter Hyperintensities in a large South Korean Cohort. RESEARCH SQUARE 2024:rs.3.rs-4473148. [PMID: 38947089 PMCID: PMC11213210 DOI: 10.21203/rs.3.rs-4473148/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Objective White matter hyperintensities (WMH) on brain MRI images are the most common feature of cerebral small vessel disease (CSVD). Studies have yielded divergent findings on the modifiable risk factors for WMH and WMH's impact on cognitive decline. Mounting evidence suggests sex differences in WMH burden and subsequent effects on cognition. Thus, we aimed to identify sex-specific modifiable risk factors for WMH. We then explored whether there were sex-specific associations of WMH to longitudinal clinical dementia outcomes. Methods Participants aged 49-89 years were recruited at memory clinics and underwent a T2-weighted fluid-attenuated inversion recovery (FLAIR) 3T MRI scan to measure WMH volume. Participants were then recruited for two additional follow-up visits, 1-2 years apart, where clinical dementia rating sum of boxes (CDR-SB) scores were measured. We first explored which known modifiable risk factors for WMH were significant when tested for a sex-interaction effect. We additionally tested which risk factors were significant when stratified by sex. We then tested to see whether WMH is longitudinally associated with clinical dementia that is sex-specific. Results The study utilized data from 713 participants (241 males, 472 females) with a mean age of 72.3 years and 72.8 years for males and females, respectively. 57.3% and 59.5% of participants were diagnosed with mild cognitive impairment (MCI) for males and females, respectively. 40.7% and 39.4% were diagnosed with dementia for males and females, respectively. Of the 713 participants, 181 participants had CDR-SB scores available for three longitudinal time points. Compared to males, females showed stronger association of age to WMH volume. Type 2 Diabetes was associated with greater WMH burden in females but not males. Finally, baseline WMH burden was associated with worse clinical dementia outcomes longitudinally in females but not in males. Discussion Elderly females have an accelerated increase in cerebrovascular burden as they age, and subsequently are more vulnerable to clinical dementia decline due to CSVD. Additionally, females are more susceptible to the cerebrovascular consequences of diabetes. These findings emphasize the importance of considering sex when examining the consequences of CSVD. Future research should explore the underlying mechanisms driving these sex differences and personalized prevention and treatment strategies. Clinical trial registration The BICWALZS is registered in the Korean National Clinical Trial Registry (Clinical Research Information Service; identifier, KCT0003391). Registration Date 2018/12/14.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sang Woon Seo
- Samsung Medical Centre, Sungkyunkwan University School of Medicine
| | | | | | | | - Minjie Wu
- University of Pittsburgh School of Medicine
| |
Collapse
|
23
|
Boa Sorte Silva NC, Barha CK, Erickson KI, Kramer AF, Liu-Ambrose T. Physical exercise, cognition, and brain health in aging. Trends Neurosci 2024; 47:402-417. [PMID: 38811309 DOI: 10.1016/j.tins.2024.04.004] [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: 11/17/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/31/2024]
Abstract
Exercise training is an important strategy to counteract cognitive and brain health decline during aging. Evidence from systematic reviews and meta-analyses supports the notion of beneficial effects of exercise in cognitively unimpaired and impaired older individuals. However, the effects are often modest, and likely influenced by moderators such as exercise training parameters, sample characteristics, outcome assessments, and control conditions. Here, we discuss evidence on the impact of exercise on cognitive and brain health outcomes in healthy aging and in individuals with or at risk for cognitive impairment and neurodegeneration. We also review neuroplastic adaptations in response to exercise and their potential neurobiological mechanisms. We conclude by highlighting goals for future studies, including addressing unexplored neurobiological mechanisms and the inclusion of under-represented populations.
Collapse
Affiliation(s)
- Nárlon C Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Cindy K Barha
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA; AdventHealth Research Institute, Neuroscience, Orlando, FL, USA
| | - Arthur F Kramer
- Center for Cognitive and Brain Health, Northeastern University, Boston, MA, USA; Beckman Institute, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.
| |
Collapse
|
24
|
Xie J, You Y, Zheng P, Chen Y, Guo S, Xu Y, Huang J, Liu Z, Tao J. Gender differences in the association between physical activity and cognitive subdomains among elders with type 2 diabetes and mild cognitive impairment: a cross-sectional study. BMJ Open 2024; 14:e080789. [PMID: 38806426 PMCID: PMC11138272 DOI: 10.1136/bmjopen-2023-080789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/21/2024] [Indexed: 05/30/2024] Open
Abstract
OBJECTIVES The objective of this study was to evaluate the gender differences in the correlation between physical activity (PA) and cognitive subdomains in elderly individuals with type 2 diabetes (T2D) and mild cognitive impairment (MCI). DESIGN Cross-sectional study. SETTING The research was carried out in communities located in Fuzhou, Fujian Province and Beijing Municipality. PARTICIPANTS Community-dwelling elders with T2D and MCI aged 60 years or older were eligible for this study. PRIMARY OUTCOME MEASURES AND ANALYSES The weekly PA score was assessed using the International Physical Activity Questionnaire (IPAQ). The cognitive subdomains were evaluated through a battery of cognitive assessments, including the Rey Auditory Verbal Learning Test (RAVLT), Trail Making Test Part B, Digit Symbol Substitution Test (DSST) and the Stroop Color-Word Test (SCWT). Multiple linear regression models were employed to examine the association between PA and cognitive subdomains in both male and female individuals. RESULTS In older men, higher total IPAQ score was positively correlated with higher RAVLT (P=0.011) and SCWT (P=0.049). There was a significant interaction between the total PA score and gender in relation to RAVLT (P=0.008) and SCWT (P=0.027). Moreover, there was a positive correlation between moderate-vigorous PA level and RAVLT in older men (P=0.007). Additionally, a positive correlation was found between moderate-vigorous PA level and DSST in older women (P=0.038). CONCLUSION In older individuals with T2D and MCI, the association between PA and cognitive subdomains differs between men and women. This discrepancy may impact the customisation of exercise recommendations.
