1
|
Khan B, Iqbal MK, Khan MA, Khan H, Kiyani MM, Bashir S, Li S. Unraveling the Complexity of Alzheimer's Disease: Insights into Etiology and Advancements in Treatment Strategies. J Mol Neurosci 2025; 75:57. [PMID: 40279003 DOI: 10.1007/s12031-025-02337-4] [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: 12/08/2024] [Accepted: 03/20/2025] [Indexed: 04/26/2025]
Abstract
Alzheimer's disease, a complex and progressive neurological disorder, is the leading cause of late-life dementia. Pathologically, it is marked by the presence of amyloid plaques and neurofibrillary tangles in the brain. Over the past two decades, advancements in understanding the disease's pathogenesis have spurred research into new pharmacological treatments that target its underlying mechanisms. Currently available drugs, such as acetylcholinesterase inhibitors (rivastigmine, galantamine, donepezil) and the NMDA receptor antagonist memantine, primarily address symptoms and are effective only in the later stages of the disease. While these medications can slow disease progression and provide symptomatic relief, they do not offer a cure. Despite having a clear understanding of Alzheimer's neuropathology, the precise mechanisms driving the disease remain elusive. The lack of effective treatments that can stop the start and progression of the disease may be caused by our incomplete understanding of the pathogenic process. New therapeutic targets are now available due to the significant advancements made in pathophysiology over the past few years, which should allow for a direct attack on the underlying illness process. The various pathophysiological pathways that underlie Alzheimer's disease and how it is managed by conventional medication therapy, including current exploratory therapeutic options, are covered in this review article. Innovative, beneficial policies are essential to determine and progress therapeutic molecules to defend against AD.
Collapse
Affiliation(s)
- Bakhtawar Khan
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Muhammad Khalid Iqbal
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Muhammad Ajmal Khan
- Division of Life Sciences, Center for Cancer Researchand, State Key Laboratory of Molecular Neurosciencesaq, The Hong Kong University of Science and Technology, Clear Water Bay Hong Kong, China
| | - Hamid Khan
- Department of Biological Science, International Islamic University, Islamabad, Pakistan
| | - Mubin Mustafa Kiyani
- Shifa College of Medical Technology, Shifa Tameer-E-Millat University, Islamabad, Pakistan
| | - Shahid Bashir
- Department of Neuroscience, King Fahad Hospital, Dammam, Saudi Arabia.
- King Salman Center for Disability Research, 11614, Riyadh, Saudi Arabia.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
VandeBunte AM, Ortiz BL, Paolillo EW, Saloner R, Diaz V, Dutt S, Cadwallader CJ, Chen C, Lago AL, Rojas JC, Chan B, Sible I, Kramer JH, Casaletto KB. Relationships between blood pressure indicators and fluid biomarkers of brain aging in functionally intact older adults. Alzheimers Res Ther 2025; 17:85. [PMID: 40259431 PMCID: PMC12010523 DOI: 10.1186/s13195-025-01731-9] [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: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Dementia risk is significantly shaped by cardiovascular health, with elevated blood pressure emerging as a key risk factor for adverse brain aging. Blood biomarkers such as pTau181, Aβ42/40, NfL, and GFAP have improved our understanding of dementia pathophysiology, however, few studies have explored how specific blood pressure metrics relate to biomarker levels, which could inform personalized dementia prevention strategies as these biomarkers move into clinic. We examined how different blood pressure metrics associated with molecular markers of astrocytic activation (GFAP), neuronal axon breakdown (NfL), and Alzheimer's disease pathobiology (pTau181, Aβ42/40) in plasma. METHODS 109 functionally intact (Clinical Dementia Rating Scale = 0) older adults completed blood draws with plasma assayed for Aβ42/40, GFAP, NfL, and pTau181 (Quanterix Simoa) and in-lab blood pressure quantification. Blood pressure metrics included diastolic blood pressure, systolic blood pressure, and pulse pressure (systolic minus diastolic). Separate regression models evaluated plasma biomarkers as a function of each blood pressure metric, adjusting for age and biological sex. Interaction models tested whether relationships between blood pressure metrics and plasma biomarkers differed by sex, age, or APOE-ε4 status. RESULTS With the exception of Aβ42/40, higher pulse pressure related to higher levels of all plasma biomarkers examined (pTau181, NfL, GFAP). Additionally, higher systolic blood pressure related to higher pTau181, while diastolic blood pressure did not meaningfully associate with any biomarker. Interaction models revealed a significantly stronger relationship between elevated pulse pressure and higher GFAP concentrations in females compared to males, as well as a significantly stronger association between elevated pulse pressure and lower Aβ42/40 plasma concentrations in APOE-ε4 carriers compared to non-carriers. CONCLUSIONS Our findings suggest that elevated pulse pressure, and to a lesser extent systolic blood pressure, are associated with increased Alzheimer's disease and neurodegenerative (axonal and astrocytic health) biology among typically aging adults. These associations underscore the importance of blood pressure management, particularly pulse pressure, for reducing dementia risk. Cardiovascular health may be incorporated with biomarkers to further personalize dementia prevention and management strategies.
Collapse
Affiliation(s)
- Anna M VandeBunte
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
- Palo Alto University, Palo Alto, CA, 94304, USA
| | - Bailey L Ortiz
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
- Palo Alto University, Palo Alto, CA, 94304, USA
| | - Emily W Paolillo
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Valentina Diaz
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Shubir Dutt
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Claire J Cadwallader
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Coty Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Argentina Lario Lago
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Brandon Chan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Isabel Sible
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
| |
Collapse
|
3
|
Butler HM, Keller E, McCrorey M, Keceli G, Combs CK, Kayed R, Namakkal-S R, Paolocci N, Jacobs Wolf B, Wold LE, Del Monte F. Particulate matter and co-occurring genetic risk induce oxidative stress and cardiac and brain Alzheimer's pathology. Commun Biol 2025; 8:603. [PMID: 40221628 PMCID: PMC11993720 DOI: 10.1038/s42003-025-07701-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/07/2025] [Indexed: 04/14/2025] Open
Abstract
Amyloid-beta (Aβ) aggregates, an Alzheimer's disease (AD) pathological hallmark, extend beyond the brain to the heart of heart failure (HF) and AD patients. Being diseases of the elderly, increased prevalence is expected as the population ages. However, changes in the incidence and prevalence of dementia over the past decades, and the independent association of exposure to air particulate matter (PM) with poor cognitive function, adverse cardiovascular effects, and oxidative stress hint to the contribution of other factors beyond senescence. Therefore we evaluate whether, and by which mechanism(s), PM exposure affects heart and brain proteinopathy with/without genetic predisposition.AD-prone and control mice are exposed for three months to filtered air (FA) or concentrated ambient PM < 2.5μm in diameter (PM2.5), and evaluated for Aβ pathology, cognitive and cardiac function, and markers of oxidative stress. Aβ pathology become noticeable in AD hearts and worsens with PM2.5 in AD brains. Functionally, PM2.5 lead to anxiety and memory deficits and worsens diastolic function. Redox homeostasis is negatively impacted by genotype and PM2.5. This study identifies environmental pollution as a potential key contributor to early progression of heart and brain proteinopathy, delineating a crucial timepoint for early interventions to limit multiorgan damage in vulnerable patients.
Collapse
Affiliation(s)
- Helen M Butler
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Everette Keller
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marice McCrorey
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Gizem Keceli
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Colin K Combs
- School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rajasekaran Namakkal-S
- Department of Pathology/Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nazareno Paolocci
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Bethany Jacobs Wolf
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Loren E Wold
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Medicine and Surgery, University of Bologna Alma Mater, Bologna, Italy.
| |
Collapse
|
4
|
Wang X, Feng S, Deng Q, Wu C, Duan R, Yang L. The role of estrogen in Alzheimer's disease pathogenesis and therapeutic potential in women. Mol Cell Biochem 2025; 480:1983-1998. [PMID: 39088186 DOI: 10.1007/s11010-024-05071-4] [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: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Estrogens are pivotal regulators of brain function throughout the lifespan, exerting profound effects from early embryonic development to aging. Extensive experimental evidence underscores the multifaceted protective roles of estrogens on neurons and neurotransmitter systems, particularly in the context of Alzheimer's disease (AD) pathogenesis. Studies have consistently revealed a greater risk of AD development in women compared to men, with postmenopausal women exhibiting heightened susceptibility. This connection between sex factors and long-term estrogen deprivation highlights the significance of estrogen signaling in AD progression. Estrogen's influence extends to key processes implicated in AD, including amyloid precursor protein (APP) processing and neuronal health maintenance mediated by brain-derived neurotrophic factor (BDNF). Reduced BDNF expression, often observed in AD, underscores estrogen's role in preserving neuronal integrity. Notably, hormone replacement therapy (HRT) has emerged as a sex-specific and time-dependent strategy for primary cardiovascular disease (CVD) prevention, offering an excellent risk profile against aging-related disorders like AD. Evidence suggests that HRT may mitigate AD onset and progression in postmenopausal women, further emphasizing the importance of estrogen signaling in AD pathophysiology. This review comprehensively examines the physiological and pathological changes associated with estrogen in AD, elucidating the therapeutic potential of estrogen-based interventions such as HRT. By synthesizing current knowledge, it aims to provide insights into the intricate interplay between estrogen signaling and AD pathogenesis, thereby informing future research directions and therapeutic strategies for this debilitating neurodegenerative disorder.
Collapse
Affiliation(s)
- Xinyi Wang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
5
|
VandeBunte AM, Ortiz BL, Paolillo EW, Saloner R, Diaz V, Dutt S, Cadwallader CJ, Chen C, Lago AL, Rojas JC, Chan B, Sible I, Kramer JH, Casaletto KB. Relationships between blood pressure indicators and fluid biomarkers of brain aging in functionally intact older adults. RESEARCH SQUARE 2025:rs.3.rs-6018137. [PMID: 40196000 PMCID: PMC11975004 DOI: 10.21203/rs.3.rs-6018137/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background : Dementia risk is significantly shaped by cardiovascular health, with elevated blood pressure emerging as a key risk factor for adverse brain aging. Blood biomarkers such as pTau181, Aβ42/40, NfL, and GFAP have improved our understanding of dementia pathophysiology, however, few studies have explored how specific blood pressure metrics relate to biomarker levels, which could inform personalized dementia prevention strategies as these biomarkers move into clinic. We examined how different blood pressure metrics associated with molecular markers of astrocytic activation (GFAP), neuronal axon breakdown (NfL), and Alzheimer's disease pathobiology (pTau181, Aβ42/40) in plasma. Methods : 109 functionally intact (Clinical Dementia Rating Scale=0) older adults completed blood draws with plasma assayed for Aβ42/40, GFAP, NfL, and pTau181 (Quanterix Simoa) and in-lab blood pressure quantification. Blood pressure metrics included diastolic blood pressure, systolic blood pressure, and pulse pressure (systolic minus diastolic). Separate regression models evaluated plasma biomarkers as a function of each blood pressure metric, adjusting for age and biological sex. Interaction models tested whether relationships between blood pressure metrics and plasma biomarkers differed by sex, age, or APOE -ε4 status. Results : With the exception of Aβ42/40, higher pulse pressure related to higher levels of all plasma biomarkers examined (pTau181, NfL, GFAP). Additionally, higher systolic blood pressure related to higher pTau181, while diastolic blood pressure did not meaningfully associate with any biomarker. Interaction models revealed a significantly stronger relationship between elevated pulse pressure and higher GFAP concentrations in females compared to males, as well as a significantly stronger association between elevated pulse pressure and lower Aβ42/40 plasma concentrations in APOE -ε4 carriers compared to non-carriers. Conclusions : Our findings suggest that elevated pulse pressure, and to a lesser extent systolic blood pressure, are associated with increased Alzheimer's disease and neurodegenerative (axonal and astrocytic health) biology among typically aging adults. These associations underscore the importance of blood pressure management, particularly pulse pressure, for reducing dementia risk. Cardiovascular health may be incorporated with biomarkers to further personalize dementia prevention and management strategies.
