1
|
Yu SP, Choi E, Jiang MQ, Wei L. Acute and chronic excitotoxicity in ischemic stroke and late-onset Alzheimer's disease. Neural Regen Res 2025; 20:1981-1988. [PMID: 39101641 PMCID: PMC11691467 DOI: 10.4103/nrr.nrr-d-24-00398] [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: 04/11/2024] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
Stroke and Alzheimer's disease are common neurological disorders and often occur in the same individuals. The comorbidity of the two neurological disorders represents a grave health threat to older populations. This review presents a brief background of the development of novel concepts and their clinical potentials. The activity of glutamatergic N-methyl-D-aspartate receptors and N-methyl-D-aspartate receptor-mediated Ca 2+ influx is critical for neuronal function. An ischemic insult induces prompt and excessive glutamate release and drastic increases of intracellular Ca 2+ mainly via N-methyl-D-aspartate receptors, particularly of those at the extrasynaptic site. This Ca 2+ -evoked neuronal cell death in the ischemic core is dominated by necrosis within a few hours and days known as acute excitotoxicity. Furthermore, mild but sustained Ca 2+ increases under neurodegenerative conditions such as in the distant penumbra of the ischemic brain and early stages of Alzheimer's disease are not immediately toxic, but gradually set off deteriorating Ca 2+ -dependent signals and neuronal cell loss mostly because of activation of programmed cell death pathways. Based on the Ca 2+ hypothesis of Alzheimer's disease and recent advances, this Ca 2+ -activated "silent" degenerative excitotoxicity evolves from years to decades and is recognized as a unique slow and chronic neuropathogenesis. The N-methyl-D-aspartate receptor subunit GluN3A, primarily at the extrasynaptic site, serves as a gatekeeper for the N-methyl-D-aspartate receptor activity and is neuroprotective against both acute and chronic excitotoxicity. Ischemic stroke and Alzheimer's disease, therefore, share an N-methyl-D-aspartate receptor- and Ca 2+ -mediated mechanism, although with much different time courses. It is thus proposed that early interventions to control Ca 2+ homeostasis at the preclinical stage are pivotal for individuals who are susceptible to sporadic late-onset Alzheimer's disease and Alzheimer's disease-related dementia. This early treatment simultaneously serves as a preconditioning therapy against ischemic stroke that often attacks the same individuals during abnormal aging.
Collapse
Affiliation(s)
- Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Emily Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Yuan J, Huang R, Nao J, Dong X. The role of semaphorin 3A in the pathogenesis and progression of Alzheimer's disease and other aging-related diseases: A comprehensive review. Pharmacol Res 2025; 215:107732. [PMID: 40222695 DOI: 10.1016/j.phrs.2025.107732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Aging serves as a pivotal factor in the etiology of numerous diseases, such as Alzheimer's disease (AD), Parkinson's disease, diabetes, osteoarthritis, atherosclerosis and aging-related macular degeneration. Notably, these diseases often interact with AD through various pathways, facilitating the onset or progression of one another. Semaphorin 3 A (Sema3A), a protein that is essential for axonal guidance during neural development, has recently been identified as a novel regulator in the pathogenesis and progression of multiple aging-related diseases. This article provides a comprehensive review of the expression patterns and mechanisms of action of Sema3A in these diseases. Specifically, Sema3A influences the occurrence and development of aging-related diseases by participating in oxidative stress, inflammatory responses, apoptosis, and synaptic plasticity. Therefore, therapeutic strategies targeting Sema3A present promising avenues for delaying the progression of aging-related diseases and offer novel insights and strategies for their treatment.
Collapse
Affiliation(s)
- Jiayu Yuan
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| |
Collapse
|
3
|
Pan W, Teng Y, Han X, Liu S, Pang X, Wang L, Zhao M. Value of blood neural cell-derived small extracellular vesicles in the diagnosis and prediction of Alzheimer's disease: A systematic revie. J Prev Alzheimers Dis 2025:100193. [PMID: 40316481 DOI: 10.1016/j.tjpad.2025.100193] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Blood neural cell-derived small extracellular vesicles (sEVs) can directly reflect changes in brain tissue and are easier to obtain than cerebrospinal fluid. This article systematically reviews the alterations of proteins and miRNAs from neural cell-derived sEVs in patients with Alzheimer's disease (AD), and summarizes the biomarkers with clinical diagnostic and predictive value. PubMed, Web of Science, Embase, and Cochrane Library were searched for studies in blood neural cell-derived sEVs in AD patients up to May 2024. According to the inclusion and exclusion criteria, the literature was screened, the information was extracted and the quality was evaluated. Proteins and miRNAs from neural cell-derived sEVs were classified and summarized, focusing on target molecules with high diagnostic and predictive values for AD. A final 34 articles reporting 5601 participants were included. In cross-sectional studies, Aβ- and Tau-related proteins (Aβ42, Aβ42/40, p-S396-Tau, p-Tau181), p-S312-IRS-1, and cathepsin D were increased, conversely, synaptic proteins (neurogranin, synaptotagmin, synaptophysin, synaptopodin, NMDAR2A) and REST were decreased in blood neuron-derived sEVs (NDsEVs) of patients with AD. While miR-29c-3p was increased in blood NDsEVs and glial cell-derived sEVs. Each of these proteins and miRNAs demonstrated high AD diagnostic value. Additionally, blood astrocyte-derived sEVs (ADsEVs) showed increased complement effector proteins and decreased complement regulatory proteins with a moderate diagnostic value. In longitudinal cohort studies, three composite models displayed high predictive efficacy for early AD prediction, and could predict the occurrence of AD within 1-10 years. Therefore, Aβ- and Tau-related proteins, synaptic proteins, and miRNA in blood neural cell-derived sEVs demonstrate high AD diagnostic and predictive values serving as important biomarkers. Especially, synaptic proteins showed significant changes in the early clinical stage, which has early predictive value.
Collapse
Affiliation(s)
- Weibing Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaojiao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxue Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Mingjing Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
de Rijke TJ, Vasseur D, van der Flier WM, Minkman MM, Rhodius-Meester HF, Verwey NA, Smets EM, Visser LN. Exploring interdisciplinary perspectives on the implementation of personalized medicine and patient-orchestrated care in Alzheimer's disease: A qualitative study within the ABOARD research project. J Alzheimers Dis 2025; 105:120-133. [PMID: 40116704 DOI: 10.1177/13872877251326166] [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: 03/23/2025]
Abstract
BackgroundThe concepts of 'personalized medicine' and 'patient-orchestrated care' in Alzheimer's disease (AD) lack standard conceptualization, which presents challenges for collaborative and interdisciplinary care.ObjectiveWe explored the interpretations and perspectives of professionals involved in interdisciplinary work on a large-scale project, "ABOARD", with the aim to implement personalized medicine and patient-orchestrated care in AD.MethodsSemi-structured interviews were conducted with 30 professionals and audio-recorded. Two researchers independently coded the data inductively, followed by a thematic analysis.ResultsAccording to professionals across different disciplinary backgrounds (mean age 45.7 years; 53.3% female), personalized medicine pertains to the relevant options that an individual has, informed by biomedical and psychosocial factors, whereas patient-orchestrated care captures factors relevant to the decision-making process. Professionals differed in their views on patient-orchestrated care regarding its desirability and feasibility. The concepts were viewed as similar by professionals, as both involve personal preferences while ultimately assigning responsibility to the clinician. However, implementation challenges persist, and no thematic differences were found between clinicians and other AD-related professionals.ConclusionsAD professionals have shared interpretations and perspectives on implementation of personalized medicine but differed in their views on patient-orchestrated care. Personal preferences are seen as part of personalized medicine, but not yet reflected in definitions in the AD field and beyond. Critical discussions on the challenges and existing doubts are necessary for both personalized medicine and patient-orchestrated care. Multi-level implementation changes are needed for both concepts, which warrants stakeholder involvement as well as support and resources from the entire AD field.
Collapse
Affiliation(s)
- Tanja J de Rijke
- Amsterdam UMC, University of Amsterdam, Medical Psychology, Meibergdreef 9, Amsterdam, the Netherlands
- Alzheimercentrum Amsterdam, Neurologie, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Amsterdam Public Health, Personalized Medicine & Quality of Care, Amsterdam, the Netherlands
| | - Dianne Vasseur
- Vilans, the national Centre of expertise for care and support, Utrecht, the Netherlands
| | - Wiesje M van der Flier
- Alzheimercentrum Amsterdam, Neurologie, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Epidemiology & Data Science, Vrije Universiteit Amsterdam UMC, Amsterdam, the Netherlands
| | - Mirella Mn Minkman
- Vilans, the national Centre of expertise for care and support, Utrecht, the Netherlands
- Tilburg University, TIAS School for business and society, Tilburg, the Netherlands
| | - Hanneke Fm Rhodius-Meester
- Alzheimercentrum Amsterdam, Neurologie, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Internal medicine, Geriatric Medicine section, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, Oslo, Norway
| | - Nicolaas A Verwey
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Neurology, Memory Clinic, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Ellen Ma Smets
- Amsterdam UMC, University of Amsterdam, Medical Psychology, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Public Health, Personalized Medicine & Quality of Care, Amsterdam, the Netherlands
| | - Leonie Nc Visser
- Amsterdam UMC, University of Amsterdam, Medical Psychology, Meibergdreef 9, Amsterdam, the Netherlands
- Alzheimercentrum Amsterdam, Neurologie, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Amsterdam Public Health, Personalized Medicine & Quality of Care, Amsterdam, the Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Liu W, Zuo C, Chen L, Lan H, Luo C, Li X, Kemp GJ, Lui S, Suo X, Gong Q. The whole-brain structural and functional connectome in Alzheimer's disease spectrum: A multimodal Bayesian meta-analysis of graph theoretical characteristics. Neurosci Biobehav Rev 2025:106174. [PMID: 40280288 DOI: 10.1016/j.neubiorev.2025.106174] [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: 11/28/2024] [Revised: 03/19/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Alzheimer's disease (AD) spectrum is increasingly recognized as a progressive network-disconnection syndrome. Neuroimaging studies using graph theoretical analysis (GTA) have reported alterations in the topological properties of whole-brain structural and functional connectomes in both preclinical AD and AD patients, though findings remain inconsistent. This study aimed to identify robust changes in multimodal GTA metrics across the AD spectrum through a comprehensive literature search and Bayesian random-effects meta-analyses. The analysis included 53 studies (17 functional and 37 structural), involving 1743 AD patients, 1502 preclinical AD patients, and 1824 healthy controls (HC). Results revealed lower structural network integration (evidenced by higher characteristic path length and/or normalized characteristic path length) and segregation (evidenced by lower clustering coefficient and local efficiency) in AD and preclinical AD patients compared to HC. Functional network segregation was also lower in AD patients, while preclinical AD showed preserved functional topology despite structural changes. Moderator analyses identified potential methodological moderators, including neuroimaging technique, node and edge definitions, and network type, although further validation is needed. These findings support the progressive disconnection hypothesis in the AD spectrum and suggest that structural network alterations may precede functional network changes. Furthermore, the results help clarify inconsistencies in previous studies and highlight the utility of graph-based metrics as biomarkers for staging AD progression.
Collapse
Affiliation(s)
- Wenxiong Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China; Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chao Zuo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Li Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Huan Lan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Chunyan Luo
- Department of Neurology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Graham J Kemp
- Liverpool Magnetic Resonance Imaging Centre (LiMRIC) and Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Su Lui
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Xueling Suo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China.
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China; Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361022, Fujian, China.
| |
Collapse
|
6
|
Busto GU, Hirtz C, Carriere I, Bennys K, Gutierrez LA, Kindermans J, Helmer C, Gabelle A, Lehmann S, Berr C. A six-year risk assessment for dementia and Alzheimer's disease in the general population through immunoprecipitation-mass spectrometry plasma amyloid quantification. J Prev Alzheimers Dis 2025:100186. [PMID: 40254499 DOI: 10.1016/j.tjpad.2025.100186] [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: 12/17/2024] [Revised: 02/13/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Identifying individuals at risk for dementia and Alzheimer's disease (AD) in the general population (GP) is increasingly essential due to new diagnostic criteria and opportunities for effective interventions. Plasma-based biomarkers (pBB) offer a promising approach for detecting positive amyloid profile. However, their effectiveness in predicting clinical dementia and AD risk at the GP level remains largely unexplored. OBJECTIVES To assess the risk of clinical dementia and AD using pBB amyloid biomarkers in GP using the most up-to-date proteomic techniques. DESIGN Case-cohort study randomly selected from a prospective cohort. SETTING The three-city community-living study. PARTICIPANTS Over 65 years recruited from the electoral rolls of three French cities. MEASUREMENTS pBB amyloid levels (Aβ42, Aβ40 and APP669-711) were measured in the plasma using the mass spectrometry-based (IPMS)-Shimadzu modified technology. Patients were monitored for up to 6 years for incident dementia and AD according to DSM-IV and NINCDS/ADRDA criteria. Cox proportional hazard models adjusted for multiple covariables, including age and renal function, were used to estimate hazard ratios. RESULTS Plasma samples from 327 participants were analyzed with a mean age 83 years (80-87), 64.8 % females and a median follow-up time of 2.7 years (0.8-4.8) and including 121 incident dementia cases. Our findings indicate that the Aβ42/Aβ40 ratio, along with a composite score that encompasses APP669-711 and Aβ40/Aβ42 ratios, serves as significant predictors of clinical dementia [HR(95 %CI) = 3.52 (1.69-7.32), p-value<0.001 and 4.34 (2.06-9.17), p-value<0.001, respectively] and AD risk over a six-year period, while also accounting for age and sex interactions. Furthermore, elevated Aβ40 levels correlate with an increased risk of developing dementia (HR=2.56, 95 % CI 1.22-5.35, p = 0.01) and AD (HR=2.60, 95 %CI 1.06-6.36, p = 0.04), and our study confirms that Aβ42 concentrations are significantly influenced by renal function. CONCLUSIONS This research advances the potential application of plasma amyloid biomarkers for assessing the risk of clinical dementia and AD in the general population within short period of time, positioning it as a valuable tool alongside existing plasma PT217 biomarkers or using ratio of both of them.
