1
|
Sharma M, Goswami I. Multivariate decomposition of gender differentials in cognitive impairment among older adults in India based on Longitudinal Ageing Study in India, 2017-2018. BMC Psychiatry 2025; 25:385. [PMID: 40241039 PMCID: PMC12004875 DOI: 10.1186/s12888-025-06811-6] [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: 10/07/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Increasing life expectancy and declining fertility rates have increased the ageing population around the world. The literature lacks a consensus regarding the risk of cognitive impairments by gender. OBJECTIVE Our study aims to examine the differences in cognition impairments between male and female older adults in India. METHODOLOGY We utilized data from the first wave of the Longitudinal Ageing Study in India (LASI) (2017-18), analyzing 31,464 older adults aged 60 years and above (15,098 males and 16,366 females). Cognitive impairment is measured using the Harmonized Cognitive Assessment Protocol (HCAP) which includes five broad domains (memory, orientation, arithmetic function, executive function, and object naming). A multivariate decomposition analysis was performed using STATA 17 software to identify covariates'contributions, which explain the group differences to average predictions. FINDINGS The prevalence of cognitive impairment was significantly higher among females (19.8%) than males (6.4%) (p < 0.001). Gender disparities were more pronounced among the oldest-old (41.5% vs. 15.9%), widowed individuals (24.6% vs. 9.8%), those with no education (25.1% vs. 11.8%), and individuals living alone (23.4% vs. 5.0%). Decomposition analysis revealed that 62% of the gender gap in cognitive impairment was attributable to differences in compositional factors, primarily education (42%), marital status (6%), working status (6%), difficulty in instrumental activities of daily living (3%), and physical activity (2%). The remaining 38% of the disparity was due to differences in how these factors impacted men and women. CONCLUSION The findings indicate that cognitive impairments are more pronounced among women. Gender-responsive interventions improving education access among the female gender would bring relevant and desired results.
Collapse
Affiliation(s)
- Madhurima Sharma
- International Institute for Population Sciences, Mumbai, 400088, India.
| | - Indrajit Goswami
- International Institute for Population Sciences, Mumbai, 400088, India
| |
Collapse
|
2
|
Roy KK, Mehta DK, Das R. Reevaluating Alzheimer's disease treatment: Can phytochemicals bridge the therapeutic Gap? Neuroscience 2025; 575:1-18. [PMID: 40216186 DOI: 10.1016/j.neuroscience.2025.04.014] [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: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a growing neurological disorder giving impact cognition and memory, posing a global health challenge with over 55 million individuals affected. It is the 7th foremost cause of dying worldwide, and its pervasiveness is expected to twofold in each five years, reaching 115 million by 2050. AD is characterized by neurofibrillary tangles, senile plaques, and oxidative stress, leading to synaptic failure and cognitive decline. Currently, there is no cure, and available FDA-approved drugs provide only symptomatic relief. The disease progresses through five phases- mild cognitive impairment (MCI), very severe, severe, moderate and mild AD. Research on AD focuses on various neurodegenerative pathways, including inflammation, oxidative stress, genetic factors, environmental variables, and amyloid-beta accumulation. Existing FDA-accepted drugs, like rivastigmine, memantine, galantamine, and donepezil, primarily address early symptoms but have limitations, including side effects and high costs. In this context, phytochemicals from plants, such as resveratrol, huperzine, quercetin, galantamine, and rosmarinic acid, show promise as potential treatments for AD and overcome the challenges and limitation of conventional treatment. These natural substances are being investigated for their ability to lower the risk of AD safely. However, there is a lack of comprehensive knowledge about their application, necessitating further research and clinical trials to explore their potential benefits and limitations. This review serves as an essential reference for advancing future studies on Alzheimer's disease. By thoroughly analyzing neurodegenerative pathways, addressing drug limitations, and highlighting the potential of phytochemicals, we establish a strong foundation for developing innovative therapeutic strategies. Closing the knowledge gap related to the use of phytochemicals in Alzheimer's management is not just important; it is critical for creating novel and more effective treatments for this challenging neurological condition.
Collapse
Affiliation(s)
- Kishor Kumar Roy
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Dinesh Kumar Mehta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Rina Das
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India.
| |
Collapse
|
3
|
Beydoun MA, Beydoun HA, Li Z, Hu YH, Noren Hooten N, Ding J, Hossain S, Maino Vieytes CA, Launer LJ, Evans MK, Zonderman AB. Alzheimer's Disease polygenic risk, the plasma proteome, and dementia incidence among UK older adults. GeroScience 2025; 47:2507-2523. [PMID: 39586964 PMCID: PMC11978584 DOI: 10.1007/s11357-024-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex polygenic neurodegenerative disorder. Its genetic risk's relationship with all-cause dementia may be influenced by the plasma proteome. Up to 40,139 UK Biobank participants aged ≥ 50y at baseline assessment (2006-2010) were followed-up for ≤ 15 y for dementia incidence. Plasma proteomics were performed on a sub-sample of UK Biobank participants (k = 1,463 plasma proteins). AD polygenic risk scores (PRS) were used as the primary exposure and Cox proportional hazards models were conducted to examine the AD PRS-dementia relationship. A four-way decomposition model then partitioned the total effect (TE) of AD PRS on dementia into an effect due to mediation only, an effect due to interaction only, neither or both. The study found that AD PRS tertiles significantly increased the risk for all-cause dementia, particularly among women. The study specifically found that AD PRS was associated with a 79% higher risk for all-cause dementia for each unit increase (HR = 1.79, 95% CI: 1.70-1.87, P < 0.001). Eighty-six plasma proteins were significantly predicted by AD PRS, including a positive association with PLA2G7, BRK1, the glial acidic fibrillary protein (GFAP), neurofilament light chain (NfL), and negative with TREM2. Both GFAP and NfL significantly interacted synergistically with AD PRS to increase all-dementia risk (> 10% of TE is pure interaction), while GFAP was also an important consistent mediator in the AD PRS-dementia relationship. In summary, we detected significant interactions of NfL and GFAP with AD PRS, in relation to dementia incidence, suggesting potential for personalized dementia prevention and management.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center On Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jun Ding
- Translational Gerontology Branch, National Institute On Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Sharmin Hossain
- Department of Human Services (DHS), State of Maryland, Baltimore, MD, 21202, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
4
|
Beydoun MA, Beydoun HA, Fanelli-Kuczmarski MT, Hu YH, Shaked D, Weiss J, Waldstein SR, Launer LJ, Evans MK, Zonderman AB. Uncovering mediational pathways behind racial and socioeconomic disparities in brain volumes: insights from the UK Biobank study. GeroScience 2025; 47:1837-1858. [PMID: 39388067 PMCID: PMC11979012 DOI: 10.1007/s11357-024-01371-1] [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: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 10/15/2024] Open
Abstract
Mediation pathways explaining racial/ethnic and socioeconomic (SES) disparities in structural MRI markers of brain health remain underexplored. We examined racial/ethnic and SES disparities in sMRI markers and tested total, direct, and indirect effects through lifestyle, health-related, and cognition factors using a structural equations modeling approach among 36,184 UK Biobank participants aged 40-70 years at baseline assessment (47% men). Race (non-White vs. White) and lower SES-predicted poorer brain sMRI volumetric outcomes at follow-up, with racial/ethnic disparities in sMRI outcomes involving multiple pathways and SES playing a central role in those pathways. Mediational patterns differed across outcomes, with the SES-sMRI total effect being partially mediated for all outcomes. Over 20% of the total effect (TE) of race/ethnicity on WMH was explained by the indirect effect (IE), by a combination of different pathways going through SES, lifestyle, health-related, and cognition factors. This is in contrast to < 10% for total brain, gray matter (GM), white matter (WM), and frontal GM left/right. Another significant finding is that around 57% of the total effect for SES and the normalized white matter hyperintensity (WMH) was attributed to an indirect effect. This effect encompasses many pathways that involve lifestyle, health-related, and cognitive aspects. Aside from WMH, the percent of TE of SES mediated through various pathways ranged from ~ 5% for WM to > 15% up to 36% for most of the remaining sMRI outcomes, which are composed mainly of GM phenotypes. Race and SES were important determinants of brain volumetric outcomes, with partial mediation of racial/ethnic disparities through SES, lifestyle, health-related, and cognition factors.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, 22060, USA
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | | | - Jordan Weiss
- Stanford Center On Longevity, Stanford University, Stanford, CA, 94305, USA
| | - Shari R Waldstein
- Department of Psychology, University of Maryland Baltimore County, Catonsville, MD, 21250, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| |
Collapse
|
5
|
Ware EB, Zhu P, Noppert G, Fu M, Benbow M, Kobayashi LC, Ryan LH, Bakulski KM. Associations of Perceived Neighborhood Factors and Alzheimer's Disease Polygenic Score with Cognition: Evidence from the Health and Retirement Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.14.25324002. [PMID: 40162253 PMCID: PMC11952623 DOI: 10.1101/2025.03.14.25324002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
We examined the relationships between neighborhood characteristics, cumulative genetic risk for Alzheimer's disease (polygenic scores for Alzheimer's disease), and cognitive function using data from the Health and Retirement Study (2008-2020, age>50). Baseline perceived neighborhood characteristics were combined into a subjective neighborhood disadvantage index. Cognitive function was assessed at baseline and measured biennially over a 10-year follow-up period. Analyses were stratified by genetic ancestry. Cox proportional hazard models analyzed associations between neighborhood characteristics, Alzheimer's disease polygenic scores, and their interactions on cognitive impairment. In the European ancestries sample, a one standard deviation higher score on the subjective neighborhood disadvantage index was associated with a higher hazard of any cognitive impairment (HR:1.09; CI:1.03-1.15). Similarly, a one standard deviation increase in Alzheimer's disease polygenic score was associated with a higher risk of cognitive impairment (HR:1.10; CI:1.05-1.16). Similar effect sizes were observed when examining cognitive impairment without dementia and dementia separately. No significant interactions were found. Comparable but nonsignificant trends were noted in the African ancestries sample. Subjective neighborhood disadvantage index and Alzheimer's disease polygenic score were independently associated with incident cognitive impairment. Preventing dementia by addressing modifiable risk factors is essential.
Collapse
Affiliation(s)
- Erin B Ware
- Survey Research Center, Institute for Social Research Center, University of Michigan, 426 Thompson St, Ann Arbor, MI, 48104, USA
- Population Studies Center, Institute for Social Research Center, University of Michigan, 426 Thompson St, Ann Arbor, MI, 48104, USA
| | - Peiyao Zhu
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Grace Noppert
- Survey Research Center, Institute for Social Research Center, University of Michigan, 426 Thompson St, Ann Arbor, MI, 48104, USA
| | - Mingzhou Fu
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Mikayla Benbow
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Lindsay C Kobayashi
- Survey Research Center, Institute for Social Research Center, University of Michigan, 426 Thompson St, Ann Arbor, MI, 48104, USA
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Lindsay H Ryan
- Survey Research Center, Institute for Social Research Center, University of Michigan, 426 Thompson St, Ann Arbor, MI, 48104, USA
| | - Kelly M Bakulski
- School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| |
Collapse
|
6
|
Abdi H, Sanchez-Molina D, Garcia-Vilana S, Rahimi-Movaghar V. Biomechanical perspectives on traumatic brain injury in the elderly: a comprehensive review. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2025; 7:022001. [PMID: 39761631 DOI: 10.1088/2516-1091/ada654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025]
Abstract
Traumatic brain injuries (TBIs) pose a significant health concern among the elderly population, influenced by age-related physiological changes and the prevalence of neurodegenerative diseases. Understanding the biomechanical dimensions of TBIs in this demographic is vital for developing effective preventive strategies and optimizing clinical management. This comprehensive review explores the intricate biomechanics of TBIs in the elderly, integrating medical and aging studies, experimental biomechanics of head tissues, and numerical simulations. Research reveals that global brain atrophy in normal aging occurs at annual rates of -0.2% to -0.5%. In contrast, neurodegenerative diseases such as Alzheimer's, Parkinson's, and multiple sclerosis are associated with significantly higher rates of brain atrophy. These variations in atrophy rates underscore the importance of considering differing brain atrophy patterns when evaluating TBIs among the elderly. Experimental studies further demonstrate that age-related changes in the mechanical properties of critical head tissues increase vulnerability to head injuries. Numerical simulations provide insights into the biomechanical response of the aging brain to traumatic events, aiding in injury prediction and preventive strategy development tailored to the elderly. Biomechanical analysis is essential for understanding injury mechanisms and forms the basis for developing effective preventive strategies. By incorporating local atrophy and age-specific impact characteristics into biomechanical models, researchers can create targeted interventions to reduce the risk of head injuries in vulnerable populations. Future research should focus on refining these models and integrating clinical data to better predict outcomes and enhance preventive care. Advancements in this field promise to improve health outcomes and reduce injury risks for the aging population.
Collapse
Affiliation(s)
- Hamed Abdi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Sunil N, Shikkandar A, Durai BRR, Unnathpadi R, Sankar V, Pullithadathil B. Label-Free Surface-Enhanced Raman Spectroscopy Detection of Amyloid Beta on Silver Nanostructured Substrates for Alzheimer's Diagnosis. JOURNAL OF BIOPHOTONICS 2025; 18:e202400314. [PMID: 39632462 DOI: 10.1002/jbio.202400314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/13/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
Alzheimer's disease (AD) is a public health concern, for which an early diagnosis is essential. Biomass-derived carbon fibres with surface-decorated silver nanoparticles (Ag@CFs) have been utilized as surface enhanced raman spectroscopy (SERS) sensor platform for the detection of the amyloid beta Aβ (25-35) for the pre-diagnosis of Alzheimer's disease. Structural and morphological characterizations confirmed the distribution of plasmonic silver nanoparticles over the surface of carbon fibres. The SERS sensor performance of Ag@CFs was evaluated using rhodamine 6G, which showed an enhancement of the order of 106 which proved the effectiveness of the developed SERS sensor towards trace level detection of analyte. A range of amyloid beta concentrations, from 100uM to 10pM, have been analyzed as a proof-of concept for this study, showcasing the efficacy of the Ag@CFs based SERS sensor to detect trace level concentrations of amyloid beta, even as low as 10 pM. This investigation is a promising development in the field of AD diagnostics since it may turn out to be a non-invasive, economical and early diagnostic tool.