Collapse
|
6
|
Song Q, Wang Q, Wu D, Zhang Z, Chen M, Fu C, Li M, Wang X, Zhao Y, Zhu D. Association between duration, initiation time, routes, and formulations of menopausal hormone therapy use and Alzheimer disease in women: A systematic review and meta-analysis. J Pharmacol Exp Ther 2025; 392:103554. [PMID: 40220453 DOI: 10.1016/j.jpet.2025.103554] [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: 11/27/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025] Open
Abstract
The purpose of this study was to investigate the effect of menopausal hormone therapy (MHT) on the risk of Alzheimer disease (AD) by examining its duration, initiation time, routes of administration, and formulations through systematic review and meta-analysis. PubMed, Embase, Cochrane Library, Web of Science, and Scopus were searched on March 15, 2023. We selected cohort studies, case-control studies, and randomized controlled trials on the effect of MHT on AD in women. Odds ratio, relative risk, and hazard ratio were extracted. Random-effect models were used to estimate the polled estimates (relative risk [RR] or odds ratio [OR]) and their 95% confidence interval (95% CI). We included 3 randomized controlled trials, 12 cohort studies, and 16 case-control studies. A total of 7,710,379 women were included. Pooled estimates showed that MHT use for 3-5 years (cohort, RR = 0.56, 95% CI: 0.34-0.93) or initiation within 5 years of menopause (cohort, RR = 0.70, 95% CI: 0.49-0.99) reduced the risk of AD. Oral administration reduced AD risk (cohort, RR = 0.42, 95% CI: 0.40-0.44). Combining estrogen and progesterone (case-control, OR = 1.13, 95% CI: 1.05-1.21) or progesterone only (case-control, OR = 1.13, 95% CI: 1.10-1.17) increases AD risk. Tibolone increased AD risk (cohort, RR = 1.04, 95% CI: 1.01-1.07; case-control, OR = 1.07, 95% CI: 1.01-1.14). MHT-protected apolipoprotein E genotype 4 carriers (cohort, RR = 0.13, 95% CI: 0.02-0.90), depressed populations (cohort, RR = 0.85, 95% CI: 0.80-0.90), and Americas (cohort, RR = 0.54, 95% CI: 0.37-0.80; case-control, OR = 0.68, 95% CI: 0.47-0.99) from AD. Using MHT early (within 5 years after menopause) for about 5 years may protect against AD. However, combining estrogen with progesterone, or using progesterone only, could increase AD risk. Oral MHT methods are more effective than transdermal ones in reducing this risk. SIGNIFICANCE STATEMENT: Menopausal hormone therapy (MHT) use within 5 years after menopause could offer protective benefits against Alzheimer disease (AD). A combination of estrogen and progesterone, using progesterone only or tibolone usage was connected with an elevated risk of AD. Oral MHT was more effective than transdermal methods in lowering AD risk. MHT lowered AD risk in apolipoprotein E genotype 4 allele carriers, individuals with depression, and Americans. MHT regimens should be highly personalized.
Collapse
Affiliation(s)
- Qixiang Song
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qi Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; Center for Clinical Epidemiology and Evidence-Based Medicine, Shandong University, Jinan, China
| | - Dang Wu
- Institute of Health Sciences, China Medical University, Shenyang, China
| | - Zhe Zhang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengyao Chen
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunying Fu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meiling Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyi Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanqing Zhao
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongshan Zhu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; Center for Clinical Epidemiology and Evidence-Based Medicine, Shandong University, Jinan, China.
| |
Collapse
|
7
|
Marongiu R, Platholi J, Park L, Yu F, Sommer G, Woods C, Milner TA, Glass MJ. Perimenopause promotes neuroinflammation in select hippocampal regions in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643317. [PMID: 40161644 PMCID: PMC11952527 DOI: 10.1101/2025.03.14.643317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by age-dependent amyloid beta (Aβ) aggregation and accumulation, neuroinflammation, and cognitive deficits. Significantly, there are prominent sex differences in the risk, onset, progression, and severity of AD, as well as response to therapies, with disease burden disproportionally affecting women. Although menopause onset (i.e., perimenopause) may be a critical transition stage for AD susceptibility in women, the role of early ovarian decline in initial disease pathology, particularly key neuroinflammatory processes, is not well understood. To study this, we developed a unique mouse model of perimenopausal AD by combining an accelerated ovarian failure (AOF) model of menopause induced by 4-vinylcyclohexene diepoxide (VCD) with the 5xFAD transgenic AD mouse model. To target early stages of disease progression, 5xFAD females were studied at a young age (∼4 months) and at the beginning stage of ovarian failure analogous to human perimenopause (termed "peri-AOF"), and compared to age-matched males. Assessment of neuropathology was performed by immunohistochemical labeling of Aβ as well as markers of astrocyte and microglia activity in the hippocampus, a brain region involved in learning and memory that is deleteriously impacted during AD. Our results show that genotype, AOF, and sex contributed to AD-like pathology. Aggregation of Aβ was heightened in female 5xFAD mice and further increased at peri-AOF, with hippocampal subregion specificity. Further, select increases in glial activation also paralleled Aβ pathology in distinct hippocampal subregions. However, cognitive function was not affected by peri-AOF. These findings align with the hypothesis that perimenopause constitutes a period of susceptibility for AD pathogenesis in women.
Collapse
|
8
|
Bonkhoff AK, Coughlan G, Perosa V, Alhadid K, Schirmer MD, Regenhardt RW, van Veluw S, Buckley R, Fox MD, Rost NS. Sex differences in age-associated neurological diseases-A roadmap for reliable and high-yield research. SCIENCE ADVANCES 2025; 11:eadt9243. [PMID: 40043111 PMCID: PMC11881909 DOI: 10.1126/sciadv.adt9243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025]
Abstract
Once taken into consideration, sex differences in neurological diseases emerge in abundance: (i) Stroke severity is significantly higher in females than in males, (ii) Alzheimer's disease (AD) pathology is more pronounced in females, and (iii) conspicuous links with hormonal cycles led to female-specific diagnoses, such as catamenial migraines and epilepsy. While these differences receive increasing attention in isolation, they likely link to similar processes in the brain. Hence, this review aims to present an overview of the influences of sex chromosomes, hormones, and aging on male and female brains across health and disease, with a particular focus on AD and stroke. The focus here on advancements across several fields holds promise to fuel future research and to lead to an enriched understanding of the brain and more effective personalized neurologic care for all.
Collapse
Affiliation(s)
- Anna K. Bonkhoff
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Valentina Perosa
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Kenda Alhadid
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Markus D. Schirmer
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Robert W. Regenhardt
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Susanne van Veluw
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Rachel Buckley
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Michael D. Fox
- Department of Neurology, Brigham and Women’s Hospital/Harvard Medical School, Boston, MA, USA
| | - Natalia S. Rost
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
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
|
10
|
Chern H, Caruso G, Desaire H, Jarosova R. Carnosine Mitigates Cognitive Impairment and Dopamine Release in an Okadaic Acid-Induced Zebrafish Model with Alzheimer's Disease-like Symptoms. ACS Chem Neurosci 2025; 16:790-801. [PMID: 39933073 DOI: 10.1021/acschemneuro.4c00596] [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: 02/13/2025] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, affects 1 in 9 people aged 65 and older. The disease impacts patients on multiple levels, from memory and problem-solving issues to difficulties with basic functions and personality changes. Unfortunately, there is only a handful of FDA-approved drugs, and none of them offer an effective cure. Therefore, recent strategies have focused on preventing and delaying disease onset, rather than curing already developed pathological changes in the brain. In this study, we investigated the therapeutic potential of carnosine (CAR), a naturally occurring dipeptide known for its multimodal mechanism of action, such as the ability to mitigate neuroinflammation, oxidative stress, and deficiencies in neurotropic factors, all of which are connected with aging-related cognitive decline and an increased risk of developing dementia. For this purpose, we utilized an okadaic acid-induced zebrafish model of AD, which replicates some of the key features of the disease, including hyperphosphorylation of tau protein, changes in Aβ-fragments, and cognitive decline. By employing a latent learning behavioral assay and fast-scan cyclic voltammetry, we evaluated the effect of CAR on the prevention of cognitive decline and neurochemical changes in the AD-like zebrafish brain. Our findings revealed that CAR prevents impaired learning and motor dysfunction in a sex-dependent manner and reduces anxiety-like behavior. Additionally, we found that CAR inhibits dopamine release impairment. Hence, our study demonstrates the potential of CAR as a promising candidate for further investigations focused on identifying molecules that could potentially serve as therapeutics for delaying the onset of AD.
Collapse
Affiliation(s)
- Hannah Chern
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy
| | - Heather Desaire
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Romana Jarosova
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, United States
| |
Collapse
|
11
|
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
|
12
|
Fah H, Bohn L, Greiner R, Dixon RA. Comparing machine learning classifier models in discriminating cognitively unimpaired older adults from three clinical cohorts in the Alzheimer's disease spectrum: demonstration analyses in the COMPASS-ND study. Front Aging Neurosci 2025; 17:1542514. [PMID: 40103927 PMCID: PMC11913811 DOI: 10.3389/fnagi.2025.1542514] [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: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Background Research in aging, impairment, and Alzheimer's disease (AD) often requires powerful computational models for discriminating between clinical cohorts and identifying early biomarkers and key risk or protective factors. Machine Learning (ML) approaches represent a diverse set of data-driven tools for performing such tasks in big or complex datasets. We present systematic demonstration analyses to compare seven frequently used ML classifier models and two eXplainable Artificial Intelligence (XAI) techniques on multiple performance metrics for a common neurodegenerative disease dataset. The aim is to identify and characterize the best performing ML and XAI algorithms for the present data. Method We accessed a Canadian Consortium on Neurodegeneration in Aging dataset featuring four well-characterized cohorts: Cognitively Unimpaired (CU), Subjective Cognitive Impairment (SCI), Mild Cognitive Impairment (MCI), and AD (N = 255). All participants contributed 102 multi-modal biomarkers and risk factors. Seven ML algorithms were compared along six performance metrics in discriminating between cohorts. Two XAI algorithms were compared using five performance and five similarity metrics. Results Although all ML models performed relatively well in the extreme-cohort comparison (CU/AD), the Super Learner (SL), Random Forest (RF) and Gradient-Boosted trees (GB) algorithms excelled in the challenging near-cohort comparisons (CU/SCI). For the XAI interpretation comparison, SHapley Additive exPlanations (SHAP) generally outperformed Local Interpretable Model agnostic Explanation (LIME) in key performance properties. Conclusion The ML results indicate that two tree-based methods (RF and GB) are reliable and effective as initial models for classification tasks involving discrete clinical aging and neurodegeneration data. In the XAI phase, SHAP performed better than LIME due to lower computational time (when applied to RF and GB) and incorporation of feature interactions, leading to more reliable results.
Collapse
Affiliation(s)
- Harrison Fah
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada
| | - Linzy Bohn
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | - Russell Greiner
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Roger A Dixon
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Zelco A, Joshi A. Single-Cell Analysis of Sex and Gender Differences in the Human Brain During Development and Disease. Cell Mol Neurobiol 2025; 45:20. [PMID: 40016536 PMCID: PMC11868228 DOI: 10.1007/s10571-025-01536-2] [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: 08/25/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
Sex and gender (SG) differences in the human brain are of interest to society and science as numerous processes are impacted by them, including brain development, behavior, and diseases. By collecting publicly available single-cell data from the in-utero to elderly age in healthy, Alzheimer's disease and multiple sclerosis samples, we identified and characterized SG-biased genes in ten brain cell types across 9 age and disease groups. Sex and gender differences in the transcriptome were present throughout the lifespan and across all cell types. Although there was limited overlap among SG-biased genes across different age and disease groups, we observed significant functional overlap. Female-biased genes are consistently enriched for brain-related processes, while male-biased genes are enriched for metabolic pathways. Additionally, mitochondrial genes showed a consistent female bias across cell types. We also found that androgen response elements (not estrogen) were significantly enriched in both male- and female-biased genes, and thymosin hormone targets being consistently enriched only in male-biased genes. We systematically characterised SG differences in brain development and brain-related disorders at a single-cell level, by analysing a total of publicly available 419,885 single nuclei from 161 human brain samples (72 females, 89 males). The significant enrichment of androgen (not estrogen) response elements in both male- and female-biased genes suggests that androgens are important regulators likely establishing these SG differences. Finally, we provide full characterization of SG-biased genes at different thresholds for the scientific community as a web resource.
Collapse
Affiliation(s)
- Aura Zelco
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
| | - Anagha Joshi
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India.
| |
Collapse
|
14
|
Hicks TH, Magalhães TNC, Bernard JA. The Human Cerebello-Hippocampal Circuit Across Adulthood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638640. [PMID: 40027698 PMCID: PMC11870467 DOI: 10.1101/2025.02.17.638640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Direct communication between the hippocampus and cerebellum has been shown via coactivation and synchronized neuronal oscillations in animal models. Further, this novel cerebello-hippocampal circuit may be impacted by sex steroid hormones. The cerebellum and hippocampus are dense with estradiol and progesterone receptors relative to other brain regions. Females experience up to a 90% decrease in ovarian estradiol production after the menopausal transition. Postmenopausal women show lower cerebello-cortical and intracerebellar FC compared to reproductive aged females. Sex hormones are established modulators of both memory function and synaptic organization in the hippocampus in non-human animal studies. However, investigation of the cerebello-hippocampal (CB-HP) circuit has been limited to animal studies and small homogeneous samples of young adults as it relates to spatial navigation. Here, we investigate the CB-HP circuit in 138 adult humans (53% female) from 35-86 years of age, to define its FC patterns, and investigate its associations with behavior, hormone levels, and sex differences therein. We established robust FC patterns between the CB and HP in this sample. We predicted and found negative relationships between age and CB-HP FC. As expected, estradiol levels exhibited positive relationships with CB-HP. We found lower CB-HP FC with higher levels of progesterone. We provide the first characterization of the CB-HP circuit across middle and older adulthood and demonstrate that connectivity is sensitive to sex steroid hormone levels. This work provides the first clear CB-HP circuit mapping in the human brain and serves as a foundation for future work in neurological and psychiatric diseases.