Collapse
Affiliation(s)
- Germain U Busto
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France; Memory Resource and Research Center, Department of Neurology, University of Montpellier Hospital, 80 avenue Augustin Fliche, 34295, Montpellier, France
| | - Christophe Hirtz
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France; University of Montpellier, IRMB, CHU Montpellier, 80 avenue Augustin Fliche, 34295, Montpellier, France
| | - Isabelle Carriere
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France
| | - Karim Bennys
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France; Memory Resource and Research Center, Department of Neurology, University of Montpellier Hospital, 80 avenue Augustin Fliche, 34295, Montpellier, France
| | - Laure-Anne Gutierrez
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France
| | - Jana Kindermans
- University of Montpellier, IRMB, CHU Montpellier, 80 avenue Augustin Fliche, 34295, Montpellier, France
| | - Catherine Helmer
- University of Bordeaux, INSERM UMR U1219, Bordeaux Population Health (BPH) Research Centre, 146 rue Léo-Saignat. 33076, Bordeaux, France
| | - Audrey Gabelle
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France; Memory Resource and Research Center, Department of Neurology, University of Montpellier Hospital, 80 avenue Augustin Fliche, 34295, Montpellier, France
| | - Sylvain Lehmann
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France; University of Montpellier, IRMB, CHU Montpellier, 80 avenue Augustin Fliche, 34295, Montpellier, France.
| | - Claudine Berr
- INM, University of Montpellier, INSERM, 80 Av. Augustin Fliche, 34000, Montpellier, France
| |
Collapse
|
7
|
Starmans NL, Leeuwis AE, Bennink E, Meyer Viol SL, Golla SS, Dankbaar JW, Bron EE, Biessels GJ, Kappelle LJ, van der Flier WM, Tolboom N. Dynamic PET imaging in patients with unilateral carotid occlusion shows lateralized cerebral hypoperfusion, but no amyloid binding. J Alzheimers Dis 2025:13872877251329593. [PMID: 40241519 DOI: 10.1177/13872877251329593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BackgroundCarotid occlusive disease is a risk factor for cognitive decline. A possible underlying etiology is that hemodynamic impairment results in decreased cerebral perfusion, exacerbated amyloid-β accumulation (Aβ) and poorer cognitive performance.ObjectiveWe aimed to determine whether patients with unilateral internal carotid artery (ICA) occlusion have less cerebral perfusion and more Aβ in the ipsilateral than in the contralateral hemisphere, and whether perfusion and Aβ are associated with cognitive functioning.MethodsWe included 20 patients (age 67.2 ± 7.0 years, 8 females, MMSE 29 [27-29]) with unilateral ICA occlusion, which underwent neuropsychological assessment and dynamic 18F-Florbetaben positron emission tomography (PET). Global and regional relative perfusion (R1) and binding potential (BPND) were obtained from the PET-images using a simplified reference tissue model. We performed Wilcoxon signed-rank tests to examine differences between hemispheres within subjects and linear regression to investigate associations with cognitive functioning.ResultsMedian global R1 was 0.911 (0.883-0.950) and global BPND was 0.172 (0.129-0.187). R1 was lower in the hemisphere ipsilateral to the ICA occlusion than in the contralateral hemisphere (0.899 [0.876-0.921] versus 0.935 [0.889-0.970]). BPND did not differ significantly between hemispheres (ipsilateral 0.172 [0.124-0.181] versus contralateral 0.168 [0.137-0.191]). Neither cerebral perfusion nor Aβ burden were associated with cognitive functioning.ConclusionsPatients with unilateral ICA occlusion did not have more Aβ in the ipsilateral hemisphere than in the contralateral hemisphere despite ipsilateral hypoperfusion. Perfusion and Aβ were unrelated to cognitive functioning. This indicates that cognitive impairment in patients with ICA occlusion is not due to exacerbated Aβ accumulation.
Collapse
Affiliation(s)
- Naomi Lp Starmans
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna E Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Medical Psychology, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Edwin Bennink
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sebastiaan L Meyer Viol
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Sandeep Sv Golla
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Jan Willem Dankbaar
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esther E Bron
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
8
|
Filippi L, Schillaci O. Global experience in brain amyloid imaging. Semin Nucl Med 2025:S0001-2998(25)00030-3. [PMID: 40222870 DOI: 10.1053/j.semnuclmed.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025]
Abstract
Brain amyloid imaging has become a crucial tool in diagnosing and understanding Alzheimer's disease (AD) and related neurodegenerative disorders. The introduction of amyloid positron emission tomography (PET) with [¹¹C]Pittsburgh Compound-B ([¹¹C]PiB) in the early 2000s marked a breakthrough in visualizing amyloid-β (Aβ) deposition in vivo. Subsequent development of ¹⁸F-labeled tracers, such as [¹⁸F]florbetapir, [¹⁸F]flutemetamol, and [¹⁸F]florbetaben, improved accessibility and extended imaging capabilities. However, global adoption remains uneven due to disparities in healthcare infrastructure, costs, and regulatory frameworks. In high-income countries, amyloid PET is increasingly used in clinical workflows, particularly for differentiating atypical dementia cases and selecting patients for anti-amyloid therapies like aducanumab and lecanemab. Despite its high sensitivity and specificity, challenges persist regarding its clinical utility, particularly in cognitively normal individuals with amyloid accumulation. Research is focusing on integrating amyloid PET with other biomarkers-tau PET, cerebrospinal fluid analysis, and plasma assays-to improve diagnostic accuracy. Geographical variations in amyloid PET research and implementation reveal North America and Europe as leaders, while access remains limited in low- and middle-income countries. Efforts such as the Worldwide Alzheimer's Disease Neuroimaging Initiative aim to enhance global standardization and accessibility. Emerging trends in artificial intelligence (AI)-assisted imaging analysis and next-generation tracers promise further improvements. Addressing ethical concerns related to preclinical screening and ensuring equitable access to amyloid PET are critical for optimizing its role in neurology and nuclear medicine worldwide.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy.
| |
Collapse
|
9
|
Pérez-González AP, de Anda-Jáuregui G, Hernández-Lemus E. Differential Transcriptional Programs Reveal Modular Network Rearrangements Associated with Late-Onset Alzheimer's Disease. Int J Mol Sci 2025; 26:2361. [PMID: 40076979 PMCID: PMC11900169 DOI: 10.3390/ijms26052361] [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: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, genetically heterogeneous disorder. The diverse phenotypes associated with AD result from interactions between genetic and environmental factors, influencing multiple biological pathways throughout disease progression. Network-based approaches offer a way to assess phenotype-specific states. In this study, we calculated key network metrics to characterize the network transcriptional structure and organization in LOAD, focusing on genes and pathways implicated in AD pathology within the dorsolateral prefrontal cortex (DLPFC). Our findings revealed disease-specific coexpression markers associated with diverse metabolic functions. Additionally, significant differences were observed at both the mesoscopic and local levels between AD and control networks, along with a restructuring of gene coexpression and biological functions into distinct transcriptional modules. These results show the molecular reorganization of the transcriptional program occurring in LOAD, highlighting specific adaptations that may contribute to or result from cellular responses to pathological stressors. Our findings may support the development of a unified model for the causal mechanisms of AD, suggesting that its diverse manifestations arise from multiple pathways working together to produce the disease's complex clinical patho-phenotype.
Collapse
Affiliation(s)
- Alejandra Paulina Pérez-González
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado Edificio B Primer Piso, Ciudad Universitaria, Mexico City 04510, Mexico
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Guillermo de Anda-Jáuregui
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Investigadores por M’exico, Conahcyt, Mexico City 03940, Mexico
| | - Enrique Hernández-Lemus
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
10
|
Gutiérrez‐Jiménez E, Rasmussen PM, Mikkelsen IK, Kura S, Fruekilde SK, Hansen B, Bordoni L, Carlsen J, Palmfeldt J, Boas DA, Sakadžić S, Vinogradov S, Khatib ME, Ramos‐Cejudo J, Wied B, Leduc‐Galindo D, Canepa E, Mar AC, Gamallo‐Lana B, Fossati S, Østergaard L. Carbonic anhydrase inhibitors prevent presymptomatic capillary flow disturbances in a model of cerebral amyloidosis. Alzheimers Dement 2025; 21:e70023. [PMID: 40133235 PMCID: PMC11936728 DOI: 10.1002/alz.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION Disturbances in microvascular flow dynamics are hypothesized to precede the symptomatic phase of Alzheimer's disease (AD). However, evidence in presymptomatic AD remains elusive, underscoring the need for therapies targeting these early vascular changes. METHODS We employed a multimodal approach, combining in vivo optical imaging, molecular techniques, and ex vivo magnetic resonance imaging, to investigate early capillary dysfunction in C57BL/6-Tg(Thy1-APPSwDutIowa)BWevn/Mmjax (Tg-SwDI) mice without memory impairment. We also assessed the efficacy of carbonic anhydrase inhibitors (CAIs) in preventing capillary flow disturbances. RESULTS Our study revealed capillary flow disturbances associated with alterations in capillary morphology, adhesion molecule expression, and amyloid beta (Aβ) load in 9- to 10-month-old Tg-SwDI mice without memory impairment. CAI treatment ameliorated these capillary flow disturbances, enhanced oxygen availability, and reduced Aβ load. DISCUSSION These findings underscore the importance of capillary flow disturbances as early biomarkers in presymptomatic AD and highlight the potential of CAIs for preserving vascular integrity in the early stages of AD. HIGHLIGHTS Uncovered early capillary dysfunction in a presymptomatic Alzheimer's disease (AD) mouse model. Evidence linking capillary stalls and capillary dysfunction with oxygen delivery issues in AD. Novel use of carbonic anhydrase inhibitors to prevent early capillary flow disturbances in AD.
Collapse
Affiliation(s)
- Eugenio Gutiérrez‐Jiménez
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Peter Mondrup Rasmussen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Irene Klærke Mikkelsen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Sreekanth Kura
- Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
| | - Signe K. Fruekilde
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Luca Bordoni
- GliaLab and Letten Centre, Division of AnatomyDepartment of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jasper Carlsen
- Research Unit for Molecular Medicine (MMF), Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine (MMF), Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - David A. Boas
- Department of Biomedical EngineeringBoston UniversityBostonMassachusettsUSA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Sergei Vinogradov
- Department of Biochemistry and BiophysicsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of ChemistrySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Mirna El Khatib
- Department of Biochemistry and BiophysicsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of ChemistrySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jaime Ramos‐Cejudo
- Department of Psychiatry and NeurologyNew York University (NYU) Grossman School of MedicineNew York CityNew YorkUSA
| | - Boris Wied
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Desiree Leduc‐Galindo
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Elisa Canepa
- Alzheimer's Center at TempleDepartment of Neural SciencesLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Adam C. Mar
- Department of Neuroscience and PhysiologyNeuroscience InstituteNew York University (NYU) Grossman School of MedicineNew YorkNew YorkUSA
| | - Begona Gamallo‐Lana
- Department of Neuroscience and PhysiologyNeuroscience InstituteNew York University (NYU) Grossman School of MedicineNew YorkNew YorkUSA
| | - Silvia Fossati
- Alzheimer's Center at TempleDepartment of Neural SciencesLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Section of NeuroradiologyDepartment of RadiologyAarhus University HospitalAarhusDenmark
| |
Collapse
|
11
|
Stögmann E, Schmidt R. Amyloid-beta antibody treatment in Alzheimer's disease : An update on recent data and outlook on implementation in clinical routine. Wien Klin Wochenschr 2025; 137:182-188. [PMID: 39505779 PMCID: PMC11926045 DOI: 10.1007/s00508-024-02466-7] [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: 07/05/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024]
Abstract
Amyloid-beta (Aβ) antibody treatment has emerged as a promising approach for the treatment of Alzheimer's disease (AD), targeting the accumulation of Aβ plaques, which are a hallmark of the disease. This review provides an update on recent clinical trial data, highlighting the efficacy and safety of various antibodies targeting Aβ. Recent trials have demonstrated that certain Aβ antibodies can reduce amyloid plaques and slow cognitive decline in patients with early AD. Key findings from trials of drugs are discussed, including their mechanisms of action, dosing regimens, and observed side effects. The potential for Aβ antibody therapy to be integrated into routine clinical practice is also explored. While Aβ antibody therapy represents a significant advancement in AD treatment, ongoing research is needed to optimize their use and understand their long-term impact. This review underscores the importance of personalized medicine in AD and the need for continued innovation in therapeutic strategies.
Collapse
Affiliation(s)
- Elisabeth Stögmann
- Department of Neurology, Medical University of Vienna, Waehringer Gürtel 18-20, 1090, Vienna, Austria.
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, 8036, Graz, Austria
| |
Collapse
|
12
|
Kaur R, Pandey S, Gupta S, Singh J. Harnessing the potential of long non-coding RNAs in the pathophysiology of Alzheimer's disease. Exp Neurol 2025; 385:115134. [PMID: 39740737 DOI: 10.1016/j.expneurol.2024.115134] [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: 07/02/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Alzheimer's disease (AD), a diverse neurodegenerative disease, is the leading cause of dementia, accounting for 60-80 % of all cases. The pathophysiology of Alzheimer's disease is unknown, and there is no cure at this time. Recent developments in transcriptome-wide profiling have led to the identification of a number of non-coding RNAs (ncRNAs). Among these, long non-coding RNAs (lncRNAs)-long transcripts that don't seem to be able to code for proteins-have drawn attention because they function as regulatory agents in a variety of biological processes. Recent research suggests that lncRNAs play a role in the pathogenesis of Alzheimer's disease by modulating tau hyperphosphorylation, amyloid production, synaptic impairment, neuroinflammation, mitochondrial dysfunction, and oxidative stress, though their precise effects on the disorder are unknown. The biology and modes of action of the best-characterized lncRNAs in AD will be outlined here, with an emphasis on their possible involvement in the pathophysiology of the disease. As lncRNAs may offer prospective prognostic/diagnostic biomarkers and therapeutic targets for the treatment of AD, a greater comprehension of the molecular processes and the intricate network of interactions in which they are implicated could pave the way for future research.