Collapse
Affiliation(s)
- Navami Sunil
- Nanosensors and Clean Energy Laboratory, PSG Institute of Advanced Studies, Coimbatore, India
| | - Ashma Shikkandar
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, India
| | | | - Rajesh Unnathpadi
- Nanosensors and Clean Energy Laboratory, PSG Institute of Advanced Studies, Coimbatore, India
| | | | - Biji Pullithadathil
- Nanosensors and Clean Energy Laboratory, PSG Institute of Advanced Studies, Coimbatore, India
| |
Collapse
|
8
|
Beydoun MA, Beydoun HA, Noren Hooten N, Li Z, Hu Y, Georgescu MF, Hossain S, Tanaka T, Bouhrara M, Maino Vieytes CA, Fanelli‐Kuczmarski MT, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers as mediators or moderators for the association between poor cardiovascular health and white matter microstructural integrity: The UK Biobank study. Alzheimers Dement 2025; 21:e14507. [PMID: 39822062 PMCID: PMC11864230 DOI: 10.1002/alz.14507] [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: 04/26/2024] [Revised: 11/16/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION The plasma proteome's mediating or moderating roles in the association between poor cardiovascular health (CVH) and brain white matter (WM) microstructural integrity are largely unknown. METHODS Data from 3953 UK Biobank participants were used (40-70 years, 2006-2010), with a neuroimaging visit between 2014 and 2021. Poor CVH was determined using Life's Essential 8 (LE8) and reversing standardized z-scores (LE8z _rev). The plasma proteome was examined as a potential mediator or moderator of LE8z _rev's effects on quantitative diffusion-weighted magnetic resonance imaging (dMRI) metrics. RESULTS LE8z_rev was significantly associated with deteriorated WM microstructural integrity, as reflected by lower tract-averaged fractional anisotropy (dMRI-FAmean), (β ± standared error (SE): -0.00152 ± 0.0003, p < 0.001) and higher tract-averaged orientation dispersion (dMRI-ODmean), (β ± SE:+0.00081 ± 0.00017, p < 0.001). Ten strongly mediating plasma proteins of 1463 were identified, with leptin as the principal driver. DISCUSSION Poor CVH is linked to poor WM microstructural integrity measures (lower FAmean and higher ODmean), mostly mediated through leptin. HIGHLIGHTS Up to 3953 UK Biobank participants were selected for this study. Poor cardiovascular health (CVH) was determined using Life's Essential 8. The plasma proteome was examined as a potential mediator or moderator of poor CVH's effect on dMRI metrics. Ten plasma proteins were identified with strong mediating effects, with leptin being the principal driver.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Hind A. Beydoun
- VA National Center on Homelessness Among VeteransU.S. Department of Veterans AffairsWashington, DCUSA
- Department of Management, Policy, and Community Health, School of Public HealthUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Yi‐Han Hu
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Sharmin Hossain
- Department of Human Services (DHS)State of MarylandBaltimoreMarylandUSA
| | - Toshiko Tanaka
- Translational Gerontology BranchNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Mustapha Bouhrara
- Laboratory of Clinical InvestigationNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Christian A. Maino Vieytes
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Marie T. Fanelli‐Kuczmarski
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, NIA/NIH/IRPBaltimoreMarylandUSA
| |
Collapse
|
9
|
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
|
10
|
Chaudhuri I, Das S. Heterogeneous treatment effects of BCG vaccine on Alzheimer's disease risk. J Alzheimers Dis Rep 2025; 9:25424823251317955. [PMID: 40034522 PMCID: PMC11864241 DOI: 10.1177/25424823251317955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 03/05/2025] Open
Abstract
Background This project has investigated the role of the Bacillus Calmette-Guérin (BCG) vaccine as a potential treatment against Alzheimer's disease (AD) and related dementias (ADRD). Objective To further establish that BCG treatment results in lower risk of ADRD through novel machine learning methods and to analyze the heterogeneity of treatment effects. Methods This retrospective cohort study was conducted from May 28, 1987 to May 6, 2021, in patients who were 50 years or older and were diagnosed with non-muscle-invasive bladder cancer (NMIBC). Follow-up duration was 15-years. Machine learning algorithms using survival analysis and the random forest algorithm were the primary methods of data analysis. Results The research has found that on average, NMIBC patients who received BCG treatment had a 6.9% (95% CI: 0.43%, 13.4%) lower risk of developing ADRD compared to those who did not. Heterogeneous treatment effects were also detected for those with a history of mental health disorders and also for those with a history of respiratory diseases. Those with mental health disorders were at a 14.7% (95% CI: 0.6%, 28.9%) reduced risk of ADRD if they received BCG treatment compared to no BCG treatment. Additionally, those taking BCG with respiratory diseases increased risk of ADRD by 13.6% (95% CI: 1.1%, 26.1%) compared to those with no BCG treatment. Conclusions BCG is associated with a lower risk of ADRD through novel analysis methods and has detected heterogeneity of treatment effects. This presents BCG as a potential low-cost method, with few side-effects, to prevent ADRD.
Collapse
Affiliation(s)
- Irfan Chaudhuri
- Department of Epidemiology, Harvard University, Cambridge, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Lu X, Zhang Y, Tang Y, Bernick C, Shan G. Conversion to Alzheimer's disease dementia from normal cognition directly or with the intermediate mild cognitive impairment stage. Alzheimers Dement 2025; 21:e14393. [PMID: 39560003 PMCID: PMC11772698 DOI: 10.1002/alz.14393] [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: 07/15/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Progression to Alzheimer's disease (AD) dementia from normal cognition (NC) can follow different trajectories, with most progressing through a recognizable mild cognitive impairment stage (NC-MCI-AD), while some individuals transition quickly from NC to AD dementia (NC-AD). METHODS We compared demographic characteristics, health factors, and cognitive and functional assessments across three time points: the first NC visit, the last NC visit, and the first AD dementia visit. RESULTS The NC-MCI-AD group showed greater impairment in cognitive and functional scores at AD dementia diagnosis, despite maintaining better cognitive function during the NC stage. Analysis of yearly changes revealed negligible differences during NC. However, the yearly change during the AD dementia stage suggested potentially more rapid functional decline in the NC-AD group. DISCUSSION These findings highlight the heterogeneity in AD disease progression and emphasize the importance of considering diverse progression patterns in AD research and clinical practice. HIGHLIGHTS We investigated the disease progression difference between patients who converted to Alzheimer's disease (AD) dementia from normal cognition (NC) directly or through the mild cognitive impairment (MCI) stage. We found that the NC-MCI-AD group showed greater impairment in cognitive and functional scores at AD dementia diagnosis. We discovered that the NC-AD group had rapid functional decline once patients were confirmed with AD onset.
Collapse
Affiliation(s)
- Xinlin Lu
- Department of BiostatisticsUniversity of FloridaGainesvilleFloridaUSA
| | - Yahui Zhang
- Department of BiostatisticsUniversity of FloridaGainesvilleFloridaUSA
| | - Yichen Tang
- School of Information and Electrical EngineeringZhejiang University City CollegeHangzhouChina
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Guogen Shan
- Department of BiostatisticsUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
12
|
Matthews KA, Murray CT, Nguyen BL, Spears KS, Jackson EMJ, Hall DM, McGuire LC. Alzheimer's disease and related dementias among Medicare beneficiaries aged ≥ 65 years in rural America, by Census region and select demographic characteristics: 2020. J Rural Health 2025; 41:e12902. [PMID: 39840892 DOI: 10.1111/jrh.12902] [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: 04/18/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE Alzheimer's disease and related dementias (ADRD) are a significant public health concern characterized by memory decline that, over time, leads to loss of independence. This study reports ADRD diagnosis rates among Medicare beneficiaries aged ≥ 65 years in rural America. METHODS We conducted a descriptive analysis of Medicare Fee-for-Service (FFS) and Medicare Advantage enrollees using administrative Medicare data from 2020. Combining data from Medicare FFS and Medicare Advantage produces a more complete and representative sample of older adults than previous studies that used FFS data alone. Nonmetropolitan counties were used to define rural. Rural ADRD diagnosis rates, stratified by age, sex, race/ethnicity, and Census region, were adjusted using the 2000 Census population standard. FINDINGS The study population consisted of 54 million Medicare data Fee-for-Service (FFS) and Medicare Advantage enrollees aged ≥ 65; 5.3 million beneficiaries were diagnosed with ADRD, and 16.2% (n = 861,337) of beneficiaries diagnosed with ADRD resided in rural America. The age-adjusted ADRD diagnosis rate was slightly lower in rural America (9.6 per 100 beneficiaries) than in the United States (10.0 per 100 beneficiaries). The South Census region had the highest rural ADRD diagnosis rates. CONCLUSION These findings underscore the need for targeted interventions and support mechanisms to address the growing burden of ADRD in rural communities.
Collapse
Affiliation(s)
- Kevin A Matthews
- Centers for Disease Control and Prevention, Office of Rural Health, Atlanta, Georgia, USA
| | - Christian T Murray
- Centers for Disease Control and Prevention, Office of Policy, Performance, and Evaluation, Atlanta, Georgia, USA
| | - Brenda L Nguyen
- Division of Health Informatics and Surveillance, Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katie S Spears
- Centers for Disease Control and Prevention, Office of Rural Health, Atlanta, Georgia, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | | | - Diane M Hall
- Centers for Disease Control and Prevention, Office of Rural Health, Atlanta, Georgia, USA
| | - Lisa C McGuire
- Gerontological Society of America, Washington, District of Columbia, USA
| |
Collapse
|
13
|
Chaudhuri S, Dempsey DA, Huang YN, Park T, Cao S, Chumin EJ, Craft H, Crane PK, Mukherjee S, Choi SE, Scollard P, Lee M, Nakano C, Mez J, Trittschuh EH, Klinedinst BS, Hohman TJ, Lee JY, Kang KM, Sohn CH, Kim YK, Yi D, Byun MS, Risacher SL, Nho K, Saykin AJ, Lee DY. Association of amyloid and cardiovascular risk with cognition: Findings from KBASE. Alzheimers Dement 2024; 20:8527-8540. [PMID: 39511852 DOI: 10.1002/alz.14290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Limited research has explored the effect of cardiovascular risk and amyloid interplay on cognitive decline in East Asians. METHODS Vascular burden was quantified using Framingham's General Cardiovascular Risk Score (FRS) in 526 Korean Brain Aging Study (KBASE) participants. Cognitive differences in groups stratified by FRS and amyloid positivity were assessed at baseline and longitudinally. RESULTS Baseline analyses revealed that amyloid-negative (Aβ-) cognitively normal (CN) individuals with high FRS had lower cognition compared to Aβ- CN individuals with low FRS (p < 0.0001). Longitudinally, amyloid pathology predominantly drove cognitive decline, while FRS alone had negligible effects on cognition in CN and mild cognitive impairment (MCI) groups. CONCLUSION Our findings indicate that managing vascular risk may be crucial in preserving cognition in Aβ- individuals early on and before the clinical manifestation of dementia. Within the CN and MCI groups, irrespective of FRS status, amyloid-positive individuals had worse cognitive performance than Aβ- individuals. HIGHLIGHTS Vascular risk significantly affects cognition in amyloid-negative older Koreans. Amyloid-negative CN older adults with high vascular risk had lower baseline cognition. Amyloid pathology drives cognitive decline in CN and MCI, regardless of vascular risk. The study underscores the impact of vascular health on the AD disease spectrum.
Collapse
Affiliation(s)
- Soumilee Chaudhuri
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Desarae A Dempsey
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yen-Ning Huang
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tamina Park
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sha Cao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Evgeny J Chumin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hannah Craft
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Seo-Eun Choi
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Phoebe Scollard
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael Lee
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Connie Nakano
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jesse Mez
- Department of Neurology, Boston University, Boston, Massachusetts, USA
| | - Emily H Trittschuh
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
- Geriatrics Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Brandon S Klinedinst
- Department of General Internal Medicine, Harborview Medical Center, University of Washington School of Medicine, Seattle, Washington, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMGSNU Boramae Medical Center, Dongjak-gu, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMGSNU Boramae Medical Center, Dongjak-gu, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Jongno-gu, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Jongno-gu, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- School of Informatics and Computing, Indiana University, Indianapolis, Indiana, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Medical Research and Library Building, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Jongno-gu, Seoul, Republic of Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Jongno-gu, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea
| |
Collapse
|
14
|
Qi X, Nizamutdinov D, Yi SS, Wu E, Huang JH. Disease Modifying Monoclonal Antibodies and Symptomatic Pharmacological Treatment for Alzheimer's Disease. Biomedicines 2024; 12:2636. [PMID: 39595200 PMCID: PMC11592475 DOI: 10.3390/biomedicines12112636] [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: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's Disease (AD) is an irreversible, progressive syndrome characterized by neurocognitive impairment. Two neuropathological features seen in AD are extracellular amyloid plaques consisting of amyloid beta1-40 and 1-42, and intracellular neurofibrillary tangles (NFTs). For decades, neuroscience research has heavily focused on seeking to understand the primary mechanism of AD and searching for pharmacological approaches for the treatment of dementia. Three monoclonal antibodies that act against amyloid beta-aducanumab, lecanemab, and donanemab-have been approved by the Food and Drug Administration (FDA) for the treatment of mild cognitive impairment and mild AD, in addition to medications for cognitive symptom management such as acetylcholinesterase inhibitors and the N-methyl-D-aspartate (NMDA) antagonist. Further trials should focus on the combination of therapies targeting amyloid plaques and tau pathology.