Collapse
|
15
|
Huang C, Wei Z, Zheng N, Yan J, Zhang J, Ye X, Zhao W. The interaction between dysfunction of vasculature and tauopathy in Alzheimer's disease and related dementias. Alzheimers Dement 2025; 21:e14618. [PMID: 39998958 PMCID: PMC11854360 DOI: 10.1002/alz.14618] [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: 08/12/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
Tauopathy is one of the pathological features of Alzheimer's disease and related dementias (ADRD). At present, there have been many studies on the formation, deposition, and intercellular transmission of tau in neurons and immune cells. The vasculature is an important component of the central nervous system. This review discusses the interaction between vasculature and tau in detail from three aspects. (1) The vascular risk factors (VRFs) discussed in this review include diabetes mellitus (DM), abnormal blood pressure (BP), and hypercholesterolemia. (2) In ADRD pathology, the hyperphosphorylation and deposition of tau interact with disrupted vasculature, such as different cells (endothelial cells, smooth muscular cells, and pericytes), the blood-brain barrier (BBB), and the cerebral lymphatic system. (3) The functions of vasculature are regulated by various signaling transductions. Endothelial nitric oxide synthase/nitric oxide, calcium signaling, Rho/Rho-associated coiled-coil containing Kinase, and receptors for advanced glycation end products are discussed in this review. Our findings indicate that the prevention and treatment of vascular health may be a potential target for ADRD combination therapy. HIGHLIGHTS: Persistent VRFs increase early disruption of vascular mechanisms and are strongly associated with tau pathology in ADRD. Cell dysfunction in the vasculature causes BBB leakage and drainage incapacity of the cerebral lymphatic system, which interacts with tau pathology. Signaling molecules in the vasculature regulate vasodilation and contraction, angiogenesis, and CBF. Abnormal signaling transduction is related to tau hyperphosphorylation and deposition.
Collapse
Affiliation(s)
- Chuyao Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhenwen Wei
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Ningxiang Zheng
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jingsi Yan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jiayu Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xinyi Ye
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Wei Zhao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| |
Collapse
|
16
|
Wang L, Wu H, Wang C, Ma Y, Xiang Z. Triple-template surface imprinted magnetic polymers for wide-coverage extraction of steroid hormones from human serum. ANAL SCI 2025; 41:151-163. [PMID: 39527176 DOI: 10.1007/s44211-024-00690-9] [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: 09/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
In this study, novel triple-template magnetic nanospheres by surface imprinting, called triple-template magnetic molecularly imprinted polymers (tri-MMIPs), were prepared for the detection of steroid hormones in human serum. The polymers were constructed by Fe3O4 as support, estrone (E1), progesterone (PROG), and estradiol (E2) as triple templates, acrylic acid (AA) as functional monomer and ethylene glycol dimethacrylate (EGDMA) as cross-linker. The resulting tri-MMIPs were further characterized and applied as sorbents in human serum pretreatment. Coupled with ultra-high performance liquid chromatography-triple quadrupole tandem mass spectrometry (UHPLC-MS/MS) profiling after derivatization, 15 steroids (E1, 6-ketoE1, 4-OHE1, 16α-OHE1, DHEA, 4-MeOE1, PROG, PREG, 17-OHPROG, 7-DHE2, E2, 2-OHE2, 16-epiE3, E3, 2-MeOE2) were successfully quantified. The developed method exhibited a good recovery (78.9-114%) and low limit of quantitation (LLOQ, 5-10 pg/mL), which indicates a great potential for application in the analysis of multiple steroid hormones in complex biological samples.
Collapse
Affiliation(s)
- Li Wang
- Medical School, Hangzhou City University, Hangzhou, 310015, China
| | - Hongyu Wu
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Caihong Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yunfei Ma
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zheng Xiang
- Medical School, Hangzhou City University, Hangzhou, 310015, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
17
|
Flynn CM, Omoluabi T, Janes AM, Rodgers EJ, Torraville SE, Negandhi BL, Nobel TE, Mayengbam S, Yuan Q. Targeting early tau pathology: probiotic diet enhances cognitive function and reduces inflammation in a preclinical Alzheimer's model. Alzheimers Res Ther 2025; 17:24. [PMID: 39827356 PMCID: PMC11742226 DOI: 10.1186/s13195-025-01674-1] [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: 04/05/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) remains incurable, yet its long prodromal phase offers a crucial window for early intervention. Pretangle tau, a precursor to neurofibrillary tangles, plays a key role in early AD pathogenesis. Intervening in pretangle tau pathology could significantly delay the progression of AD. The gut-brain axis, increasingly recognized as a contributor to AD, represents a promising therapeutic target due to its role in regulating neuroinflammation and neurodegeneration. While probiotics have shown cognitive benefits in amyloid-centered AD models, their effect on pretangle tau pathology remains elusive. METHODS This study evaluates the effects of probiotics in a rat model of preclinical AD, specifically targeting hyperphosphorylated pretangle tau in the locus coeruleus. TH-CRE rats (N = 47; 24 females and 23 males) received either AAV carrying pseudophosphorylated human tau (htauE14) or a control virus at 3 months of age. Probiotic or control diets were administered at 9-12 months, with blood and fecal samples collected for ELISA and 16S rRNA gene sequencing. Behavioral assessments were conducted at 13-14 months, followed by analysis of brain inflammation, blood-brain barrier integrity, and GSK-3β activation. RESULTS Rats expressing pseudophosphorylated tau displayed impairment in spatial Y-maze (F1,39 = 4.228, p = 0.046), spontaneous object location (F1,39 = 6.240, p = 0.017), and olfactory discrimination (F1,39 = 7.521, p = 0.009) tests. Phosphorylation of tau at S262 (t3 = -4.834) and S356 (t3 = -3.258) in the locus coeruleus was parallelled by GSK-3β activation in the hippocampus (F1,24 = 10.530, p = 0.003). Probiotic supplementation increased gut microbiome diversity (F1,31 = 8.065, p = 0.007) and improved bacterial composition (F1,31 = 3.4867, p = 0.001). The enhancement in gut microbiomes was associated with enhanced spatial learning (p < 0.05), reduced inflammation indexed by Iba-1 (F1,25 = 5.284, p = 0.030) and CD-68 (F1,26 = 8.441, p = 0.007) expression, and inhibited GSK-3β in female rats (p < 0.01 compared to control females). CONCLUSIONS This study underscores the potential of probiotics to modulate the gut-brain axis and mitigate pretangle tau-related pathology in preclinical AD. Probiotic supplementation could offer a novel early intervention strategy for AD, highlighting the pivotal role of gut health in neurodegeneration.
Collapse
Affiliation(s)
- Cassandra M Flynn
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Tamunotonye Omoluabi
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Alyssa M Janes
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Biochemistry Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Emma J Rodgers
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Psychology Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Sarah E Torraville
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Brenda L Negandhi
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Timothy E Nobel
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Shyamchand Mayengbam
- Biochemistry Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
18
|
Bruno F, Spadafora P, Veltri I, Cuconati ML, Condino F, Cerantonio A, De Benedittis S, Greco BM, Di Palma G, Gallo O, Citrigno L, Qualtieri A, Cundari M, Cavalcanti F. Sex and APOE genotype modulate neuropsychological profile and depression in temporal lobe epilepsy. Front Neurosci 2025; 18:1514902. [PMID: 39886338 PMCID: PMC11780593 DOI: 10.3389/fnins.2024.1514902] [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: 10/22/2024] [Accepted: 12/02/2024] [Indexed: 02/01/2025] Open
Abstract
Introduction Temporal lobe epilepsy is the most common form of focal epilepsy, often associated with cognitive impairments, particularly in memory functions, and depression. Sex and APOE ε4 genotype play a crucial role in modulating cognitive outcomes and depression in various neurological conditions like Alzheimer's disease. However, the combined effects of APOE genotype and sex on cognitive performance and depression in temporal lobe epilepsy have not been previously investigated. Objective This study aims to (i) identify impaired cognitive performance and clinically relevant depression; (ii) explore the interaction between sex and APOE ε4 genotype on cognitive performance and depression in individuals with temporal lobe epilepsy. Methods We used a comprehensive battery of neuropsychological tests to assess domains such as learning and memory, attention, executive functions, language, and visuo-spatial constructional skills and the Hamilton Depression Rating Scale. We also performed APOE genotyping to assess its role in the study. The final sample was composed by fifty-four patients (53.7% female). Cognitive performance and depression were analyzed using normative cut-off scores. To examine the main effects and interactions of sex and APOE ε4 carrier status on neuropsychological test scores and the Hamilton Depression Rating Scale, we also conducted a two-way Analysis of Variance (ANOVA). Results Female APOE ε4 carriers compared to normative cut-offs, exhibited poor performance on multiple test scores, including the MMSE, The Rey Auditory Verbal Learning Test (immediate and delayed recall), The Corsi Block-Tapping Task, The Verbal Fluency Test, The Raven's Standard Progressive Matrices and The Pentagon-copying Test. Males showed impairment only in visuo-spatial short-term memory. ANOVA analysis revealed significant main effects of APOE ε4 status and sex on the MMSE, The Rey Auditory Verbal Learning Test, The Verbal Fluency, The Raven's Standard Progressive Matrices and The Pentagon-copying Test scores. Specifically, female APOE ε4 carriers performed consistently worse than other groups on many tasks. For depression, only an effect of sex emerged. Females scored higher besides APOE genotype. Conclusions These findings underscore the importance of considering both sex and APOE genotype when assessing cognitive performance in patients with temporal lobe epilepsy. The significant cognitive deficits we observed among females carrying the APOE ε4 allele highlight previously unexplored genetic and sex-related influences on cognition. This has potential implications for personalized therapeutic strategies, emphasizing the need for targeted assessment and intervention.
Collapse
Affiliation(s)
- Francesco Bruno
- Faculty of Social and Communication Sciences, Universitas Mercatorum, Rome, Italy
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Patrizia Spadafora
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Ida Veltri
- Territorial Social-Health Company of Lodi, Lodi, Italy
| | - Mario L. Cuconati
- Department of Medical and Surgical Sciences, Science and Techniques of Cognitive Psychology Degree Course, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Francesca Condino
- Department of Economics, Statistics and Finance “Giovanni Anania”, University of Calabria, Rende, Cosenza, Italy
| | - Annamaria Cerantonio
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Selene De Benedittis
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Beatrice M. Greco
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Cosenza, Italy
| | - Gemma Di Palma
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Olivier Gallo
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Luigi Citrigno
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Antonio Qualtieri
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| | - Maurizio Cundari
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Unit of Neuropsychiatry, Hospital of Helsingborg, Helsingborg, Sweden
- Unit of Neurology, Hospital of Helsingborg, Helsingborg, Sweden
| | - Francesca Cavalcanti
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council (CNR), Cosenza, Italy
| |
Collapse
|
19
|
Atalay B, Dogan S, Gudu BO, Yilmaz E, Ayden A, Ozorhan U, Cicekdal MB, Yaltirik K, Ekici ID, Tuna BG. Neurodegeneration: Effects of calorie restriction on the brain sirtuin protein levels. Behav Brain Res 2025; 476:115258. [PMID: 39332639 DOI: 10.1016/j.bbr.2024.115258] [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: 12/16/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Calorie restriction (CR) is suggested to activate protective mechanisms in neurodegenerative diseases (NDDs). Despite existing literature highlighting the protective role of Sirtuin (SIRT) proteins against age-related neurodegeneration (ND), no study has explored the total levels of SIRT 1, 3, and 6 proteins simultaneously in brain homogenates by ELISA following intermittent calorie restriction. Applying CR protocols in mice to induce stress, we aimed to determine whether ND would be more pronounced with ad libitum (AL) or with CR. METHODS Mice were randomly assigned to ad libitum (AL), Chronic CR (CCR), or Intermittent CR (ICR) groups at 10 weeks of baseline age (BL). SIRT 1, 3, and 6 protein levels were measured in the homogenized whole-brain supernatants of 49/50 weeks old mice by the ELISA method. Neuronal morphology was evaluated by the cresyl violet on the hippocampus. Neurodegeneration (ND) was assessed by the fluoro-jade and ImageJ was used for quantifications. RESULTS In the ICR group, SIRT1 levels were elevated compared to both the AL and BL groups. Similarly, the CCR group exhibited higher SIRT1 values compared to the AL and BL groups. While SIRT3 levels were higher in both the ICR and CCR groups compared to the AL and BL groups, this disparity did not reach statistical significance. SIRT6 levels were also higher in the ICR group compared to both the BL and AL groups, with the CCR group showing higher values compared to the BL and AL groups as well. Image quantification demonstrated significant neurodegeneration in the AL group compared to the CCR and ICR group, with no observed alterations in nerve cell morphology and number. CONCLUSION This study revealed that the levels of SIRT 1, SIRT 3, and SIRT 6 in brain tissue were notably elevated, and there was less evidence of ND at the 50-week mark in groups undergoing continuous calorie restriction and intermittent calorie restriction compared to baseline and ad libitum groups. Our findings illustrate that CR promotes increased SIRT expression in the mouse brain, thereby potentially mitigating neurodegeneration.