Collapse
Affiliation(s)
- Jinjin Xie
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yue You
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Peiyun Zheng
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yannan Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Shuai Guo
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Ying Xu
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jia Huang
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhizhen Liu
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
25
|
Jiang Z, Sun Y, Liu S. Association between human blood metabolites and cerebral cortex architecture: evidence from a Mendelian randomization study. Front Neurol 2024; 15:1386844. [PMID: 38784905 PMCID: PMC11111910 DOI: 10.3389/fneur.2024.1386844] [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: 02/16/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Background Dysregulation of circulating metabolites may affect brain function and cognition, associated with alterations in the cerebral cortex architecture. However, the exact cause remains unclear. This study aimed to determine the causal effect of circulating metabolites on the cerebral cortex architecture. Methods This study utilized retrieved data from genome-wide association studies to investigate the relationship between blood metabolites and cortical architecture. A total of 1,091 metabolites and 309 metabolite ratios were used for exposure. The brain cortex surface area and cortex thickness were selected as the primary outcomes in this study. In this study, the inverse variance weighting method was used as the main analytical method, complemented by sensitivity analyses that were more robust to pleiotropy. Furthermore, metabolic pathway analysis was performed via MetaboAnalyst 6.0. Finally, reverse Mendelian randomization (MR) analysis was conducted to assess the potential for reverse causation. Results After correcting for the false discovery rate (FDR), we identified 37 metabolites and 9 metabolite ratios that showed significant causal associations with cortical structures. Among these, Oxalate was found to be most strongly associated with cortical surface area (β: 2387.532, 95% CI 756.570-4018.495, p = 0.037), while Tyrosine was most correlated with cortical thickness (β: -0.015, 95% CI -0.005 to -0.025, p = 0.025). Furthermore, pathway analysis based on metabolites identified six significant metabolic pathways associated with cortical structures and 13 significant metabolic pathways based on metabolite ratios. Conclusion The identified metabolites and relevant metabolic pathways reveal potential therapeutic pathways for reducing the risk of neurodegenerative diseases. These findings will help guide health policies and clinical practice in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Zongzhi Jiang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yining Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
26
|
Wood Alexander M, Wu CY, Coughlan GT, Puri T, Buckley RF, Palta P, Swardfager W, Masellis M, Galea LAM, Einstein G, Black SE, Rabin JS. Associations Between Age at Menopause, Vascular Risk, and 3-Year Cognitive Change in the Canadian Longitudinal Study on Aging. Neurology 2024; 102:e209298. [PMID: 38569140 DOI: 10.1212/wnl.0000000000209298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/13/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Mounting evidence supports sex differences in Alzheimer disease (AD) risk. Vascular and hormonal factors may together contribute to AD risk in female adults. We investigated whether age at menopause, vascular risk, and history of hormone therapy (HT) containing estrogens together influence cognition over a 3-year follow-up period. We hypothesized that earlier menopause and elevated vascular risk would have a synergistic association with lower cognitive scores at follow-up and that HT containing estrogens would attenuate this synergistic association to preserve cognition. METHODS We used data from postmenopausal female participants and age-matched male participants in the Canadian Longitudinal Study on Aging. Vascular risk was calculated using a summary score of elevated blood pressure, antihypertensive medications, elevated low-density lipoprotein cholesterol, diabetes, smoking, and obesity. Cognition was measured with a global cognitive composite at baseline and 3-year follow-up. Linear models tested independent and interactive associations of age at menopause, vascular risk, and HT history with cognition at 3-year follow-up, adjusting for baseline cognition, baseline age, years of education, and test language (English/French). RESULTS We included 8,360 postmenopausal female participants (mean age at baseline = 65.0 ± 8.53 years, mean age at menopause = 50.1 ± 4.62 years) and 8,360 age-matched male participants for comparison. There was an interaction between age at menopause and vascular risk, such that earlier menopause and higher vascular risk were synergistically associated with lower cognitive scores at follow-up (β = 0.013, 95% CI 0.001-0.025, p = 0.03). In stratified analyses, vascular risk was associated with lower cognitive scores in female participants with earlier menopause (menopausal ages 35-48 years; β = -0.044, 95% CI -0.066 to -0.022, p < 0.001), but not average (ages 49-52 years; β = -0.007, 95% CI -0.027 to 0.012, p = 0.46) or later menopause (ages 53-65 years; β = 0.003, 95% CI -0.020 to 0.025, p = 0.82). The negative association of vascular risk with cognition in female participants with earlier menopause was stronger than the equivalent association in age-matched male participants. HT history did not further modify the synergistic association of age at menopause and vascular risk with follow-up cognition (β = -0.005, 95% CI -0.032 to 0.021, p = 0.69). DISCUSSION Endocrine and vascular processes may synergistically contribute to increased risk of cognitive decline in female adults. These findings have implications for the development of sex-specific dementia prevention strategies.
Collapse
Affiliation(s)
- Madeline Wood Alexander
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Che-Yuan Wu
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Gillian T Coughlan
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Tanvi Puri
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Rachel F Buckley
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Priya Palta
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Walter Swardfager
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Mario Masellis
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Liisa A M Galea
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Gillian Einstein
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Sandra E Black
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Jennifer S Rabin
- From the Hurvitz Brain Sciences Program (M.W.A., C.-Y.W., W.S., M.M., S.E.B., J.S.R.), Sunnybrook Research Institute; Rehabilitation Sciences Institute (M.W.A., J.S.R.), Department of Pharmacology & Toxicology (C.-Y.W., W.S.), University of Toronto, Ontario, Canada; Department of Neurology (G.T.C., R.F.B.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Psychology (T.P.), University of British Columbia, Vancouver, Canada; Center for Alzheimer Research and Treatment (CART) (R.F.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Parkville, Victoria, Australia; Department of Neurology (P.P.), University of North Carolina at Chapel Hill School of Medicine; Division of Neurology (M.M., S.E.B., J.S.R.), Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto; Campbell Family Mental Health Research Institute (L.A.M.G.), The Centre for Addition and Mental Health; Department of Psychiatry (L.A.M.G.), Temerty Faculty of Medicine, Dalla Lana School of Public Health (G.E.), and Department of Psychology (G.E.), University of Toronto; Rotman Research Institute (G.E.), Baycrest Hospital; and Harquail Centre for Neuromodulation (J.S.R.), Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| |
Collapse
|
27
|
Yang Q, Han X, Ye M, Jiang T, Wang B, Zhang Z, Li F. Association of genetically predicted 486 blood metabolites on the risk of Alzheimer's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1372605. [PMID: 38681667 PMCID: PMC11047179 DOI: 10.3389/fnagi.2024.1372605] [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: 01/18/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Background Studies have reported that metabolic disturbance exhibits in patients with Alzheimer's disease (AD). Still, the presence of definitive evidence concerning the genetic effect of metabolites on AD risk remains insufficient. A systematic exploration of the genetic association between blood metabolites and AD would contribute to the identification of new targets for AD screening and prevention. Methods We conducted an exploratory two-sample Mendelian randomization (MR) study aiming to preliminarily identify the potential metabolites involved in AD development. A genome-wide association study (GWAS) involving 7,824 participants provided information on 486 human blood metabolites. Outcome information was obtained from a large-scale GWAS meta-analysis of AD, encompassing 21,982 cases and 41,944 controls of Europeans. The primary two-sample MR analysis utilized the inverse variance weighted (IVW) model while supplementary analyses used Weighted median (WM), MR Egger, Simple mode, and Weighted mode, followed by sensitivity analyses such as the heterogeneity test, horizontal pleiotropy test, and leave-one-out analysis. For the further identification of metabolites, replication and meta-analysis with FinnGen data, steiger test, linkage disequilibrium score regression, confounding analysis, and were conducted for further evaluation. Multivariable MR was performed to assess the direct effect of metabolites on AD. Besides, an extra replication analysis with EADB data was conducted for final evaluation of the most promising findings. Results After rigorous genetic variant selection, IVW, complementary analysis, sensitivity analysis, replication and meta-analysis with the FinnGen data, five metabolites (epiandrosterone sulfate, X-12680, pyruvate, docosapentaenoate, and 1-stearoylglycerophosphocholine) were identified as being genetically associated with AD. MVMR analysis disclosed that genetically predicted these four known metabolites can directly influence AD independently of other metabolites. Only epiandrosterone sulfate and X-12680 remained suggestive significant associations with AD after replication analysis with the EADB data. Conclusion By integrating genomics with metabonomics, this study furnishes evidence substantiating the genetic association of epiandrosterone sulfate and X-12680 with AD. These findings hold significance for the screening, prevention, and treatment strategies for AD.