Collapse
Affiliation(s)
- Rasanpreet Kaur
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Swadha Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India
| | - Saurabh Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, Mathura 281406, Uttar Pradesh, India.
| | - Jitendra Singh
- Department of Translational Medicine, All India Institute of Medical Sciences (AIIMS)Bhopal, Saket Nagar, Bhopal 462020, Madhya Pradesh, India
| |
Collapse
|
13
|
Lan H, Liu W, Zuo C, Chen L, Wang S, Luo C, Kuang W, Kemp GJ, Lui S, Suo X, Gong Q. Heterogeneous brain abnormalities in subjective cognitive decline converge on a common network and their transcriptional signature. Alzheimers Dement 2025; 21:e70073. [PMID: 40145464 PMCID: PMC11948054 DOI: 10.1002/alz.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Subjective cognitive decline (SCD) is increasingly recognized as closely related to future Alzheimer's disease (AD). Numerous neuroimaging findings in SCD are inconsistent. We tested whether the various findings localize to a common brain network. METHODS Using a novel coordinate network mapping approach, we delineated common brain damage networks that were functionally connected to reported neuroimaging findings. We then decoded these common networks using microscale transcriptomic and chemo-architectures and psychological processes. RESULTS We enrolled 45 studies comprising 2453 SCD patients and 3017 healthy controls. The identified SCD networks were largely localized in the somatosensory network (SMN) and default mode network (DMN). Both were robust to perturbations of analyzed parameters and in an independent validation dataset. Neurobiology correlation analyses identified some key biological pathways and neurotransmitters linked to these networks. DISCUSSION Our findings reconcile heterogeneous neuroimaging abnormalities in SCD and provide a richer neurobiological underpinning, which has implications for understanding patients with SCD. HIGHLIGHTS The heterogeneous neuroimaging findings on SCD were reconciled in a coordinate network mapping framework. The SCD-related functional network involves changes in the DMN, while the SCD-related structural network has changes mainly in primary sensory areas. The identified genes in the functional network were predominantly enriched in biological processes related to synaptic structure, calcium ion binding, and cellular metabolism. An ALE meta-analysis was conducted for comparison.
Collapse
Affiliation(s)
- Huan Lan
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Wenxiong Liu
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Chao Zuo
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Li Chen
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Song Wang
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Chunyan Luo
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Weihong Kuang
- Department of PsychiatryWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Graham J Kemp
- Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - Su Lui
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Xueling Suo
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
| | - Qiyong Gong
- Department of RadiologyHuaxi MR Research Center (HMRRC)Institution of Radiology and Medical ImagingWest China Hospital of Sichuan UniversityChengduSichuanChina
- Functional and Molecular lmaging Key Laboratory of Sichuan ProvinceWest China Hospital of Sichuan UniversityChengduSichuanChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Xiamen Key Lab of Psychoradiology and NeuromodulationDepartment of RadiologyWest China Xiamen Hospital of Sichuan UniversityXiamenFujianChina
| |
Collapse
|
14
|
Fułek M, Hachiya N, Gachowska M, Beszłej JA, Bartoszewska E, Kurpas D, Kurpiński T, Adamska H, Poręba R, Urban S, Fułek K, Leszek J. Cellular Prion Protein and Amyloid-β Oligomers in Alzheimer's Disease-Are There Connections? Int J Mol Sci 2025; 26:2097. [PMID: 40076721 PMCID: PMC11900156 DOI: 10.3390/ijms26052097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Pathological deposits of neurotoxin proteins within the brain, such as amyloid-β and hyperphosphorylated tau tangles, are prominent features in AD. The prion protein (PrP) is involved in neurodegeneration via its conversion from the normal cellular form (PrPC) to the infection prion protein scrapie (PrPSc) form. Some studies indicated that post-translationally modified PrPC isoforms play a fundamental role in AD pathological progression. Several studies have shown that the interaction of Aβ oligomers (Aβos) with the N-terminal residues of the PrPC protein region appears critical for neuronal toxicity. PrPC-Aβ binding always occurs in AD brains and is never detected in non-demented controls, and the binding of Aβ aggregates to PrPC is restricted to the N-terminus of PrPC. In this study, we aimed to gather all of the recent information about the connections between PrPC and AD, with potential clinical implications.
Collapse
Affiliation(s)
- Michał Fułek
- Department and Clinic of Diabetology, Hypertension and Internal Diseases, Institute of Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Naomi Hachiya
- Shonan Research Center, New-STEP Research Center, Central Glass Co., Ltd., Shonan Health Innovation Park 26-1, Muraoka Higashi, Fujisawa 251-8555, Kanagawa, Japan;
| | - Martyna Gachowska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Jan Aleksander Beszłej
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Donata Kurpas
- Division of Research Methodology, Department of Nursing, Faculty of Nursing and Midwifery, Wroclaw Medical University, 51-618 Wroclaw, Poland;
| | - Tomasz Kurpiński
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Hanna Adamska
- Department of Rheumatology and Internal Medicine, Marciniak Lower Silesian Specialist Hospital, 54-049 Wroclaw, Poland;
| | - Rafał Poręba
- Department of Biological Principles of Physical Activity, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland;
| | - Szymon Urban
- Department of Cardiology, The Copper Health Center, 59-301 Lubin, Poland;
| | - Katarzyna Fułek
- Department and Clinic of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Jerzy Leszek
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
15
|
Guo H, Yang R, Cheng W, Li Q, Du M. An Update of Salivary Biomarkers for the Diagnosis of Alzheimer's Disease. Int J Mol Sci 2025; 26:2059. [PMID: 40076682 PMCID: PMC11900270 DOI: 10.3390/ijms26052059] [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: 01/27/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive cognition and behavior impairments. Diagnosing AD early is important for clinicians to slow down AD progression and preserve brain function. Biomarkers such as tau protein and amyloid-β peptide (Aβ) are used to aid diagnosis as clinical diagnosis often lags. Additionally, biomarkers can be used to monitor AD status and evaluate AD treatment. Clinicians detect these AD biomarkers in the brain using positron emission tomography/computed tomography or in the cerebrospinal fluid using a lumbar puncture. However, these methods are expensive and invasive. In contrast, saliva collection is simple, inexpensive, non-invasive, stress-free, and repeatable. Moreover, damage to the brain parenchyma can impact the oral cavity and some pathogenic molecules could travel back and forth from the brain to the mouth. This has prompted researchers to explore biomarkers in the saliva. Therefore, this study provides an overview of the main finding of salivary biomarkers for AD diagnosis. Based on these available studies, Aβ, tau, cholinesterase enzyme activity, lactoferrin, melatonin, cortisol, proteomics, metabolomics, exosomes, and the microbiome were changed in AD patients' saliva when compared to controls. However, well-designed studies are essential to confirm the reliability and validity of these biomarkers in diagnosing and monitoring AD.
Collapse
Affiliation(s)
| | | | | | | | - Minquan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; (H.G.); (R.Y.); (W.C.); (Q.L.)
| |
Collapse
|
16
|
Aghasizadeh Sherbaf R, Kaposvári GM, Nagy K, Pakáski M, Gajdács M, Matusovits D, Baráth Z. Oral Health Status and Factors Associated with Oral Health in Patients with Alzheimer's Disease: A Matched Case-Control Observational Study. J Clin Med 2025; 14:1412. [PMID: 40094842 PMCID: PMC11900378 DOI: 10.3390/jcm14051412] [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: 01/16/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Alzheimer's disease (AD) is a chronic neurodegenerative disease, ranking as the seventh leading cause of death in both sexes. There is increasing awareness of the role of chronic periodontal disease and severe tooth loss as a modifiable risk factor for developing AD. The aim of the present observational study was to assess AD patients with non-affected healthy controls in the context of their dental and periodontal health outcomes; additionally, the potential impact of anamnestic factors and lifestyle habits on oral health outcomes was also studied. Methods: A total of n = 41 AD patients receiving treatment at the Department of Psychiatry, University of Szeged, were compared with n = 41 age- and gender-matched controls from individuals seeking dental treatment and from retirement homes (mean age was 83.32 ± 7.82 years). Dental and periodontal status indices were assessed according to World Health Organization (WHO) criteria. Results: Overall, 51.2%, 68.3%, and 87.8% of AD patients received mood stabilizers, drugs for their non-cognitive symptoms and cognitive symptoms, respectively. Severe tooth loss was observed in 43.9% of AD patients and 56.1% of controls, respectively. There were no significant differences among AD patients and controls regarding the dental status indices studied (p > 0.05 for all indicators). AD patients had significantly higher plaque indices (%) (59.06 ± 15.45 vs. 41.35 ± 7.97; p < 0.001), bleeding on probing (BOP%) (62.65 ± 12.00 vs. 40.12 ± 10.86; p < 0.001), pocket depth [PD] (2.63 ± 0.56 vs. 2.29 ± 0.13; p = 0.002) and attachment loss [AL] (2.85 ± 0.79 vs. 2.39 ± 0.41; p = 0.026) values, compared to controls. Smoking (vs. non-smokers; 56.28 ± 12.36 vs. 51.40 ± 13.23, p = 0.038) and consumption of alcohol (vs. non-drinkers; 58.68 ± 9.86 vs. 54.78 ± 14.86, p = 0.040) were associated with higher plaque indices [%], while no similar effects were shown for dental status parameters (p > 0.05). In contrast, coffee intake and vitamin supplement use had no significant effect on dental or periodontal status parameters (p > 0.05 in all cases). Conclusions: The results of our study underscore the substantial treatment needs of AD patients, calling for heightened awareness among dental healthcare professionals.
Collapse
Affiliation(s)
- Reza Aghasizadeh Sherbaf
- Department of Oral Surgery, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary; (R.A.S.); (G.M.K.); (K.N.); (D.M.)
| | - George Michael Kaposvári
- Department of Oral Surgery, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary; (R.A.S.); (G.M.K.); (K.N.); (D.M.)
| | - Katalin Nagy
- Department of Oral Surgery, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary; (R.A.S.); (G.M.K.); (K.N.); (D.M.)
| | - Magdolna Pakáski
- Department of Psychiatry, Faculty of Medicine, University of Szeged, Korányi fasor 8–10., 6720 Szeged, Hungary;
| | - Márió Gajdács
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary;
| | - Danica Matusovits
- Department of Oral Surgery, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary; (R.A.S.); (G.M.K.); (K.N.); (D.M.)
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 64–66., 6720 Szeged, Hungary
| |
Collapse
|
17
|
Pan N, Liu S, Ge X, Zheng Y. Association of hippocampal atrophy with tau pathology of temporal regions in preclinical Alzheimer's disease. J Alzheimers Dis 2025:13872877251314785. [PMID: 39956951 DOI: 10.1177/13872877251314785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
BACKGROUND Hippocampal atrophy is linked to memory and cognitive deficits, preceding clinical diagnosis of mild cognitive impairment (MCI) by decades. Morphometry changes in the hippocampal formation (HF) and their relationship to tau deposition in non-demented individuals remains unclear. OBJECTIVE To investigate morphometry changes in the HF and their association with tau deposition in a non-demented cohort. METHODS Eighty-three subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) underwent T1-weighted MRI and Tau-PET scans at baseline and longitudinal follow-up. Participants were divided into amyloid-negative (Aβ-) and amyloid-positive (Aβ+) groups. Hippocampal volume/thickness were measured, and associations with tau deposition in temporal regions were examined using multivariable linear regression. RESULTS No significant association was found between the hippocampal volume/thickness and tau deposition of temporal regions for the Aβ- group. For the Aβ+ group, the hippocampal thickness was significantly associated with tau deposition of entorhinal cortex (ERC) for both hemispheres, and temporal pole, inferior temporal, and middle temporal regions for right hippocampi with the longitudinal follow up scans, while no significant association with the baseline scans. It was interesting that there was strong association between the baseline tau deposition of ERC and temporal pole and the longitudinal follow up thickness of left hippocampi, while the associated regions for the right hemisphere were ERC, temporal pole, and inferior temporal regions. CONCLUSIONS Hippocampal atrophy may precede cognitive symptoms, with tau deposition in adjacent temporal regions contributing to hippocampal changes. The right HF appears more vulnerable than the left, indicating hemispheric differences in pathology.
Collapse
Affiliation(s)
- Ningning Pan
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Shujuan Liu
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Xinting Ge
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
18
|
Bay S, Rodina A, Haut F, Roychowdhury T, Argyrousi EK, Staniszewski A, Han K, Sharma S, Chakrabarty S, Digwal CS, Stanisavljevic A, Labuza A, Alldred MJ, Panchal P, SanthaSeela A, Tuffery L, Li Z, Hashmi A, Rosiek E, Chan E, Monetti M, Sasaguri H, Saido TC, Schneider JA, Bennett DA, Fraser PE, Erdjument-Bromage H, Neubert TA, Ginsberg SD, Arancio O, Chiosis G. Systems-Level Interactome Mapping Reveals Actionable Protein Network Dysregulation Across the Alzheimer's Disease Spectrum. RESEARCH SQUARE 2025:rs.3.rs-5930673. [PMID: 39989971 PMCID: PMC11844643 DOI: 10.21203/rs.3.rs-5930673/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) progresses as a continuum, from preclinical stages to late-stage cognitive decline, yet the molecular mechanisms driving this progression remain poorly understood. Here, we provide a systems-level map of protein-protein interaction (PPI) network dysfunction across the AD spectrum and uncover epichaperomes-stable scaffolding platforms formed by chaperones and co-factors-as central drivers of this process. Using over 100 human brain specimens, mouse models, and human neurons, we show that epichaperomes emerge early, even in preclinical AD, and progressively disrupt multiple PPI networks critical for synaptic function and neuroplasticity. Glutamatergic neurons, essential for learning and memory, exhibit heightened vulnerability, with their dysfunction driven by protein sequestration into epichaperome scaffolds, independent of changes in protein expression. Notably, pharmacological disruption of epichaperomes with PU-AD restores PPI network integrity and reverses synaptic and cognitive deficits, directly linking epichaperome-driven network dysfunction to AD pathology. These findings establish epichaperomes as key mediators of molecular collapse in AD and identify network-centric intervention strategies as a promising avenue for disease-modifying therapies.