Collapse
Affiliation(s)
- Basar Atalay
- University of Yeditepe, School of Medicine, Department of Neurosurgery, Istanbul, Turkiye; University of Miami Miller School of Medicine, Department of Neurology, Neurocriticalcare, Miami, USA; Jackson Memorial Hospital, Department of Neurology, Neurocritical Care, Miami, USA
| | - Soner Dogan
- Yeditepe University, School of Medicine, Department of Medical Biology, Istanbul, Turkiye
| | - Burhan Oral Gudu
- University of Yeditepe, School of Medicine, Department of Neurosurgery, Istanbul, Turkiye
| | - Elif Yilmaz
- Yeditepe University, School of Medicine, Department of Medical Biology, Istanbul, Turkiye
| | - Atakan Ayden
- Yeditepe University, School of Medicine, Department of Medical Biology, Istanbul, Turkiye
| | - Umit Ozorhan
- University of Lübeck, Institude of Experimental ans Clinical Pharmacology and Toxicology, Lübeck, Germany
| | - Munevver Burcu Cicekdal
- University of Ghent, Medical Biology, School of Medicine and Health Sciences, Ghent, Belgium
| | - Kaan Yaltirik
- University of Yeditepe, School of Medicine, Department of Neurosurgery, Istanbul, Turkiye
| | - Isin Dogan Ekici
- Yeditepe University, School of Medicine, Department of Pathology, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Yeditepe University, School of Medicine, Department of Biophysics, Istanbul, Turkiye.
| |
Collapse
|
20
|
Zeng J, Gao Z, Xiong X, Hou X, Qin H, Liu Y, Bowman H, Ritchie C, O'Brien JT, Su L. The effects of two Alzheimer's disease related genes APOE and MAPT in healthy young adults: An attentional blink study. J Alzheimers Dis 2025; 103:167-179. [PMID: 39639576 DOI: 10.1177/13872877241299124] [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: 12/07/2024]
Abstract
BACKGROUND Genetic risk factors start to affect the brain and behavior in Alzheimer's disease (AD) before clinical symptoms occur. Although AD is mainly associated with memory deficits, attention and executive dysfunctions can present at the early presymptomatic stages in middle age for those with non-modifiable risks. OBJECTIVE Here, we investigated whether known risk genes for AD already affected attention in young adulthood. METHODS A total of 392 healthy young adults aged around 20 years underwent genetic testing for risks of dementia (APOE and MAPT) and performed a computerized cognitive test for temporal attention called the Attentional Blink (AB) task, in which patients with dementia tested in previous studies often showed reduced performance. Here, the AB task was analyzed using repeated-measurements analysis of variance for the ability of visual perception, attention deployment and temporal memory encoding/binding performance. RESULTS The results showed that all participants exhibited AB effects. Importantly, genetic risk factors had statistically significant influence on temporal attention depending on sex in healthy young adults. APOE4 status was associated with enhanced attention deployment in males but not females, while MAPT AA carriers had poorer performance in AB but only in females. No genetic effects were found for visual perception and temporal memory binding errors between high and low risk groups. CONCLUSIONS We provided evidence that both APOE and MAPT start to affect attentional function as early as young adulthood. Furthermore, unlike previous findings in older people, these genes had a differential effect for males and females in young adults.
Collapse
Affiliation(s)
- Jianmin Zeng
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing, China
| | - Ziyun Gao
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing, China
| | - Xiong Xiong
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Xingrong Hou
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing, China
| | - Huihui Qin
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunication, Beijing, China
| | - Howard Bowman
- School of Psychology, University of Birmingham, Birmingham, UK
| | - Craig Ritchie
- School of Medicine, University of St Andrews, St Andrews, UK
- Scottish Brain Sciences, Edinburgh, UK
| | - John T O'Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Li Su
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Scheinman SB, Dong H. The impact of sex on memory during aging and Alzheimer's disease progression: Epigenetic mechanisms. J Alzheimers Dis 2024; 102:562-576. [PMID: 39539121 PMCID: PMC11721493 DOI: 10.1177/13872877241288709] [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: 11/16/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, disability, and death in the elderly. While the etiology of AD is unknown, there are several established risk factors for the disease including, aging, female sex, and genetics. However, specific genetic mutations only account for a small percentage (1-5%) of AD cases and the much more common sporadic form of the disease has no causative genetic basis, although certain risk factor genes have been identified. While the genetic code remains static throughout the lifetime, the activation and expression levels of genes change dynamically over time via epigenetics. Recent evidence has emerged linking changes in epigenetics to the pathogenesis of AD, and epigenetic alterations also modulate cognitive changes during physiological aging. Aging is the greatest risk factor for the development of AD and two-thirds of all AD patients are women, who experience an increased rate of symptom progression compared to men of the same age. In humans and other mammalian species, males and females experience aging differently, raising the important question of whether sex differences in epigenetic regulation during aging could provide an explanation for sex differences in neurodegenerative diseases such as AD. This review explores distinct epigenetic changes that impact memory function during aging and AD, with a specific focus on sexually divergent epigenetic alterations (in particular, histone modifications) as a potential mechanistic explanation for sex differences in AD.
Collapse
Affiliation(s)
- Sarah B Scheinman
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
22
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
23
|
Schworer EK, Zammit MD, Wang J, Handen BL, Betthauser T, Laymon CM, Tudorascu DL, Cohen AD, Zaman SH, Ances BM, Mapstone M, Head E, Christian BT, Hartley SL. Timeline to symptomatic Alzheimer's disease in people with Down syndrome as assessed by amyloid-PET and tau-PET: a longitudinal cohort study. Lancet Neurol 2024; 23:1214-1224. [PMID: 39577922 DOI: 10.1016/s1474-4422(24)00426-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Adults with Down syndrome are at risk for Alzheimer's disease. Natural history cohort studies have characterised the progression of Alzheimer's disease biomarkers in people with Down syndrome, with a focus on amyloid β-PET and tau-PET. In this study, we aimed to leverage these well characterised imaging biomarkers in a large cohort of individuals with Down syndrome, to examine the timeline to symptomatic Alzheimer's disease based on estimated years since the detection on PET of amyloid β-positivity, referred to here as amyloid age, and in relation to tau burden as assessed by PET. METHODS In this prospective, longitudinal, observational cohort study, data were collected at four university research sites in the UK and USA as part of the Alzheimer's Biomarker Consortium-Down Syndrome (ABC-DS) study. Eligible participants were aged 25 years or older with Down syndrome, had a mental age of at least 3 years (based on a standardised intelligence quotient test), and had trisomy 21 (full, mosaic, or translocation) confirmed through karyotyping. Participants were assessed twice between 2017 and 2022, with approximately 32 months between visits. Participants had amyloid-PET and tau-PET scans, and underwent cognitive assessment with the modified Cued Recall Test (mCRT) and the Down Syndrome Mental Status Examination (DSMSE) to assess cognitive functioning. Study partners completed the National Task Group-Early Detection Screen for Dementia (NTG-EDSD). Generalised linear models were used to assess the association between amyloid age (whereby 0 years equated to 18 centiloids) and mCRT, DSMSE, NTG-EDSD, and tau PET at baseline and the 32-month follow-up. Broken stick regression was used to identify the amyloid age that corresponded to decreases in cognitive performance and increases in tau PET after the onset of amyloid β positivity. FINDINGS 167 adults with Down syndrome, of whom 92 had longitudinal data, were included in our analyses. Generalised linear regressions showed significant quadratic associations between amyloid age and cognitive performance and cubic associations between amyloid age and tau, both at baseline and at the 32-month follow-up. Using broken stick regression models, differences in mCRT total scores were detected beginning 2·7 years (95% credible interval [CrI] 0·2 to 5·4; equating to 29·8 centiloids) after the onset of amyloid β positivity in cross-sectional models. Based on cross-sectional data, increases in tau deposition started a mean of 2·7-6·1 years (equating to 29·8-47·9 centiloids) after the onset of amyloid β positivity. Mild cognitive impairment was observed at a mean amyloid age of 7·4 years (SD 6·6; equating to 56·8 centiloids) and dementia was observed at a mean amyloid age of 12·7 years (5·6; equating to 97·4 centiloids). INTERPRETATION There is a short timeline to initial cognitive decline and dementia from onset of amyloid β positivity and tau deposition in people with Down syndrome. This newly established timeline based on amyloid age (or equivalent centiloid values) is important for clinical practice and informing the design of Alzheimer's disease clinical trials, and it avoids the limitations of timelines based on chronological age. FUNDING National Institute on Aging and the National Institute for Child Health and Human Development.
Collapse
Affiliation(s)
- Emily K Schworer
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew D Zammit
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jiebiao Wang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tobey Betthauser
- Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annie D Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shahid H Zaman
- Cambridge Intellectual Disability Research Group, University of Cambridge, Cambridge, UK
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Bradley T Christian
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Sigan L Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; School of Human Ecology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
24
|
Ohno K, Sawada S, Fujimaki N, Sakai K, Wakui S, Shibata N, Sato N, Naito H, Machida S. The Association Between Mild Cognitive Impairment and Physical Function in Older Japanese Adults Aged 75 Years or Older Living in Independent Senior Housing: A Cross-Sectional Study. Healthcare (Basel) 2024; 12:2106. [PMID: 39517319 PMCID: PMC11544811 DOI: 10.3390/healthcare12212106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objective: Although there are many reports on the association between cognitive and physical functions in older adults, little information is available on those aged ≥75 years. Therefore, this study aimed to determine whether mild cognitive impairment (MCI) in older adults over 75 years who live in independent senior housing is associated with physical function. Methods: In this study, 271 participants (174 women and 97 men) with a mean age of 85.4 ± 4.7 years were included. Cognitive function was assessed using the Japanese version of the Montreal Cognitive Assessment; a score < 26 confirmed MCI. MCI was an objective variable in univariate and multivariable logistic regression analyses. Physical function was measured using hand grip strength, normal and maximum gait speeds, and the 30 s chair stand test. Physical function was an explanatory variable adjusted for age and divided into tertiles (high, middle, and low) based on sex. The significance level was set at 5%. Results: There were 170 participants (63%) with MCI. Compared to the non-MCI group, the MCI group had significantly higher age and significantly lower normal and maximum gait speeds and 30 s chair stand test values Age-adjusted univariate analyses in women showed higher MCI rates in the low-fitness group than in the high-fitness group for maximum gait speed and 30 s chair stand test values. No variables were associated with MCI in men. Conclusions: MCI may be associated with physical function in women and older adults over 75 years who live in independent senior housing.
Collapse
Affiliation(s)
- Kanako Ohno
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (K.O.); (H.N.)
- Tokyu E-Life Design Co., Ltd., Tokyo 150-0043, Japan; (N.F.); (K.S.)
| | - Shuji Sawada
- Faculty of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (S.S.); (S.W.)
| | - Naho Fujimaki
- Tokyu E-Life Design Co., Ltd., Tokyo 150-0043, Japan; (N.F.); (K.S.)
| | - Kyoko Sakai
- Tokyu E-Life Design Co., Ltd., Tokyo 150-0043, Japan; (N.F.); (K.S.)
| | - Sawako Wakui
- Faculty of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (S.S.); (S.W.)
| | - Nobuto Shibata
- Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan;
| | - Nobuhiro Sato
- Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan;
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (K.O.); (H.N.)
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (K.O.); (H.N.)
| |
Collapse
|
25
|
Pannangrong W, Nillert N, Boonyarat C, Welbat JU, Yannasithinon S, Choowong-In P. Clausena harmandiana root extract ameliorates Aβ 1-42 induced cognitive deficits, oxidative stress, and apoptosis in rats. BMC Complement Med Ther 2024; 24:364. [PMID: 39390478 PMCID: PMC11465876 DOI: 10.1186/s12906-024-04662-4] [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: 10/30/2023] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Clausena harmandiana (CH), commonly known as song fa dong, was a medicinal plant traditionally used to treat illnesses and as a health tonic. CH root extract (CHRE) exhibited various bioactivities, including neuroprotective, antioxidant, antimicrobial, antifungal, anti-inflammatory, and anti-cancer effects. However, CHRE data on neuroprotective in AD-like animal models were still scarce. OBJECTIVES This study aimed to investigate the effects of CHRE on Aβ1-42-induced cognitive deficits, free radical damage, and neuronal death in rats. METHODS Forty-eight adult male Sprague-Dawley rats (250-300 g) were classified as sham control (SC), V+Aβ, Vit C+Aβ, CHRE125+Aβ, CHRE250+Aβ, and CHRE500+Aβ (n = 8 in each group). Animals were orally administered with 0.5% sodium carboxymethylcellulose, vitamin C (200 mg/kg BW), or CHRE (125, 250, and 500 mg/kg BW) and were untreated for 35 days. On day 21, all treated rats were injected with 1 µl of aggregated Aβ1-42 (1 µg/µl) into the lateral ventricles, bilaterally, whereas untreated rats were injected with sterilized normal saline (NS). The Morris water maze test estimated the rat's learning and memory one week later. At the end of the treatment, all rats were sacrificed, and their brains were removed and divided into two hemispheres. On the left, morphological changes and neuronal density were observed in hippocampal CA1 and CA3 regions. While, on the right, changes in free radical damage markers (SOD, CAT, GPx, MDA, and Nrf2) and protein expression of active caspase-3 were evaluated in the hippocampus. RESULTS Pretreatment with CHRE at all doses could alleviate spatial learning and memory defects. CHRE also improved morphological changes and a decrease in neuronal density in CA1 and CA3 regions. Additionally, CHRE significantly increased the activities of antioxidant enzymes (SOD, CAT, GPx) and Nrf2 expression. This was coupled with significantly decreased MDA levels and active caspase-3 expression in the hippocampus of Aβ1-42-induced rats, which was similar to vitamin C exposure. CONCLUSIONS Our findings suggested that CHRE ameliorated cognitive deficits and exhibited neuroprotective effects by reducing free radical damage and mitigating neuronal abnormality and neuronal death.