Collapse
Affiliation(s)
- Qiqi Yang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Xinyu Han
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Min Ye
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Tianxin Jiang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Baoguo Wang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhenfeng Zhang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Fei Li
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Intelligent Manufacturing Institute, Hefei University of Technology, Hefei, China
| |
Collapse
|
28
|
Cha WJ, Yi D, Ahn H, Byun MS, Chang YY, Choi JM, Kim K, Choi H, Jung G, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Association between brain amyloid deposition and longitudinal changes of white matter hyperintensities. Alzheimers Res Ther 2024; 16:50. [PMID: 38454444 PMCID: PMC10918927 DOI: 10.1186/s13195-024-01417-8] [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: 11/04/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence suggests that not only cerebrovascular disease but also Alzheimer's disease (AD) pathological process itself cause cerebral white matter degeneration, resulting in white matter hyperintensities (WMHs). Some preclinical evidence also indicates that white matter degeneration may precede or affect the development of AD pathology. This study aimed to clarify the direction of influence between in vivo AD pathologies, particularly beta-amyloid (Aβ) and tau deposition, and WMHs through longitudinal approach. METHODS Total 282 older adults including cognitively normal and cognitively impaired individuals were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. The participants underwent comprehensive clinical and neuropsychological assessment, [11C] Pittsburgh Compound B PET for measuring Aβ deposition, [18F] AV-1451 PET for measuring tau deposition, and MRI scans with fluid-attenuated inversion recovery image for measuring WMH volume. The relationships between Aβ or tau deposition and WMH volume were examined using multiple linear regression analysis. In this analysis, baseline Aβ or tau were used as independent variables, and change of WMH volume over 2 years was used as dependent variable to examine the effect of AD pathology on increase of WMH volume. Additionally, we set baseline WMH volume as independent variable and longitudinal change of Aβ or tau deposition for 2 years as dependent variables to investigate whether WMH volume could precede AD pathologies. RESULTS Baseline Aβ deposition, but not tau deposition, had significant positive association with longitudinal change of WMH volume over 2 years. Baseline WMH volume was not related with any of longitudinal change of Aβ or tau deposition for 2 years. We also found a significant interaction effect between baseline Aβ deposition and sex on longitudinal change of WMH volume. Subsequent subgroup analyses showed that high baseline Aβ deposition was associated with increase of WMH volume over 2 years in female, but not in male. CONCLUSIONS Our findings suggest that Aβ deposition accelerates cerebral WMHs, particularly in female, whereas white matter degeneration appears not influence on longitudinal Aβ increase. The results also did not support any direction of influence between tau deposition and WMHs.
Collapse
Affiliation(s)
- Woo-Jin Cha
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jung-Min Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Umoh IO, dos Reis HJ, de Oliveira ACP. Molecular Mechanisms Linking Osteoarthritis and Alzheimer's Disease: Shared Pathways, Mechanisms and Breakthrough Prospects. Int J Mol Sci 2024; 25:3044. [PMID: 38474288 PMCID: PMC10931612 DOI: 10.3390/ijms25053044] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease mostly affecting the elderly population. It is characterized by cognitive decline that occurs due to impaired neurotransmission and neuronal death. Even though deposition of amyloid beta (Aβ) peptides and aggregation of hyperphosphorylated TAU have been established as major pathological hallmarks of the disease, other factors such as the interaction of genetic and environmental factors are believed to contribute to the development and progression of AD. In general, patients initially present mild forgetfulness and difficulty in forming new memories. As it progresses, there are significant impairments in problem solving, social interaction, speech and overall cognitive function of the affected individual. Osteoarthritis (OA) is the most recurrent form of arthritis and widely acknowledged as a whole-joint disease, distinguished by progressive degeneration and erosion of joint cartilage accompanying synovitis and subchondral bone changes that can prompt peripheral inflammatory responses. Also predominantly affecting the elderly, OA frequently embroils weight-bearing joints such as the knees, spine and hips leading to pains, stiffness and diminished joint mobility, which in turn significantly impacts the patient's standard of life. Both infirmities can co-occur in older adults as a result of independent factors, as multiple health conditions are common in old age. Additionally, risk factors such as genetics, lifestyle changes, age and chronic inflammation may contribute to both conditions in some individuals. Besides localized peripheral low-grade inflammation, it is notable that low-grade systemic inflammation prompted by OA can play a role in AD pathogenesis. Studies have explored relationships between systemic inflammatory-associated diseases like obesity, hypertension, dyslipidemia, diabetes mellitus and AD. Given that AD is the most common form of dementia and shares similar risk factors with OA-both being age-related and low-grade inflammatory-associated diseases, OA may indeed serve as a risk factor for AD. This work aims to review literature on molecular mechanisms linking OA and AD pathologies, and explore potential connections between these conditions alongside future prospects and innovative treatments.
Collapse
Affiliation(s)
| | - Helton Jose dos Reis
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| | - Antonio Carlos Pinheiro de Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil;
| |
Collapse
|
30
|
Panghal A, Flora SJS. Nanotechnology in the diagnostic and therapy for Alzheimer's disease. Biochim Biophys Acta Gen Subj 2024; 1868:130559. [PMID: 38191034 DOI: 10.1016/j.bbagen.2024.130559] [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: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder primarily characterized by β-amyloid plaque, intraneuronal tangles, significant neuronal loss and cognitive deficit. Treatment in the early stages of the disease is crucial for preventing or perhaps reversing the neurodegeneration in the AD cases. However, none of the current diagnostic procedures are capable of early diagnosis of AD. Further, the available treatments merely provide symptomatic alleviation in AD and do not address the underlying illness. Therefore, there is no permanent cure for AD currently. Better therapeutic outcomes need the optimum drug concentration in the central nervous system (CNS) by traversing blood-brain-barrier (BBB). Nanotechnology offers enormous promise to transform the treatment and diagnostics of neurodegenerative diseases. Nanotechnology based diagnostic tools, drug delivery systems and theragnostic are capable of highly sensitive molecular detection, effective drug targeting and their combination. Significant work has been done in this area over the last decade and prospective results have been obtained in AD therapy. This review explores the various applications of nanotechnology in addressing the varied facets of AD, ranging from early detection to therapeutic interventions. This review also looks at how nanotechnology can help with the development of disease-modifying medicines, such as the delivery of anti-amyloid, anti-tau, cholinesterase inhibitors, antioxidants and hormonal drugs. In conclusion, this paper discusses the role of nanotechnology in the early detection of AD, effective drug targeting to the CNS and theragnostic applications in the management of AD.
Collapse
Affiliation(s)
- Archna Panghal
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India; Institute of Pharmaceutical Sciences, Era Medical University, Safarajganj, Lucknow 226003, U.P., India.
| |
Collapse
|
31
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
32
|
Tang S, Huang L, Lin F, Chen X, Wang Y, Xu J, Wang Y, Gao J, Xiao Q. Sex steroid and cognitive function among community-dwelling older men with or without vascular risk factors: a cross-sectional study. BMC Geriatr 2024; 24:147. [PMID: 38350861 PMCID: PMC10865607 DOI: 10.1186/s12877-024-04727-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The relationship of testosterone and estradiol concentrations with cognitive function among community-dwelling older men was inconclusive. To examine the association of serum testosterone and estradiol concentrations with cognitive function in older men with or without vascular risk factors (VRFs). METHODS This cross-sectional study consisted of 224 community-dwelling men aged 65-90 years in the Songjiang District of Shanghai, China. Serum testosterone and estradiol were measured by electrochemiluminescence immunoassay. The following five factors were defined as VRFs in this study: obesity, history of hypertension, diabetes, stroke, and coronary heart disease. Multivariable linear regression was used to examine the association of testosterone and estradiol with the Mini-Mental State Examination (MMSE) in participants with or without VRF. Restricted cubic spline (RCS) regression was performed to account for the nonlinearity of these associations. RESULTS An inverted "U" shaped non-linear relationship was found between testosterone concentration and MMSE score in men with one VRF (P overall =.003, non-linear P =.002). Estradiol showed an inverted "U" shaped non-linear relationship with MMSE score independent of VRFs (men without VRF, P overall =.049, non-linear P =.015; men with one VRF, overall P =.007, non-linear P =.003; men with two or more VRFs, overall P =.009, non-linear P =.005). CONCLUSION In older men, an optimal level of sex steroid concentration may be beneficial to cognitive function and the VRFs should be considered when interpreting the relationship between sex steroid and cognitive function.
Collapse
Affiliation(s)
- Shuning Tang
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China
- Health Communication Institute, Fudan University, 200032, Shanghai, China
| | - Limei Huang
- Songjiang Center of Disease Prevention and Control, 201620, Shanghai, China
| | - Fangting Lin
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China
- Health Communication Institute, Fudan University, 200032, Shanghai, China
| | - Xiuqin Chen
- Songjiang Center of Disease Prevention and Control, 201620, Shanghai, China
| | - Yunhui Wang
- Songjiang District Xinqiao Town Community Health Service Center, 201600, Shanghai, China
| | - Jixiang Xu
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China
- Health Communication Institute, Fudan University, 200032, Shanghai, China
| | - Yujie Wang
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China
- Health Communication Institute, Fudan University, 200032, Shanghai, China
| | - Junling Gao
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China.