Collapse
Affiliation(s)
- Xiaoming Qi
- Department of Neurology, Baylor Scott & White Health, Temple, TX 78508, USA;
| | - Damir Nizamutdinov
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Song Stephen Yi
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Erxi Wu
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
- College of Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- LIVESTRONG Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jason H. Huang
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
15
|
Can KC, Ozel-Kizil ET, Onar D, Duman B, Kırıcı S, Baştuğ G, Baştuğ M. Exploring the Effect of Single-Session Transcranial Direct Current Stimulation on Attention, Verbal Fluency, and Working Memory in Patients With Alzheimer's Disease-Related Dementia. J ECT 2024:00124509-990000000-00220. [PMID: 39531323 DOI: 10.1097/yct.0000000000001075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Alzheimer's disease (AD) is a leading cause of morbidity and mortality among the elderly. Transcranial direct current stimulation (tDCS) applies low-intensity currents to the brain, resulting in short-term neurocognitive effects and long-term neuroplasticity enhancement. Limited research reported on the impact of tDCS on cognitive functions in dementia due to AD. This study aims to compare changes in verbal fluency and working memory following a single tDCS application to the left dorsolateral prefrontal cortex (DLPFC) in AD patients. METHODS Patients with mild dementia due to AD underwent cognitive assessment using the Standardized Mini-Mental Test, Clock Drawing Test, Rey Auditory Verbal Learning Test, Functional Activities Questionnaire, Informant Questionnaire on Cognitive Decline in the Elderly, and Montreal Cognitive Assessment Scale. A single-session tDCS was administered by applying anodal tDCS to the left DLPFC for a duration of 30 minutes. Verbal fluency and working memory were evaluated before and after tDCS using the WAIS-R Digit Span Test forward and backward subscales, Trail Making Test (TMT) A and B, and Verbal Fluency Test. RESULTS Regarding cognitive test scores before and after tDCS application, there were statistically significant reductions in the durations of TMT-A and TMT-B. However, there were no significant differences observed for TMT B-A, VFT, DST-forward, and DST-backward performances. CONCLUSIONS The findings indicate that a single anodal tDCS targeting the left DLPFC enhances attention and processing speed in AD patients but has no effect on working memory or verbal fluency.
Collapse
Affiliation(s)
- Kazım Cihan Can
- From the Geriatric Psychiatry Unit, Department of Psychiatry, Ankara University School of Medicine, Ankara, Turkey
| | | | - Deha Onar
- From the Geriatric Psychiatry Unit, Department of Psychiatry, Ankara University School of Medicine, Ankara, Turkey
| | - Berker Duman
- Consultation-Liaison Unit, Department of Psychiatry, Ankara University School of Medicine
| | - Sevinç Kırıcı
- From the Geriatric Psychiatry Unit, Department of Psychiatry, Ankara University School of Medicine, Ankara, Turkey
| | | | - Metin Baştuğ
- Department of Physiology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
16
|
Su M, Wang T, Zou C, Cao K, Liu F. Global, regional, and national burdens of Alzheimer's disease and other forms of dementia in the elderly population from 1999 to 2019: A trend analysis based on the Global Burden of Disease Study 2019. IBRAIN 2024; 10:488-499. [PMID: 39691425 PMCID: PMC11649385 DOI: 10.1002/ibra.12181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 12/19/2024]
Abstract
Dementia represents a significant health issue, afflicting both patients and their families. To assess the global trends in the incidence, prevalence, mortality, and disability-adjusted life years (DALYs) of Alzheimer's disease (AD) and other dementias in the elderly population, the Global Burden of Disease Study (1999-2019) was used. The average annual percentage change (AAPC) was estimated using linear regression. Stratified analysis of the global trends by age, sex, region, national level, and social development index (SDI) were also performed. The global incidence of AD and other dementias increased from 507.96 per 100,000 in 1990 to 569.39 per 100,000 in 2019, showing a significant increase in this period. In males, the incidence increased from 387.56 per 100,000 population in 1990 to 462.40 per 100,000 in 2019 (AAPC = 0.61), whereas females experienced a slower rise (AAPC = 0.31) and had a higher incidence in 2019 (662.93 per 100,000 population). The most significant increase was observed in individuals aged 60-64 and those in the middle-SDI quintile. Regionally, the high-income Asia Pacific had the highest incidence (890.01 per 100,000 population) and DALYs (3043.86 per 100,000) in AD and other dementias in 2019. As for national trends, Japan had the most pronounced increase in the incidence and DALYs of AD and other dementias during the 1990-2019 period. These findings highlight the growing burden of dementias on life expectancy at a population level, which is significant for healthcare professionals and decision-makers to conduct the ongoing debate on the policy of AD and other dementias.
Collapse
Affiliation(s)
- Mengdan Su
- Department of Anesthesiology, West China HospitalSichuan UniversityChengduChina
| | - Tianhong Wang
- Department of Anesthesiology, West China HospitalSichuan UniversityChengduChina
| | - Congcong Zou
- Department of Anesthesiology, West China HospitalSichuan UniversityChengduChina
| | - Keyu Cao
- Department of Nursing, West China HospitalSichuan UniversityChengduChina
- West China School of NursingSichuan UniversityChengduChina
| | - Fei Liu
- Department of Anesthesiology, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
17
|
Ifebirinachi C, Park MS, Kim ST. Evaluating disparity of subjective cognitive decline between male veterans and non-veterans in the United States using propensity score matching estimation: A behavioral risk factor surveillance system survey cross-sectional study. PLoS One 2024; 19:e0310102. [PMID: 39269972 PMCID: PMC11398683 DOI: 10.1371/journal.pone.0310102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Subjective cognitive decline (SCD) is one of the most important early onset symptoms of Alzheimer's Disease. Previous studies consistently reported that SCD is associated with quality of life, socio-economic factors, and related health comorbidities. However, the impact of veteran status on SCD has been little investigated. This study conducted a cross-sectional study to address disproportionate effects in subjective cognitive decline between veterans and non-veterans in the United States. Propensity score matching (PSM) was applied in this observational study to achieve covariate balancing and reduce selection bias, providing a more accurate estimate of the isolated effect of veteran status on SCD. Our study utilized 32,431 forty-five years or older non-institutionalized White, Black or African-American, and Hispanic or Latin-American male population from the 2019 Behavioral Risk Factor Surveillance System data. We first identified 10,685 paired PSM samples for the binary veteran status using the preselected covariates. Next, we performed a logistic regression for modeling the relationship between the veteran status and the SCD status using the PSM samples along with the covariates selected by a BIC-based stepwise selection. Our analyses revealed a statistically significant causal association between veteran status and SCD after PSM (odds ratio (OR): 1.16 and 95% confidence interval (CI): 1.06-1.27). We obtained a similar result before PSM with an OR of 1.20 and 95% CI of 1.10-1.31. When we focused on a minority group (Black or African-American males), we found a significantly increased veterans' risk of SCD, especially after propensity score matching (OR: 1.69, 95% CI: 1.16-2.45). We also found several factors such as employment status, difficulty dressing/walking/running errands, general health status, physical health status, unaffordability of medical costs, mental health status, and comorbid conditions including stroke, blindness, high cholesterol, and arthritis as statistically significantly associated with SCD (P-value < 0.05). Similar to post-traumatic stress disorder and traumatic brain injury, our study demonstrated a causal association between SCD and military-related activities in the United States, which has a disproportionate impact on the minority population. This study sets the groundwork to further research in this domain to diagnose neurological diseases early among veterans.
Collapse
Affiliation(s)
- Chinenye Ifebirinachi
- Department of Mathematics and Statistics, North Carolina Agricultural & Technical State University, Greensboro, North Carolina, United States of America
| | - Man Sik Park
- Department of Statistics, Sungshin Women's University, Seoul, Republic of Korea
| | - Seong-Tae Kim
- Department of Mathematics and Statistics, North Carolina Agricultural & Technical State University, Greensboro, North Carolina, United States of America
| |
Collapse
|
18
|
Beydoun MA, Tate R, Georgescu MF, Gamaldo AA, Maino Vieytes CA, Beydoun HA, Noren Hooten N, Evans MK, Zonderman AB. Poor sleep quality, dementia status and their association with all-cause mortality among older US adults. Aging (Albany NY) 2024; 16:12138-12167. [PMID: 39237306 PMCID: PMC11424588 DOI: 10.18632/aging.206102] [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: 01/23/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Evidence points to associations between sleep quality, dementia, and mortality. We examined whether poor sleep quality mediated or moderated the association between dementia and mortality risk among older US adults and vice versa, and whether these associations differed by sex and by race. METHODS The study investigated bi-directional associations between sleep quality, dementia and mortality in older US adults using data from the Health and Retirement Study (N = 6,991, mean age = 78.1y, follow-up: 2006-2020, number of deaths = 4,938). It tested interactions and mediating effects, using Cox proportional hazards models and four-way decomposition models. RESULTS Poor sleep quality was associated with increased mortality risk, particularly among male and White older adults. However, the association was reversed in the fully adjusted model, with a 7% decrease in risk per tertile. Probable dementia was associated with a two-fold increase in mortality risk, with a stronger association found among White adults. The association was markedly attenuated in the fully adjusted models. Sleep quality-stratified models showed a stronger positive association between dementia and mortality among individuals with better sleep quality. Both mediation and interaction were involved in explaining the total effects under study, though statistically significant total effects were mainly composed of controlled direct effects. CONCLUSIONS Poor sleep quality is directly related to mortality risk before lifestyle and health-related factors are adjusted. Dementia is linked to mortality risk, especially in individuals with better sleep quality, males, and White older adults. Future research should explore the underlying mechanisms.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Rio Tate
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Alyssa A. Gamaldo
- Department of Psychology, Clemson University, Clemson, SC 29634, USA
| | - Christian A. Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Hind A. Beydoun
- Department of Veterans Affairs, VA National Center on Homelessness Among Veterans, Washington, DC 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, NIA/NIH/IRP, Baltimore, MD 21224, USA
| |
Collapse
|
19
|
Bohra A, Jyotsna M, Pereddy SDGR, Sampath A, Rozatkar AR, Coulter RWS, Gupta S. Training Needs Analysis for Medical Students in Assessing Cognitive Functioning: An Observational Study. Indian J Psychol Med 2024; 46:417-424. [PMID: 39421058 PMCID: PMC11483722 DOI: 10.1177/02537176241274143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Backgrounds Research shows that medical students' knowledge and skills concerning the assessment of cognition in various neuropsychiatric conditions (e.g., Alzheimer's disease and schizophrenia) are unsatisfactory. This research aims to conduct a training needs analysis (TNA) for medical students to identify and refer patients with neuropsychiatric conditions. Methods The study comprised two phases. First, developing a TNA toolkit to assess training needs in clinical tasks related to cognitive function assessment (by adopting the Hennessy-Hicks TNA toolkit); and second, through a self-reported survey, their training needs in cognitive function assessment were assessed. Data analysis involved calculating training gaps, importance scores, performance scores, training scores, and organization scores for various clinical tasks-stratified participants' study year; for training needs and trends, factor analysis and post-hoc analyses were conducted. Results A total of 153 medical students from a tertiary care center participated in the survey. The participants rated their performance lower than the perceived importance of tasks, indicating a need for improvement in all competencies (P < .01). Pre-final-year students had the highest training needs, particularly in accessing literature, planning, and organizing care for patients with cognitive impairment, performing mental status examination, screening patients for cognitive deficits, and counseling them/caregivers about interventions (P < .01). Factor analysis identified a single dominant factor, suggesting a correlation among these skills. Conclusion Pre-final-year students require targeted training, whereas students beyond this stage can benefit from special training modules and awareness of available resources for cognitive assessment. The findings also suggest the importance of a hybrid approach involving training and organizational modifications.
Collapse
Affiliation(s)
- Arwa Bohra
- Dept. of Psychiatry, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, Madhya Pradesh, India
| | - Mili Jyotsna
- Dept. of Psychiatry, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, Madhya Pradesh, India
| | | | - Ananyan Sampath
- Dept. of Psychiatry, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, Madhya Pradesh, India
| | - Abhijit R Rozatkar
- Dept. of Behavioral and Community Health Sciences, School of Public Health, University of Pittsburgh, Pennsylvania, USA
| | - Rober WS Coulter
- Dept. of Behavioral and Community Health Sciences, School of Public Health, University of Pittsburgh, Pennsylvania, USA
| | - Snehil Gupta
- Dept. of Psychiatry, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
20
|
Alshabrmi FM, Aba Alkhayl FF, Rehman A. Novel drug discovery: Advancing Alzheimer's therapy through machine learning and network pharmacology. Eur J Pharmacol 2024; 976:176661. [PMID: 38795753 DOI: 10.1016/j.ejphar.2024.176661] [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: 03/21/2024] [Revised: 04/28/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
Alzheimer's disease (AD), marked by tau tangles and amyloid-beta plaques, leads to cognitive decline. Despite extensive research, its complex etiology remains elusive, necessitating new treatments. This study utilized machine learning (ML) to analyze compounds with neuroprotective potential. This approach exposed the disease's complexity and identified important proteins, namely MTOR and BCL2, as central to the pathogenic network of AD. MTOR regulates neuronal autophagy and survival, whereas BCL2 regulates apoptosis, both of which are disrupted in AD. The identified compounds, including Armepavine, Oprea1_264702,1-cyclopropyl-7-fluoro-8-methoxy-4-oxoquinoline-3-carboxylic acid,(2S)-4'-Hydroxy-5,7,3'-trimethoxyflavan,Oprea1_130514,Sativanone,5-hydroxy-7,8-dimethoxyflavanone,7,4'-Dihydroxy-8,3'-dimethoxyflavanone,N,1-dicyclopropyl-6,Difluoro-Methoxy-Gatifloxacin,6,8-difluoro-1-(2-fluoroethyl),1-ethyl-6-fluoro-7-(4-methylpiperidin-1-yl),Avicenol C, demonstrated potential modulatory effects on these proteins. The potential for synergistic effects of these drugs in treating AD has been revealed via network pharmacology. By targeting numerous proteins at once, these chemicals may provide a more comprehensive therapeutic approach, addressing many aspects of AD's complex pathophysiology. A Molecular docking, dynamic simulation, and Principle Component Analysis have confirmed these drugs' efficacy by establishing substantial binding affinities and interactions with important proteins such as MTOR and BCL2. This evidence implies that various compounds may interact within the AD pathological framework, providing a sophisticated and multifaceted therapy strategy. In conclusion, our study establishes a solid foundation for the use of these drugs in AD therapy. Thus current study highlights the possibility of multi-targeted, synergistic therapeutic approaches in addressing the complex pathophysiology of AD by integrating machine learning, network pharmacology, and molecular docking simulations. This holistic technique not only advances drug development but also opens up new avenues for developing more effective treatments for this difficult and widespread disease.