Collapse
Affiliation(s)
- Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nutchareeporn Nillert
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Faculty of Nursing Sciences and Allied Health, Phetchaburi Rajabhat University, Phetchaburi, 76000, Thailand
| | - Chantana Boonyarat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | - Pannawat Choowong-In
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Faculty of Science and Technology, Uttaradit Rajabhat University, Uttaradit, 53000, Thailand.
| |
Collapse
|
26
|
López Hanotte J, Peralta F, Reggiani PC, Zappa Villar MF. Investigating the Impact of Intracerebroventricular Streptozotocin on Female Rats with and without Ovaries: Implications for Alzheimer's Disease. Neurochem Res 2024; 49:2785-2802. [PMID: 38985243 DOI: 10.1007/s11064-024-04204-x] [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: 05/03/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
To contribute to research on female models of Alzheimer's disease (AD), our aim was to study the effect of intracerebroventricular (ICV) injection of streptozotocin (STZ) in female rats, and to evaluate a potential neuroprotective action of ovarian steroids against STZ. Female rats were either ovariectomized (OVX) or kept with ovaries (Sham) two weeks before ICV injections. Animals were injected with either vehicle (artificial cerebrospinal fluid, aCSF) or STZ (3 mg/kg) and separated into four experimental groups: Sham + aCSF, Sham + STZ, OVX + aCSF and OVX + STZ. Nineteen days post-injection, we assessed different behavioral aspects: burying, anxiety and exploration, object recognition memory, spatial memory, and depressive-like behavior. Immunohistochemistry and Immunoblot analyses were performed in the hippocampus to examine changes in AD-related proteins and neuronal and microglial populations. STZ affected burying and exploratory behavior depending on ovarian status, and impaired recognition but not spatial memory. STZ and ovariectomy increased depressive-like behavior. Interestingly, STZ did not alter the expression of β-amyloid peptide or Tau phosphorylated forms. STZ affected the neuronal population from the Dentate Gyrus, where immature neurons were more vulnerable to STZ in OVX rats. Regarding microglia, STZ increased reactive cells, and the OVX + STZ group showed an increase in the total cell number. In sum, STZ partially affected female rats, compared to what was previously reported for males. Although AD is more frequent in women, reports about the effect of ICV-STZ in female rats are scarce. Our work highlights the need to deepen into the effects of STZ in the female brain and study possible sex differences.
Collapse
Affiliation(s)
- Juliette López Hanotte
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Facundo Peralta
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Paula Cecilia Reggiani
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
| | - María Florencia Zappa Villar
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina.
| |
Collapse
|
27
|
Tábuas-Pereira M, Bernardes C, Durães J, Lima M, Nogueira AR, Saraiva J, Tábuas T, Coelho M, Paquette K, Westra K, Kun-Rodrigues C, Almeida MR, Baldeiras I, Brás J, Guerreiro R, Santana I. Exploring first-degree family history in a cohort of Portuguese Alzheimer's disease patients: population evidence for X-chromosome linked and recessive inheritance of risk factors. J Neurol 2024; 271:6983-6990. [PMID: 39235525 PMCID: PMC11447147 DOI: 10.1007/s00415-024-12673-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) heritability is estimated to be around 70-80%. Yet, much of it remains to be explained. Studying transmission patterns may help in understanding other factors contributing to the development of AD. OBJECTIVE In this study, we aimed to search for evidence of autosomal recessive or X- and Y-linked inheritance of risk factors in a large cohort of Portuguese AD patients. METHODS We collected family history from patients with AD and cognitively healthy controls over 75 years of age. We compared the proportions of maternal and paternal history in male and female patients and controls (to search for evidence of X-linked and Y-linked inherited risk factors). We compared the risk of developing AD depending on parents' birthplace (same vs. different), as a proxy of remote consanguinity. We performed linear regressions to study the association of these variables with different endophenotypes. RESULTS We included 3090 participants, 2183 cognitively healthy controls and 907 patients with AD. Men whose mother had dementia have increased odds of developing AD comparing to women whose mother had dementia. In female patients with a CSF biomarker-supported diagnosis of AD, paternal history of dementia is associated with increased CSF phosphorylated Tau levels. People whose parents are from the same town have higher risk of dementia. In multivariate analysis, this proxy is associated with a lower age of onset and higher CSF phosphorylated tau. CONCLUSIONS Our study gives evidence supporting an increased risk of developing AD associated with an X-linked inheritance pattern and remote consanguinity.
Collapse
Affiliation(s)
- Miguel Tábuas-Pereira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal.
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal.
| | - Catarina Bernardes
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - João Durães
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Marisa Lima
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Jorge Saraiva
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Teresa Tábuas
- Instituto Politécnico de Bragança, Bragança, Portugal
| | - Mariana Coelho
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Kimberly Paquette
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Kaitlyn Westra
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Célia Kun-Rodrigues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Maria Rosário Almeida
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| |
Collapse
|
28
|
Lin HY, Feng YH, Kao TJ, Chen HC, Chen GY, Ko CY, Hsu TI. Exploring neuron-specific steroid synthesis and DHEAS therapy in Alzheimer's disease. J Steroid Biochem Mol Biol 2024; 243:106585. [PMID: 39019196 DOI: 10.1016/j.jsbmb.2024.106585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/22/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by cognitive decline and memory loss. Recent studies have suggested a potential role for steroid synthesis in AD pathology. This study investigated the co-localization of steroidogenic enzymes in neuronal cells, changes in enzyme expression in an AD mouse model, and steroid expressions in human AD samples. Additionally, we conducted a steroidomic metabolomics analysis and evaluated the effects of dehydroepiandrosterone sulfate (DHEAS) treatment in an AD mouse model. Immunofluorescence analysis revealed significant co-localization of cytochrome P450 family 17 subfamily A member 1 (CYP17A1) and steroidogenic acute regulatory protein (StAR) proteins with α-synuclein in presynaptic neurons, suggesting active steroid synthesis in these cells. Conversely, such co-localization was absent in astrocytes. In the AD mouse model, a marked decrease in the expression of steroidogenic enzymes (Cyp11a1, Cyp17a1, Star) was observed, especially in areas with amyloid beta plaque accumulation. Human AD and MS brain tissues showed similar reductions in StAR and CYP17A1 expressions. Steroidomic analysis indicated a downregulation of key steroids in the serum of AD patients. DHEAS treatment in AD mice resulted in improved cognitive function and reduced Aβ accumulation. Our findings indicate a neuron-specific pathway for steroid synthesis, potentially playing a crucial role in AD pathology. The reduction in steroidogenic enzymes and key steroids in AD models and human samples suggests that impaired steroid synthesis is a feature of neurodegenerative diseases. The therapeutic potential of targeting steroid synthesis pathways, as indicated by the positive effects of DHEAS treatment, warrants further investigation.
Collapse
Affiliation(s)
- Hong-Yi Lin
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan
| | | | - Tzu-Jen Kao
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Research Center for Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Hsien-Chung Chen
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Guan-Yuan Chen
- Graduate Institute of Forensic Medicine, National Taiwan University, Taipei, Taiwan
| | - Chiung-Yuan Ko
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan; Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Biomedical Science and Environment Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Research Center for Neuroscience, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
29
|
Pan J, Yao Q, Wang Y, Chang S, Li C, Wu Y, Shen J, Yang R. The role of PI3K signaling pathway in Alzheimer's disease. Front Aging Neurosci 2024; 16:1459025. [PMID: 39399315 PMCID: PMC11466886 DOI: 10.3389/fnagi.2024.1459025] [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: 07/03/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating progressively neurodegenerative disease. The best-characterized hallmark of AD, which is marked by behavioral alterations and cognitive deficits, is the aggregation of deposition of amyloid-beta (Aβ) and hyper-phosphorylated microtubule-associated protein Tau. Despite decades of experimental progress, the control rate of AD remains poor, and more precise deciphering is needed for potential therapeutic targets and signaling pathways involved. In recent years, phosphoinositide 3-kinase (PI3K) and Akt have been recognized for their role in the neuroprotective effect of various agents, and glycogen synthase kinase 3 (GSK3), a downstream enzyme, is also crucial in the tau phosphorylation and Aβ deposition. An overview of the function of PI3K/Akt pathway in the pathophysiology of AD is provided in this review, along with a discussion of recent developments in the pharmaceuticals and herbal remedies that target the PI3K/Akt signaling pathway. In conclusion, despite the challenges and hurdles, cumulative findings of novel targets and agents in the PI3K/Akt signaling axis are expected to hold promise for advancing AD prevention and treatment.
Collapse
Affiliation(s)
- Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yankai Wang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Suyan Chang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Chenlong Li
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongjiang Wu
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jianhong Shen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
30
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
31
|
Castillo-Mendieta T, Bautista-Poblet G, Coyoy-Salgado A, Castillo-García EL, Pinto-Almazán R, Fuentes-Venado CE, Neri-Gómez T, Guerra-Araiza C. Effect of Chronic Tibolone Administration on Memory and Choline Acetyltransferase and Tryptophan Hydroxylase Content in Aging Mice. Brain Sci 2024; 14:903. [PMID: 39335399 PMCID: PMC11430777 DOI: 10.3390/brainsci14090903] [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: 07/11/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Gonadal steroids exert different effects on the central nervous system (CNS), such as preserving neuronal function and promoting neuronal survival. Estradiol, progesterone, and testosterone reduce neuronal loss in the CNS in animal models of neurodegeneration. However, hormone replacement therapy has been associated with higher rates of endometrial, prostate, and breast cancer. Tibolone (TIB), the metabolites of which show estrogenic and progestogenic effects, is an alternative to reduce this risk. However, the impact of TIB on memory and learning, as well as on choline acetyltransferase (ChAT) and tryptophan hydroxylase (TPH) levels in the hippocampus of aging males, is unknown. We administered TIB to aged C57BL/6J male mice at different doses (0.01 or 1.0 mg/kg per day for 12 weeks) and evaluated its effects on memory and learning and the content of ChAT and TPH. We assessed memory and learning with object recognition and elevated T-maze tasks. Additionally, we determined ChAT and TPH protein levels in the hippocampus by Western blotting. TIB administration increased the percentage of time spent on the novel object in the object recognition task. In addition, the latency of leaving the enclosed arm increased in both TIB groups, suggesting an improvement in fear-based learning. We also observed decreased ChAT content in the group treated with the 0.01 mg/kg TIB dose. In the case of TPH, no changes were observed with either TIB dose. These results show that long-term TIB administration improves memory without affecting locomotor activity and modulates cholinergic but not serotonergic systems in the hippocampus of aged male mice.
Collapse
Affiliation(s)
- Tzayaka Castillo-Mendieta
- CONAHCyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 03940, Mexico
| | - Guadalupe Bautista-Poblet
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 06720, Mexico (A.C.-S.)
- Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City C.P. 09340, Mexico
| | - Angélica Coyoy-Salgado
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 06720, Mexico (A.C.-S.)
| | - Emily L. Castillo-García
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 06720, Mexico (A.C.-S.)
- Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City C.P. 09340, Mexico
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City C.P. 11340, Mexico; (R.P.-A.)
| | - Claudia Erika Fuentes-Venado
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City C.P. 11340, Mexico; (R.P.-A.)
- Servicio de Medicina Física y Rehabilitación, Hospital General de Zona No 197 IMSS, Texcoco C.P. 56108, Mexico
| | - Teresa Neri-Gómez
- Laboratorio de Patología Molecular, Unidad de Investigación Biomolecular en Cardiología, Hospital de Cardiología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 03940, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Mexico City C.P. 06720, Mexico (A.C.-S.)
| |
Collapse
|
32
|
Li T, Chen J, Zhao B, Garden GA, Giovanello KS, Wu G, Zhu H. The Interaction Effects of Sex, Age, APOE and Common Health Risk Factors on Human Brain Functions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.05.24311482. [PMID: 39148839 PMCID: PMC11326347 DOI: 10.1101/2024.08.05.24311482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Importance Nonlinear changes in brain function during aging are shaped by a complex interplay of factors, including sex, age, genetics, and modifiable health risk factors. However, the combined effects and underlying mechanisms of these factors on brain functional connectivity remain poorly understood. Objective To comprehensively investigate the combined associations of sex, age, APOE genotypes, and ten common modifiable health risk factors with brain functional connectivities during aging. Design Setting and Participants This analysis used data from 36,630 UK Biobank participants, aged 44-81, who were assessed for sex, age, APOE genotypes, 10 health risk factors, and brain functional connectivities through resting-state functional magnetic resonance imaging. Main Outcomes and Measures Brain functional connectivities were evaluated through within- and between-network functional connectivities and connectivity strength. Associations between risk factors and brain functional connectivities, including their interaction effects, were analyzed. Results Hypertension, BMI, and education were the top three influential factors. Sex-specific effects were also observed in interactions involving APOE4 gene, smoking, alcohol consumption, diabetes, BMI, and education. Notably, a negative sex-excessive alcohol interaction showed a stronger negative effect on functional connectivities in males, particularly between the dorsal attention network and the language network, while moderate alcohol consumption appeared to have protective effects. A significant negative interaction between sex and APOE4 revealed a greater reduction in functional connectivity between the cingulo-opercular network and the posterior multimodal network in male APOE4 carriers. Additional findings included a negative age-BMI interaction between the visual and dorsal attention networks, and a positive age-hypertension interaction between the frontoparietal and default mode networks. Conclusions and Relevance The findings highlight significant sex disparities in the associations between age, the APOE-ε4 gene, modifiable health risk factors, and brain functional connectivity, emphasizing the necessity of jointly considering these factors to gain a deeper understanding of the complex processes underlying brain aging.