- Health Communication Institute, Fudan University, 200032, Shanghai, China.
| | - Qianyi Xiao
- Department of Preventive Medicine and Health Education, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, 200032, Shanghai, China.
- Health Communication Institute, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
33
|
Kommaddi RP, Gowaikar R, P A H, Diwakar L, Singh K, Mondal A. Akt activation ameliorates deficits in hippocampal-dependent memory and activity-dependent synaptic protein synthesis in an Alzheimer's disease mouse model. J Biol Chem 2024; 300:105619. [PMID: 38182004 PMCID: PMC10839450 DOI: 10.1016/j.jbc.2023.105619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/07/2024] Open
Abstract
Protein kinase-B (Akt) and the mechanistic target of rapamycin (mTOR) signaling pathways are implicated in Alzheimer's disease (AD) pathology. Akt/mTOR signaling pathways, activated by external inputs, enable new protein synthesis at the synapse and synaptic plasticity. The molecular mechanisms impeding new protein synthesis at the synapse in AD pathogenesis remain elusive. Here, we aimed to understand the molecular mechanisms prior to the manifestation of histopathological hallmarks by characterizing Akt1/mTOR signaling cascades and new protein synthesis in the hippocampus of WT and amyloid precursor protein/presenilin-1 (APP/PS1) male mice. Intriguingly, compared to those in WT mice, we found significant decreases in pAkt1, pGSK3β, pmTOR, pS6 ribosomal protein, and p4E-BP1 levels in both post nuclear supernatant and synaptosomes isolated from the hippocampus of one-month-old (presymptomatic) APP/PS1 mice. In synaptoneurosomes prepared from the hippocampus of presymptomatic APP/PS1 mice, activity-dependent protein synthesis at the synapse was impaired and this deficit was sustained in young adults. In hippocampal neurons from C57BL/6 mice, downregulation of Akt1 precluded synaptic activity-dependent protein synthesis at the dendrites but not in the soma. In three-month-old APP/PS1 mice, Akt activator (SC79) administration restored deficits in memory recall and activity-dependent synaptic protein synthesis. C57BL/6 mice administered with an Akt inhibitor (MK2206) resulted in memory recall deficits compared to those treated with vehicle. We conclude that dysregulation of Akt1/mTOR and its downstream signaling molecules in the hippocampus contribute to memory recall deficits and loss of activity-dependent synaptic protein synthesis. In AD mice, however, Akt activation ameliorates deficits in memory recall and activity-dependent synaptic protein synthesis.
Collapse
Affiliation(s)
| | - Ruturaj Gowaikar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India; Manipal Academy of Higher Education, Manipal, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Kunal Singh
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Amrita Mondal
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
34
|
Ekanayake A, Peiris S, Ahmed B, Kanekar S, Grove C, Kalra D, Eslinger P, Yang Q, Karunanayaka P. A Review of the Role of Estrogens in Olfaction, Sleep and Glymphatic Functionality in Relation to Sex Disparity in Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2024; 39:15333175241272025. [PMID: 39116421 PMCID: PMC11311174 DOI: 10.1177/15333175241272025] [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] [Indexed: 08/10/2024]
Abstract
Several risk factors contribute to the development of Alzheimer's disease (AD), including genetics, metabolic health, cardiovascular history, and diet. It has been observed that women appear to face a higher risk of developing AD. Among the various hypotheses surrounding the gender disparity in AD, one pertains to the potential neuroprotective properties of estrogen. Compared to men, women are believed to be more susceptible to neuropathology due to the significant decline in circulating estrogen levels following menopause. Studies have shown, however, that estrogen replacement therapies in post-menopausal women do not consistently reduce the risk of AD. While menopause and estrogen levels are potential factors in the elevated incidence rates of AD among women, this review highlights the possible roles estrogen has in other pathways that may also contribute to the sex disparity observed in AD such as olfaction, sleep, and glymphatic functionality.
Collapse
Affiliation(s)
- Anupa Ekanayake
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Grodno State Medical University, Grodno, Belarus
| | - Senal Peiris
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Biyar Ahmed
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Sangam Kanekar
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Cooper Grove
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Deepak Kalra
- Department of Neurology, Penn State University College of Medicine, Hershey, PA, USA
| | - Paul Eslinger
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Department of Neurology, Penn State University College of Medicine, Hershey, PA, USA
| | - Qing Yang
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Department of Neurosurgery, Penn State University College of Medicine, Hershey, PA, USA
| | - Prasanna Karunanayaka
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
35
|
Abstract
Androgens play a key biological role in libido and sexual arousal in women, and knowledge about their complex role in other systems remains ambiguous and incomplete. This narrative review examines the role of endogenous androgens in women's health throughout the life span before focusing on evidence surrounding the use of androgen-based therapies to treat postmenopausal women. The role of testosterone as a therapeutic agent in women continues to attract controversy as approved preparations are rare, and use of off-label and compounded formulations is widespread. Despite this androgen therapy has been used for decades in oral, injectable, and transdermal formulations. Responses to androgen therapy have been demonstrated to improve aspects of female sexual dysfunction, notably hypoactive sexual desire disorder, in a dose related manner. Substantial research has also been conducted into the role of androgens in treating aspects of the genitourinary syndrome of menopause (GSM). Evidence for benefits beyond these is mixed and more research is required regarding long-term safety. However, It remains biologically plausible that androgens will be effective in treating hypoestrogenic symptoms related to menopause, either through direct physiological effects or following aromatization to estradiol throughout the body.
Collapse
Affiliation(s)
- Kath Whitton
- North Shore Private Hospital, Level 3, Suite 7, 3 Westbourne Street, St Leonards, NSW 2065, Australia
| | - Rodney Baber
- Obstetrics, Gynaecology and Neonatology, Northern Clinical School, The University of Sydney, Australia.
| |
Collapse
|
36
|
Reed EG, Keller-Norrell PR. Minding the Gap: Exploring Neuroinflammatory and Microglial Sex Differences in Alzheimer's Disease. Int J Mol Sci 2023; 24:17377. [PMID: 38139206 PMCID: PMC10743742 DOI: 10.3390/ijms242417377] [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: 11/20/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Research into Alzheimer's Disease (AD) describes a link between AD and the resident immune cells of the brain, the microglia. Further, this suspected link is thought to have underlying sex effects, although the mechanisms of these effects are only just beginning to be understood. Many of these insights are the result of policies put in place by funding agencies such as the National Institutes of Health (NIH) to consider sex as a biological variable (SABV) and the move towards precision medicine due to continued lackluster therapeutic options. The purpose of this review is to provide an updated assessment of the current research that summarizes sex differences and the research pertaining to microglia and their varied responses in AD.