Collapse
Affiliation(s)
- Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A & F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
21
|
Beydoun HA, Szymkowiak D, Kinney R, Beydoun MA, Zonderman AB, Tsai J. Is the Risk of Alzheimer's Disease and Related Dementias Among U.S. Veterans Influenced by the Intersectionality of Housing Status, HIV/AIDS, Hepatitis C, and Psychiatric Disorders? J Gerontol A Biol Sci Med Sci 2024; 79:glae153. [PMID: 38869510 PMCID: PMC11519038 DOI: 10.1093/gerona/glae153] [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: 02/29/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Homelessness and housing instability disproportionately affect U.S. veterans with psychiatric disorders, human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), hepatitis C, and Alzheimer's disease and related disorders (ADRD). We examined housing status and/or HIV/AIDS in relation to ADRD risk and evaluated hepatitis C, substance use, and mental health disorders as mediators and/or moderators of hypothesized relationships, among U.S. veterans ≥50 years of age seeking Department of Veterans Affairs (VA) healthcare services. METHODS A retrospective cohort study was conducted using linked VA Homeless Operations Management and Evaluation System and Corporate Data Warehouse databases (2017-2023) on 3 275 098 eligible veterans yielding 133 388 ADRD cases over 5 years of follow-up. Multivariable regression and causal mediation analyses were performed, controlling for demographic and clinical characteristics. RESULTS Taking stably housed veterans without HIV/AIDS as referent, ADRD risk was higher among veterans with homelessness/housing instability alone (adjusted hazard ratio [aHR] = 1.67, 95% confidence interval [CI]: 1.63,1.72), lower among veterans with HIV/AIDS alone (aHR = 0.65, 95% CI: 0.58,0.73), but similar to veterans with homelessness/housing instability and HIV/AIDS (aHR = 1.01, 95% CI: 0.79,1.29). In adjusted models, hepatitis C and psychiatric disorders were positively related to homelessness/housing instability and ADRD risk, but negatively related to HIV/AIDS. Statistically significant mediation and/or moderation of hepatitis C and psychiatric disorders were observed, although <10% of total effects were explained by these characteristics, controlling for confounders. CONCLUSIONS Among older veterans, ADRD diagnoses over 5 years were less among those with HIV/AIDS, but more among those with homelessness/housing instability, and these relationships were partly explained by hepatitis C and psychiatric disorders.
Collapse
Affiliation(s)
- Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, District of Columba, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Dorota Szymkowiak
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, District of Columba, USA
| | - Rebecca Kinney
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, District of Columba, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Jack Tsai
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, District of Columba, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
22
|
Abdulhameed N, Babin A, Hansen K, Weaver R, Banks WA, Talbot K, Rhea EM. Comparing regional brain uptake of incretin receptor agonists after intranasal delivery in CD-1 mice and the APP/PS1 mouse model of Alzheimer's disease. Alzheimers Res Ther 2024; 16:173. [PMID: 39085976 PMCID: PMC11293113 DOI: 10.1186/s13195-024-01537-1] [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: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
Targeting brain insulin resistance (BIR) has become an attractive alternative to traditional therapeutic treatments for Alzheimer's disease (AD). Incretin receptor agonists (IRAs), targeting either or both of the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors, have proven to reverse BIR and improve cognition in mouse models of AD. We previously showed that many, but not all, IRAs can cross the blood-brain barrier (BBB) after intravenous (IV) delivery. Here we determined if widespread brain uptake of IRAs could be achieved by circumventing the BBB using intranasal (IN) delivery, which has the added advantage of minimizing adverse gastrointestinal effects of systemically delivered IRAs. Of the 5 radiolabeled IRAs tested (exenatide, dulaglutide, semaglutide, DA4-JC, and DA5-CH) in CD-1 mice, exenatide, dulaglutide, and DA4-JC were successfully distributed throughout the brain following IN delivery. We observed significant sex differences in uptake for DA4-JC. Dulaglutide and DA4-JC exhibited high uptake by the hippocampus and multiple neocortical areas. We further tested and found the presence of AD-associated Aβ pathology minimally affected uptake of dulaglutide and DA4-JC. Of the 5 tested IRAs, dulaglutide and DA4-JC are best capable of accessing brain regions most vulnerable in AD (neocortex and hippocampus) after IN administration. Future studies will need to be performed to determine if IN IRA delivery can reduce BIR in AD or animal models of that disorder.
Collapse
Affiliation(s)
- Noor Abdulhameed
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Alice Babin
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Kim Hansen
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Riley Weaver
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA.
| |
Collapse
|
23
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [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: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
24
|
Beydoun MA, Beydoun HA, Hu YH, Maino Vieytes CA, Noren Hooten N, Song M, Georgescu MF, Fanelli-Kuczmarski MT, Meirelles O, Launer LJ, Evans MK, Zonderman AB. Plasma proteomic biomarkers and the association between poor cardiovascular health and incident dementia: The UK Biobank study. Brain Behav Immun 2024; 119:995-1007. [PMID: 38710337 PMCID: PMC11285716 DOI: 10.1016/j.bbi.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The study examined how plasma proteome indicators may explain the link between poor cardiovascular health (CVH) and dementia risk. METHODS The present study involved 28,974 UK Biobank participants aged 50-74y at baseline (2006-2010) who were followed-up for ≤ 15 y for incidence of dementia. CVH was calculated using Life's Essential 8 (LE8) total scores. The scores were standardized and reverse coded to reflect poor CVH (LE8z_rev). OLINK proteomics was available on this sample (k = 1,463 plasma proteins). The study primarily tested the mediating effects of the plasma proteome in LE8z_rev-dementia effect. The total effect was decomposed into "mediation only" or pure indirect effect (PIE), "interaction only" or interaction referent (INTREF), "neither mediation nor interaction" or controlled direct effect (CDE), and "both mediation and interaction" or mediated interaction (INTMED). RESULTS The study found poorer CVH assessed by LE8z_rev increased the risk of all-cause dementia by 11 % [per 1 SD, hazard ratio, (HR) = 1.11, 95 % CI: 1.03-1.20, p = 0.005). The study identified 11 plasma proteins with strong mediating effects, with GDF15 having the strongest association with dementia risk (per 1 SD, HR = 1.24, 95 % CI: 1.16, 1.33, P < 0.001 when LE8z_rev is set at its mean value) and the largest proportion mediated combining PIE and INTMED (62.6 %; 48 % of TE is PIE), followed by adrenomedullin or ADM. A first principal component with 10 top mediators (TNFRSF1A, GDF15, FSTL3, COL6A3, PLAUR, ADM, GFRAL, ACVRL1, TNFRSF6B, TGFA) mediated 53.6 % of the LE8z_rev-dementia effect. Using all the significant PIE (k = 526) proteins, we used OLINK Insight pathway analysis to identify key pathways, which revealed the involvement of the immune system, signal transduction, metabolism, disease, protein metabolism, hemostasis, membrane trafficking, extracellular matrix organization, developmental biology, and gene expression among others. STRING analysis revealed that five top consistent proteomic mediators were represented in two larger clusters reflecting numerous interconnected biological gene ontology pathways, most notably cytokine-mediated signaling pathway for GDF15 cluster (GO:0019221) and regulation of peptidyl-tyrosine phosphorylation for the ADM cluster (GO:0050730). CONCLUSION Dementia is linked to poor CVH mediated by GDF15 and ADM among several key proteomic markers which collectively explained ∼ 54 % of the total effect.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States.
| | - Hind A Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC 20420, United States; Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Minkyo Song
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, United States
| |
Collapse
|
25
|
Zhang Y, Zhang P, Yin D. Association between a body shape index and cognitive impairment among us older adults from a cross-sectional survey of the NHANES 2011-2014. Lipids Health Dis 2024; 23:169. [PMID: 38840158 PMCID: PMC11151546 DOI: 10.1186/s12944-024-02165-2] [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: 04/12/2024] [Accepted: 05/26/2024] [Indexed: 06/07/2024] Open
Abstract
PURPOSE This study aimed to assess the relationship between A Body Shape Index (ABSI) and cognitive impairment among older adults in the United States. METHODS This cross-sectional study analyzed cognitive function in 2,752 individuals aged 60 and older using data from the 2011-2014 National Health and Nutrition Examination Survey (NHANES). Cognitive assessments were conducted using the Immediate Recall Test (IRT), Delayed Recall Test (DRT), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST). A Body Shape Index (ABSI) was calculated from waist circumference (WC), weight, and height. The relationship between ABSI and cognitive outcomes was examined through multifactorial linear regression, smooth curve fitting, and subgroup and interaction analyses. RESULTS With complete data, 2752 persons 60 and older participated in the study. After adjusting for covariables, these results showed statistically significant negative relationships between ABSI, IRT, and DSST scores. The negative correlation between DSST and ABSI is more substantial in males than females. There is less of a negative link between ABSI, AFT, and DSST among drinkers who consume 12 or more drinks annually compared to those who consume less. Furthermore, compared to individuals without high blood pressure(HBP), those who suffered HBP showed a more significant negative connection between ABSI and AFT. CONCLUSION Lower cognitive function was linked to higher ABSI.
Collapse
Affiliation(s)
- Yanwei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dekun Yin
- Department of Anesthesiology, Funing People's Hospital of Jiangsu, Yancheng, Jiangsu province, China.
| |
Collapse
|
26
|
Weiss J, Beydoun MA, Beydoun HA, Georgescu MF, Hu YH, Noren Hooten N, Banerjee S, Launer LJ, Evans MK, Zonderman AB. Pathways explaining racial/ethnic and socio-economic disparities in brain white matter integrity outcomes in the UK Biobank study. SSM Popul Health 2024; 26:101655. [PMID: 38562403 PMCID: PMC10982559 DOI: 10.1016/j.ssmph.2024.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/14/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Pathways explaining racial/ethnic and socio-economic status (SES) disparities in white matter integrity (WMI) reflecting brain health, remain underexplored, particularly in the UK population. We examined racial/ethnic and SES disparities in diffusion tensor brain magnetic resonance imaging (dMRI) markers, namely global and tract-specific mean fractional anisotropy (FA), and tested total, direct and indirect effects through lifestyle, health-related and cognition factors using a structural equations modeling approach among 36,184 UK Biobank participants aged 40-70 y at baseline assessment (47% men). Multiple linear regression models were conducted, testing independent associations of race/ethnicity, socio-economic and other downstream factors in relation to global mean FA, while stratifying by Alzheimer's Disease polygenic Risk Score (AD PRS) tertiles. Race (Non-White vs. White) and lower SES predicted poorer WMI (i.e. lower global mean FA) at follow-up, with racial/ethnic disparities in FAmean involving multiple pathways and SES playing a central role in those pathways. Mediational patterns differed across tract-specific FA outcomes, with SES-FAmean total effect being partially mediated (41% of total effect = indirect effect). Furthermore, the association of poor cognition with FAmean was markedly stronger in the two uppermost AD PRS tertiles compared to the lower tertile (T2 and T3: β±SE: -0.0009 ± 0.0001 vs. T1: β±SE: -0.0005 ± 0.0001, P < 0.001), independently of potentially confounding factors. Race and lower SES were generally important determinants of adverse WMI outcomes, with partial mediation of socio-economic disparities in global mean FA through lifestyle, health-related and cognition factors. The association of poor cognition with lower global mean FA was stronger at higher AD polygenic risk.
Collapse
Affiliation(s)
- Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA, USA
| | - May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Hind A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Sri Banerjee
- Public Health Doctoral Programs, Walden University, Minneapolis, MN, USA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, USA
| |
Collapse
|
27
|
Abukuri DN. Novel Biomarkers for Alzheimer's Disease: Plasma Neurofilament Light and Cerebrospinal Fluid. Int J Alzheimers Dis 2024; 2024:6668159. [PMID: 38779175 PMCID: PMC11111307 DOI: 10.1155/2024/6668159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent an increasingly significant public health concern. As clinical diagnosis faces challenges, biomarkers are becoming increasingly important in research, trials, and patient assessments. While biomarkers like amyloid-β peptide, tau proteins, CSF levels (Aβ, tau, and p-tau), and neuroimaging techniques are commonly used in AD diagnosis, they are often limited and invasive in monitoring and diagnosis. For this reason, blood-based biomarkers are the optimal choice for detecting neurodegeneration in brain diseases due to their noninvasiveness, affordability, reliability, and consistency. This literature review focuses on plasma neurofilament light (NfL) and CSF NfL as blood-based biomarkers used in recent AD diagnosis. The findings revealed that the core CSF biomarkers of neurodegeneration (T-tau, P-tau, and Aβ42), CSF NFL, and plasma T-tau were strongly associated with Alzheimer's disease, and the core biomarkers were strongly associated with mild cognitive impairment due to Alzheimer's disease. Elevated levels of plasma and cerebrospinal fluid NfL were linked to decreased [18F]FDG uptake in corresponding brain areas. In participants with Aβ positivity (Aβ+), NfL correlated with reduced metabolism in regions susceptible to Alzheimer's disease. In addition, CSF NfL levels correlate with brain atrophy and predict cognitive changes, while plasma total tau does not. Plasma P-tau, especially in combination with Aβ42/Aβ40, is promising for symptomatic AD stages. Though not AD-exclusive, blood NfL holds promise for neurodegeneration detection and assessing treatment efficacy. Given the consistent levels of T-tau, P-tau, Aβ42, and NFL in CSF, their incorporation into both clinical practice and research is highly recommended.