Collapse
Affiliation(s)
- Tengfei Li
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, the Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Gwenn A. Garden
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kelly S. Giovanello
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Guorong Wu
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Statistics and Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Insititute for Developmental Disabilities, Chapel Hill, NC, USA
| | - Hongtu Zhu
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Statistics and Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Departments of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
33
|
He R, Cheng J, Qiu Y, Hu Y, Liu J, Wang TH, Cao X. IGF1R and FLT1 in female endothelial cells and CHD2 in male microglia play important roles in Alzheimer's disease based on gender difference analysis. Exp Gerontol 2024; 194:112512. [PMID: 38971545 DOI: 10.1016/j.exger.2024.112512] [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: 11/24/2023] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE This study investigated sex-specific pathogenesis mechanisms in Alzheimer's disease (AD) using single-nucleus RNA sequencing (snRNA-seq) data. METHODS Data from the Gene Expression Omnibus (GEO) were searched using terms "Alzheimer's Disease", "single cell", and "Homo sapiens". Studies excluding APOE E4 and including comprehensive gender information with 10× sequencing methods were selected, resulting in GSE157827 and GSE174367 datasets from human prefrontal cortex samples. Sex-stratified analyses were conducted on these datasets, and the outcomes of the analysis for GSE157827 were compared with those of GSE174367. The findings were validated using expression profiling from the mouse dataset GSE85162. Furthermore, real-time PCR experiments in mice further confirmed these findings. The Seurat R package was used to identify cell types, and batch effects were mitigated using the Harmony R package. Cell proportions by sex were compared using the Mann-Whitney-Wilcoxon test, and gene expression variability was displayed with an empirical cumulative distribution plot. Differentially expressed genes were identified using the FindMarkers function with the MAST test. Transcription factors were analyzed using the RcisTarget R package. RESULTS Seven cell types were identified: astrocytes, endothelial cells, excitatory neurons, inhibitory neurons, microglia, oligodendrocytes, and oligodendrocyte progenitor cells. Additionally, five distinct subpopulations of both endothelial and microglial cells were also identified, respectively. Key findings included: (1) In endothelial cells, genes involved in synapse organization, such as Insulin Like Growth Factor 1 Receptor (IGF1R) and Fms Related Receptor Tyrosine Kinase 1(FLT1), showed higher expression in females with AD. (2) In microglial cells, genes in the ribosome pathway exhibited higher expression in males without AD compared to females (with or without AD) and males with AD. (3) Chromodomain Helicase DNA Binding Protein 2 (CHD2) negatively regulated gene expression in the ribosome pathway in male microglia, suppressing AD, this finding was further validated in mice. (4) Differences between Asians and Caucasians were observed based on sex and disease status stratification. CONCLUSIONS IGF1R and FLT1 in endothelial cells contribute to AD in females, while CHD2 negatively regulates ribosome pathway gene expression in male microglia, suppressing AD in humans and mice.
Collapse
Affiliation(s)
- Rong He
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jishuai Cheng
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Yue Qiu
- Dermatology Department of Xiangya Hospital, Central South University, Changsha, China
| | - Yiwen Hu
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jia Liu
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| | - Ting-Hua Wang
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| | - Xue Cao
- Laboratory Animal Department, Kunming Medical University, Kunming 650500, Yunnan, China.
| |
Collapse
|
34
|
Cao D, Zhang S, Zhang Y, Shao M, Yang Q, Wang P. Association between gynecologic cancer and Alzheimer's disease: a bidirectional mendelian randomization study. BMC Cancer 2024; 24:1032. [PMID: 39169299 PMCID: PMC11337634 DOI: 10.1186/s12885-024-12787-5] [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: 01/02/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) manifests with a higher rate of occurrence in women. Previous epidemiological studies have suggested a potential association between AD and gynecological cancers, but the causal relationship between them remains unclear. This study aims to explore the causal link between 12 types of gynecological cancers and AD using a bidirectional Mendelian randomization (MR) approach. METHODS We obtained genetic correlation tools for AD using data from the most extensive genome-wide association study. Genetic correlation data for 12 types of gynecological cancers were also sourced from the Finnish Biobank. These cancers include breast cancer (BC), cervical adenocarcinoma (CA), cervical squamous cell carcinoma (CSCC), cervical cancer (CC), endometrial cancer (EC), ovarian endometrioid carcinoma (OEC), ovarian cancer (OC), ovarian serous carcinoma (OSC), breast carcinoma in situ (BCIS), cervical carcinoma in situ (CCIS), endometrial carcinoma in situ (ECIS), and vulvar carcinoma in situ (VCIS). We used the inverse-variance weighted (IVW) model for causal analysis and conducted horizontal pleiotropy tests, heterogeneity tests, MR-PRESSO tests, and leave-one-out analyses to ensure the robustness of our results. We also applied replication analysis and meta-analysis to further validate our experimental results. RESULTS The study found that EC (P_IVW =0.037, OR [95% CI] = 1.032 [1.002, 1.064]) and CCIS (P_IVW = 0.046, OR [95% CI] = 1.032 [1.011, 1.064]) increase the risk of AD, whereas OC was negatively correlated with AD (P_IVW = 0.016, OR [95% CI] = 0.974[0.954, 0.995]). In reverse MR analysis, AD increased the risk of CC (P_IVW = 0.039, OR [95% CI] = 1.395 [1.017, 1.914]) and VCIS (P_IVW = 0.041, OR [95% CI] = 1.761 [1.027, 2.021]), but was negatively correlated with OEC (P_IVW = 0.034, OR [95% CI] = 0.634 [0.417, 0.966]). Sensitivity analysis results demonstrated robustness. These findings were further substantiated through replication and meta-analyses. CONCLUSIONS Our MR study supports a causal relationship between AD and gynecological cancers. This encourages further research into the incidence of gynecological cancers in female Alzheimer's patients and the active prevention of AD.
Collapse
Affiliation(s)
- Di Cao
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430065, China
| | - Shaobo Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, 130000, China
| | - Yini Zhang
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430065, China
| | - Ming Shao
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210000, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210000, China
| | - Qiguang Yang
- The Second Affiliated Hospital of Changchun University of Chinese Medicine, Changchun Hospital of Chinese Medicine, Changchun, Jilin, 130000, China
| | - Ping Wang
- Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, Hubei, 430065, China.
| |
Collapse
|
35
|
Chen G, Jin Y, Chu C, Zheng Y, Chen Y, Zhu X. Genetic prediction of blood metabolites mediating the relationship between gut microbiota and Alzheimer's disease: a Mendelian randomization study. Front Microbiol 2024; 15:1414977. [PMID: 39224217 PMCID: PMC11366617 DOI: 10.3389/fmicb.2024.1414977] [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: 04/10/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Background Observational studies have suggested an association between gut microbiota and Alzheimer's disease (AD); however, the causal relationship remains unclear, and the role of blood metabolites in this association remains elusive. Purpose To elucidate the causal relationship between gut microbiota and AD and to investigate whether blood metabolites serve as potential mediators. Materials and methods Univariable Mendelian randomization (UVMR) analysis was employed to assess the causal relationship between gut microbiota and AD, while multivariable MR (MVMR) was utilized to mitigate confounding factors. Subsequently, a two-step mediation MR approach was employed to explore the role of blood metabolites as potential mediators. We primarily utilized the inverse variance-weighted method to evaluate the causal relationship between exposure and outcome, and sensitivity analyses including Contamination mixture, Maximum-likelihood, Debiased inverse-variance weighted, MR-Egger, Bayesian Weighted Mendelian randomization, and MR pleiotropy residual sum and outlier were conducted to address pleiotropy. Results After adjustment for reverse causality and MVMR correction, class Actinobacteria (OR: 1.03, 95% CI: 1.01-1.06, p = 0.006), family Lactobacillaceae (OR: 1.03, 95% CI: 1.00-1.05, p = 0.017), genus Lachnoclostridium (OR: 1.03, 95% CI: 1.00-1.06, p = 0.019), genus Ruminiclostridium9 (OR: 0.97, 95% CI: 0.94-1.00, p = 0.027) and genus Ruminiclostridium6 (OR: 1.03, 95% CI: 1.01-1.05, p = 0.009) exhibited causal effects on AD. Moreover, 1-ribosyl-imidazoleacetate levels (-6.62%), Metabolonic lactone sulfate levels (2.90%), and Nonadecanoate (19:0) levels (-12.17%) mediated the total genetic predictive effects of class Actinobacteria on AD risk. Similarly, 2-stearoyl-GPE (18:0) levels (-9.87%), Octadecanedioylcarnitine (C18-DC) levels (4.44%), 1-(1-enyl-stearoyl)-2-oleoyl-GPE (p-18:0/18:1) levels (38.66%), and X-23639 levels (13.28%) respectively mediated the total genetic predictive effects of family Lactobacillaceae on AD risk. Furthermore, Hexadecanedioate (C16-DC) levels (5.45%) mediated the total genetic predictive effects of genus Ruminiclostridium 6 on AD risk; Indole-3-carboxylate levels (13.91%), X-13431 levels (7.08%), Alpha-ketoglutarate to succinate ratio (-13.91%), 3-phosphoglycerate to glycerate ratio (15.27%), and Succinate to proline ratio (-14.64%) respectively mediated the total genetic predictive effects of genus Ruminiclostridium 9 on AD risk. Conclusion Our mediation MR analysis provides genetic evidence suggesting the potential mediating role of blood metabolites in the causal relationship between gut microbiota and AD. Further large-scale randomized controlled trials are warranted to validate the role of blood metabolites in the specific mechanisms by which gut microbiota influence AD.
Collapse
Affiliation(s)
- Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yaxian Jin
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Cancan Chu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yuhao Zheng
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yunzhi Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xing Zhu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
36
|
Handschuh PA, Reed MB, Murgaš M, Vraka C, Kaufmann U, Nics L, Klöbl M, Ozenil M, Konadu ME, Patronas EM, Spurny-Dworak B, Hahn A, Hacker M, Spies M, Baldinger-Melich P, Kranz GS, Lanzenberger R. Effects of gender-affirming hormone therapy on gray matter density, microstructure and monoamine oxidase A levels in transgender subjects. Neuroimage 2024; 297:120716. [PMID: 38955254 DOI: 10.1016/j.neuroimage.2024.120716] [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: 01/26/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024] Open
Abstract
MAO-A catalyzes the oxidative degradation of monoamines and is thus implicated in sex-specific neuroplastic processes that influence gray matter (GM) density (GMD) and microstructure (GMM). Given the exact monitoring of plasma hormone levels and sex steroid intake, transgender individuals undergoing gender-affirming hormone therapy (GHT) represent a valuable cohort to potentially investigate sex steroid-induced changes of GM and concomitant MAO-A density. Here, we investigated the effects of GHT over a median time period of 4.5 months on GMD and GMM as well as MAO-A distribution volume. To this end, 20 cisgender women, 11 cisgender men, 20 transgender women and 10 transgender men underwent two MRI scans in a longitudinal design. PET scans using [11C]harmine were performed before each MRI session in a subset of 35 individuals. GM changes determined by diffusion weighted imaging (DWI) metrics for GMM and voxel based morphometry (VBM) for GMD were estimated using repeated measures ANOVA. Regions showing significant changes of both GMM and GMD were used for the subsequent analysis of MAO-A density. These involved the fusiform gyrus, rolandic operculum, inferior occipital cortex, middle and anterior cingulum, bilateral insula, cerebellum and the lingual gyrus (post-hoc tests: pFWE+Bonferroni < 0.025). In terms of MAO-A distribution volume, no significant effects were found. Additionally, the sexual desire inventory (SDI) was applied to assess GHT-induced changes in sexual desire, showing an increase of SDI scores among transgender men. Changes in the GMD of the bilateral insula showed a moderate correlation to SDI scores (rho = - 0.62, pBonferroni = 0.047). The present results are indicative of a reliable influence of gender-affirming hormone therapy on 1) GMD and GMM following an interregional pattern and 2) sexual desire specifically among transgender men.
Collapse
Affiliation(s)
- P A Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M B Reed
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - C Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - U Kaufmann
- Department of Obstetrics and Gynecology, Medical University of Vienna, Austria
| | - L Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M Klöbl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Ozenil
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M E Konadu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - E M Patronas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - B Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - A Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - M Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - M Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - P Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - G S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria; Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria.
| |
Collapse
|
37
|
Lima VDS, Ferreira YG, de Oliveira JC, Raia VDA, Rodrigues Emerick LBB, Albiero LR, Sinhorin VDG, Emerick GL. Alzheimer's Disease Mortality Rate: Correlation with Socio-Economic and Environmental Factors. TOXICS 2024; 12:586. [PMID: 39195688 PMCID: PMC11359452 DOI: 10.3390/toxics12080586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
The progressive increase in the number of deaths caused by Alzheimer's disease (AD) in Brazil and around the world between 2010 and 2020 raises questions in scientific society. At the same time, there is also an increase in life expectancy at birth (LEB). Thus, the aim of this study was, for the first time, to compare the increase in AD mortality rate (ADMR) in Brazilian regions over the years 2010 to 2020 with the increase in LEB, and investigate the possible correlation between these demographic transition phenomena and pesticide sales and exposure during this period. Data were extracted from the Brazilian Institute of Geography and Statistics (IBGE), from the Department of Informatics and Technology of the Brazilian Ministry of Health (DATASUS) and from the Brazilian Institute of the Environment and Renewable Natural Resources (IBAMA). There was a significant increase in life expectancy at birth and in ADMR over the years between 2010 and 2020 in all Brazilian regions, with the female population in the South region being the most affected. In conclusion, there is a strong positive correlation between the increase in ADMR and LEB; ADMR and Human Development Index (HDI) and ADMR and pesticide sales and exposure in Brazil over the years studied.