Collapse
Affiliation(s)
- Erin G. Reed
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44242, USA
| | | |
Collapse
|
37
|
Jiang C, Wang Y, Yang W, Yang X. New evidence for the effect of type 2 diabetes and glycemic traits on testosterone levels: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1238090. [PMID: 37900148 PMCID: PMC10600375 DOI: 10.3389/fendo.2023.1238090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Objective Type 2 diabetes mellitus (T2DM) is an endocrine-related disease with an increasing incidence worldwide. Male sexual dysfunction is common in diabetic patients. Therefore, we designed a Mendelian randomization (MR) study to investigate the association of type 2 diabetes and 3 glycemic traits with testosterone levels. Methods Uncorrelated single nucleotide polymorphisms (SNPs) associated with T2DM (N = 228), fasting insulin (N = 38), fasting glucose (N = 71), and HbA1c (N = 75) at the genome-wide significance were selected as instrument variables. Genetic associations with testosterone levels (total testosterone, TT, bioavailable testosterone, BT, and sex hormone-binding globulin, SHBG) were obtained from the UK Biobank studies and other large consortia. Two-sample MR analysis was used to minimize the bias caused by confounding factors and response causality. Multivariable MR analysis was performed using Body mass index (BMI), Triglycerides (TG), LDL cholesterol (LDL), and adiponectin to adjust for the effects of potential confounders. Results Type 2 diabetes mellitus was associated with the decrease of total testosterone (β: -0.021,95%CI: -0.032, -0.010, p<0.001) and sex hormone binding globulin (β: -0.048,95%CI: -0.065, -0.031, p<0.001). In males, total testosterone (β: 0.058, 95% CI: 0.088, 0.028, p < 0.001) decreased. In females, it was associated with an increase in bioavailable testosterone (β: 0.077,95%CI: 0.058,0.096, p<0.001). Each unit (pmol/L) increase in fasting insulin was associated with 0.283nmol/L decrease in sex hormone-binding globulin (95%CI: -0.464, -0.102, p=0.002) and 0.260nmol/L increase in bioavailable testosterone (95%CI: -0.464, -0.102, p= 0.002). In males, sex hormone binding globulin decreased by 0.507nmol/L (95%CI: -0.960, -0.054, p= 0.028) and bioavailable testosterone increased by 0.216nmol/L (95%CI: 0.087,0.344, p= 0.001). In females, sex hormone binding globulin decreased by 0.714 nmol/L (95%CI: -1.093, -0.335, p<0.001) and bioavailable testosterone increased by 0.467nmol/L (95%CI: 0.286,0.648, p<0.001). Each unit (%) increase in HbA1c was associated with 0.060nmol/L decrease in sex hormone-binding globulin (95%CI: -0.113, -0.007, p= 0.026). In males, total testosterone decreased by 0.171nmol/L (95%CI: -0.288, -0.053, p=0.005) and sex hormone binding globulin decreased by 0.206nmol/L (95%CI: -0.340, -0.072, p=0.003). Total testosterone increased by 0.122nmol/L (95%CI: 0.012,0.233, p=0.029) and bioavailable testosterone increased by 0.163nmol/L (95%CI: 0.042,0.285, p=0.008) in females. Conclusions Using MR Analysis, we found independent effects of type 2 diabetes, fasting insulin, and HbA1c on total testosterone and sex hormone-binding globulin after maximum exclusion of the effects of obesity, BMI, TG, LDL and Adiponectin.
Collapse
Affiliation(s)
- Chengyang Jiang
- Department of Pediatric Surgery, Maternal and Child Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuwei Wang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Wenqiang Yang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xinghai Yang
- Department of Pediatric Surgery, Maternal and Child Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
38
|
Islam RM, Bell RJ, Berk M, Handelsman DJ, McNeil JJ, Wolfe R, Woods RL, Davis SR. Associations between low sex hormone concentrations and depression in older women: An observational study. Maturitas 2023; 176:107822. [PMID: 37591034 DOI: 10.1016/j.maturitas.2023.107822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/22/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVE We investigated whether low sex hormone concentrations are associated with depression in older women. STUDY DESIGN This was a cross-sectional study of Australian women, aged at least 70 years, not taking medications modulating sex hormone levels. Associations between hormones, measured by liquid chromatography-tandem mass spectrometry, and depression were examined by logistic regression adjusted for potential confounders. MAIN OUTCOME MEASURES The primary outcome was a Center for Epidemiologic Studies Depression score >10, designated as 'depression', with an expanded definition that included anti-depressant use as a secondary outcome. RESULTS For the 5535 participants in the analysis, median age 74.0 years (interquartile range 71.7-77.7), depression prevalence was 5.8 % (95 % CI 5.2-6.4 %). In the adjusted models, a statistically significantly greater likelihood of depression was seen for women with testosterone and oestrone blood concentrations in quartile 1 compared with quartiles 2-4 (odds ratio 1.33, 95 % CI 1.04 to 1.70, p = 0.022; and 1.37, 95 % CI 1.06 to 1.78, p = 0.017, respectively). For the expanded definition, the odds ratios for the lowest testosterone and oestrone quartile compared with other quartiles were 1.47 (95 % CI 1.24 to 1.75, p < 0.001) and 1.31 (95 % CI 1.09 to 1.58, p < 0.001), respectively. A significant association for low DHEA was seen only for the expanded definition of depression (1.36, 95 % CI 1.13 to 1.64, p = 0.001). Receiver operating characteristic curves showed that the contribution of each sex hormone to the likelihood of depression was small. CONCLUSIONS Amongst older women not taking medications that influence sex hormone concentrations, low testosterone and oestrone levels are associated with a greater likelihood of depression, but the effects are small. TRIAL REGISTRATION International Standard Randomized Controlled Trial Number Register (ISRCTN83772183) and clinicaltrials.gov (NCT01038583).
Collapse
Affiliation(s)
- Rakibul M Islam
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - Robin J Bell
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation Strategy Research Centre, Deakin University School of Medicine, Geelong, Australia; Department of Psychiatry, University of Melbourne, Parkville, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, New South Wales 2139, Australia.
| | - John J McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.
| | - Susan R Davis
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; Department of Endocrinology and Diabetes, Alfred Health, Melbourne 3004, Australia.
| |
Collapse
|
39
|
Schmidt M, Vaskova M, Rotterova A, Fiandova P, Miskerikova M, Zemanova L, Benek O, Musilek K. Physiologically relevant fluorescent assay for identification of 17β-hydroxysteroid dehydrogenase type 10 inhibitors. J Neurochem 2023; 167:154-167. [PMID: 37458164 DOI: 10.1111/jnc.15917] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial enzyme 17β-hydroxysteroid dehydrogenase type 10 (HSD10) is a potential molecular target for treatment of mitochondrial-related disorders such as Alzheimer's disease (AD). Its over-expression in AD brains is one of the critical factors disturbing the homeostasis of neuroprotective steroids and exacerbating amyloid beta (Aβ)-mediated mitochondrial toxicity and neuronal stress. This study was focused on revalidation of the most potent HSD10 inhibitors derived from benzothiazolyl urea scaffold using fluorescent-based enzymatic assay with physiologically relevant substrates of 17β-oestradiol and allopregnanolone. The oestradiol-based assay led to the identification of two nanomolar inhibitors (IC50 70 and 346 nM) differing from HSD10 hits revealed from the formerly used assay. Both identified inhibitors were found to be effective also in allopregnanolone-based assay with non-competitive or uncompetitive mode of action. In addition, both inhibitors were confirmed to penetrate the HEK293 cells and they were able to inhibit the HSD10 enzyme in the cellular environment. Both molecules seem to be potential lead structures for further research and development of HDS10 inhibitors.
Collapse
Affiliation(s)
- Monika Schmidt
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Michaela Vaskova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Aneta Rotterova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Pavlina Fiandova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marketa Miskerikova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Lucie Zemanova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Benek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
40
|
Davis SR, Pinkerton J, Santoro N, Simoncini T. Menopause-Biology, consequences, supportive care, and therapeutic options. Cell 2023; 186:4038-4058. [PMID: 37678251 DOI: 10.1016/j.cell.2023.08.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/06/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023]
Abstract
Menopause is the cessation of ovarian function, with loss of reproductive hormone production and irreversible loss of fertility. It is a natural part of reproductive aging. The physiology of the menopause is complex and incompletely understood. Globally, menopause occurs around the age of 49 years, with geographic and ethnic variation. The hormonal changes of the menopause transition may result in both symptoms and long-term systemic effects, predominantly adverse effects on cardiometabolic and musculoskeletal health. The most effective treatment for bothersome menopausal symptoms is evidence-based, menopausal hormone therapy (MHT), which reduces bone loss and may have cardiometabolic benefits. Evidence-based non-hormonal interventions are also available for symptom relief. Treatment should be individualized with shared decision-making. Most MHT regimens are not regulator approved for perimenopausal women. Studies that include perimenopausal women are needed to determine the efficacy and safety of treatment options. Further research is crucial to improve menopause care, along with research to guide policy and clinical practice.