Collapse
Affiliation(s)
- Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Florence Haut
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elentina K Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Kyung Han
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Souparna Chakrabarty
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Amanda Labuza
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anand SanthaSeela
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Laura Tuffery
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zhuoning Li
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arsalan Hashmi
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Rosiek
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Chan
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mara Monetti
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
19
|
Benson GS, Bartels C, Stamatis F, Belz M, Esselmann H, Frölich L, Hausner L. The Use and Understanding of Mild Cognitive Impairment in Routine Specialist Care: A Survey Among German Memory Clinics. Geriatrics (Basel) 2025; 10:21. [PMID: 39997520 PMCID: PMC11855903 DOI: 10.3390/geriatrics10010021] [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: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 02/26/2025] Open
Abstract
Objectives: Mild cognitive impairment (MCI) is a heterogeneous clinical syndrome and is important for the diagnosis and management of Alzheimer's disease (AD). With the expansion of biomarker-based diagnostics, the aim of this study is to clarify the current attitudes towards and the use of MCI, and MCI due to AD, in German memory clinics. Methods: An online survey (50 items) was performed in 2022 among specialized clinicians (N = 45) in German memory clinics to assess the use of MCI and biomarkers in current diagnosis and treatment. Attitudinal and frequency items were assessed with a five-point numeric scale (strongly disagree = 1 to completely agree = 5 and never = 1 to always = 5, respectively). Results: All respondents used MCI as a clinical diagnosis. The benefits of diagnosing MCI were labeling deficits as disease symptoms (M = 4.4, SD = 0.7), improving coping with symptoms (M = 4.1, SD = 0.9), and motivating risk reduction activities (M = 4.0, SD = 0.9). Overall, 37 respondents used specialized diagnostic criteria for MCI due to AD, and all had access to biomarker diagnostics. Patients with MCI due to AD received more frequent counseling on memory training (p < 0.001), other non-pharmacological treatments (p < 0.001), and antidementive drug treatment (p < 0.001) than patients with MCI of other etiologies. Acetylcholinesterase inhibitors were prescribed significantly more frequently to patients with MCI due to AD (p < 0.001) compared to other MCI patients. Conclusions: MCI is commonly used as a clinical diagnosis in German memory clinics. AD biomarker assessment is well established and influences patient counseling and treatment recommendations.
Collapse
Affiliation(s)
- Gloria S. Benson
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany; (G.S.B.); (F.S.); (L.F.)
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; (C.B.); (M.B.); (H.E.)
| | - Feride Stamatis
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany; (G.S.B.); (F.S.); (L.F.)
| | - Michael Belz
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; (C.B.); (M.B.); (H.E.)
| | - Hermann Esselmann
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; (C.B.); (M.B.); (H.E.)
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany; (G.S.B.); (F.S.); (L.F.)
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J5, 68159 Mannheim, Germany; (G.S.B.); (F.S.); (L.F.)
| |
Collapse
|
20
|
Guillén N, Contador J, Buongiorno M, Álvarez I, Culell N, Alcolea D, Lleó A, Fortea J, Piñol-Ripoll G, Carnes-Vendrell A, Lourdes Ispierto M, Vilas D, Puig-Pijoan A, Fernández-Lebrero A, Balasa M, Sánchez-Valle R, Lladó A. Agreement of cerebrospinal fluid biomarkers and amyloid-PET in a multicenter study. Eur Arch Psychiatry Clin Neurosci 2025; 275:257-266. [PMID: 37898567 PMCID: PMC11799063 DOI: 10.1007/s00406-023-01701-y] [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: 02/13/2023] [Accepted: 10/02/2023] [Indexed: 10/30/2023]
Abstract
Core Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers have shown incomplete agreement with amyloid-positron emission tomography (PET). Our goal was to analyze the agreement between AD CSF biomarkers and amyloid-PET in a multicenter study. Retrospective multicenter study (5 centers). Participants who underwent both CSF biomarkers and amyloid-PET scan within 18 months were included. Clinical diagnoses were made according to latest diagnostic criteria by the attending clinicians. CSF Amyloid Beta1-42 (Aβ1-42, A), phosphorliated tau 181 (pTau181, T) and total tau (tTau, N) biomarkers were considered normal (-) or abnormal ( +) according to cutoffs of each center. Amyloid-PET was visually classified as positive/negative. Agreement between CSF biomarkers and amyloid-PET was analyzed by overall percent agreement (OPA). 236 participants were included (mean age 67.9 years (SD 9.1), MMSE score 24.5 (SD 4.1)). Diagnoses were mild cognitive impairment or dementia due to AD (49%), Lewy body dementia (22%), frontotemporal dementia (10%) and others (19%). Mean time between tests was 5.1 months (SD 4.1). OPA between single CSF biomarkers and amyloid-PET was 74% for Aβ1-42, 75% for pTau181, 73% for tTau. The use of biomarker ratios improved OPA: 87% for Aβ1-42/Aβ1-40 (n = 155), 88% for pTau181/Aβ1-42 (n = 94) and 82% for tTau/Aβ1-42 (n = 160). A + T + N + cases showed the highest agreement between CSF biomarkers and amyloid-PET (96%), followed by A-T-N- cases (89%). Aβ1-42/Aβ1-40 was a better marker of cerebral amyloid deposition, as identified by amyloid tracers, than Aβ1-42 alone. Combined biomarkers in CSF predicted amyloid-PET result better than single biomarkers.
Collapse
Affiliation(s)
- Núria Guillén
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - Mariateresa Buongiorno
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Ignacio Álvarez
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Natalia Culell
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Gerard Piñol-Ripoll
- Clinical Neuroscience Research, Unitat Trastorns Cognitius, IRBLleida, Santa Maria University Hospital, Lleida, Spain
| | - Anna Carnes-Vendrell
- Clinical Neuroscience Research, Unitat Trastorns Cognitius, IRBLleida, Santa Maria University Hospital, Lleida, Spain
| | - María Lourdes Ispierto
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Badalona, Spain
| | - Dolores Vilas
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Badalona, Spain
| | - Albert Puig-Pijoan
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Aida Fernández-Lebrero
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
- Institute of Neurosciences, Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain.
- Institute of Neurosciences, Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
21
|
Cantu PA, Al Snih S, Markides K, Raji M. Neuropsychiatric Symptoms and Mortality in Cognitively Normal Older Mexican Americans. HISPANIC JOURNAL OF BEHAVIORAL SCIENCES 2025; 47:49-67. [PMID: 40308647 PMCID: PMC12040322 DOI: 10.1177/07399863241310476] [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] [Indexed: 05/02/2025]
Abstract
Neuropsychiatric symptoms (NPS) present in older adults with Alzheimer's disease (AD) and other dementias are related to mortality. Research on the relationship between NPS and mortality in a non-dementia population is limited. This study examines NPS as a predictor of six-year mortality among community dwelling Mexican Americans aged 80 years and older. Data included 466 cognitively normal participants from Wave 7 of the Hispanic Established Population for the Epidemiological Study of Elderly. NPS were measured using the Neuropsychiatric inventory (NPI). Cox proportional hazard models were used to estimate the hazard ratio (HR) of mortality. The HR of death at 6 years was 1.02 (95% Confidence Interval-CI [1.00, 1.04]) as a function of NPI score and 1.09 (95% CI [1.02, 1.17]) for the number of NPI conditions, controlling for demographic and health characteristics. Apathy, irritability, and aberrant motor behavior were all independently predictors of mortality. NPS may be modifiable risk factors to increase survival time or may be indicative of underlying health problems. NPS may be related to underlying health conditions among older adults with normal cognitive functioning.
Collapse
Affiliation(s)
| | | | | | - Mukaila Raji
- University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
22
|
Petkus AJ, Sonti AN, Montoya L, Rowe B, Sagare A, Ringman JM. Sequence of decline on the NIH-toolbox cognitive battery in a predominantly Latino sample with autosomal dominant Alzheimer's disease. J Alzheimers Dis 2025; 103:1150-1160. [PMID: 39865680 DOI: 10.1177/13872877241312934] [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: 01/28/2025]
Abstract
BACKGROUND Understanding the sequential progression of cognitive decline in autosomal dominant Alzheimer's disease (ADAD) in the Latino population is crucial for enhancing early identification for targeted interventions. Given the tablet-based administration and increasing frequency of use in epidemiological research, validating this progression within the NIH Toolbox cognitive battery (NIHTB-CB) is important. OBJECTIVE The first aim was to utilize an innovative Event-Based Modeling (EBM) analytic approach to estimate the sequence of cognitive declines in persons at risk for ADAD enriched for being of Latino origin. The second aim was to examine associations between EBM-derived estimates of cognitive disease severity and independent cognitive outcomes within carriers and noncarriers. METHODS This cross-sectional observational study (N = 30) included 16 ADAD mutation carriers and 14 noncarriers who completed the NIHTB-CB in their primary language (n = 8 Spanish; n = 22 English). An EBM was constructed to compare ADAD mutation carriers and noncarriers on NIHTB-CB performance. We utilized linear regression to examine the associations between the EBM-derived cognitive-decline disease stage and independent outcomes (e.g., performance on the Cognitive Abilities Screening Instrument (CASI) and estimated years to dementia diagnosis). RESULTS The EBM estimated that tests assessing episodic memory were the first to become abnormal in the sequence of ADAD-related cognitive decline. Each higher estimated cognitive-decline disease stage was associated with approximately a three-point decline in the CASI and two years closer to dementia diagnosis. CONCLUSIONS Findings support the EBM applied to the tablet-based NIHTB-CB to estimate the likely progression of cognitive decline in Latinos with ADAD.
Collapse
Affiliation(s)
- Andrew J Petkus
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anup N Sonti
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lucy Montoya
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bryan Rowe
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay Sagare
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
23
|
Zilioli A, Pancaldi B, Baumeister H, Busi G, Misirocchi F, Mutti C, Florindo I, Morelli N, Mohanty R, Berron D, Westman E, Spallazzi M. Unveiling the hippocampal subfield changes across the Alzheimer's disease continuum: a systematic review of neuroimaging studies. Brain Imaging Behav 2025; 19:253-267. [PMID: 39443362 DOI: 10.1007/s11682-024-00952-0] [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] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Studies exploring the hippocampal subfield atrophy in Alzheimer's disease (AD) have shown contradictory results. This review aims to disentangle such heterogeneity by investigating the dynamic changes of hippocampal subfields across the AD continuum. We systematically searched the PubMed and EMBASE databases for case-control studies. Selected studies included investigations of biomarker-based amyloid status and reported data on hippocampal subfield atrophy using advanced MRI techniques. Twelve studies were included. Despite high heterogeneity, a distinguishable pattern of vulnerability of hippocampal subfields can be recognized from the cognitively unimpaired phase to the dementia stage, shedding light on hippocampal changes with disease progression. Consistent findings revealed atrophy in the subiculum and presubiculum, along with a potential increase in volume in the cornu ammonis (CA) among the cognitively unimpaired group, a feature not observed in patients experiencing subjective cognitive decline. Atrophy in the subiculum, presubiculum, CA 1-4, and the dentate gyrus characterized the mild cognitive impairment stage, with a more pronounced severity in the progression to dementia.