Collapse
Affiliation(s)
- Valfran da Silva Lima
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
- Programa de Pós-Graduação em Ciências em Saúde, Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Sinop 78550-728, MT, Brazil
| | - Yasmin Gabriele Ferreira
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
| | - Júlio Cezar de Oliveira
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
- Programa de Pós-Graduação em Ciências em Saúde, Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Sinop 78550-728, MT, Brazil
| | - Vanessa de Almeida Raia
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
| | - Ludmila Barbosa Bandeira Rodrigues Emerick
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
- Programa de Pós-Graduação em Ciências em Saúde, Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Sinop 78550-728, MT, Brazil
| | - Lucinéia Reuse Albiero
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
| | | | - Guilherme Luz Emerick
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Campus Sinop, Avenida Alexandre Ferronato, 1200, Cidade Jardim, Sinop 78550-728, MT, Brazil; (V.d.S.L.); (Y.G.F.); (J.C.d.O.); (V.d.A.R.); (L.B.B.R.E.); (L.R.A.)
- Programa de Pós-Graduação em Ciências em Saúde, Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso—ICS/CUS/UFMT, Sinop 78550-728, MT, Brazil
| |
Collapse
|
38
|
Schworer EK, Zammit MD, Wang J, Handen BL, Betthauser T, Laymon CM, Tudorascu DL, Cohen AD, Zaman SH, Ances BM, Mapstone M, Head E, Klunk WE, Christian BT, Hartley SL. Amyloid age and tau PET timeline to symptomatic Alzheimer's disease in Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311702. [PMID: 39211859 PMCID: PMC11361254 DOI: 10.1101/2024.08.08.24311702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Adults with Down syndrome (DS) are at risk for Alzheimer's disease (AD). Recent natural history cohort studies have characterized AD biomarkers, with a focus on PET amyloid-beta (Aβ) and PET tau. Leveraging these well-characterized biomarkers, the present study examined the timeline to symptomatic AD based on estimated years since reaching Aβ+, referred to as "amyloid age", and in relation to tau in a large cohort of individuals with DS. Methods In this multicenter cohort study, 25 - 57-year-old adults with DS (n = 167) were assessed twice from 2017 to 2022, with approximately 32 months between visits as part of the Alzheimer Biomarker Consortium - Down Syndrome. Adults with DS completed amyloid and tau PET scans, and were administered the modified Cued Recall Test and the Down Syndrome Mental Status Examination. Study partners completed the National Task Group-Early Detection Screen for Dementia. Findings Mixed linear regressions showed significant quadratic associations between amyloid age and cognitive performance and cubic associations between amyloid age and tau, both at baseline and across 32 months. Using broken stick regression models, differences in mCRT scores were detected beginning 2.7 years following Aβ+ in cross-sectional models, with an estimated decline of 1.3 points per year. Increases in tau began, on average, 2.7 - 6.1 years following Aβ+. On average, participants with mild cognitive impairment were 7.4 years post Aβ+ and those with dementia were 12.7 years post Aβ+. Interpretation There is a short timeline to initial cognitive decline and dementia from Aβ+ (Centiloid = 18) and tau deposition in DS relative to late onset AD. The established timeline based on amyloid age (or equivalent Centiloid values) is important for clinical practice and informing AD clinical trials, and avoids limitations of timelines based on chronological age. Funding. National Institute on Aging and the National Institute for Child Health and Human Development. Research in Context Evidence before this study: We searched PubMed for articles published involving the progression of Aβ and tau deposition in adults with Down syndrome from database inception to March 1, 2024. Terms included "amyloid", "Down syndrome", "tau", "Alzheimer's disease", "cognitive decline", and "amyloid chronicity," with no language restrictions. One previous study outlined the progression of tau in adults with Down syndrome without consideration of cognitive decline or clinical status. Other studies reported cognitive decline associated with Aβ burden and estimated years to AD symptom onset in Down syndrome. Amyloid age estimates have also been created for older neurotypical adults and compared to cognitive performance, but this has not been investigated in Down syndrome.Added value of this study: The timeline to symptomatic Alzheimer's disease in relation to amyloid, expressed as duration of Aβ+, and tau has yet to be described in adults with Down syndrome. Our longitudinal study is the first to provide a timeline of cognitive decline and transition to mild cognitive impairment and dementia in relation to Aβ+.Implications of all the available evidence: In a cohort study of 167 adults with Down syndrome, cognitive decline began 2.7 - 5.4 years and tau deposition began 2.7 - 6.1 years following Aβ+ (Centiloid = 18). Adults with Down syndrome converted to MCI after ~7 years and dementia after ~12-13 years of Aβ+. This shortened timeline to AD symptomology from Aβ+ and tau deposition in DS based on amyloid age (or corresponding Centiloid values) can inform clinical AD intervention trials and is of use in clinical settings.
Collapse
|
39
|
Johansen LL, Christensen K, Hallas J, Osler M, Thinggaard M. Postmenopausal hormone therapy and cognition in twins. Eur J Obstet Gynecol Reprod Biol 2024; 298:116-122. [PMID: 38754277 DOI: 10.1016/j.ejogrb.2024.05.006] [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: 03/19/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE Mild cognitive impairment may be caused by pathophysiological changes occurring decades prior to symptom development. It has been hypothesised that oestrogen can prevent such changes. We aimed to investigate the association between postmenopausal hormone therapy and cognition in Danish female twins and to examine differences in this association before and after publication of the findings from the Women's Health Initiative study in 2002. STUDY DESIGN This study includes cognitive assessment of 4510 twins aged 50+ years. Information on hormone therapy was obtained through Danish health registries. The association between current hormone therapy use and cognition was analysed in twins aged 50+ using both cross-sectional, intrapair and longitudinal analysis, adjusting for age, education, social class, and unobserved familial confounding. RESULTS Cross-sectionally, systemic HT users aged 70+ had a significantly lower cognitive function than non-users, whereas systemic HT users aged 50-69 did not differ from non-users before 2002. Longitudinal data in younger twins aged 50-69 showed a significantly lower cognitive function in systemic HT users after 2002 compared to non-users. Systemic HT users aged 70+ showed that the lower cognitive function was most explicit before 2002, whereas after 2002 the cognitive function was closer to non-users. Twins aged 50-69 who changed from systemic HT to local HT after 2002, or dropped it altogether, performed cognitively better. CONCLUSIONS Our findings cautiously indicate a change in the association between cognition and hormone therapy use after 2002, which suggests an alteration in the hormone therapy user profile in the wake of the 2002 WHI publication.
Collapse
Affiliation(s)
- Laura L Johansen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jesper Hallas
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Merete Osler
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospitals, Denmark
| | - Mikael Thinggaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark; The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark; Danish Aging Research Center, Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
40
|
Burmistrov DE, Gudkov SV, Franceschi C, Vedunova MV. Sex as a Determinant of Age-Related Changes in the Brain. Int J Mol Sci 2024; 25:7122. [PMID: 39000227 PMCID: PMC11241365 DOI: 10.3390/ijms25137122] [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] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.
Collapse
Affiliation(s)
- Dmitriy E. Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
41
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [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] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
42
|
Iqbal I, Saqib F, Mubarak Z, Latif MF, Wahid M, Nasir B, Shahzad H, Sharifi-Rad J, Mubarak MS. Alzheimer's disease and drug delivery across the blood-brain barrier: approaches and challenges. Eur J Med Res 2024; 29:313. [PMID: 38849950 PMCID: PMC11161981 DOI: 10.1186/s40001-024-01915-3] [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: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
Alzheimer's disease (AD) is a diverse disease with a complex pathophysiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyper-phosphorylated tau as neurofibrillary tangles remain the core neuropathologic criteria for diagnosing Alzheimer's disease. Nonetheless, several recent basic discoveries have revealed significant pathogenic roles for other essential cellular and molecular processes. Previously, there were not so many disease-modifying medications (DMT) available as drug distribution through the blood-brain barrier (BBB) is difficult due to its nature, especially drugs of polypeptides nature and proteins. Recently FDA has approved lecanemab as DMT for its proven efficacy. It is also complicated to deliver drugs for diseases like epilepsy or any brain tumor due to the limitations of the BBB. After the advancements in the drug delivery system, different techniques are used to transport the medication across the BBB. Other methods are used, like enhancement of brain blood vessel fluidity by liposomes, infusion of hyperosmotic solutions, and local intracerebral implants, but these are invasive approaches. Non-invasive approaches include the formulation of nanoparticles and their coating with polymers. This review article emphasizes all the above-mentioned techniques, procedures, and challenges to transporting medicines across the BBB. It summarizes the most recent literature dealing with drug delivery across the BBB.
Collapse
Affiliation(s)
- Iram Iqbal
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Zobia Mubarak
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Muhammad Farhaj Latif
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Bushra Nasir
- Department of Pharmaceutics, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hamna Shahzad
- Department of Biochemistry, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
43
|
Chen YH, Wang ZB, Liu XP, Xu JP, Mao ZQ. Sex differences in the relationship between depression and Alzheimer's disease-mechanisms, genetics, and therapeutic opportunities. Front Aging Neurosci 2024; 16:1301854. [PMID: 38903903 PMCID: PMC11188317 DOI: 10.3389/fnagi.2024.1301854] [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: 09/25/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024] Open
Abstract
Depression and Alzheimer's disease (AD) are prevalent neuropsychiatric disorders with intriguing epidemiological overlaps. Their interrelation has recently garnered widespread attention. Empirical evidence indicates that depressive disorders significantly contribute to AD risk, and approximately a quarter of AD patients have comorbid major depressive disorder, which underscores the bidirectional link between AD and depression. A growing body of evidence substantiates pervasive sex differences in both AD and depression: both conditions exhibit a higher incidence among women than among men. However, the available literature on this topic is somewhat fragmented, with no comprehensive review that delineates sex disparities in the depression-AD correlation. In this review, we bridge these gaps by summarizing recent progress in understanding sex-based differences in mechanisms, genetics, and therapeutic prospects for depression and AD. Additionally, we outline key challenges in the field, holding potential for improving treatment precision and efficacy tailored to male and female patients' distinct needs.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North, Zhangjiakou, China
| | - Jun-Peng Xu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
44
|
Glaesser D, Iwig M. Increased molar ratio of free fatty acids to albumin in blood as cause and early biomarker for the development of cataracts and Alzheimer's disease. Exp Eye Res 2024; 243:109888. [PMID: 38583754 DOI: 10.1016/j.exer.2024.109888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Cataracts and Alzheimer's disease (AD) are closely linked and are associated with aging and with systemic diseases that increase the molar ratio of free fatty acids to albumin (mFAR) in the blood. From the results of our earlier studies on the development of senile cataracts and from results recently published in the literature on the pathogenesis of Alzheimer's disease, we suggest that there is a common lipotoxic cascade for both diseases, explaining the strong connection between aging, an elevated mFAR in the blood, cataract formation, and AD. Long-chain free fatty acids (FFA) are transported in the blood as FFA/albumin complexes. In young people, vascular albumin barriers in the eyes and brain, very similar in their structure and effect, reduce the FFA/albumin complex concentration from around 650 μmol/l in the blood to 1-3 μmol/l in the aqueous humour of the eyes as well as in the cerebrospinal fluid of the brain. At such low concentrations the fatty acid uptake of the target cells - lens epithelial and brain cells - rises with increasing FFA/albumin complex concentrations, especially when the fatty acid load of albumin molecules is mFAR>1. At higher albumin concentrations, for instance in blood plasma or the interstitial tissue spaces, the fatty acid uptake of the target cells becomes increasingly independent of the FFA/albumin complex concentration and is mainly a function of the mFAR (Richieri et al., 1993). In the blood plasma of young people, the mFAR is normally below 1.0. In people over 40 years old, aging increases the mFAR by decreasing the plasma concentration of albumin and enhancing the plasma concentrations of FFA. The increase in the mFAR in association with C6-unsaturated FFA are risk factors for the vascular albumin barriers (Hennig et al., 1984). Damage to the vascular albumin barrier in the eyes and brain increases the concentration of FFA/albumin complex in the aqueous humour as well as in the cerebrospinal fluid, leading to mitochondrial dysfunction and the death of lens epithelial and brain cells, the development of cataracts, and AD. An age-dependent increase in the concentration of FFA/albumin complex has been found in the aqueous humour of 177 cataract patients, correlating with the mitochondria-mediated apoptotic death of lens epithelial cells, lens opacification and cataracts (Iwig et al., 2004). Mitochondrial dysfunction is also an early crucial event in Alzheimer's pathology, closely connected with the generation of amyloid beta peptides (Leuner et al., 2012). Very recently, amyloid beta production has also been confirmed in the lenses of Alzheimer's patients, causing cataracts (Moncaster et al., 2022). In view of this, we propose that there is a common lipotoxic cascade for senile cataract formation and senile AD, initiated by aging and/or systemic diseases, leading to an mFAR>1 in the blood.