Collapse
Affiliation(s)
- Susan R Davis
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Endocrinology and Diabetes, Alfred Health, Commercial Rd., Melbourne, VIC 3004, Australia.
| | - JoAnn Pinkerton
- Department of Obstetrics and Gynecology, Division of Midlife Health, The University of Virginia Health System, Charlottesville, VA, USA
| | | | - Tommaso Simoncini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
41
|
Zahedi E, Sanaeierad A, Nikbakhtzadeh M, Roghani M, Zamani E. Simvastatin improves learning and memory impairment via gut-brain axis regulation in an ovariectomized/D-galactose Alzheimer's rat model. Behav Brain Res 2023; 453:114611. [PMID: 37541447 DOI: 10.1016/j.bbr.2023.114611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023]
Abstract
AIM Alzheimer's disease (AD) is the most prevalent form of dementia with multiple etiology and no effective remedy. Statins are a group of medicines that are basically used to lower cholesterol. However, several studies have recently done to assess the potential relationship between statins use and dementia but presented controversial results. METHODS In this study, using ovariectomy and D-galactose injection, a model of AD was induced in female rats, and then the protective effects of oral administration of simvastatin were investigated. shuttle box and Y-maze tests were done to assess the animals' learning and memory performance. Using GC-MC, ELISA, Immunohistochemistry and tissue staining techniques, changes in the amount of short-chain fatty acids (SCFAs), plasma and hippocampus neuroinflammatory markers and histological changes in the intestine and hippocampus were assessed in sham, disease and treatment groups. KEY FINDINGS Oral administration of simvastatin improved the gut microbiome activity (increased the amount of SCFAs in fecal samples) and strengthened the tight junctions of intestinal cells. Moreover, simvastatin reduced the amount of TNF-α and IL-1β in plasma and hippocampus. Also, cell death and Amyloid plaques notably decreased in the simvastatin-treated hippocampal tissue. All these physiological changes led to better performance in behavioral tasks in the treatment group in comparison to the disease group. SIGNIFICANCE These findings provide evidence that simvastatin may improve gut-brain axis followed by improvement in learning and memory via an anti-inflammatory effect.
Collapse
Affiliation(s)
- Elham Zahedi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Sanaeierad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Elham Zamani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Psychology, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
42
|
Kusters CDJ, Paul KC, Romero T, Sinsheimer JS, Ritz BR. Among men, androgens are associated with a decrease in Alzheimer's disease risk. Alzheimers Dement 2023; 19:3826-3834. [PMID: 36938850 PMCID: PMC10509321 DOI: 10.1002/alz.13013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 03/21/2023]
Abstract
INTRODUCTION Increased levels of sex hormones have been hypothesized to decrease Alzheimer's disease (AD) risk. We assessed the association between sex steroid hormones with AD using a Mendelian randomization (MR) approach. METHODS An inverse-variance weighting (IVW) MR analysis was performed using effect estimates from external genome-wide association study (GWAS) summary statistics. We included independent variants (linkage disequilibrium R2 < 0.001) and a p-value threshold of 5 × 10-8 . RESULTS An increase in androgens was associated with a decreased AD risk among men: testosterone (odds ratio [OR]: 0.53; 95% confidence interval [CI]: 0.32-0.88; p-value: 0.01; false discovery rate [FDR] p-value: 0.03); dehydroepiandrosterone sulfate (DHEAS; OR: 0.56; 95% CI: 0.38-0.85; p-value: 0.01; FDR p-value: 0.03); and androsterone sulfate (OR: 0.69; 95% CI: 0.46-1.02; p-value: 0.06; FDR p-value: 0.10). There was no association between sex steroid hormones and AD among women, although analysis for estradiol had limited statistical power. DISCUSSION A higher concentration of androgens was associated with a decreased risk of AD among men of European ancestry, suggesting that androgens among men might be neuroprotective and could potentially prevent or delay an AD diagnosis. HIGHLIGHTS Sex hormones are hypothesized to play a role in developing Alzheimer's disease (AD). The effect of sex hormones on AD was assessed using Mendelian randomization (MR) analysis. Among women, genetically determined effects of sex hormones were limited or null. Among men, a higher concentration of androgens decreased AD risk. This study suggests a causal relationship between androgens and AD among men.
Collapse
Affiliation(s)
- Cynthia D J Kusters
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, California, USA
| | - Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine, Los Angeles, California, USA
| | - Tahmineh Romero
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Janet S Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine, Los Angeles, California, USA
- Department of Biostatistics, School of Public Health, University of California, Los Angeles, California, USA
- Department of Computational Medicine, David Geffen School of Medicine, Los Angeles, California, USA
| | - Beate R Ritz
- Department of Neurology, David Geffen School of Medicine, Los Angeles, California, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
- Department of Environmental Health, UCLA Fielding School of Public Health, Los Angeles, California, USA
| |
Collapse
|
43
|
Chen XL, Fortes JM, Hu YT, van Iersel J, He KN, van Heerikhuize J, Balesar R, Swaab D, Bao AM. Sexually dimorphic age-related molecular differences in the entorhinal cortex of cognitively intact elderly: Relation to early Alzheimer's changes. Alzheimers Dement 2023; 19:3848-3857. [PMID: 36960685 DOI: 10.1002/alz.13037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Women are more vulnerable to Alzheimer's disease (AD) than men. The entorhinal cortex (EC) is one of the earliest structures affected in AD. We identified in cognitively intact elderly different molecular changes in the EC in relation to age. METHODS Changes in 12 characteristic molecules in relation to age were determined by quantitative immunohistochemistry or in situ hybridization in the EC. They were arbitrarily grouped into sex steroid-related molecules, markers of neuronal activity, neurotransmitter-related molecules, and cholinergic activity-related molecules. RESULTS The changes in molecules indicated increasing local estrogenic and neuronal activity accompanied by a higher and faster hyperphosphorylated tau accumulation in women's EC in relation to age, versus a mainly stable local estrogenic/androgenic and neuronal activity in men's EC. DISCUSSION EC employs a different neurobiological strategy in women and men to maintain cognitive function, which seems to be accompanied by an earlier start of AD in women. HIGHLIGHTS Local estrogen system is activated with age only in women's entorhinal cortex (EC). EC neuronal activity increased with age only in elderly women with intact cognition. Men and women have different molecular strategies to retain cognition with aging. P-tau accumulation in the EC was higher and faster in cognitively intact elderly women.
Collapse
Affiliation(s)
- Xin-Lu Chen
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jennifer Monteiro Fortes
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Yu-Ting Hu
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Juliet van Iersel
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Kang-Ning He
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Joop van Heerikhuize
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rawien Balesar
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Dick Swaab
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Vila-Castelar C, Chen Y, Langella S, Lopera F, Zetterberg H, Hansson O, Dage JL, Janelidzde S, Su Y, Chen K, McDowell CP, Martinez JE, Ramirez-Gomez L, Garcia G, Aguillon D, Baena A, Giraldo-Chica M, Protas HD, Ghisays V, Rios-Romenets S, Tariot PN, Blennow K, Reiman EM, Quiroz YT. Sex differences in blood biomarkers and cognitive performance in individuals with autosomal dominant Alzheimer's disease. Alzheimers Dement 2023; 19:4127-4138. [PMID: 37279390 PMCID: PMC10527358 DOI: 10.1002/alz.13314] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Plasma tau phosphorylated at threonine 217 (P-tau217) and neurofilament light (NfL) have emerged as markers of Alzheimer's disease (AD) pathology. Few studies have examined the role of sex in plasma biomarkers in sporadic AD, yielding mixed findings, and none in autosomal dominant AD. METHODS We examined the effects of sex and age on plasma P-tau217 and NfL, and their association with cognitive performance in a cross-sectional study of 621 Presenilin-1 E280A mutation carriers (PSEN1) and non-carriers. RESULTS As plasma P-tau217 levels increase, cognitively unimpaired female carriers showed better cognitive performance than cognitively unimpaired male carriers. Yet, as disease progresses, female carriers had a greater plasma NfL increase than male carriers. There were no sex differences in the association between age and plasma biomarkers among non-carriers. DISCUSSION Our findings suggest that, among PSEN1 mutation carriers, females had a greater rate of neurodegeneration than males, yet it did not predict cognitive performance. HIGHLIGHTS We examined sex differences in plasma P-tau217 and NfL in Presenilin-1 E280A (PSEN1) mutation carriers and non-carriers. Female carriers had a greater plasma NfL increase, but not P-tau217, than male carriers. As plasma P-tau217 levels increase, cognitively unimpaired female carriers showed better cognitive performance than cognitively unimpaired male carriers. The interaction effect of sex by plasma NfL levels did not predict cognition among carriers.