Collapse
|
28
|
Altomare D, Libri I, Alberici A, Rivolta J, Padovani A, Ashton NJ, Zetterberg H, Blennow K, Borroni B. Plasma biomarkers increase diagnostic confidence in patients with Alzheimer's disease or frontotemporal lobar degeneration. Alzheimers Res Ther 2024; 16:107. [PMID: 38734612 PMCID: PMC11088144 DOI: 10.1186/s13195-024-01474-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: 11/29/2023] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The recent development of techniques to assess plasma biomarkers has changed the way the research community envisions the future of diagnosis and management of Alzheimer's disease (AD) and other neurodegenerative disorders. This work aims to provide real world evidence on the clinical impact of plasma biomarkers in an academic tertiary care center. METHODS Anonymized clinical reports of patients diagnosed with AD or Frontotemporal Lobar Degeneration with available plasma biomarkers (Aβ42, Aβ42/Aβ40, p-tau181, p-tau231, NfL, GFAP) were independently assessed by two neurologists who expressed diagnosis and diagnostic confidence three times: (T0) at baseline based on the information collected during the first visit, (T1) after plasma biomarkers, and (T2) after traditional biomarkers (when available). Finally, we assessed whether clinicians' interpretation of plasma biomarkers and the consequent clinical impact are consistent with the final diagnosis, determined after the conclusion of the diagnostic clinical and instrumental work-up by the actual managing physicians who had complete access to all available information. RESULTS Clinicians assessed 122 reports, and their concordance ranged from 81 to 91% at the three time points. At T1, the presentation of plasma biomarkers resulted in a change of diagnosis in 2% (2/122, p = 1.00) of cases, and in increased diagnostic confidence in 76% (91/120, p < 0.001) of cases with confirmed diagnosis. The change in diagnosis and the increase in diagnostic confidence after plasma biomarkers were consistent with the final diagnosis in 100% (2/2) and 81% (74/91) of cases, respectively. At T2, the presentation of traditional biomarkers resulted in a further change of diagnosis in 13% (12/94, p = 0.149) of cases, and in increased diagnostic confidence in 88% (72/82, p < 0.001) of cases with confirmed diagnosis. CONCLUSIONS In an academic tertiary care center, plasma biomarkers supported clinicians by increasing their diagnostic confidence in most cases, despite a negligible impact on diagnosis. Future prospective studies are needed to assess the full potential of plasma biomarkers on clinical grounds.
Collapse
Affiliation(s)
- Daniele Altomare
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Ilenia Libri
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Antonella Alberici
- Department of Continuity of Care and Frailty, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Jasmine Rivolta
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Department of Continuity of Care and Frailty, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, UCL, London, W1T 7NF, UK
- Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy.
- Department of Continuity of Care and Frailty, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy.
| |
Collapse
|
29
|
Ma’arif B, Anwar MF, Hidayatullah H, Muslikh FA, Suryadinata A, Sugihantoro H, Mirza DM, Maulina N, Taek MM. Effect of polar fractions of Marsilea crenata C. Presl. leaves in zebrafish locomotor activity. J Adv Pharm Technol Res 2024; 15:125-129. [PMID: 38903546 PMCID: PMC11186547 DOI: 10.4103/japtr.japtr_241_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 11/01/2023] [Accepted: 02/02/2024] [Indexed: 06/22/2024] Open
Abstract
Neurodegenerative diseases (NDs) are pathological conditions initiated by the loss of neuronal cell structure and the progressive decline in function caused by prolonged neuroinflammation. Postmenopausal women are at a high risk of experiencing NDs due to estrogen deficiency in their bodies, necessitating the administration of phytoestrogens as a replacement for estrogen in the body. One alternative therapy is administering phytoestrogens, estrogen-like substances from plants, which can be obtained from Marsilea crenata C. Presl. leaves. The purpose of this study was to determine whether administration of the n-butanol fraction (BF) and water fraction (WF) of M. crenata leaves could increase locomotor activity in rotenone-induced zebrafish. Treatment was given to each group of zebrafish with BF and WF at doses of 2.5; 5; 10; and 20 ppm to determine the locomotor activity. Then an analysis was carried out by looking at each movement of the zebrafish swimming for 1 min at the time of observation on days 0, 7, 14, 21, and 28. The result showed that BF and WF significantly increased the locomotor activity of zebrafish at the optimum dose of 20 ppm for BF and 5 ppm for WF compared to the negative control. This concludes that the polar fraction of M. crenata leaves is proven to have the potential to prevent ND progressivity.
Collapse
Affiliation(s)
- Burhan Ma’arif
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Mohamad Fajrul Anwar
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Hidayatullah Hidayatullah
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Faisal Akhmal Muslikh
- Department of Pharmacy, Faculty of Pharmacy, Bhakti Wiyata Institute of Health Sciences, Kediri, East Java, Indonesia
| | - Arief Suryadinata
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Hajar Sugihantoro
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Denis Mery Mirza
- Department of Pharmacy, Faculty of Medicine, Islamic University of Malang, Malang, Indonesia
| | - Novia Maulina
- Department of Pharmacy, Faculty of Medicine and Health Science, Maulana Malik Ibrahim State Islamic University, Kediri, East Java, Indonesia
| | - Maximus M. Taek
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Widya Mandira Catholic University, Kupang, Indonesia
| |
Collapse
|
30
|
Krut' VG, Kalinichenko AL, Maltsev DI, Jappy D, Shevchenko EK, Podgorny OV, Belousov VV. Optogenetic and chemogenetic approaches for modeling neurological disorders in vivo. Prog Neurobiol 2024; 235:102600. [PMID: 38548126 DOI: 10.1016/j.pneurobio.2024.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Animal models of human neurological disorders provide valuable experimental tools which enable us to study various aspects of disorder pathogeneses, ranging from structural abnormalities and disrupted metabolism and signaling to motor and mental deficits, and allow us to test novel therapies in preclinical studies. To be valid, these animal models should recapitulate complex pathological features at the molecular, cellular, tissue, and behavioral levels as closely as possible to those observed in human subjects. Pathological states resembling known human neurological disorders can be induced in animal species by toxins, genetic factors, lesioning, or exposure to extreme conditions. In recent years, novel animal models recapitulating neuropathologies in humans have been introduced. These animal models are based on synthetic biology approaches: opto- and chemogenetics. In this paper, we review recent opto- and chemogenetics-based animal models of human neurological disorders. These models allow for the creation of pathological states by disrupting specific processes at the cellular level. The artificial pathological states mimic a range of human neurological disorders, such as aging-related dementia, Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, and ataxias. Opto- and chemogenetics provide new opportunities unavailable with other animal models of human neurological disorders. These techniques enable researchers to induce neuropathological states varying in severity and ranging from acute to chronic. We also discuss future directions for the development and application of synthetic biology approaches for modeling neurological disorders.
Collapse
Affiliation(s)
- Viktoriya G Krut'
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Andrei L Kalinichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry I Maltsev
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Evgeny K Shevchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Oleg V Podgorny
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia.
| | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow 143025, Russia.
| |
Collapse
|
31
|
Brigo F, Lattanzi S. Diagnosing epileptic seizures in patients with Alzheimer's disease and deciding on the appropriate treatment plan. Expert Rev Neurother 2024; 24:361-370. [PMID: 38426448 DOI: 10.1080/14737175.2024.2325038] [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: 11/09/2023] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the predominant cause of dementia and a significant contributor to morbidity among the elderly. Patients diagnosed with AD face an increased risk of epileptic seizures. AREAS COVERED Herein, the authors review the challenges in the diagnosis of seizures in patients with AD, the risks of seizures related to medications used in AD and the pharmacological treatment of seizures in AD. The authors also provide the reader with their expert opinion on the subject matter and future perspectives. EXPERT OPINION Healthcare professionals should maintain a vigilant approach to suspecting seizures in AD patients. Acute symptomatic seizures triggered by metabolic disturbances, infections, toxins, or drug-related factors often have a low risk of recurrence. In such cases, addressing the underlying cause may suffice without initiating antiseizure medications (ASMs). However, unprovoked seizures in certain AD patients carry a higher risk of recurrence over time, warranting the use of ASMs. Although data is limited, both lamotrigine and levetiracetam appear to be reasonable choices for controlling seizures in elderly AD patients. Decisions should be informed by the best available evidence, the treating physician's clinical experience, and the patient's preferences.
Collapse
Affiliation(s)
- Francesco Brigo
- Innovation, Research and Teaching Service (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Bolzano, Italy
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
32
|
Azargoonjahromi A, Abutalebian F. Unraveling the therapeutic efficacy of resveratrol in Alzheimer's disease: an umbrella review of systematic evidence. Nutr Metab (Lond) 2024; 21:15. [PMID: 38504306 PMCID: PMC10953289 DOI: 10.1186/s12986-024-00792-1] [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: 11/16/2023] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
CONTEXT Resveratrol (RV), a natural compound found in grapes, berries, and peanuts, has been extensively studied for its potential in treating Alzheimer's disease (AD). RV has shown promise in inhibiting the formation of beta-amyloid plaques (Aβ) and neurofibrillary tangles (NFTs), protecting against neuronal damage and oxidative stress, reducing inflammation, promoting neuroprotection, and improving the function of the blood-brain barrier (BBB). However, conflicting results have been reported, necessitating a comprehensive umbrella review of systematic reviews to provide an unbiased conclusion on the therapeutic effectiveness of RV in AD. OBJECTIVE The objective of this study was to systematically synthesize and evaluate systematic and meta-analysis reviews investigating the role of RV in AD using data from both human and animal studies. DATA SOURCES AND EXTRACTION Of the 34 systematic and meta-analysis reviews examining the association between RV and AD that were collected, six were included in this study based on specific selection criteria. To identify pertinent studies, a comprehensive search was conducted in English-language peer-reviewed journals without any restrictions on the publication date until October 15, 2023. The search was carried out across multiple databases, including Embase, MEDLINE (PubMed), Cochrane Library, Web of Science, and Google Scholar, utilizing appropriate terms relevant to the specific research field. The AMSTAR-2 and ROBIS tools were also used to evaluate the quality and risk of bias of the included systematic reviews, respectively. Two researchers independently extracted and analyzed the data, resolving any discrepancies through consensus. Of note, the study adhered to the PRIOR checklist. DATA ANALYSIS This umbrella review presented robust evidence supporting the positive impacts of RV in AD, irrespective of the specific mechanisms involved. It indeed indicated that all six systematic and meta-analysis reviews unanimously concluded that the consumption of RV can be effective in the treatment of AD. CONCLUSION RV exhibits promising potential for benefiting individuals with AD through various mechanisms. It has been observed to enhance cognitive function, reduce Aβ accumulation, provide neuroprotection, protect the BBB, support mitochondrial function, facilitate synaptic plasticity, stabilize tau proteins, mitigate oxidative stress, and reduce neuroinflammation commonly associated with AD.