Collapse
Affiliation(s)
- Dietmar Glaesser
- Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, D-06097, Halle, Germany.
| | - Martin Iwig
- Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, D-06097, Halle, Germany
| |
Collapse
|
45
|
Miramontes S, Pereda Serras C, Woldemariam SR, Khan U, Li Y, Tang AS, Tsoy E, Oskotsky TT, Sirota M. Alzheimer's disease as a women's health challenge: a call for action on integrative precision medicine approaches. NPJ WOMEN'S HEALTH 2024; 2:17. [PMID: 38778871 PMCID: PMC11106001 DOI: 10.1038/s44294-024-00021-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's Disease (AD) is marked by pronounced sex differences in pathophysiology and progression. However, the field has yet to fully recognize AD as a women's health issue, delaying the development of targeted preventative strategies and treatments. This perspective explores the elements impacting AD in women, identifying sex specificity in risk factors, highlighting new diagnostic approaches with electronic health records, and reviewing key molecular studies to underscore the need for integrative precision medicine approaches. Established AD risk factors such as advancing age, the apolipoprotein E4 allele, and poorer cardiovascular health affect women differently. We also shed light on sociocultural risk factors, focusing on the gender disparities that may play a role in AD development. From a biological perspective, sex differences in AD are apparent in biomarkers and transcriptomics, further emphasizing the need for targeted diagnostics and treatments. The convergence of novel multiomics data and cutting-edge computational tools provides a unique opportunity to study the molecular underpinnings behind sex dimorphism in AD. Thus, precision medicine emerges as a promising framework for understanding AD pathogenesis through the integration of genetics, sex, environment, and lifestyle. By characterizing AD as a women's health challenge, we can catalyze a transformative shift in AD research and care, marked by improved diagnostic accuracy, targeted interventions, and ultimately, enhanced clinical outcomes.
Collapse
Affiliation(s)
- S. Miramontes
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - C. Pereda Serras
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - S. R. Woldemariam
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - U. Khan
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - Y. Li
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - A. S. Tang
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - E. Tsoy
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA USA
| | - T. T. Oskotsky
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - M. Sirota
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
46
|
Fernández A, Cuesta P, Marcos A, Montenegro-Peña M, Yus M, Rodríguez-Rojo IC, Bruña R, Maestú F, López ME. Sex differences in the progression to Alzheimer's disease: a combination of functional and structural markers. GeroScience 2024; 46:2619-2640. [PMID: 38105400 PMCID: PMC10828170 DOI: 10.1007/s11357-023-01020-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023] Open
Abstract
Mild cognitive impairment (MCI) has been frequently interpreted as a transitional phase between healthy cognitive aging and dementia, particularly of the Alzheimer's disease (AD) type. Of note, few studies explored that transition from a multifactorial perspective, taking into consideration the effect of basic factors such as biological sex. In the present study 96 subjects with MCI (37 males and 59 females) were followed-up and divided into two subgroups according to their clinical outcome: "progressive" MCI (pMCI = 41), if they fulfilled the diagnostic criteria for AD at the end of follow-up; and "stable" MCI (sMCI = 55), if they remained with the initial diagnosis. Different markers were combined to characterize sex differences between groups, including magnetoencephalography recordings, cognitive performance, and brain volumes derived from magnetic resonance imaging. Results indicated that the pMCI group exhibited higher low-frequency activity, lower scores in neuropsychological tests and reduced brain volumes than the sMCI group, being these measures significantly correlated. When sex was considered, results revealed that this pattern was mainly due to the influence of the females' sample. Overall, females exhibited lower cognitive scores and reduced brain volumes. More interestingly, females in the pMCI group showed an increased theta activity that correlated with a more abrupt reduction of cognitive and volumetric scores as compared with females in the sMCI group and with males in the pMCI group. These findings suggest that females' brains might be more vulnerable to the effects of AD pathology, since regardless of age, they showed signs of more pronounced deterioration than males.
Collapse
Affiliation(s)
- Alberto Fernández
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid, Spain
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
| | - Pablo Cuesta
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Radiology, Rehabilitation and Physiotherapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Alberto Marcos
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Neurology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Mercedes Montenegro-Peña
- Centre for the Prevention of Cognitive Impairment, Madrid Salud, Madrid City Council, Madrid, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Miguel Yus
- Radiology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Inmaculada Concepción Rodríguez-Rojo
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Department of Nursing and Psysiotherapy, Universidad de Alcalá, Madrid, Spain
| | - Ricardo Bruña
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Radiology, Rehabilitation and Physiotherapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Maestú
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - María Eugenia López
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, Madrid, Spain.
- Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain.
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
47
|
Bramwell LR, Frankum R, Harries LW. Repurposing Drugs for Senotherapeutic Effect: Potential Senomorphic Effects of Female Synthetic Hormones. Cells 2024; 13:517. [PMID: 38534362 PMCID: PMC10969307 DOI: 10.3390/cells13060517] [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: 01/26/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Repurposing previously approved drugs may fast track the route to the clinic for potential senotherapeutics and improves the inefficiency of the clinical drug development pipeline. We performed a repurposing screen of 240 clinically approved molecules in human primary dermal fibroblasts for their effects on CDKN2A expression. Molecules demonstrating effects on CDKN2A expression underwent secondary screening for senescence-associated beta galactosidase (SAB) activity, based on effect size, direction, and/or molecule identity. Selected molecules then underwent a more detailed assessment of senescence phenotypes including proliferation, apoptosis, DNA damage, senescence-associated secretory phenotype (SASP) expression, and regulators of alternative splicing. A selection of the molecules demonstrating effects on senescence were then used in a new bioinformatic structure-function screen to identify common structural motifs. In total, 90 molecules displayed altered CDKN2A expression at one or other dose, of which 15 also displayed effects on SAB positivity in primary human dermal fibroblasts. Of these, 3 were associated with increased SAB activity, and 11 with reduced activity. The female synthetic sex hormones-diethylstilboestrol, ethynyl estradiol and levonorgestrel-were all associated with a reduction in aspects of the senescence phenotype in male cells, with no effects visible in female cells. Finally, we identified that the 30 compounds that decreased CDKN2A activity the most had a common substructure linked to this function. Our results suggest that several drugs licensed for other indications may warrant exploration as future senotherapies, but that different donors and potentially different sexes may respond differently to senotherapeutic compounds. This underlines the importance of considering donor-related characteristics when designing drug screening platforms.
Collapse
Affiliation(s)
| | | | - Lorna W. Harries
- RNA-Mediated Mechanisms of Disease Group, Department of Clinical and Biomedical Sciences (Medical School), Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK; (L.R.B.); (R.F.)
| |
Collapse
|
48
|
Xu Q, Ji M, Huang S, Guo W. Association between serum estradiol levels and cognitive function in older women: a cross-sectional analysis. Front Aging Neurosci 2024; 16:1356791. [PMID: 38450384 PMCID: PMC10915044 DOI: 10.3389/fnagi.2024.1356791] [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: 12/16/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Estradiol is a sex steroid hormone, which has been implicated in the pathogenesis of Alzheimer's disease and cognitive impairment. This cross-sectional study aimed to examine the relationship between serum estradiol levels and cognitive performance in older American women. Methods Data were obtained from the National Health and Nutrition Examination Survey 2013-2014. A total of 731 women aged ≥60 years who met the inclusion criteria were included in this study. Serum estradiol levels were measured using the isotope dilution liquid chromatography tandem mass spectrometry (ID-LC-MS/MS) method developed by the Centers for Disease Control and Prevention for routine analysis. All measured serum levels were further divided into three parts: T1, <3.68 pg./mL; T2, 3.68-7.49 pg./mL; T3, >7.49 pg./mL, and analyzed. Participants' cognitive abilities were tested using the Vocabulary Learning Subtest (CERAD), Animal Fluency Test (AFS), and digital symbol substitution test (DSST). Scores for each test were calculated based on the sample mean and standard deviation (SD). To examine the relationship between serum estradiol level tertiles and cognitive scores, multiple linear regression models were developed, controlling for race/ethnicity, education level, hypertension, diabetes, and insomnia. Results The mean age of the participants was 69.57 ± 6.68 years. The non-Hispanic whites were 78.95%, and those who had completed at least some college-level education were 60.62%. The mean BMI of the participants was 29.30 ± 6.79, and 10.85% had a history of smoking. Further, 73.41% did not have a history of alcohol consumption, and 63.03% had hypertension (63.03%). In addition, 81.81 and 88.3% did not have a history of diabetes mellitus and did not have sleep disorders, respectively. The mean serum estradiol level was 8.48 ± 0.77 pg./mL. Multivariate linear regression of the reference group consisting of participants in tertiles of serum estradiol levels revealed that one unit increase in serum estradiol levels increased DSST scores by 0.61 (0.87, 6.34) in the T3 group. However, no significant correlation was found in the CERAD and AFS tests. Conclusion Participants with higher estradiol levels had higher DSST scores and better processing speed, sustained attention, and working memory, suggesting that serum estradiol may serve as a biomarker for cognitive decline in older women.
Collapse
Affiliation(s)
- Qian Xu
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Suzhou, China
| | - Meng Ji
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine (Suzhou Wujiang District Second People's Hospital), Suzhou, China
| | - Shicai Huang
- Kunshan Integrated TCM and Western Medicine Hospital, Kunshan, China
| | - Weifeng Guo
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
49
|
Mensah-Kane P, Davis DL, Shi HS, Trinh OT, Vann PH, Dory L, Sumien N. Hyperbaric oxygen alleviates selective domains of cognitive and motor deficits in female 5xFAD mice. GeroScience 2024; 46:517-530. [PMID: 38153668 PMCID: PMC10828284 DOI: 10.1007/s11357-023-01047-2] [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: 09/25/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023] Open
Abstract
Treatment of Alzheimer's disease (AD) has been limited to managing of symptoms or anti-amyloid therapy with limited results and uncertainty. Seeking out new therapies that can reverse the effects of this devastating disease is important. Hyperbaric oxygen (HBO) therapy could be such a candidate as it has been shown to improve brain function in certain neurological conditions. Furthermore, the role sex plays in the vulnerability/resilience to AD remains equivocal. An understanding of what makes one sex more vulnerable to AD could unveil new pathways for therapy development. In this study, we investigated the effects of HBO on cognitive, motor, and affective function in a mouse model of AD (5xFAD) and assessed protein oxidation in peripheral tissues as a safety indicator. The motor and cognitive abilities of 5xFAD mice were significantly impaired. HBO therapy improved cognitive flexibility and associative learning of 5xFAD females but not males, but HBO had no effect other aspects of cognition. HBO also reversed AD-related declines in balance but had no impact on gait and anxiety-like behavior. HBO did not affect body weights or oxidative stress in peripheral tissues. Our study provides further support for HBO therapy as a potential treatment for AD and emphasizes the importance of considering sex as a biological variable in therapeutic development. Further investigations into the underlying mechanisms of HBO's sex-specific responses are warranted, as well as optimizing treatment protocols for maximum benefits.
Collapse
Affiliation(s)
- Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Helen S Shi
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Oanh T Trinh
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Ladislav Dory
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA
| | - Nathalie Sumien
- Department of Pharmacology & Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie, Fort Worth, TX, 76107, USA.
| |
Collapse
|
50
|
Foley KE, Winder Z, Sudduth TL, Martin BJ, Nelson PT, Jicha GA, Harp JP, Weekman EM, Wilcock DM. Alzheimer's disease and inflammatory biomarkers positively correlate in plasma in the UK-ADRC cohort. Alzheimers Dement 2024; 20:1374-1386. [PMID: 38011580 PMCID: PMC10917006 DOI: 10.1002/alz.13485] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 11/29/2023]
Abstract
INTRODUCTION Protein-based plasma assays provide hope for improving accessibility and specificity of molecular diagnostics to diagnose dementia. METHODS Plasma was obtained from participants (N = 837) in our community-based University of Kentucky Alzheimer's Disease Research Center cohort. We evaluated six Alzheimer's disease (AD)- and neurodegeneration-related (Aβ40, Aβ42, Aβ42/40, p-tau181, total tau, and NfLight) and five inflammatory biomarkers (TNF𝛼, IL6, IL8, IL10, and GFAP) using the SIMOA-based protein assay platform. Statistics were performed to assess correlations. RESULTS Our large cohort reflects previous plasma biomarker findings. Relationships between biomarkers to understand AD-inflammatory biomarker correlations showed significant associations between AD and inflammatory biomarkers suggesting peripheral inflammatory interactions with increasing AD pathology. Biomarker associations parsed out by clinical diagnosis (normal, MCI, and dementia) reveal changes in strength of the correlations across the cognitive continuum. DISCUSSION Unique AD-inflammatory biomarker correlations in a community-based cohort reveal a new avenue for utilizing plasma-based biomarkers in the assessment of AD and related dementias. HIGHLIGHTS Large community cohorts studying sex, age, and APOE genotype effects on biomarkers are few. It is unknown how biomarker-biomarker associations vary through aging and dementia. Six AD (Aβ40, Aβ42, Aβ42/40, p-tau181, total tau, and NfLight) and five inflammatory biomarkers (TNFα, IL6, IL8, IL10, and GFAP) were used to examine associations between biomarkers. Plasma biomarkers suggesting increasing cerebral AD pathology corresponded to increases in peripheral inflammatory markers, both pro-inflammatory and anti-inflammatory. Strength of correlations, between pairs of classic AD and inflammatory plasma biomarker, changes throughout cognitive progression to dementia.
Collapse
Affiliation(s)
- Kate E. Foley
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Zachary Winder
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- College of MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Tiffany L. Sudduth
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Barbara J. Martin
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Peter T. Nelson
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Pathology and Laboratory MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Gregory A. Jicha
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Neurology, College of Public HealthUniversity of KentuckyLexingtonKentuckyUSA
| | - Jordan P. Harp
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Neurology, College of Public HealthUniversity of KentuckyLexingtonKentuckyUSA
| | - Erica M. Weekman
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Donna M. Wilcock
- Sanders Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|