Collapse
Affiliation(s)
- Clara Vila-Castelar
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Yinghua Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Stephanie Langella
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 405 30, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Oskar Hansson
- Memory Clinic, Skåne University Hospital, Malmö, 214 28, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 205 02, Sweden
| | - Jeffrey L. Dage
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, 85718, USA
| | - Celina Pluim McDowell
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, 02215, MA
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA
| | - Jairo E. Martinez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Psychological and Brain Sciences, Boston University, Boston, 02215, MA
| | | | - Gloria Garcia
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - David Aguillon
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | - Ana Baena
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | | | | | | | - Silvia Rios-Romenets
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 405 30, Sweden
| | | | - Yakeel T. Quiroz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, 1226, Colombia
| |
Collapse
|
45
|
Siegmann EM, Olm P, Lenz B, Mühle C, Oberstein TJ, Maler JM, Kornhuber J. Digit Ratio (2D:4D) Is Not Associated with Alzheimer's Disease in the Elderly. Brain Sci 2023; 13:1229. [PMID: 37759830 PMCID: PMC10526128 DOI: 10.3390/brainsci13091229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
The development of Alzheimer's disease (AD) is influenced by sex hormones-estrogens and androgens in particular. However, the impact of prenatal sex hormone exposure is less clear; very few investigations have examined the relationship between the second-to-fourth digit length ratio (2D:4D), a putative proxy for the ratio of prenatal estrogens to androgens, and AD, with inconsistent results among the few that have. Therefore, we aimed to investigate this relationship using methodologically robust metrics. In a 2 (sex) × 4 (group) MANOVA incorporating 108 participants (30 AD patients, 19 patients with tauopathy but no amyloidopathy, 31 clinical and 28 healthy age- and education-matched controls), the effects of sex and group on the dependent variables right and left 2D:4D were examined. We also explored the association between 2D:4D and the severity of AD symptoms assessed via neuropsychological examination. We did not find any significant differences in the right- and left-hand 2D:4D between patients with AD and the other groups; no significant associations between 2D:4D and neuropsychological task performances were found in the dementia groups. The 2D:4D of healthy women was significantly lower than that of depressed women without AD, i.e., clinical controls, but not significantly different from depressed female patients with AD. This investigation does not support the role of 2D:4D in the development or severity of AD in general, but suggests a potential role of 2D:4D for depression in women. Future studies are warranted to clarify whether 2D:4D can distinguish between early- and late-onset depression in women.
Collapse
Affiliation(s)
- Eva-Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
46
|
Ali N, Sohail R, Jaffer SR, Siddique S, Kaya B, Atowoju I, Imran A, Wright W, Pamulapati S, Choudhry F, Akbar A, Khawaja UA. The Role of Estrogen Therapy as a Protective Factor for Alzheimer's Disease and Dementia in Postmenopausal Women: A Comprehensive Review of the Literature. Cureus 2023; 15:e43053. [PMID: 37680393 PMCID: PMC10480684 DOI: 10.7759/cureus.43053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
The complete cessation of menstruation for 12 months with associated vasomotor symptoms is termed menopause. Apart from playing a role in reproduction, estrogen significantly affects the central nervous system (CNS). Population-based studies highlighted a substantial difference in the prevalence of dementia between men and women, with Alzheimer-associated dementia being more prevalent in women, indicating that estrogen deficiency might be a risk factor for neurodegenerative diseases. Patients with dementia experience a progressive decline in neurocognitive function, beginning with short-term memory loss that progresses to long-term memory loss and the inability to perform everyday activities, leading ultimately to death. There is currently no cure for dementia, so preventing or slowing the disease's progression is paramount. Accordingly, researchers have widely studied the role of estrogen as a neuroprotective agent. Estrogen prevents dementia by augmenting Hippocampal and prefrontal cortex function, reducing neuroinflammation, preventing degradation of estrogen receptors, decreasing oxidative damage to the brain, and increasing cholinergic and serotonergic function. According to the window phase hypothesis, estrogen's effect on preventing dementia is more pronounced if therapy is started early, during the first five years of menopause. Other studies like The Woman's Health Initiative Memory Study (WHIMS) showed unfavorable effects of estrogen on the brain. This review aims to establish an understanding of the currently available data on estrogen's effect on neurodegeneration, namely, dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- Noor Ali
- Obstetrics and Gynecology, Thumbay University Hospital, Ajman, ARE
- General Physician, Dubai Medical College, DXB, ARE
| | - Rohab Sohail
- Internal Medicine, Quaid-e-Azam Medical College, Bahawalpur, PAK
| | | | - Sadia Siddique
- Gastroenterology, Blackpool Victoria Hospital National Health Services (NHS) Foundation Trust, Blackpool, GBR
| | - Berfin Kaya
- Obstetrics and Gynaecology, Izmir Ataturk Research and Training Hospital, Izmir, TUR
- Obstetrics and Gynaecology, Izmir Kâtip Celebi University, Faculty of Medicine, Izmir, TUR
| | - Inioluwa Atowoju
- Obstetrics and Gynecology, Kharkiv National Medical University, Kharkiv, UKR
| | - Alizay Imran
- Surgery, Windsor University School of Medicine, Chicago, USA
| | - Whitney Wright
- Obstetrics and Gynecology, Texila American University, Georgetown, GUY
| | - Spandana Pamulapati
- Obstetrics and Gynecology, Alluri Sita Rama Raju Academy of Medical Sciences, Eluru, IND
| | - Faiza Choudhry
- Medicine and Surgery, Liaquat University of Medical and Health Sciences, Sindh, PAK
| | - Anum Akbar
- Pediatrics, University of Nebraska Medical Center, Omaha, USA
| | - Uzzam Ahmed Khawaja
- Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Clinical and Translational Research, Dr Ferrer BioPharma, South Miami, USA
| |
Collapse
|
47
|
Manna PR, Kshirsagar S, Pradeepkiran JA, Rawat P, Kumar S, Reddy AP, Reddy PH. Protective function of StAR in amyloid-β accumulated hippocampal neurotoxicity and neurosteroidogenesis: Mechanistic insights into Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166738. [PMID: 37142132 DOI: 10.1016/j.bbadis.2023.166738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
The steroidogenic acute regulatory (StAR) protein principally mediates steroid hormone biosynthesis by governing the transport of intramitochondrial cholesterol. Neurosteroids progressively decrease during aging, the key risk factor for Alzheimer's disease (AD), which is triggered by brain-region specific accumulation of amyloid beta (Aβ) precursor protein (APP), a key pathological factor. We demonstrate that hippocampal neuronal cells overexpressing wild-type (WtAPP) and mutant APP (mAPP) plasmids, conditions mimetic to AD, resulted in decreases in StAR mRNA, free cholesterol, and pregnenolone levels. The magnitude of suppression of the steroidogenic response was more pronounced with mAPP than that of WtAPP. While mAPP-waned assorted anomalies correlate to AD pathology, deterioration of APP/Aβ laden StAR expression and neurosteroid biosynthesis was enhanced by retinoid signaling. An abundance of mitochondrially targeted StAR expression partially restored APP/Aβ accumulated diverse neurodegenerative vulnerabilities. Immunofluorescence analyses revealed that overexpression of StAR diminishes mAPP provoked Aβ aggregation. Co-expression of StAR and mAPP in hippocampal neurons substantially reversed the declines in mAPP mediated cell survival, mitochondrial oxygen consumption rate, and ATP production. Concurrently, induction of mAPP induced Aβ loading showed an increase in cholesterol esters, but decrease in free cholesterol, concomitant with pregnenolone biosynthesis, events that were inversely regulated by StAR. Moreover, retinoid signaling was found to augment cholesterol content for facilitating neurosteroid biosynthesis in an AD mimetic condition. These findings provide novel insights into the molecular events by which StAR acts to protect mAPP-induced hippocampal neurotoxicity, mitochondrial dysfunction, and neurosteroidogenesis, and these measures are fundamental for ameliorating and/or delaying dementia in individuals with AD.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Priyanka Rawat