Collapse
Affiliation(s)
| | - Fatemeh Abutalebian
- Department of Biotechnology and Medicine, Islamic Azad University of Tehran Central Branch, Tehran, Iran
| |
Collapse
|
33
|
Beydoun MA, Beydoun HA, Hu YH, El-Hajj ZW, Georgescu MF, Noren Hooten N, Li Z, Weiss J, Lyall DM, Waldstein SR, Hedges DW, Gale SD, Launer LJ, Evans MK, Zonderman AB. Helicobacter pylori, persistent infection burden and structural brain imaging markers. Brain Commun 2024; 6:fcae088. [PMID: 38529358 PMCID: PMC10961948 DOI: 10.1093/braincomms/fcae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 01/11/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024] Open
Abstract
Persistent infections, whether viral, bacterial or parasitic, including Helicobacter pylori infection, have been implicated in non-communicable diseases, including dementia and other neurodegenerative diseases. In this cross-sectional study, data on 635 cognitively normal participants from the UK Biobank study (2006-21, age range: 40-70 years) were used to examine whether H. pylori seropositivity (e.g. presence of antibodies), serointensities of five H. pylori antigens and a measure of total persistent infection burden were associated with selected brain volumetric structural MRI (total, white, grey matter, frontal grey matter (left/right), white matter hyperintensity as percent intracranial volume and bi-lateral sub-cortical volumes) and diffusion-weighted MRI measures (global and tract-specific bi-lateral fractional anisotropy and mean diffusivity), after an average 9-10 years of lag time. Persistent infection burden was calculated as a cumulative score of seropositivity for over 20 different pathogens. Multivariable-adjusted linear regression analyses were conducted, whereby selected potential confounders (all measures) and intracranial volume (sub-cortical volumes) were adjusted, with stratification by Alzheimer's disease polygenic risk score tertile when exposures were H. pylori antigen serointensities. Type I error was adjusted to 0.007. We report little evidence of an association between H. pylori seropositivity and persistent infection burden with various volumetric outcomes (P > 0.007, from multivariable regression models), unlike previously reported in past research. However, H. pylori antigen serointensities, particularly immunoglobulin G against the vacuolating cytotoxin A, GroEL and outer membrane protein antigens, were associated with poorer tract-specific white matter integrity (P < 0.007), with outer membrane protein serointensity linked to worse outcomes in cognition-related tracts such as the external capsule, the anterior limb of the internal capsule and the cingulum, specifically at low Alzheimer's disease polygenic risk. Vacuolating cytotoxin A serointensity was associated with greater white matter hyperintensity volume among individuals with mid-level Alzheimer's disease polygenic risk, while among individuals with the highest Alzheimer's disease polygenic risk, the urease serointensity was consistently associated with reduced bi-lateral caudate volumes and the vacuolating cytotoxin A serointensity was linked to reduced right putamen volume (P < 0.007). Outer membrane protein and urease were associated with larger sub-cortical volumes (e.g. left putamen and right nucleus accumbens) at middle Alzheimer's disease polygenic risk levels (P < 0.007). Our results shed light on the relationship between H. pylori seropositivity, H. pylori antigen levels and persistent infection burden with brain volumetric structural measures. These data are important given the links between infectious agents and neurodegenerative diseases, including Alzheimer's disease, and can be used for the development of drugs and preventive interventions that would reduce the burden of those diseases.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA 22060, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Ziad W El-Hajj
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Michael F Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA 94305, USA
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Shari R Waldstein
- Department of Psychology, University of Maryland, Catonsville, MD 21250, USA
- Division of Gerontology, Geriatrics, and Palliative Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dawson W Hedges
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA
| | - Shawn D Gale
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| |
Collapse
|
34
|
Thapa S, Shah S, Bhattarai A, Yazdan Panah M, Chand S, Mirmosayyeb O. Risk of dementia following herpes zoster infection among patients undertreatment versus those not: A systematic review and meta-analysis. Health Sci Rep 2024; 7:e1941. [PMID: 38482136 PMCID: PMC10935877 DOI: 10.1002/hsr2.1941] [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: 07/07/2023] [Revised: 12/02/2023] [Accepted: 02/17/2024] [Indexed: 11/02/2024] Open
Abstract
Background and Aims According to the previous studies, herpes zoster (HZ) has been associated with cognitive function and dementia. There is a hypothesis claiming that dementia risk may be reduced by receiving the antiviral treatment for HZ. The purpose of this systematic review and meta-analysis was to shed light on the association between dementia and HZ in individuals receiving and not receiving antiviral medications. Methods Studies investigating the association between HZ and dementia were identified through a systematic search in PubMed/MEDLINE, Scopus, Embase, Google Scholar, and Cochrane Library databases from January, 2000 to April, 2022. Data on the risk of dementia in HZ-infected patients under and not under antiviral treatment were extracted. The meta-analysis was conducted using a random-effects model. The modified ROBIN-I tool was used to evaluate the risk of bias assessment. By utilizing the funnel plots, publication bias was investigated. Results Six cohort studies on 538,531 patients were included. The overall risk of bias assessment was moderate. According to evidence-based cohort studies, there was a significant direct association between HZ and risk of dementia in patients with HZ, who did not receive antiviral treatments (hazard ratio [HR]: 1.15, 95% confidence interval [CI]: 1.03 to 1.28, p = 0.01). On the other hand, there was an inverse relationship between HZ and risk of dementia among patients with HZ, who received antiviral treatments (HR: 0.68, 95% CI: 0.59 to 0.77, p < 0.001). Conclusions This study demonstrated that antiviral therapies may significantly lower the risk of dementia in patients with HZ. This study also confirmed that patients with HZ, without receiving antiviral therapies, may have an increased risk of developing dementia. Further longitudinal research is warranted in this area.
Collapse
Affiliation(s)
| | - Sangam Shah
- Tribhuvan UniversityInstitute of MedicineMaharjgunjNepal
| | | | | | - Swati Chand
- Westchester Medical CenterNew York Medical CollegeValhallaNYUSA
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research CenterIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
35
|
Verma A, Waiker DK, Singh N, Roy A, Singh N, Saraf P, Bhardwaj B, Krishnamurthy S, Trigun SK, Shrivastava SK. Design, Synthesis, and Biological Investigation of Quinazoline Derivatives as Multitargeting Therapeutics in Alzheimer's Disease Therapy. ACS Chem Neurosci 2024; 15:745-771. [PMID: 38327209 DOI: 10.1021/acschemneuro.3c00653] [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: 02/09/2024] Open
Abstract
An efficient and promising method of treating complex neurodegenerative diseases like Alzheimer's disease (AD) is the multitarget-directed approach. Here in this work, a series of quinazoline derivatives (AV-1 to AV-21) were rationally designed, synthesized, and biologically evaluated as multitargeted directed ligands against human cholinesterase (hChE) and human β-secretase (hBACE-1) that exhibit moderate to good inhibitory effects. Compounds AV-1, AV-2, and AV-3 from the series demonstrated balanced and significant inhibition against these targets. These compounds also displayed excellent blood-brain barrier permeability via the PAMPA-BBB assay. Compound AV-2 significantly displaced propidium iodide (PI) from the acetylcholinesterase-peripheral anionic site (AChE-PAS) and was found to be non-neurotoxic at the maximum tested concentration (80 μM) against differentiated SH-SY5Y cell lines. Compound AV-2 also prevented AChE- and self-induced Aβ aggregation in the thioflavin T assay. Additionally, compound AV-2 significantly ameliorated scopolamine and Aβ-induced cognitive impairments in the in vivo behavioral Y-maze and Morris water maze studies, respectively. The ex vivo and biochemical analysis further revealed good hippocampal AChE inhibition and the antioxidant potential of the compound AV-2. Western blot and immunohistochemical (IHC) analysis of hippocampal brain revealed reduced Aβ, BACE-1, APP/Aβ, and Tau molecular protein expressions levels. The pharmacokinetic analysis of compound AV-2 demonstrated significant oral absorption with good bioavailability. The in silico molecular modeling studies of lead compound AV-2 moreover demonstrated a reasonable binding profile with AChE and BACE-1 enzymes and stable ligand-protein complexes throughout the 100 ns run. Compound AV-2 can be regarded as the lead candidate and could be explored more for AD therapy.
Collapse
Affiliation(s)
- Akash Verma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Digambar Kumar Waiker
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Neha Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Anima Roy
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Namrata Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Poorvi Saraf
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Bhagwati Bhardwaj
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Surendra Kumar Trigun
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Sushant Kumar Shrivastava
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
36
|
Wang J, Chen T, Xie J, Zhao S, Jiang Y, Zhang H, Zhu W. A bibliometric analysis of international publication trends in brain atrophy research (2008-2023). Front Neurol 2024; 15:1348778. [PMID: 38356880 PMCID: PMC10864491 DOI: 10.3389/fneur.2024.1348778] [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: 12/03/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Background Brain atrophy is a type of neurological and psychiatric disorder characterized by a decrease in brain tissue volume and weight for various reasons and can have a serious impact on the quality of life of patients. Although there are many studies on brain atrophy, there is a lack of relevant bibliometric studies. Therefore, this study aims to provide a visual analysis of global trends in brain atrophy research over the past 16 years. Methods CiteSpace and VOSviewer were used to visually analyze publication output, scientific collaborations, cocitations, publishing journals, and keywords to determine the current status and future trends of brain atrophy research. Materials published from 2008 to 2023 were collected from the Web of Science Core Collection (WoSCC) database. This study placed no restrictions on the types of literature and focused on English language publications. Results A total of 3,371 publications were included in the analysis. From 2008 to 2023, the number of publications increased annually. In terms of national and academic institutions, universities in the United States and University College London rank first in publication out. Barkhof Frederik and Zivadinov Robert are the most prolific researchers in this field. The publication with the highest cocitation strength is "Deep gray matter volume loss drives disability worsening in multiple sclerosis." Keyword clustering analysis showed that "Alzheimer's disease" and "multiple sclerosis" are current popular topics. The analysis of emergent words indicates that "cerebral small vessel disease," "neurodegeneration," and "cortex/gray matter volume" may become hot research topics in the coming years. Conclusion This study analyses papers on brain atrophy from the past 16 years, providing a new perspective for research in this field. In the past 16 years, research on brain atrophy has received increasing attention. The quality of articles in this field is generally high. Extensive national cooperation already exists. The statistical results indicate that a stable core author group in the field of brain atrophy has almost formed.
Collapse
Affiliation(s)
- Juwei Wang
- Zhejiang Chinese Medical University, Department of Graduate College, Hangzhou, China
| | - Tingting Chen
- Zhejiang Chinese Medical University, Department of Graduate College, Hangzhou, China
| | - Jiayi Xie
- Zhejiang Chinese Medical University, Department of Graduate College, Hangzhou, China
| | - Sheng Zhao
- Zhejiang Chinese Medical University, Department of Graduate College, Hangzhou, China
| | - Yue Jiang
- Department of Acupuncture, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huihe Zhang
- Department of Neurology, Wenzhou Hospital of Traditional Chinese Medicine, Wenzhou, China
| | - Wenzong Zhu
- Department of Neurology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang Chinese Medical University, Wenzhou, China
| |
Collapse
|
37
|
Wang QY, Gao Y, Yao JN, Zhou L, Chen HP, Liu JK. Penisimplicins A and B: Novel Polyketide-Peptide Hybrid Alkaloids from the Fungus Penicillium simplicissimum JXCC5. Molecules 2024; 29:613. [PMID: 38338359 PMCID: PMC10856265 DOI: 10.3390/molecules29030613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
In this study, two previously undescribed nitrogen-containing compounds, penisimplicins A (1) and B (2), were isolated from Penicillium simplicissimum JXCC5. The structures of 1 and 2 were elucidated on the basis of comprehensive spectroscopic data analysis, including 1D and 2D NMR and HRESIMS data. The absolute configuration of 2 was determined by Marfey's method, ECD calculation, and DP4+ analysis. Both structures of 1 and 2 feature an unprecedented manner of amino acid-derivatives attaching to a polyketide moiety by C-C bond. The postulated biosynthetic pathways for 1 and 2 were discussed. Additionally, compound 1 exhibited significant acetylcholinesterase inhibitory activity, with IC50 values of 6.35 μM.
Collapse
Affiliation(s)
- Qing-Yuan Wang
- School of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - Yang Gao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Jian-Neng Yao
- Yunnan Key Laboratory of Pharmacology for Natural Products & School of Pharmaceutical Science, Kunming Medical University, Kunming 650500, China;
| | - Li Zhou
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - He-Ping Chen
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Ji-Kai Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China
| |
Collapse
|
38
|
Xu X, Chen P, Li W, Xiang Y, Xie Z, Yu Q, Tang Y, Wang P. Topological properties analysis and identification of mild cognitive impairment based on individual morphological brain network connectome. Cereb Cortex 2024; 34:bhad450. [PMID: 38012122 DOI: 10.1093/cercor/bhad450] [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: 09/28/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023] Open
Abstract
Mild cognitive impairment is considered the prodromal stage of Alzheimer's disease. Accurate diagnosis and the exploration of the pathological mechanism of mild cognitive impairment are extremely valuable for targeted Alzheimer's disease prevention and early intervention. In all, 100 mild cognitive impairment patients and 86 normal controls were recruited in this study. We innovatively constructed the individual morphological brain networks and derived multiple brain connectome features based on 3D-T1 structural magnetic resonance imaging with the Jensen-Shannon divergence similarity estimation method. Our results showed that the most distinguishing morphological brain connectome features in mild cognitive impairment patients were consensus connections and nodal graph metrics, mainly located in the frontal, occipital, limbic lobes, and subcortical gray matter nuclei, corresponding to the default mode network. Topological properties analysis revealed that mild cognitive impairment patients exhibited compensatory changes in the frontal lobe, while abnormal cortical-subcortical circuits associated with cognition were present. Moreover, the combination of multidimensional brain connectome features using multiple kernel-support vector machine achieved the best classification performance in distinguishing mild cognitive impairment patients and normal controls, with an accuracy of 84.21%. Therefore, our findings are of significant importance for developing potential brain imaging biomarkers for early detection of Alzheimer's disease and understanding the neuroimaging mechanisms of the disease.
Collapse
Affiliation(s)
- Xiaowen Xu
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| | - Peiying Chen
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| | - Weikai Li
- School of Mathematics and Statistics, Chongqing Jiaotong University, Chongqing 400064, China
- MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing 276800, China
| | - Yongsheng Xiang
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| | - Zhongfeng Xie
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| | - Qiang Yu
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| | - Ying Tang
- Department of Electrical and Computer Engineering, Rowan University, Glassboro, New Jersey 08028, USA
| | - Peijun Wang
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
39
|
Zhao X, Hu X, Xie Q, Qi S, Xiang Z, Sun X, Xie Z, Dang R, Zhou L, Liu W, Cheng X, Wang C. Ameliorative effect of scopolamine-induced cognitive dysfunction by Fufangmuniziqi formula: The roles of alkaloids, saponins, and flavonoids. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116792. [PMID: 37356745 DOI: 10.1016/j.jep.2023.116792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufangmuniziqi formula (FFMN), a traditional Uyghur medicine used in China, is derived from an ancient Uyghur medical book and consists of 13 herbs. The herbs of FFMN, such as Peganum harmala L., Glycyrrhiza uralensis Fisch., and Nigella glandulifera, have been demonstrated to have acetylcholinesterase (AChE) inhibitory, anti-neuroinflammatory, or antioxidant effects. Therefore, FFMN may have a good anti-Alzheimer's disease (AD) effect, but its specific action and mechanism need to be further proven. AIM OF THE STUDY This study aims to investigate the anti-AD effects of FFMN and the role played by alkaloids, flavonoids, and saponins in anti-AD. MATERIALS AND METHODS The alkaloids, flavonoids, and saponins fractions of FFMN were prepared by macroporous resin chromatography. The absorbed ingredients in the drug-containing serum were identified by UPLC⁃Q⁃TOF⁃MS. An AD mouse model was established by intraperitoneal injection of scopolamine (SCO). The role of different fractions of FFMN in the anti-AD process was examined by Morris water maze (MWM), in-vitro cell, and AChE inhibition assay. RESULTS A total of 20 ingredients were identified in the serum samples collected after oral administration of FFMN, and seven compounds were selected as candidate active compounds. MWM experiments showed that different fractions of FFMN could significantly improve SCO-induced learning memory impairment in mice. The alkaloids fraction (ALK) regulated cholinergic function by inhibiting AChE activity, activating choline acetyltransferase activity, and protein expression. Flavonoids and saponins were more potent than the ALK in downregulating pro-inflammatory factors or inflammatory mediators, such as TNF-α, MPO, and nitric oxide. Western blot results further confirmed that flavonoids and saponins attenuated neuroinflammation by inhibiting the phosphorylation of IκB and NF-κB p65. This result was also verified by in-vitro cellular assays. FFMN enhanced antioxidant defense by increasing the activity of superoxide dismutase and reducing the production of MDA. Combined with cellular experiments, flavonoids and saponins were proven more protective against oxidative damage. CONCLUSION FFMN improved cognitive and memory impairment in the SCO-induced AD mouse model. ALK mainly enhanced the function of the cholinergic system. Flavonoid and saponin fractions mainly attenuated neuroinflammation and oxidative stress by modulating the NF-κB pathway. All these findings strongly suggested that the combination of alkaloid, flavonoid, and saponin fractions derived from FFMN is a promising anti-AD agent that deserves further development.