Collapse
Affiliation(s)
- Alessandro Zilioli
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Beatrice Pancaldi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Hannah Baumeister
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Gabriele Busi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Francesco Misirocchi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Carlotta Mutti
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
- Sleep Disorders Center, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Irene Florindo
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
| | - Nicola Morelli
- Department of Neurology, G. da Saliceto Hospital, Piacenza, Italy
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
| | - David Berron
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
- Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marco Spallazzi
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
| |
Collapse
|
24
|
Du W, Yu S, Liu R, Kong Q, Hao X, Liu Y. Precision Prediction of Alzheimer's Disease: Integrating Mitochondrial Energy Metabolism and Immunological Insights. J Mol Neurosci 2025; 75:5. [PMID: 39806062 DOI: 10.1007/s12031-024-02291-7] [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: 09/18/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD), a prevalent neurodegenerative disorder, is characterized by mitochondrial dysfunction and immune dysregulation. This study is aimed at developing a risk prediction model for AD by integrating multi-omics data and exploring the interplay between mitochondrial energy metabolism-related genes (MEMRGs) and immune cell dynamics. We integrated four GEO datasets (GSE132903, GSE29378, GSE33000, GSE5281) for differential gene expression analysis, functional enrichment, and weighted gene co-expression network analysis (WGCNA). We identified two key gene modules (turquoise and magenta) significantly correlated with AD. Subsequently, we constructed a risk prediction model incorporating five MEMRGs (MRPL15, RBP4, ABCA1, MPV17, and MRPL37) and clinical factors using LASSO regression. The model demonstrated robust predictive performance (AUC > 0.815) in both internal and external validation (GSE44770) cohorts. Downregulation of MRPL15, RBP4, MPV17, and MRPL37 in AD brain regions (validated using AlzData and qRT-PCR) suggests impaired mitochondrial function. Conversely, ABCA1 upregulation may represent a compensatory response. Furthermore, significant differences in immune cell proportions, particularly gamma delta T cells (p = 0.002) and activated CD4 memory T cells (p = 0.027), were found between AD and non-demented samples. We observed significant correlations between MEMRG expression and specific immune cell fractions, indicating a potential link between mitochondrial dysfunction and immune dysregulation in AD. Our study provides a reliable risk prediction model for AD and highlights the crucial roles of MEMRGs and immune responses in disease pathogenesis, offering potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Wenlong Du
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Shihui Yu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ruiyao Liu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Qingqing Kong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xin Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yi Liu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
25
|
Kang JM, Manjavong M, Jin C, Diaz A, Ashford MT, Eichenbaum J, Thorp E, Wragg E, Zavitz KH, Cormack F, Aaronson A, Mackin RS, Tank R, Landavazo B, Cavallone E, Truran D, Farias ST, Weiner MW, Nosheny RL. Subjective cognitive decline predicts longitudinal neuropsychological test performance in an unsupervised online setting in the Brain Health Registry. Alzheimers Res Ther 2025; 17:10. [PMID: 39773247 PMCID: PMC11706033 DOI: 10.1186/s13195-024-01641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUNDS Digital, online assessments are efficient means to detect early cognitive decline, but few studies have investigated the relationship between remotely collected subjective cognitive change and cognitive decline. We hypothesized that the Everyday Cognition Scale (ECog), a subjective change measure, predicts longitudinal change in cognition in the Brain Health Registry (BHR), an online registry for neuroscience research. METHODS This study included BHR participants aged 55 + who completed both the baseline ECog and repeated administrations of the CANTAB® Paired Associates Learning (PAL) visual learning and memory test. Both self-reported ECog (Self-ECog) and study partner-reported ECog (SP-ECog), and two PAL scores (first attempt memory score [FAMS] and total errors adjusted [TEA]) were assessed. We estimated associations between multiple ECog scoring outputs (ECog positive [same or above cut-off score], ECog consistent [report of consistent decline in any item], and total score) and longitudinal change in PAL. Additionally we assessed the ability of ECog to identify 'decliners', who exhibited the worst PAL progression slopes corresponding to the fifth percentile and below. RESULTS Participants (n = 16,683) had an average age of 69.07 ± 7.34, 72.04% were female, and had an average of 16.66 ± 2.26 years of education. They were followed for an average of 2.52 ± 1.63 visits over a period of 11.49 ± 11.53 months. Both Self-ECog positive (estimate = -0.01, p < 0.001, R²m = 0.56) and Self-ECog consistent (estimate=-0.01, p = 0.002, R²m = 0.56) were associated with longitudinal change in PAL FAMS after adjusting demographics and clinical confounders. Those who were Self-ECog total (Odds ratio [95% confidence interval] = 1.390 [1.121-1.708]) and SP-ECog consistent (2.417 [1.591-3.655]) had higher probability of being decliners based on PAL FAMS. CONCLUSION In the BHR's unsupervised online setting, baseline subjective change was feasible in predicting longitudinal decline in neuropsychological tests. Online, self-administered measures of subjective cognitive change might have a potential to predict objective subjective change and identify individuals with cognitive impairments.
Collapse
Affiliation(s)
- Jae Myeong Kang
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Manchumad Manjavong
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Division of Geriatric Medicine, Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chengshi Jin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Adam Diaz
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Miriam T Ashford
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
| | - Joseph Eichenbaum
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Francesca Cormack
- Cambridge Cognition, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Anna Aaronson
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - R Scott Mackin
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
| | - Rachana Tank
- Dementia Research Centre, UCL Institute of Neurology, University College London, London, WC1E 6BT, UK
| | - Bernard Landavazo
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Erika Cavallone
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Diana Truran
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | | | - Michael W Weiner
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rachel L Nosheny
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
- VA Advanced Imaging Research Center, San Francisco Veteran's Administration Medical Center, San Francisco, CA, USA.
- Northern California Institute for Research and Education (NCIRE), San Francisco, CA, USA.
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Heydari R, Khosravifar M, Abiri S, Dashtbin S, Alvandi A, Nedaei SE, Salimi Z, Zarei F, Abiri R. A domestic strain of Lactobacillus rhamnosus attenuates cognitive deficit and pro-inflammatory cytokine expression in an animal model of Alzheimer's disease. Behav Brain Res 2025; 476:115277. [PMID: 39343242 DOI: 10.1016/j.bbr.2024.115277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/30/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Microbiome dysbiosis plays a significant role in neuroinflammation and Alzheimer's disease (AD). Therefore, gut microbiome restoration using appropriate probiotics may be beneficial in alleviating AD features. In this study, we investigated the effects of a domestic strain of Lactobacillus rhamnosus (L. rhamnosus) on spatial memory, and cytokines expression in an inflammation-based AD model. METHOD Male Wistar rats were randomly divided into four groups (six animals per group) of control, L. rhamnosus-only, D-galactose (D-gal)-only, and D-gal + L. rhamnosus. Spatial learning and memory were assessed using the Morris water maze test. IL-1β, IL-6, and TNF-α expression levels were measured using Real-Time qPCR. A significance level of 0.05 was used for statistical analysis. RESULTS In contrast to the D-gal + L. rhamnosus-treated group, D-gal only treated group showed impaired memory in MWM test compared to the control group. Additionally, D-gal treatment resulted in an increase in IL-1β and TNF-α levels and a decrease in IL-6 levels, which was not statistically significant. However, the TNF-α level was significantly decreased in D-gal + L. rhamnosus-treated group compared to D-gal-only treated group (P < 0.05). Also, IL-6 level was significantly lower in D-gal + L. rhamnosus-treated group compared to control group (P < 0.05). CONCLUSION These results suggest that the domestic L. rhamnosus might positively impact cognitive deficit and neuroinflammation. Further studies are suggested to investigate the specific mechanisms mediating the effects of L. rhamnosus on cognitive functions and neuroinflammation in animal models of AD.
Collapse
Affiliation(s)
- Ruhollah Heydari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mina Khosravifar
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shervin Abiri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhoushang Alvandi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Ershad Nedaei
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Salimi
- Department of Biology, Faculty of Science, University of Qom, Qom, Iran
| | - Fatemeh Zarei
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Ramin Abiri
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
27
|
Zeng T, Zhang C, Sun L, Xu H. Water-Soluble Ginseng Oligosaccharides Prevent Scopolamine-Induced Cholinergic Dysfunction and Inflammatory Cytokine Overexpression. Cell Biochem Biophys 2025:10.1007/s12013-024-01660-8. [PMID: 39751741 DOI: 10.1007/s12013-024-01660-8] [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] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Cholinergic deficiency and neuroinflammation are the two main factors of Alzheimer's disease. Recent studies have shown that water-soluble ginseng oligosaccharides (WGOS) derived from Panax ginseng roots can protect against scopolamine-induced impairments in learning and memory. However, the fundamental mechanisms remain unclear for the most part. The purpose of this study was to examine the effect of WGOS on cholinergic function and protein levels of proinflammatory cytokines in the hippocampus of mice. Mice were first pretreated with WGOS or saline, and then treated with scopolamine to establish an Alzheimer's disease model. The cognition memory of the mice was assessed through the behavioral test. The effect of WGOS on the cholinergic system was evaluated by measuring acetylcholine (ACh) neurotransmitter concentration and acetylcholinesterase (AChE) activity in the hippocampus. Using ELISA, the inflammatory cytokines IL-1β and TNF-α in the hippocampus were identified. This study found that WGOS treatment prevented the scopolamine-induced impairment of mice's recognition memory, as seen by their enhanced object recognition. In addition, WGOS prevented the scopolamine-induced decrease in ACh concentration and increase in AChE activity. Moreover, WGOS treatment inhibited scopolamine-induced upregulation of the inflammatory proteins IL-1β and TNF-α. These findings suggest that the amelioration of scopolamine-induced cognitive impairment in mice by WGOS was a consequence of the control of cholinergic function and inflammatory response in the hippocampus. Our findings suggest that WGOS should be investigated as a dietary supplement or medication for the treatment of learning and memory disorders in humans.
Collapse
Affiliation(s)
- Ting Zeng
- Department of Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Chengwei Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lili Sun
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China
| | - Haiyan Xu
- Department of Rehabilitation Therapeutics, School of Nursing, Jilin University, Changchun, Jilin, China.
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
28
|
Al Amin M, Dehbia Z, Nafady MH, Zehravi M, Kumar KP, Haque MA, Baig MS, Farhana A, Khan SL, Afroz T, Koula D, Tutone M, Nainu F, Ahmad I, Emran TB. Flavonoids and Alzheimer’s disease: reviewing the evidence for neuroprotective potential. Mol Cell Biochem 2025; 480:43-73. [PMID: 38568359 DOI: 10.1007/s11010-023-04922-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2025]
|
29
|
Kozin MS, Kulakova OG, Kiselev IS, Semina EV, Kakotkin VV, Agapov MA, Favorova OO. Mitochondrial Genome Variants and Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S146-S163. [PMID: 40164157 DOI: 10.1134/s0006297924603174] [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: 02/22/2024] [Revised: 03/04/2024] [Accepted: 05/14/2024] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD), a severe neurodegenerative disease of the central nervous system, is the most common cause of cognitive impairment in people over the age of 60. The etiology and pathogenesis of Alzheimer's disease are still unclear despite decades of active research. Numerous studies have shown that neurodegenerative processes in AD are associated with the mitochondrial dysfunction. In this review, we briefly discuss the results of these studies and present the reported evidence that mitochondrial dysfunction in AD is associated with mitochondrial DNA (mtDNA) variations. The results of association analysis of mtDNA haplogroups and individual polymorphic variants, including those whose combinations define haplogroups, with AD are described in detail. These data clearly indicate the role of variations in the mitochondrial genome in the susceptibility to AD, although the problem of significance of individual mtDNA variants is far from being resolved.
Collapse
Affiliation(s)
- Maxim S Kozin
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia.
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Olga G Kulakova
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Ivan S Kiselev
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | | | - Viktor V Kakotkin
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
| | - Mikhail A Agapov
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
| | - Olga O Favorova
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| |
Collapse
|
30
|
Zhang L, An H, Zhen R, Zhang T, Ding M, Zhang M, Sun Y, Gu C. Di Huang Yi Zhi Fang improves cognitive function in APP/PS1 mice by inducing neuronal mitochondrial autophagy through the PINK1-parkin pathway. J Alzheimers Dis 2025; 103:372-382. [PMID: 39873362 DOI: 10.1177/13872877241299832] [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: 01/30/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is an irreversible age-related neurodegenerative condition characterized by the deposition of amyloid-β (Aβ) peptides and neurofibrillary tangles. Di Huang Yi Zhi (DHYZ) formula, a traditional Chinese herbal compound comprising several prescriptions, demonstrates properties that improve cognitive abilities in clinical. Nonetheless, its molecular mechanisms on treating AD through improving neuron cells mitochondria function have not been deeply investigated. OBJECTIVE This study administered DHYZ to APP/PS1 mice to explore its potential therapeutic mechanisms in AD treatment. METHODS APP/PS1 transgenic mice were given DHYZ (L, M, H), donepezil, or distilled water for a consecutive 12-week period. The Morris water maze test was used to assess memory capacity, transmission electron microscopy was used to observe mitochondrial and synaptic structures, immunohistochemistry and western blot detected proteins involved in the mitochondrial autophagy pathway, ELISA measured serum Aβ content, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assessed neuronal cell apoptosis. RESULTS DHYZ demonstrates a notable therapeutic impact on mice with AD, effectively improving cognitive and memory impairments. DHYZ decreases Aβ accumulation in the hippocampus by reducing BACE1 activity and enhancing Aβ clearance through the blood-brain barrier. Additionally, DHYZ significantly suppresses neuronal apoptosis, enhances synaptic structure, and increases synapse numbers, processes strongly linked to the activation of mitochondrial PINK1-Parkin autophagy. CONCLUSIONS DHYZ enhances cognitive function in APP/PS1 mice by stimulating neuronal mitochondrial autophagy through the PINK1-Parkin pathway.
Collapse
Affiliation(s)
- Limin Zhang
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongmei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rongrong Zhen
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Minrui Ding
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengxue Zhang
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiguo Sun
- Department of General Internal Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Gu
- Department of General Internal Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Zhang Y, Sun K, Liu Y, Xie F, Guo Q, Shen D. A Modality-Flexible Framework for Alzheimer's Disease Diagnosis Following Clinical Routine. IEEE J Biomed Health Inform 2025; 29:535-546. [PMID: 39352829 DOI: 10.1109/jbhi.2024.3472011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Dementia has high incidence among the elderly, and Alzheimer's disease (AD) is the most common dementia. The procedure of AD diagnosis in clinics usually follows a standard routine consisting of different phases, from acquiring non-imaging tabular data in the screening phase to MR imaging and ultimately to PET imaging. Most of the existing AD diagnosis studies are dedicated to a specific phase using either single or multi-modal data. In this paper, we introduce a modality-flexible classification framework, which is applicable for different AD diagnosis phases following the clinical routine. Specifically, our framework consists of three branches corresponding to three diagnosis phases: 1) a tabular branch using only tabular data for screening phase, 2) an MRI branch using both MRI and tabular data for uncertain cases in screening phase, and 3) ultimately a PET branch for the challenging cases using all the modalities including PET, MRI, and tabular data. To achieve effective fusion of imaging and non-imaging modalities, we introduce an image-tabular transformer block to adaptively scale and shift the image and tabular features according to modality importance determined by the network. The proposed framework is extensively validated on four cohorts containing 6495 subjects. Experiments demonstrate that our framework achieves superior diagnostic performance than the other representative methods across various AD diagnosis tasks, and shows promising performance for all the diagnosis phases, which exhibits great potential for clinical application.