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Subodh Kumar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
48
|
Calvo N, Einstein G. Steroid hormones: risk and resilience in women's Alzheimer disease. Front Aging Neurosci 2023; 15:1159435. [PMID: 37396653 PMCID: PMC10313425 DOI: 10.3389/fnagi.2023.1159435] [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: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
More women have Alzheimer disease (AD) than men, but the reasons for this phenomenon are still unknown. Including women in clinical research and studying their biology is key to understand not just their increased risk but also their resilience against the disease. In this sense, women are more affected by AD than men, but their reserve or resilience mechanisms might delay symptom onset. The aim of this review was to explore what is known about mechanisms underlying women's risk and resilience in AD and identify emerging themes in this area that merit further research. We conducted a review of studies analyzing molecular mechanisms that may induce neuroplasticity in women, as well as cognitive and brain reserve. We also analyzed how the loss of steroid hormones in aging may be linked to AD. We included empirical studies with human and animal models, literature reviews as well as meta-analyses. Our search identified the importance of 17-b-estradiol (E2) as a mechanism driving cognitive and brain reserve in women. More broadly, our analysis revealed the following emerging perspectives: (1) the importance of steroid hormones and their effects on both neurons and glia for the study of risk and resilience in AD, (2) E2's crucial role in women's brain reserve, (3) women's verbal memory advantage as a cognitive reserve factor, and (4) E2's potential role in linguistic experiences such as multilingualism and hearing loss. Future directions for research include analyzing the reserve mechanisms of steroid hormones on neuronal and glial plasticity, as well as identifying the links between steroid hormone loss in aging and risk for AD.
Collapse
Affiliation(s)
- Noelia Calvo
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Women’s College Research Institute, Toronto, ON, Canada
- Centre for Life Course and Aging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Wrigglesworth J, Harding IH, Islam RM, Ward PGD, Woods RL, Bell RJ, McNeil JJ, Storey E, Egan G, Murray AM, Trevaks RE, Ward SA, Davis SR, Ryan J. The association between sex hormones and the change in brain-predicted age difference in older women. Clin Endocrinol (Oxf) 2023; 98:692-699. [PMID: 36807922 PMCID: PMC10073334 DOI: 10.1111/cen.14898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/17/2023] [Accepted: 02/19/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE The role of circulating sex hormones on structural brain ageing is yet to be established. This study explored whether concentrations of circulating sex hormones in older women are associated with the baseline and longitudinal changes in structural brain ageing, defined by the brain-predicted age difference (brain-PAD). DESIGN Prospective cohort study using data from NEURO and Sex Hormones in Older Women; substudies of the ASPirin in Reducing Events in the Elderly clinical trial. PATIENTS Community-dwelling older women (aged 70+ years). MEASUREMENTS Oestrone, testosterone, dehydroepiandrosterone (DHEA), and sex-hormone binding globulin (SHBG) were quantified from plasma samples collected at baseline. T1-weighted magnetic resonance imaging was performed at baseline, 1 and 3 years. Brain age was derived from whole brain volume using a validated algorithm. RESULTS The sample comprised of 207 women not taking medications known to influence sex hormone concentrations. A statistically higher baseline brain-PAD (older brain age relative to chronological age) was seen for women in the highest DHEA tertile compared with the lowest in the unadjusted analysis (p = .04). This was not significant when adjusted for chronological age, and potential confounding health and behavioural factors. Oestrone, testosterone and SHBG were not associated with brain-PAD cross-sectionally, nor were any of the examined sex hormones or SHBG associated with brain-PAD longitudinally. CONCLUSION No strong evidence of an association between circulating sex hormones and brain-PAD. Given there is prior evidence to suggests sex hormones may be important for brain ageing, further studies of circulating sex hormones and brain health in postmenopausal women are warranted.
Collapse
Affiliation(s)
- Jo Wrigglesworth
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Ian H. Harding
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne Victoria, Australia
| | - Rakibul M. Islam
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Phillip G. D. Ward
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800, Victoria Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Australia
| | - Robyn L. Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Robin J. Bell
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - John J. McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Elsdon Storey
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Gary Egan
- Monash Biomedical Imaging, Monash University, Melbourne, 3800, Victoria, Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function, Australia
| | - Anne M. Murray
- Berman Centre for Outcomes & Clinical Research, Hennepin Healthcare Research Institute, Hennepin, Minneapolis, MN, 55404, USA
- Department of Medicine, Division of Geriatrics, Hennepin Healthcare, University of Minnesota, Minneapolis, MN, 55404, USA
| | - Ruth E. Trevaks
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Stephanie A. Ward
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
- Department of Geriatric Medicine, Prince of Wales Hospital, Randwick, 2031, New South Wales, Australia
| | - Susan R. Davis
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
- Department of Endocrinology and Diabetes, Alfred Health, Melbourne, 3004, Victoria, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| |
Collapse
|
50
|
Sultana F, Davis SR, Murray AM, Woods RL, McNeil JJ, Islam RM. Sex hormones, SHBG and cognitive performance among older Australian women: an observational study. Climacteric 2023; 26:121-128. [PMID: 36716780 PMCID: PMC10033445 DOI: 10.1080/13697137.2023.2166824] [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: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVE This study aims to explore the associations between sex hormones and cognitive performance in older women. METHODS Associations between sex hormones, sex hormone binding globulin (SHBG) and cognitive performance were examined in women aged at least 70 years, without dementia and not using medications that influence sex hormones. Linear and generalized linear regression models included age, body mass index, education, smoking, alcohol, living circumstances, diabetes, hypertension, depression and impaired renal function. RESULTS The included 5511 women had a median (interquartile range) age of 73.9 (71.6-77.6) years. No associations were found for estrone, estradiol, testosterone or dehydroepiandrosterone and cognitive performance. SHBG concentrations above quartile 1 (Q1) were significantly inversely associated with processing speed (Q2, β = -0.94, 95% confidence interval [CI] - 1.64 to -0.24, p = 0.009; Q3, β = -0.82, 95% CI -1.53 to -0.10, p = 0.025; and Q4, β = -0.95, 95% CI -1.70 to -0.20, p = 0.013). CONCLUSIONS Sex hormones were not associated with cognitive performance. The finding that low SHBG is associated with better processing speed warrants further investigation. The null findings for the sex hormones establish a firm baseline to confidently explore the association between sex hormones and longitudinal cognitive performance in this population. TRIAL REGISTRATION International Standard Randomized Controlled Trial Number Register (ISRCTN83772183) and ClinicalTrials.gov (NCT01038583).
Collapse
Affiliation(s)
- F Sultana
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Environmental Interventions Unit, Infectious Diseases Division, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, Bangladesh
| | - S R Davis
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Endocrinology and Diabetes, Alfred Health, Melbourne, Vic 3004, Australia
| | - A M Murray
- Berman Center for Outcomes and Clinical Research, Hennepin-Health Research Institute and Division of Geriatrics, Department of Medicine, Hennepin Healthcare, Minneapolis, MN, USA
| | - R L Woods
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - J J McNeil
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - R M Islam
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|