Collapse
Affiliation(s)
- Xiang Zhao
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Xianrun Hu
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Qi Xie
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Shenglan Qi
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Zedong Xiang
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Xin Sun
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Zhejun Xie
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Rui Dang
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Limei Zhou
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Wei Liu
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rood, Shanghai, 201203, China.
| |
Collapse
|
40
|
Cao W, Ji Z, Zhu S, Wang M, Sun R. Bioinformatic identification and experiment validation reveal 6 hub genes, promising diagnostic and therapeutic targets for Alzheimer's disease. BMC Med Genomics 2024; 17:6. [PMID: 38167011 PMCID: PMC10763315 DOI: 10.1186/s12920-023-01775-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease that can cause dementia. We aim to screen out the hub genes involved in AD based on microarray datasets. METHODS Gene expression profiles GSE5281 and GSE28146 were retrieved from Gene Expression Omnibus database to acquire differentially expressed genes (DEGs). Gene Ontology and pathway enrichment were conducted using DAVID online tool. The STRING database and Cytoscape tools were employed to analyze protein-protein interactions and identify hub genes. The predictive value of hub genes was assessed by principal component analysis and receiver operating characteristic curves. AD mice model was constructed, and histology was then observed by hematoxylin-eosin staining. Gene expression levels were finally determined by real-time quantitative PCR. RESULTS We obtained 197 overlapping DEGs from GSE5281 and GSE28146 datasets. After constructing protein-protein interaction network, three highly interconnected clusters were identified and 6 hub genes (RBL1, BUB1, HDAC7, KAT5, SIRT2, and ITGB1) were selected. The hub genes could be used as basis to predict AD. Histological abnormalities of brain were observed, suggesting successful AD model was constructed. Compared with the control group, the mRNA expression levels of RBL1, BUB1, HDAC7, KAT5 and SIRT2 were significantly increased, while the mRNA expression level of ITGB1 was significantly decreased in AD groups. CONCLUSION RBL1, BUB1, HDAC7, KAT5, SIRT2 and ITGB1 are promising gene signatures for diagnosis and therapy of AD.
Collapse
Affiliation(s)
- Wenyuan Cao
- Department of Neurology Second Ward, Zibo Municipal Hospital, No. 139, Huangong Road, Linzi District, Zibo City, 255400, Shandong Province, China
| | - Zhangge Ji
- Department of Neurology Second Ward, Zibo Municipal Hospital, No. 139, Huangong Road, Linzi District, Zibo City, 255400, Shandong Province, China
| | - Shoulian Zhu
- Department of Neurology Second Ward, Zibo Municipal Hospital, No. 139, Huangong Road, Linzi District, Zibo City, 255400, Shandong Province, China
| | - Mei Wang
- Department of Rehabilitation, Zibo Municipal Hospital, No. 139, Huangong Road, Linzi District, Zibo City, 255400, Shandong Province, China
| | - Runming Sun
- Department of Neurology Second Ward, Zibo Municipal Hospital, No. 139, Huangong Road, Linzi District, Zibo City, 255400, Shandong Province, China.
| |
Collapse
|
41
|
Platero C, Tohka J, Strange B. Estimating Dementia Onset: AT(N) Profiles and Predictive Modeling in Mild Cognitive Impairment Patients. Curr Alzheimer Res 2024; 20:778-790. [PMID: 38425106 DOI: 10.2174/0115672050295317240223162312] [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: 01/04/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Mild Cognitive Impairment (MCI) usually precedes the symptomatic phase of dementia and constitutes a window of opportunities for preventive therapies. OBJECTIVES The objective of this study was to predict the time an MCI patient has left to reach dementia and obtain the most likely natural history in the progression of MCI towards dementia. METHODS This study was conducted on 633 MCI patients and 145 subjects with dementia through 4726 visits over 15 years from Alzheimer Disease Neuroimaging Initiative (ADNI) cohort. A combination of data from AT(N) profiles at baseline and longitudinal predictive modeling was applied. A data-driven approach was proposed for categorical diagnosis prediction and timeline estimation of cognitive decline progression, which combined supervised and unsupervised learning techniques. RESULTS A reduced vector of only neuropsychological measures was selected for training the models. At baseline, this approach had high performance in detecting subjects at high risk of converting from MCI to dementia in the coming years. Furthermore, a Disease Progression Model (DPM) was built and also verified using three metrics. As a result of the DPM focused on the studied population, it was inferred that amyloid pathology (A+) appears about 7 years before dementia, and tau pathology (T+) and neurodegeneration (N+) occur almost simultaneously, between 3 and 4 years before dementia. In addition, MCI-A+ subjects were shown to progress more rapidly to dementia compared to MCI-A- subjects. CONCLUSION Based on proposed natural histories and cross-sectional and longitudinal analysis of AD markers, the results indicated that only a single cerebrospinal fluid sample is necessary during the prodromal phase of AD. Prediction from MCI into dementia and its timeline can be achieved exclusively through neuropsychological measures.
Collapse
Affiliation(s)
- Carlos Platero
- Health Science Technology Group, Technical University of Madrid, 28012 Madrid, Spain
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Bryan Strange
- Laboratory for Clinical Neuroscience, CTB, Technical University of Madrid, IdISSC, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| |
Collapse
|
42
|
Xue D, Blue EE, Conomos MP, Fohner AE. The power of representation: Statistical analysis of diversity in US Alzheimer's disease genetics data. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12462. [PMID: 38500778 PMCID: PMC10945594 DOI: 10.1002/trc2.12462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a complex disease influenced by genetics and environment. More than 75 susceptibility loci have been linked to late-onset AD, but most of these loci were discovered in genome-wide association studies (GWAS) exclusive to non-Hispanic White individuals. There are wide disparities in AD risk across racially stratified groups, and while these disparities are not due to genetic differences, underrepresentation in genetic research can further exacerbate and contribute to their persistence. We investigated the racial/ethnic representation of participants in United States (US)-based AD genetics and the statistical implications of current representation. METHODS We compared racial/ethnic data of participants from array and sequencing studies in US AD genetics databases, including National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site (NIAGADS) and NIAGADS Data Sharing Service (dssNIAGADS), to AD and related dementia (ADRD) prevalence and mortality. We then simulated the statistical power of these datasets to identify risk variants from non-White populations. RESULTS There is insufficient statistical power (probability <80%) to detect single nucleotide polymorphisms (SNPs) with low to moderate effect sizes (odds ratio [OR]<1.5) using array data from Black and Hispanic participants; studies of Asian participants are not powered to detect variants OR <= 2. Using available and projected sequencing data from Black and Hispanic participants, risk variants with OR = 1.2 are detectable at high allele frequencies. Sample sizes remain insufficiently powered to detect these variants in Asian populations. DISCUSSION AD genetics datasets are largely representative of US ADRD burden. However, there is a wide discrepancy between proportional representation and statistically meaningful representation. Most variation identified in GWAS of non-Hispanic White individuals have low to moderate effects. Comparable risk variants in non-White populations are not detectable given current sample sizes, which could lead to disparities in future studies and drug development. We urge AD genetics researchers and institutions to continue investing in recruiting diverse participants and use community-based participatory research practices.
Collapse
Affiliation(s)
- Diane Xue
- Institute for Public Health GeneticsUniversity of Washington School of Public HealthSeattleWashingtonUSA
| | - Elizabeth E. Blue
- Institute for Public Health GeneticsUniversity of Washington School of Public HealthSeattleWashingtonUSA
- Division of Medical GeneticsDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Brotman Baty InstituteSeattleWashingtonUSA
| | - Matthew P. Conomos
- Department of BiostatisticsUniversity of Washington School of Public HealthSeattleWashingtonUSA
| | - Alison E. Fohner
- Institute for Public Health GeneticsUniversity of Washington School of Public HealthSeattleWashingtonUSA
- Department of EpidemiologyUniversity of Washington School of Public HealthSeattleWashingtonUSA
| |
Collapse
|
43
|
Zempel H. Genetic and sporadic forms of tauopathies-TAU as a disease driver for the majority of patients but the minority of tauopathies. Cytoskeleton (Hoboken) 2024; 81:66-70. [PMID: 37795931 DOI: 10.1002/cm.21793] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
Ageing-associated tauopathies like frontotemporal dementia (FTD), variants thereof (like progressive supranuclear palsy (PSP), pick diseases (PiD), corticobasal degeneration (CBD)), and of course the most prevalent form of dementia, Alzheimer Disease (AD), are widely recognized forms of tauopathies. The list of tauopathies is expanding. We now include: (i) tauopathies where the disease cause or trigger is clearly either physical, such as in Traumatic Brain Injury (TBI) or Chronic Traumatic Encephalopathy (CTE), and (ii) genetic diseases that result in tauopathy but have pathogenic genetic variants in genes not related to TAU. Examples of the latter are myotonic dystrophy Type 1 and Type 2 (DM1, DM2, due to pathogenic genetic variants in the genes DMPK and CNBP, respectively), Niemann-Pick Disease Type C (NPD, due to mutations in NPC1 or NPC2), Kufs Disease (CLN6), Christianson Syndrome (SLC9A6), familial forms of Parkinson Disease (PD), and many others. In terms of affected brain regions and cell types, intracellular distribution of TAU pathology/aggregates, age of disease onset, velocity of disease progression and spreading of TAU pathology, there is, however, little in common in most of these disease entities. Here, I reason that TAU/MAPT is causative for the minority of tauopathies (e.g., MAPT-related FTD/PSP and Vacuolar Tauopathy (VCP)) and a critical mediator for others, like shown by overwhelming evidence for AD. However, TAU may also be a mere bystander or even protective in other settings. Improved understanding of rare tauopathies is necessary to develop specific treatments, but also to improve our understanding of the pathomechanistic role of TAU and to identify diseases that may profit from TAU-based therapies.
Collapse
Affiliation(s)
- Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Ashebir N, Hailesilassie H, Girma S, Nigusu E, Ezo E. Prevalence of Cognitive Impairment and Associated Factors Among Diabetes Mellitus Patients Attending Follow-up Treatment at Fiche General Hospital, North Ethiopia. SAGE Open Nurs 2024; 10:23779608241227752. [PMID: 38292043 PMCID: PMC10826377 DOI: 10.1177/23779608241227752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/22/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024] Open
Abstract
INTRODUCTION Cognitive impairment is having trouble remembering, learning new things, concentrating, or making decisions that affect the daily life of diabetic patients. The worldwide prevalence of diabetes mellitus (DM) was 2.8% in 2000 and is estimated to be 4.4% by 2030. OBJECTIVE To assess the prevalence of cognitive impairment and associated factors among DM patients attending follow-up treatment at Fiche General Hospital, North Ethiopia, 2022. METHODS A hospital-based cross-sectional study was conducted from July 15 to September 15, 2022. The total sample size was 421 and a systematic random sampling technique was used. Data were collected through a face-to-face interview. Data were entered using EpiData Version 3.1 and exported to SPSS Version 24 for analysis. Statistically significant was declared at a P-value of less than .05 with an adjusted odds ratio (AOR) and 95% confidence interval (CI). RESULT The prevalence of cognitive impairment in the current study was 56.3% with (95% CI: 51.5-60.8). Primary educational status (AOR 6.73, 95% CI: 2.92-15.51), having Type II DM (AOR 4.93, 95% CI: 2.84-8.56), uncontrolled blood sugar (AOR 6.24, 95% CI: 3.84-10.17), and current alcohol use (AOR 1.94, 95% CI: 1.11-3.36) were significantly associated. CONCLUSION About three in 5 DM patients attending follow-up treatment at Fiche General Hospital were suffering from cognitive impairment. Educational status, type of DM, status of fasting blood sugar, and current alcohol use were associated with cognitive impairment among DM patients. Therefore, improving educational status, controlling blood sugar, and avoiding alcohol use may reduce the risk of cognitive impairment in DM patients.