Collapse
|
32
|
Luo L, Pan Y, Chen F, Zhang Z. Exploring the potential mechanism of Polygonatum sibiricum for Alzheimer's disease based on network pharmacology and molecular docking: An observational study. Medicine (Baltimore) 2024; 103:e40726. [PMID: 39969345 PMCID: PMC11688029 DOI: 10.1097/md.0000000000040726] [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: 05/15/2024] [Accepted: 11/08/2024] [Indexed: 02/20/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, and there have been no systematic studies of Polygonatum against Alzheimer's disease. Therefore, our study will elucidate the mechanism of Polygonatum against AD based on network pharmacology and molecular docking. The active ingredients and corresponding targets of Polygonatum were identified using the traditional Chinese medicine systematic pharmacology database and analysis platform. Disease targets of AD were retrieved from the therapeutic target database, Online Mendelian Inheritance in Man, GeneCards, and Disgenet databases. Using the STRING database, we constructed protein interaction networks and performed gene ontology functional enrichment analysis as well as Kyoto encyclopedia of genes and genomes pathway enrichment analysis on common targets. We then drew drug-component-target-pathway-disease network maps using Cytoscape 3.10.1 software and validated the molecular docking using AutoDock4. A total of 10 active ingredients and 108 common targets were screened from Polygonatum, 29 genes (including AKT1 and STAT3) were identified as core genes. According to gene ontology analysis, the core targets were found to be mainly involved in signal transduction, positive regulation of gene expression, negative regulation of the apoptotic process, and so on. The Kyoto encyclopedia of genes and genomes analysis revealed that the signaling pathways comprised pathways in cancer, pathways of neurodegeneration - multiple diseases, and PI3K-Akt signaling pathway. The molecular docking results indicated that 10 of active ingredients from Polygonatum exhibited strong binding affinity with the 6 core targets that were screened before. The activity of Polygonatum against AD could be attributed to the regulation of multiple biological effects via multi-pathways (pathways in cancer, pathways of neurodegeneration - multiple diseases, and PI3K-Akt signaling pathway). The binding activities were estimated as good level by molecular docking. These discoveries disclosed the multi-component, multi-target, and multi-pathway characteristics of Polygonatum against AD, providing a new strategy for such medical problem.
Collapse
Affiliation(s)
- Liangliang Luo
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yao Pan
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| | - Fang Chen
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| | - Zhihong Zhang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| |
Collapse
|
33
|
Vicente-Alba J, Gutiérrez-Botella J, García-Mahía C, Mateos R. Needs assessment of people living with cognitive impairment/dementia, a requirement of comprehensive psychogeriatric assessment and person-centered care. Empirical validation of the model in a community study. Front Psychiatry 2024; 15:1481898. [PMID: 39758450 PMCID: PMC11697431 DOI: 10.3389/fpsyt.2024.1481898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Cognitive impairment and dementia are part of a continuum that progressively leads to functional impairment and dependency. Dementia is a paradigmatic example of chronic and complex psychogeriatric diseases, requiring a comprehensive assessment. The authors underline the importance of implementing a formal assessment of needs (whether met or unmet) as an essential element of comprehensive assessment. The aim of this paper is to empirically validate this model of approach towards dementia, the needs assessment, demonstrating the relationship between needs and functionality/dependency in people with cognitive impairment/dementia in the community. Material and methods Community-based, cross-sectional, descriptive epidemiological study based on the reanalysis of data from a two-phase community epidemiological study conducted in Santiago de Compostela, Spain, of 800 people over 65 years of age. The present study reanalyzes a subsample of 368 people, including those with dementia/cognitive impairment. The comprehensive assessment of the sample included sociodemographic variables, the presence of chronic diseases, health self-perception, assessment of affective, cognitive and functional state, as well as the needs assessment. The main instruments used were the MMSE, the Geriatric Depression Scale (GDS), the Barthel and Katz Indices, the Lawton Scale, and, for the needs assessment, the Camberwell Assessment of Need for the Elderly (CANE). For the clinical diagnosis of dementia, the ICD-10 criteria were followed. Statistical analysis: A study of the association between variables was carried out through hypothesis testing and a multivariate study was performed using regression models to analyze the relationship between the different variables defining disability/dependency, other health conditions and sociodemographic variables and the MMSE score as an expression of the cognitive impairment/dementia continuum. Results People with cognitive impairment/dementia had a higher number of needs compared to the healthy population. The severity of cognitive impairment is a significant predictor of dependency in Basic Activities of Daily Living (BADL) and is also a predictor of a greater number of needs (both met and unmet). Discussion The present study provides empirical evidence of the importance of implementing scales to assess the needs of people with cognitive impairment, as part of the process of comprehensive biopsychosocial assessment and person-centered care for dementia.
Collapse
Affiliation(s)
- Javier Vicente-Alba
- Psychiatry Service, Vigo’s Health Area, Vigo, Pontevedra, Spain
- Psychiatry Department, University of Santiago de Compostela (USC), Santiago de Compostela, A Coruña, Spain
| | - Jesús Gutiérrez-Botella
- Biostatech Advice Training and Innovation in Biostatistics Limited Society (SL) (USC), Santiago de Compostela, A Coruña, Spain
| | - Carmen García-Mahía
- Psychiatry Department, University of Santiago de Compostela (USC), Santiago de Compostela, A Coruña, Spain
- Psychiatry Service, Coruña’s Health Area, A Coruña, Spain
| | - Raimundo Mateos
- Psychiatry Department, University of Santiago de Compostela (USC), Santiago de Compostela, A Coruña, Spain
- Psychogeriatric Unit, Psychiatry Service, University Complex Hospital of Santiago de Compostela (CHUS) University Hospital, Santiago de Compostela, Spain
| |
Collapse
|
34
|
Gutiérrez-Jiménez E, Rasmussen PM, Mikkelsen IK, Kura S, Fruekilde SK, Hansen B, Bordoni L, Carlsen J, Palmfeldt J, Boas DA, Sakadžić S, Vinogradov S, Khatib ME, Ramos-Cejudo J, Wied B, Leduc-Galindo D, Canepa E, Mar AC, Gamallo-Lana B, Fossati S, Østergaard L. Carbonic anhydrase inhibitors prevent presymptomatic capillary flow disturbances in a model of cerebral amyloidosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609091. [PMID: 39229198 PMCID: PMC11370441 DOI: 10.1101/2024.08.22.609091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Disturbances in microvascular flow dynamics are hypothesized to precede the symptomatic phase of Alzheimer's disease (AD). However, evidence in presymptomatic AD remains elusive, underscoring the need for therapies targeting these early vascular changes. METHODS We employed a multimodal approach, combining in vivo optical imaging, molecular techniques, and ex vivo MRI, to investigate early capillary dysfunction in Tg-SwDI mice without memory impairment. We also assessed the efficacy of carbonic anhydrase inhibitors (CAIs) in preventing capillary flow disturbances. RESULTS Our study revealed capillary flow disturbances associated with alterations in capillary morphology, adhesion molecule expression, and Amyloid-β (Aβ) load in 9-10-month-old Tg-SwDI mice without memory impairment. CAI treatment ameliorated these capillary flow disturbances, enhanced oxygen availability, and reduced Aβ load. DISCUSSION These findings underscore the importance of capillary flow disturbances as early biomarkers in presymptomatic AD and highlight the potential of CAIs for preserving vascular integrity in the early stages of AD.
Collapse
|
35
|
Zhai Y, Li N, Zhang Y, Li H, Wu L, Wei C, Ji J, Zheng D. Identification of JAZF1, KNOP1, and PLEKHA1 as causally associated genes and drug targets for Alzheimer's disease: a summary data-based Mendelian randomization study. Inflammopharmacology 2024; 32:3913-3923. [PMID: 39455528 DOI: 10.1007/s10787-024-01583-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: 08/28/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND There is a growing body of evidence indicating the significant role of the immune system and immune cells in the progression of Alzheimer's disease (AD). However, the exact role of genes from various immune cell types in AD remains unclear. We aimed to utilize summary data-based Mendelian randomization (SMR) to explore the potential causal relationships between genes in specific immune cells and the risk of AD. METHODS By utilizing data sets of expression quantitative trait loci (eQTL) for 14 different immune cell types and large-scale AD genome-wide association study (GWAS), we employed SMR to identify key genes associated with AD within specific immune cells. Sensitivity analyses, including F-statistic, colocalization, and assessment of horizontal pleiotropy, were further conducted to validate the discovered genes. In addition, replication analyses were performed in AD GWAS from the FinnGen consortium. Finally, we further identified existing drugs that target or interact with the druggable genes and reviewed the studies about the associations between these drugs and AD. RESULTS SMR analysis revealed 342 genes associated with AD across 14 immune cell types. Further sensitivity analyses identified nine genes, CTSH, FCER1G, FNBP4, HLA-E, JAZF1, KNOP1, PLEKHA1, RP11-960L18.1, and ZNF638 that had significant associations with AD across nine specific immune cell types. JAZF1, KNOP1 and PLEKHA1 were replicated in an independent analysis using the GWAS data. The review on gene-related drugs also supported these findings. CONCLUSIONS Our research suggests that the expression of the genes JAZF1, KNOP1, and PLEKHA1 in specific immune cell types is related to the risk of AD.
Collapse
Affiliation(s)
- Yuhan Zhai
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Ning Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yujie Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Haibin Li
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lijuan Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Cuibai Wei
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jianguang Ji
- Faculty of Health Science, University of Macau, Taipa, Macao SAR, China.
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| |
Collapse
|
36
|
Yakubu A, Adedeji I, Maduka OC, Jegede A, Adebamowo C. Ethical issues in genomics research in persons with Alzheimer's Disease/Alzheimer's Disease-related dementia (AD/ADRD): a systematic review. BMC Med Ethics 2024; 25:138. [PMID: 39587628 PMCID: PMC11587778 DOI: 10.1186/s12910-024-01141-w] [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: 06/07/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
INTRODUCTION Given the growing number of Alzheimer's Disease and Alzheimer's Disease Related Dementias (AD/ADRD) genomics research projects and the vulnerabilities of study participants, it is critical to evaluate the literature on the ethical challenges in such studies to ensure high ethical standards. METHODS We conducted a systematic review of the literature on ethical issues in AD/ADRD genomics research. We searched Embase, PsycINFO, CiNAHL, Scopus, and Ovid Medline for empirical and normative papers published in peer-reviewed journals on the ethical issues involved in conducting genomics research among persons with AD/ADRD. We used ethical principles from an existing framework as a priori codes to categorize the ethical issues and adapted another framework of Dementia Research Ethical Issues (DREI) as subcategories for our synthesis. We used the 2021 PRISMA guidelines to guide our study. RESULTS We screened 5,509 papers and included 27 of these papers in the systematic review after deduplication, title, and full-text review. The papers contained 109 ethical issues that were mapped against 42 out of 75 relevant DREIs. The highest number of DREIs were mapped to "respect for persons and communities", "favorable risk-benefit ratio", "informed consent" and "scientific validity". The least mapped principles to the DREIs were "fair participant selection", "independent review", "social value", and "collaborative partnership". CONCLUSION Our review showed that there is a dearth of literature on the ethical principles of "fair participant selection", "independent review", "social value" and "collaborative partnership" in genomics research on AD/ADRDs. It is difficult to draw firm conclusions from the distribution of attention paid to specific principles because these may only reflect the concerns of AD/ADRD genomics research ethicists in high-income countries. There is need for more research on the ethics of AD/ADRD genomics research in low and middle-income countries for a more balanced account of the important ethical considerations in this field.
Collapse
Affiliation(s)
- Aminu Yakubu
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Bioethics, Faculty of Multi-Disciplinary Studies, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Isaac Adedeji
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Sociology, Olabisi Onabanjo University, Ago-Iwoye, Ogun State, Nigeria
| | - Oluchi C Maduka
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Bioethics, Faculty of Multi-Disciplinary Studies, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Ayodele Jegede
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Sociology, Faculty of the Social Sciences, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Clement Adebamowo
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria.
- Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
37
|
Reshma A, Subramanian A, Kumarasamy V, Tamilanban T, Sekar M, Gan S, Subramaniyan V, Wong L, Rani N, Wu Y. Neurocognitive effects of proanthocyanidin in Alzheimer's disease: a systematic review of preclinical evidence. Braz J Med Biol Res 2024; 57:e13587. [PMID: 39504064 PMCID: PMC11540257 DOI: 10.1590/1414-431x2024e13587] [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: 12/04/2023] [Accepted: 09/17/2024] [Indexed: 11/08/2024] Open
Abstract
Cognitive disorders and dementia largely influence individual independence and orientation. Based on the Alzheimer's Disease International (ADI) estimation, approximately 75% of individuals with dementia are undiagnosed. In fact, in some low- and middle-income countries, the percentage is as high as 90%. In this systematic review, which is based on PRISMA guidelines, we aim to identify the mechanism of action of proanthocyanidin. Finding a natural product alternative as a potential nootropic can help increase the number of armamentariums against dementia and other cognitive impairments. In this preclinical research, we determined the effect of proanthocyanidins on Alzheimer's disease (AD) by searching electronic bibliographic databases like Scopus, Proquest, ScienceDirect, PubMed, and Google. There was no imposed time limit. However, the search was limited to only English articles. The review protocol is registered on PROSPERO as CRD42022356301. A population, intervention, control, and outcomes (PICO) technique was utilized for report inclusion, and all reports were assessed for risk of bias by using the SYRCLE's RoB tool. The article's bibliographic information, induction model, type of proanthocyanidins, animal strain/weight/age, and outcome measurements were acquired from ten papers and are reported here. Further analysis was validated and determined for the review. The included studies met the review's inclusion criteria and suggested that proanthocyanidins have a neurocognitive effect against AD. Additionally, the effectiveness of proanthocyanidins in reducing oxidative stress, acetylcholinesterase activity, amyloid beta, its efficacy in alleviating superoxide dismutase, cognitive properties, and in facilitating cholinergic transmission in various models of AD has been collectively observed in ten studies.
Collapse
Affiliation(s)
- A. Reshma
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
| | - A. Subramanian
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
| | - V. Kumarasamy
- Department of Parasitology & Medical Entomology, Faculty of
Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur,
Malaysia
| | - T. Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
- Department of Occupational Safety and Health, Faculty of Public
Health, Universitas Airlangga, Surabaya, Indonesia
- Faculty of Health and Life Sciences, INTI International
University, Nilai, Malaysia
- Department of Pharmacology, Faculty of Medicine, MAHSA
University, Bandar Saujana Putra, Selangor, Malaysia
| | - M. Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway,
Selangor, Malaysia
| | - S.H. Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway,
Selangor, Malaysia
| | - V. Subramaniyan
- Department of Medical Sciences, School of Medical and Life
Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - L.S. Wong
- Faculty of Health and Life Sciences, INTI International
University, Nilai, Malaysia
| | - N.N.I.M. Rani
- Faculty of Pharmacy and Health Sciences, Royal College of
Medicine Perak, Universiti Kuala Lumpur, Perak, Malaysia
| | - Y.S. Wu
- Sunway Microbiome Centre & Department of Biological
Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya,
Selangor, Malaysia
| |
Collapse
|
38
|
Galluzzi S, Marizzoni M, Gatti E, Bonfiglio NS, Cattaneo A, Epifano F, Frisoni GB, Genovese S, Geviti A, Marchetti L, Sgrò G, Solorzano CS, Pievani M, Fiorito S. Citrus supplementation in subjective cognitive decline: results of a 36-week, randomized, placebo-controlled trial. Nutr J 2024; 23:135. [PMID: 39482712 PMCID: PMC11529263 DOI: 10.1186/s12937-024-01039-8] [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: 06/29/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Developing interventions for older adults with subjective cognitive decline (SCD) has the potential to prevent dementia in this at-risk group. Preclinical models indicate that Citrus-derived phytochemicals could benefit cognition and inflammatory processes, but results from clinical trials are still preliminary. The aim of this study is to determine the effects of long-term supplementation with Citrus peel extract on cognitive performance and inflammation in individuals with SCD. METHODS Eighty participants were randomly assigned to active treatment (400 mg of Citrus peel extract containing 3.0 mg of naringenin and 0.1 mg of auraptene) or placebo at 1:1 ratio for 36 weeks. The primary endpoint was the change in the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) total score across the 36-week trial period. Other cognitive outcomes included tests and scales evaluating verbal memory, attention, executive and visuospatial functions, and memory concerns. The secondary endpoint was the change of interleukin-8 (IL-8) levels over the 36-week trial period in a subsample of 60 consecutive participants. An Intention-to-treat approach with generalized linear mixed models was used for data analysis. RESULTS The RBANS total score showed significant improvement in both Citrus peel extract and placebo groups at 36 weeks (p for time < .001, d = 0.36, p time x treatment = .910). Significant time effects were also found in cognitive domains of short- and long-term verbal memory (p < .001) and scales of subjective memory (p < .01), with no significant time x treatment interaction. The largest effect sizes were observed in verbal memory in the placebo group (d = 0.69 in short-term, and d = 0.78 in long-term verbal memory). Increased IL-8 levels were found at 36-week follow-up in both Citrus peel extract and placebo groups (p for time = .010, d = 0.21, p time x treatment = .772). Adverse events were balanced between groups. CONCLUSIONS In this randomized clinical trial, long-term Citrus peel extract supplementation did not show cognitive benefits over placebo in participants with SCD, possibly due to high placebo response. These findings might have specific implications for designing future nutraceutical trials in individuals experiencing SCD. TRIAL REGISTRATION The trial has been registered at the United States National Library of Medicine at the National Institutes of Health Registry of Clinical Trials under the code NCT04744922 on February 9th, 2021 ( https://www. CLINICALTRIALS gov/ct2/show/NCT04744922 ).
Collapse
Affiliation(s)
- Samantha Galluzzi
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy.
| | - Elena Gatti
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | | | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesco Epifano
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giovanni B Frisoni
- Memory Center, Geneva University and University Hospitals, Geneva, Switzerland
| | - Salvatore Genovese
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Andrea Geviti
- Service of Statistics, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Lorenzo Marchetti
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giovanni Sgrò
- Clinical Trial Service, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Claudio Singh Solorzano
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Michela Pievani
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Serena Fiorito
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
39
|
Daly T, Mastroleo I. Reducing confusion surrounding expert conceptions of Alzheimer's and dementia: A practical analysis. J Neuropsychol 2024. [PMID: 39450469 DOI: 10.1111/jnp.12398] [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: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
Biological, clinicobiological and clinical conceptions of Alzheimer's disease and related dementias are being promoted simultaneously to different practical ends. The co-existence of contemporary conceptions and the 'scary label' associated with older diagnostic criteria create the possibility of misunderstanding and harm. In this comment, we argue in favour of socio-ethical interventions targeted to health workers and the general public so as to lower the uncertainties introduced by contemporary diagnostic criteria and to articulate how they relate to established criteria.
Collapse
Affiliation(s)
- Timothy Daly
- Bordeaux Population Health CNRS UMR 1219, University of Bordeaux and INSERM, Bordeaux, France
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
| | - Ignacio Mastroleo
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
- National Council of Science and Technology (CONICET), Buenos Aires, Argentina
| |
Collapse
|
40
|
Andrade K, Pacella V. The unique role of anosognosia in the clinical progression of Alzheimer's disease: a disorder-network perspective. Commun Biol 2024; 7:1384. [PMID: 39448784 PMCID: PMC11502706 DOI: 10.1038/s42003-024-07076-7] [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: 06/20/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Alzheimer's disease (AD) encompasses a long continuum from a preclinical phase, characterized by neuropathological alterations albeit normal cognition, to a symptomatic phase, marked by its clinical manifestations. Yet, the neural mechanisms responsible for cognitive decline in AD patients remain poorly understood. Here, we posit that anosognosia, emerging from an error-monitoring failure due to early amyloid-β deposits in the posterior cingulate cortex, plays a causal role in the clinical progression of AD by preventing patients from being aware of their deficits and implementing strategies to cope with their difficulties, thus fostering a vicious circle of cognitive decline.
Collapse
Affiliation(s)
- Katia Andrade
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne University, Pitié-Salpêtrière Hospital, 75013, Paris, France.
- FrontLab, Paris Brain Institute (Institut du Cerveau, ICM), AP-HP, Pitié-Salpêtrière Hospital, 75013, Paris, France.
| | - Valentina Pacella
- IUSS Cognitive Neuroscience (ICON) Center, Scuola Universitaria Superiore IUSS, Pavia, 27100, Italy
- Brain Connectivity and Behaviour Laboratory, Paris, France
| |
Collapse
|
41
|
Hayashi M, Kudo C, Hanamoto H, Maegawa H, Usami N, Niwa H. Effects of hippocampal damage on pain perception in a rat model of Alzheimer's disease induced by amyloid-β and ibotenic acid injection into the hippocampus. Physiol Behav 2024; 285:114652. [PMID: 39096985 DOI: 10.1016/j.physbeh.2024.114652] [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: 03/27/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/05/2024]
Abstract
Patients with Alzheimer's disease (AD) present with a variety of symptoms, including core symptoms as well as behavioral and psychological symptoms. Somatosensory neural systems are generally believed to be relatively unaffected by AD until late in the course of the disease; however, somatosensory perception in patients with AD is not yet well understood. One factor that may complicate the assessment of somatosensory perception in humans centers on individual variations in pathological and psychological backgrounds. It is therefore necessary to evaluate somatosensory perception using animal models with uniform status. In the current study, we focused on the hippocampus, the primary site of AD. We first constructed a rat model of AD model using bilateral hippocampal injections of amyloid-β peptide 1-40 and ibotenic acid; sham rats received saline injections. The Morris water maze test was used to evaluate memory impairment, and the formalin test (1 % or 4 % formalin) and upper lip von Frey test were performed to compare pain perception between AD model and sham rats. Finally, histological and immunohistochemical methods were used to evaluate tissue damage and neuronal activity, respectively, in the hippocampus. AD model rats showed bilateral hippocampal damage and had memory impairment in the Morris water maze test. Furthermore, AD model rats exhibited significantly less pain-related behavior in phase 2 (the last 50 min of the 60-minute observation) of the 4 % formalin test compared with the sham rats. However, no significant changes were observed in the von Frey test. Immunohistochemical observations of the trigeminal spinal subnucleus caudalis after 4 % formalin injection revealed significantly fewer c-Fos-immunoreactive cells in AD model rats than in sham rats, reflecting reduced neuronal activity. These results indicate that AD model rats with hippocampal damage have reduced responsiveness to persistent inflammatory chemical stimuli to the orofacial region.
Collapse
Affiliation(s)
- Masayoshi Hayashi
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-871, Japan.
| | - Chiho Kudo
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-871, Japan
| | - Hiroshi Hanamoto
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Hiroharu Maegawa
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-871, Japan
| | - Nayuka Usami
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-871, Japan
| | - Hitoshi Niwa
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-871, Japan
| |
Collapse
|
42
|
Zhuang X, Cordes D, Caldwell JZK, Bender AR, Miller JB. Disparities in structural brain imaging in older adults from rural communities in Southern Nevada. Front Aging Neurosci 2024; 16:1465744. [PMID: 39430976 PMCID: PMC11486705 DOI: 10.3389/fnagi.2024.1465744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Identifying the associations between rural-living or neighborhood disadvantage and neurobiology may clarify rural-urban disparities in older adults with cognitive impairment related to Alzheimer's disease. Methods We examined rural-urban differences and neighborhood disadvantages in brain cortical thickness (CT) measures among 71 rural and 87 urban-dwelling older adults. Analysis of covariance was used to test each FreeSurfer-derived CT measures' associations with rural-urban living, clinical impairment status, and their interactions. Post-hoc linear regressions were used to test the association between CT measures and neighborhood disadvantage index. Results Rural-dwelling older adults had thinner cortices in temporal and inferior frontal regions compared to urban participants, especially among clinically normal participants, where the thinner temporal cortex further correlated with higher neighborhood disadvantage. Conversely, rural participants had thicker cortices in superior frontal, parietal and occipital regions. Discussion Our results suggest a complex interplay between community contexts and neurobiology. For memory-related regions, rural-living and neighborhood disadvantage might be negatively associated with subjects' brain structures.
Collapse
Affiliation(s)
- Xiaowei Zhuang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Interdisciplinary Neuroscience PhD Program, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Institute of Cognitive Science, University of Colorado Boulder, Boulder, CO, United States
| | | | - Andrew R. Bender
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
| | - Justin B. Miller
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
| |
Collapse
|
43
|
Kwon HS, Yu HJ, Koh SH. Revolutionizing Alzheimer's Diagnosis and Management: The Dawn of Biomarker-Based Precision Medicine. Dement Neurocogn Disord 2024; 23:188-201. [PMID: 39512700 PMCID: PMC11538857 DOI: 10.12779/dnd.2024.23.4.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, presents a formidable global health challenge intensified by the aging population. This review encapsulates the evolving landscape of AD diagnosis and treatment with a special focus on the innovative role of fluid biomarkers. Pathologically, AD is marked by amyloid beta (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau, which lead to synaptic dysfunction, neuronal loss, and cognitive decline. These pathological changes, commencing decades before symptom onset, underscore the need for early detection and intervention. Diagnosis traditionally relies on clinical assessment, neuropsychological testing, and neuroimaging techniques. However, fluid biomarkers in cerebrospinal fluid and blood, such as various forms of Aβ, total tau, phosphorylated tau, and neurofilament light chain, are emerging as less invasive, cost-effective diagnostic tools. These biomarkers are pivotal for early diagnosis, differential diagnosis, disease progression monitoring, and treatment response evaluation. The treatment landscape is shifting toward personalized medicine, highlighted by advancements in Aβ immunotherapies, such as lecanemab and donanemab. Demonstrating efficacy in phase III clinical trials, these therapies hold promise as tailored treatment strategies based on individual biomarker profiles. The integration of fluid biomarkers into clinical practice represents a significant advance in AD management, providing the potential for early and precise diagnosis, coupled with personalized therapeutic approaches. This heralds a new era in combating this debilitating disease.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun-Jung Yu
- Department of Neurology, Bundang Jesaeng General Hospital, Seongnam, Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
44
|
Luo JS, Zhai WH, Ding LL, Zhang XJ, Han J, Ning JQ, Chen XM, Jiang WC, Yan RY, Chen MJ. MAMs and Mitochondrial Quality Control: Overview and Their Role in Alzheimer's Disease. Neurochem Res 2024; 49:2682-2698. [PMID: 39002091 DOI: 10.1007/s11064-024-04205-w] [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: 04/25/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) represents the most widespread neurodegenerative disorder, distinguished by a gradual onset and slow progression, presenting a substantial challenge to global public health. The mitochondrial-associated membrane (MAMs) functions as a crucial center for signal transduction and material transport between mitochondria and the endoplasmic reticulum, playing a pivotal role in various pathological mechanisms of AD. The dysregulation of mitochondrial quality control systems is considered a fundamental factor in the development of AD, leading to mitochondrial dysfunction and subsequent neurodegenerative events. Recent studies have emphasized the role of MAMs in regulating mitochondrial quality control. This review will delve into the molecular mechanisms underlying the imbalance in mitochondrial quality control in AD and provide a comprehensive overview of the role of MAMs in regulating mitochondrial quality control.