Collapse
Affiliation(s)
- Nimona Ashebir
- Department of Psychiatry, Faculty of Medical Sciences, Institute of Health, Jimma University Medical Center, Jimma, Ethiopia
| | - Hailemariam Hailesilassie
- Department of Psychiatry, Faculty of Medical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Shimelis Girma
- Department of Psychiatry, Faculty of Medical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Elias Nigusu
- Department of Comprehensive Nursing, School of Nursing, College of Medicine and Health Sciences, Wachemo University, Hosanna, Ethiopia
| | - Elias Ezo
- Department of Comprehensive Nursing, School of Nursing, College of Medicine and Health Sciences, Wachemo University, Hosanna, Ethiopia
| |
Collapse
|
45
|
Ruggiero C, Baroni M, Xenos D, Parretti L, Macchione IG, Bubba V, Laudisio A, Pedone C, Ferracci M, Magierski R, Boccardi V, Antonelli-Incalzi R, Mecocci P. Dementia, osteoporosis and fragility fractures: Intricate epidemiological relationships, plausible biological connections, and twisted clinical practices. Ageing Res Rev 2024; 93:102130. [PMID: 38030092 DOI: 10.1016/j.arr.2023.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Dementia, osteoporosis, and fragility fractures are chronic diseases, often co-existing in older adults. These conditions pose severe morbidity, long-term disability, and mortality, with relevant socioeconomic implications. While in the research arena, the discussion remains on whether dementia is the cause or the consequence of fragility fractures, healthcare professionals need a better understanding of the interplay between such conditions from epidemiological and physiological standpoints. With this review, we summarized the available literature surrounding the relationship between cognitive impairment, dementia, and both low bone mineral density (BMD) and fragility fractures. Given the strength of the bi-directional associations and their impact on the quality of life, we shed light on the biological connections between brain and bone systems, presenting the main mediators, including gut microbioma, and pathological pathways leading to the dysregulation of bone and brain metabolism. Ultimately, we synthesized the evidence about the impact of available pharmacological treatments for the prevention of fragility fractures on cognitive functions and individuals' outcomes when dementia coexists. Vice versa, the effects of symptomatic treatments for dementia on the risk of falls and fragility fractures are explored. Combining evidence alongside clinical practice, we discuss challenges and opportunities related to the management of older adults affected by cognitive impairment or dementia and at high risk for fragility fracture prevention, which leads to not only an improvement in patient health-related outcomes and survival but also a reduction in healthcare cost and socio-economic burden.
Collapse
Affiliation(s)
- C Ruggiero
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy.
| | - M Baroni
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - D Xenos
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - L Parretti
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - I G Macchione
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - V Bubba
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - A Laudisio
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - C Pedone
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - M Ferracci
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - R Magierski
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - V Boccardi
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - R Antonelli-Incalzi
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - P Mecocci
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| |
Collapse
|
46
|
Begum F, Yousaf M, Iqbal S, Ullah N, Hussain A, Khan M, Khalid A, Algarni AS, Abdalla AN, Khan A, Lodhi MA, Al-Harrasi A. Inhibition of Acetylcholinesterase with Novel 1, 3, 4, Oxadiazole Derivatives: A Kinetic, In Silico, and In Vitro Approach. ACS OMEGA 2023; 8:46816-46829. [PMID: 38107974 PMCID: PMC10719919 DOI: 10.1021/acsomega.3c06298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023]
Abstract
Alzheimer's disease (AD) is a neurological disease that disturbs the memory, thinking skills, and behavior of the affected person. AD is a complex disease caused by the breakdown of acetylcholine via acetylcholinesterase (AChE). The present study aimed to assess the synthetic inhibitors of AChE that could be used to treat AD. For this purpose, synthetic compounds of oxadiazole derivatives (15-35) were evaluated and identified as promising inhibitors of AChE, exhibiting IC50 varying between 41.87 ± 0.67 and 1580.25 ± 0.7 μM. The kinetic parameters indicated that all the studied compounds bind to the allosteric site and decrease the efficiency of the AChE enzyme. In silico docking analysis showed that the majority of the compounds interact with the anionic subsite and Per-Arnt-Sim domain of AChE and are stabilized by various bonds including π-π and hydrogen bonding. The stability of the most potent compounds 16 and 17 with AChE interaction was confirmed by molecular dynamics simulations. Moreover, all compounds exhibited concentration-dependent calcium (Ca2+) antagonistic and spasmolytic activities. Among the whole series of oxadiazole derivatives, compounds 16 and 17 displayed the highest activities on spontaneous and potassium (K+)-induced contraction. Therefore, the AChE inhibitory potential, cytotoxicity safe profile, and Ca2+ antagonistic ability of these compounds make them potential therapeutic agents against AD and its associated problems in the future.
Collapse
Affiliation(s)
- Farida Begum
- Department
of Biochemistry, Abdul Wali Khan University
Mardan, Khyber
Pakhtunkhwa 23200, Pakistan
| | - Muhammad Yousaf
- Department
of Chemistry, Government Post Graduate College
Mardan, Khyber
Pakhtunkhwa 23200, Pakistan
| | - Sajid Iqbal
- Atta-ur-Rahman
School of Applied Biosciences (ASAB), National
University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Nazif Ullah
- Department
of Biotechnology, Abdul Wali Khan University
Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Anwar Hussain
- Department
of Botany, Garden Campus, Abdul Wali Khan
University Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Momin Khan
- Department
of Chemistry, Abdul Wali Khan University
Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Asaad Khalid
- Substance
Abuse and Toxicology Research Center, Jazan
University, P.O. Box: 114, Jazan 45142, Saudi Arabia
| | - Alanood S. Algarni
- Department
of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ashraf N. Abdalla
- Department
of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ajmal Khan
- Natural
and Medical Sciences Research Centre, University
of Nizwa, Birkat-ul-Mouz, Nizwa 616, Sultanate of Oman
| | - Muhammad Arif Lodhi
- Department
of Biochemistry, Abdul Wali Khan University
Mardan, Khyber
Pakhtunkhwa 23200, Pakistan
| | - Ahmed Al-Harrasi
- Natural
and Medical Sciences Research Centre, University
of Nizwa, Birkat-ul-Mouz, Nizwa 616, Sultanate of Oman
| |
Collapse
|
47
|
Weiss J, Beydoun MA, Beydoun HA, Fanelli-Kuczmarski MT, Banerjee S, Hamrah A, Evans MK, Zonderman AB. Pathways explaining racial/ethnic disparities in incident all-cause dementia among middle-aged US adults. Alzheimers Dement 2023; 19:4299-4310. [PMID: 36868873 PMCID: PMC10475144 DOI: 10.1002/alz.12976] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/16/2022] [Accepted: 01/13/2023] [Indexed: 03/05/2023]
Abstract
INTRODUCTION Racial disparities in dementia incidence exist, but less is known about their presence and drivers among middle-aged adults. METHODS We used time-to-event analysis among a sample of 4378 respondents (age 40-59 years at baseline) drawn from the third National Health and Nutrition Examination Surveys (NHANES III) with administrative linkage-spanning the years 1988-2014-to evaluate potential mediating pathways through socioeconomic status (SES), lifestyle, and health-related characteristics. RESULTS Compared with Non-Hispanic White (NHW) adults, Non-White adults had a higher incidence of AD-specific (hazard ratio [HR] = 2.05, 95% confidence interval [CI]: 1.21, 3.49) and all-cause dementia (HR = 2.01, 95% CI: 1.36, 2.98). Diet, smoking, and physical activity were among characteristics on the pathway between race/ethnicity, SES, and dementia, with health-mediating effects of smoking and physical activity on dementia risk. DISCUSSION We identified several pathways that may generate racial disparities in incident all-cause dementia among middle-aged adults. No direct effect of race was observed. More studies are needed to corroborate our findings in comparable populations.
Collapse
Affiliation(s)
- Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA, USA
| | - May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institutes on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | - Hind A. Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA
| | - Marie T. Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institutes on Aging, NIA/NIH/IRP, Baltimore, MD, USA
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA
| | - Sri Banerjee
- Public Health Doctoral Programs, Walden University, Minneapolis, MN, USA
| | | | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institutes on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institutes on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| |
Collapse
|
48
|
Beydoun MA, Beydoun HA, Fanelli-Kuczmarski MT, Weiss J, Georgescu MF, Meirelles O, Lyall DM, Evans MK, Zonderman AB. Pathways explaining racial/ethnic and socio-economic disparities in dementia incidence: the UK Biobank study. Aging (Albany NY) 2023; 15:9310-9340. [PMID: 37751591 PMCID: PMC10564412 DOI: 10.18632/aging.205058] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Pathways explaining racial/ethnic disparities in dementia risk are under-evaluated. METHODS We examine those disparities and their related pathways among UK Biobank study respondents (50-74 y, N = 323,483; 3.6% non-White minorities) using a series of Cox proportional hazards and generalized structural equations models (GSEM). RESULTS After ≤15 years, 5,491 all-cause dementia cases were diagnosed. Racial minority status (RACE_ETHN, Non-White vs. White) increased dementia risk by 24% (HR = 1.24, 95% CI: 1.07-1.45, P = 0.005), an association attenuated by socio-economic status (SES), (HR = 1.12, 95% CI: 0.96-1.31). Total race-dementia effect was mediated through both SES and Life's Essential 8 lifestyle sub-score (LE8LIFESTYLE), combining diet, smoking, physical activity, and sleep factors. SES was inversely related to dementia risk (HR = 0.69, 95% CI: 0.67, 0.72, P < 0.001). Pathways explaining excess dementia risk among racial minorities included 'RACE_ETHN(-) → SES(-) → DEMENTIA', 'RACE_ETHN(-) → SES(-) → Poor cognitive performance, COGN(+) → DEMENTIA' and 'RACE_ETHN(-) → SES(+) → LE8LIFESTYLE(-) → DEMENTIA'. CONCLUSIONS Pending future interventions, lifestyle factors including diet, smoking, physical activity, and sleep are crucial for reducing racial and socio-economic disparities in dementia.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Hind A. Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA 22060, USA
| | - Marie T. Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Jordan Weiss
- Stanford Center on Longevity, Stanford University, Stanford, CA 94305, USA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Donald M. Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow, Scottland, UK
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| |
Collapse
|
49
|
Kim SE, Shin C, Yim J, Seo K, Ryu H, Choi H, Park J, Min BK. Resting-state electroencephalographic characteristics related to mild cognitive impairments. Front Psychiatry 2023; 14:1231861. [PMID: 37779609 PMCID: PMC10539934 DOI: 10.3389/fpsyt.2023.1231861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Alzheimer's disease (AD) causes a rapid deterioration in cognitive and physical functions, including problem-solving, memory, language, and daily activities. Mild cognitive impairment (MCI) is considered a risk factor for AD, and early diagnosis and treatment of MCI may help slow the progression of AD. Electroencephalography (EEG) analysis has become an increasingly popular tool for developing biomarkers for MCI and AD diagnosis. Compared with healthy elderly, patients with AD showed very clear differences in EEG patterns, but it is inconclusive for MCI. This study aimed to investigate the resting-state EEG features of individuals with MCI (n = 12) and cognitively healthy controls (HC) (n = 13) with their eyes closed. EEG data were analyzed using spectral power, complexity, functional connectivity, and graph analysis. The results revealed no significant difference in EEG spectral power between the HC and MCI groups. However, we observed significant changes in brain complexity and networks in individuals with MCI compared with HC. Patients with MCI exhibited lower complexity in the middle temporal lobe, lower global efficiency in theta and alpha bands, higher local efficiency in the beta band, lower nodal efficiency in the frontal theta band, and less small-world network topology compared to the HC group. These observed differences may be related to underlying neuropathological alterations associated with MCI progression. The findings highlight the potential of network analysis as a promising tool for the diagnosis of MCI.
Collapse
Affiliation(s)
- Seong-Eun Kim
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Chanwoo Shin
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Junyeop Yim
- Department of Applied Mathematics, Kongju National University, Gongju-si, Republic of Korea
| | - Kyoungwon Seo
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Hokyoung Ryu
- Graduate School of Technology and Innovation Management, Hanyang University, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Byoung-Kyong Min
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| |
Collapse
|
50
|
Iakunchykova O, Schirmer H, Vangberg T, Wang Y, Benavente ED, van Es R, van de Leur RR, Lindekleiv H, Attia ZI, Lopez-Jimenez F, Leon DA, Wilsgaard T. Machine-learning-derived heart and brain age are independently associated with cognition. Eur J Neurol 2023; 30:2611-2619. [PMID: 37254942 DOI: 10.1111/ene.15902] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/03/2023] [Accepted: 05/28/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND AND PURPOSE A heart age biomarker has been developed using deep neural networks applied to electrocardiograms. Whether this biomarker is associated with cognitive function was investigated. METHODS Using 12-lead electrocardiograms, heart age was estimated for a population-based sample (N = 7779, age 40-85 years, 45.3% men). Associations between heart delta age (HDA) and cognitive test scores were studied adjusted for cardiovascular risk factors. In addition, the relationship between HDA, brain delta age (BDA) and cognitive test scores was investigated in mediation analysis. RESULTS Significant associations between HDA and the Word test, Digit Symbol Coding Test and tapping test scores were found. HDA was correlated with BDA (Pearson's r = 0.12, p = 0.0001). Moreover, 13% (95% confidence interval 3-36) of the HDA effect on the tapping test score was mediated through BDA. DISCUSSION Heart delta age, representing the cumulative effects of life-long exposures, was associated with brain age. HDA was associated with cognitive function that was minimally explained through BDA.
Collapse
Affiliation(s)
- Olena Iakunchykova
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Henrik Schirmer
- Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Torgil Vangberg
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- PET Imaging Center, University Hospital of North Norway, Tromsø, Norway
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Ernest D Benavente
- Department of Experimental Cardiology, University Medical Center, Utrecht, The Netherlands
| | - René van Es
- Department of Cardiology, University Medical Center, Utrecht, The Netherlands
| | | | - Haakon Lindekleiv
- University Hospital of North Norway, Tromsø, Norway
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Zachi I Attia
- Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | - David A Leon
- Department of Noncommunicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Tom Wilsgaard
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|