Collapse
Affiliation(s)
- Jian-Sheng Luo
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Wen-Hu Zhai
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Ling-Ling Ding
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xian-Jie Zhang
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Jia Han
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Jia-Qi Ning
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Xue-Meng Chen
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Wen-Cai Jiang
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Ru-Yu Yan
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Meng-Jie Chen
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| |
Collapse
|
45
|
Muñoz-Neira C, Zeng J, Kucikova L, Huang W, Xiong X, Muniz-Terrera G, Ritchie C, O'Brien JT, Su L. Differences in Grey Matter Concentrations and Functional Connectivity between Young Carriers and Non-Carriers of the APOE ε4 Genotype. J Clin Med 2024; 13:5228. [PMID: 39274441 PMCID: PMC11396314 DOI: 10.3390/jcm13175228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Background: The pathophysiology of Alzheimer's disease (AD) may begin developing years or even decades prior to the manifestation of its first symptoms. The APOE ε4 genotype is a prominent genetic risk for AD that has been found to be associated with brain changes across the lifespan since early adulthood. Thus, studying brain changes that may occur in young adults with an APOE ε4 status is highly relevant. Objective: Examine potential differences in grey matter (GM) and functional connectivity (FC) in brains of cognitively healthy young APOE ε4 carriers and non-carriers, denoted here as ε4(+) and ε4(-), respectively. Methods: Three Tesla magnetic resonance imaging (MRI) brain scans were acquired from cognitively healthy young participants aged approximately 20 years (n = 151). Voxel-based morphometry (VBM) analysis was employed to identify potential structural differences in GM between ε4(+) and ε4(-). In a subsequent seed-based connectivity (SBC) analysis, brain regions that structurally differed in the VBM analysis were considered as seeds and correlated with all the remaining voxels across the brains to then measure the differences in FC between groups. Results: The VBM analysis suggested that ε4(+) (n = 28) had greater GM densities relative to ε4(-) (n = 123) in the left hippocampus and the left posterior insula (puncorr < 0.001). However, the effect did not survive the correction for multiple comparisons, suggesting minimal structural differences in this age range. In contrast, the SBC analysis indicated that ε4(+) exhibited significantly decreased FC between the left hippocampus and areas of the left middle temporal gyrus (n = 27) compared to ε4(-) (n = 102). These results remained significant after multiple comparisons (pFDR < 0.05). Lastly, no statistically significant differences in FC between groups were observed for the left insular seed (pFDR > 0.05). Discussion: These results suggest early structural and functional brain changes associated with the APOE ε4 genotype on young adults. Yet, they must be cautiously interpreted and contrasted with both older adults with genetic risk for AD and patients diagnosed with AD.
Collapse
Affiliation(s)
- Carlos Muñoz-Neira
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Jianmin Zeng
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing 400715, China
| | - Ludmila Kucikova
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK
| | - Weijie Huang
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Xiong Xiong
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- School of Information and Communication Engineering, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Craig Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Scottish Brain Sciences, Edinburgh EH12 9DQ, UK
| | - John T O'Brien
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Li Su
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
46
|
Mohammadzadeh M, Khoshakhlagh AH, Grafman J. Air pollution: a latent key driving force of dementia. BMC Public Health 2024; 24:2370. [PMID: 39223534 PMCID: PMC11367863 DOI: 10.1186/s12889-024-19918-4] [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: 06/05/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Many researchers have studied the role of air pollutants on cognitive function, changes in brain structure, and occurrence of dementia. Due to the wide range of studies and often contradictory results, the present systematic review was conducted to try and clarify the relationship between air pollutants and dementia. To identify studies for this review, a systematic search was conducted in Scopus, PubMed, and Web of Science databases (without historical restrictions) until May 22, 2023. The PECO statement was created to clarify the research question, and articles that did not meet the criteria of this statement were excluded. In this review, animal studies, laboratory studies, books, review articles, conference papers and letters to the editors were avoided. Also, studies focused on the effect of air pollutants on cellular and biochemical changes (without investigating dementia) were also excluded. A quality assessment was done according to the type of design of each article, using the checklist developed by the Joanna Briggs Institute (JBI). Finally, selected studies were reviewed and discussed in terms of Alzheimer's dementia and non-Alzheimer's dementia. We identified 14,924 articles through a systematic search in databases, and after comprehensive reviews, 53 articles were found to be eligible for inclusion in the current systematic review. The results showed that chronic exposure to higher levels of air pollutants was associated with adverse effects on cognitive abilities and the presence of dementia. Studies strongly supported the negative effects of PM2.5 and then NO2 on the brain and the development of neurodegenerative disorders in old age. Because the onset of brain structural changes due to dementia begins decades before the onset of disease symptoms, and that exposure to air pollution is considered a modifiable risk factor, taking preventive measures to reduce air pollution and introducing behavioral interventions to reduce people's exposure to pollutants is advisable.
Collapse
Affiliation(s)
- Mahdiyeh Mohammadzadeh
- Department of Health in Emergencies and Disasters, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Climate Change and Health Research Center (CCHRC), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Khoshakhlagh
- Department of Occupational Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran.
| | - Jordan Grafman
- Department of Physical Medicine & Rehabilitation, Neurology, Cognitive Neurology and Alzheimer's Center, Department of Psychiatry, Feinberg School of Medicine & Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| |
Collapse
|
47
|
Sathe A, Yang Y, Schilling KG, Shashikumar N, Moore E, Dumitrescu L, Pechman KR, Landman BA, Gifford KA, Hohman TJ, Jefferson AL, Archer DB. Free-water: A promising structural biomarker for cognitive decline in aging and mild cognitive impairment. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-16. [PMID: 39512598 PMCID: PMC11540062 DOI: 10.1162/imag_a_00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 11/15/2024]
Abstract
Diffusion MRI derived free-water (FW) metrics show promise in predicting cognitive impairment and decline in aging and Alzheimer's disease (AD). FW is sensitive to subtle changes in brain microstructure, so it is possible these measures may be more sensitive than traditional structural neuroimaging biomarkers. In this study, we examined the associations among FW metrics (measured in the hippocampus and two AD signature meta-ROIs) with cognitive performance, and compared FW findings to those from more traditional neuroimaging biomarkers of AD. We leveraged data from a longitudinal cohort (nparticipants = 296, nobservations = 870, age at baseline: 73 ± 7 years, 40% mild cognitive impairment [MCI]) of older adults who underwent serial neuropsychological assessment (episodic memory, information processing speed, executive function, language, and visuospatial skills) and brain MRI over a maximum of four time points, including baseline (n = 284), 18-month (n = 246), 3-year (n = 215), and 5-year (n = 125) visits. The mean follow-up period was 2.8 ± 1.3 years. Structural MRI was used to quantify hippocampal volume, in addition to Schwarz and McEvoy AD Signatures. FW and FW-corrected fractional anisotropy (FAFWcorr) were quantified in the hippocampus (hippocampal FW) and the AD signature areas (SchwarzFW, McEvoyFW) from diffusion-weighted (dMRI) images using bi-tensor modeling (FW elimination and mapping method). Linear regression assessed the association of each biomarker with baseline cognitive performance. Additionally, linear mixed-effects regression assessed the association between baseline biomarker values and longitudinal cognitive performance. A subsequent competitive model analysis was conducted on both baseline and longitudinal data to determine how much additional variance in cognitive performance was explained by each biomarker compared to the covariate only model, which included age, sex, race/ethnicity, apolipoprotein-ε4 status, cognitive status, and modified Framingham Stroke Risk Profile scores. All analyses were corrected for multiple comparisons using an FDR procedure. Cross-sectional results indicate that hippocampal volume, hippocampal FW, Schwarz and McEvoy AD Signatures, and the SchwarzFW and McEvoyFW metrics are all significantly associated with memory performance. Baseline competitive model analyses showed that the McEvoy AD Signature and SchwarzFW explain the most unique variance beyond covariates for memory (ΔRadj 2 = 3.47 ± 1.65%) and executive function (ΔRadj 2 = 2.43 ± 1.63%), respectively. Longitudinal models revealed that hippocampal FW explained substantial unique variance for memory performance (ΔRadj 2 = 8.13 ± 1.25%), and outperformed all other biomarkers examined in predicting memory decline (pFDR = 1.95 x 10-11). This study shows that hippocampal FW is a sensitive biomarker for cognitive impairment and decline, and provides strong evidence for further exploration of this measure in aging and AD.
Collapse
Affiliation(s)
- Aditi Sathe
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Yisu Yang
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Kurt G. Schilling
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Niranjana Shashikumar
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Elizabeth Moore
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kimberly R. Pechman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Bennett A. Landman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Katherine A. Gifford
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Derek B. Archer
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
48
|
Hu J, Zhang M, Zhang Y, Zhuang H, Zhao Y, Li Y, Jin W, Qian X, Wang L, Ye G, Tang H, Liu J, Li B, Nachev P, Liang Z, Li Y. Neurometabolic topography and associations with cognition in Alzheimer's disease: A whole-brain high-resolution 3D MRSI study. Alzheimers Dement 2024; 20:6407-6422. [PMID: 39073196 PMCID: PMC11497670 DOI: 10.1002/alz.14137] [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: 03/04/2024] [Revised: 05/29/2024] [Accepted: 06/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Altered neurometabolism, detectable via proton magnetic resonance spectroscopic imaging (1H-MRSI), is spatially heterogeneous and underpins cognitive impairments in Alzheimer's disease (AD). However, the spatial relationships between neurometabolic topography and cognitive impairment in AD remain unexplored due to technical limitations. METHODS We used a novel whole-brain high-resolution 1H-MRSI technique, with simultaneously acquired 18F-florbetapir positron emission tomography (PET) imaging, to investigate the relationship between neurometabolic topography and cognitive functions in 117 participants, including 22 prodromal AD, 51 AD dementia, and 44 controls. RESULTS Prodromal AD and AD dementia patients exhibited spatially distinct reductions in N-acetylaspartate, and increases in myo-inositol. Reduced N-acetylaspartate and increased myo-inositol were associated with worse global cognitive performance, and N-acetylaspartate correlated with five specific cognitive scores. Neurometabolic topography provides biological insights into diverse cognitive dysfunctions. DISCUSSION Whole-brain high-resolution 1H-MRSI revealed spatially distinct neurometabolic topographies associated with cognitive decline in AD, suggesting potential for noninvasive brain metabolic imaging to track AD progression. HIGHLIGHTS Whole-brain high-resolution 1H-MRSI unveils neurometabolic topography in AD. Spatially distinct reductions in NAA, and increases in mI, are demonstrated. NAA and mI topography correlates with global cognitive performance. NAA topography correlates with specific cognitive performance.
Collapse
Affiliation(s)
- Jialin Hu
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Miao Zhang
- Department of Nuclear MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yaoyu Zhang
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Huixiang Zhuang
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yibo Zhao
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Yudu Li
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- National Center for Supercomputing ApplicationsUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Wen Jin
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Xiao‐Hang Qian
- Department of GeriatricsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lijun Wang
- Department of Neurovascular CenterChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Guanyu Ye
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huidong Tang
- Department of GeriatricsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jun Liu
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Biao Li
- Department of Nuclear MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Parashkev Nachev
- High‐Dimensional Neurology GroupInstitute of NeurologyUniversity College LondonLondonUK
| | - Zhi‐Pei Liang
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Yao Li
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- Institute of Medical RoboticsShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
49
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
50
|
Salazar R, Dwivedi AK, Alvarado LA, Escamilla M. An 11-Fold Higher Risk of Incident Mild Cognitive Impairment With Hispanic Ethnicity and Baseline Neuropsychiatric Symptoms. J Neuropsychiatry Clin Neurosci 2024; 37:29-37. [PMID: 39169739 DOI: 10.1176/appi.neuropsych.20230180] [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] [Indexed: 08/23/2024]
Abstract
OBJECTIVE Neuropsychiatric symptoms (NPSs) have been linked to cognitive decline. This study explored ethnic differences and the effects of baseline NPSs on incident mild cognitive impairment (MCI) among 386 Hispanic and non-Hispanic participants from the Texas Harris Alzheimer's Research Study. METHODS Data on NPSs from the Neuropsychiatric Inventory Questionnaire were available for all participants. Cox proportional hazards regression models were used to estimate the effect of ≥1 NPS at baseline and Hispanic ethnicity on incident MCI over a 7-year follow-up period. RESULTS NPSs at baseline were associated with incident MCI for Hispanic participants but not non-Hispanic participants. Being Hispanic with at least one NPS at baseline had an 11-times higher risk of incident MCI. CONCLUSIONS The Hispanic participants converted to MCI to a greater extent than the non-Hispanic participants. Only depressive symptoms increased the risk of MCI among non-Hispanics. Being of Hispanic ethnicity and having NPSs appeared to jointly increase the risk of progressing to MCI. To better understand the Alzheimer's disease continuum, further studies should explore other cultural, genetic, and medical risk factors influencing disease progression. Our findings strongly suggest the need to incorporate NPSs as outcomes of disease progression in future clinical trials involving Hispanic participants.
Collapse
Affiliation(s)
- Ricardo Salazar
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Neurosciences (Salazar), and Department of Molecular and Translational Medicine, Division of Biostatistics and Epidemiology (Dwivedi, Alvarado), Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso; Department of Psychiatry, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (Escamilla)
| | - Alok K Dwivedi
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Neurosciences (Salazar), and Department of Molecular and Translational Medicine, Division of Biostatistics and Epidemiology (Dwivedi, Alvarado), Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso; Department of Psychiatry, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (Escamilla)
| | - Luis A Alvarado
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Neurosciences (Salazar), and Department of Molecular and Translational Medicine, Division of Biostatistics and Epidemiology (Dwivedi, Alvarado), Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso; Department of Psychiatry, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (Escamilla)
| | - Michael Escamilla
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Neurosciences (Salazar), and Department of Molecular and Translational Medicine, Division of Biostatistics and Epidemiology (Dwivedi, Alvarado), Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso; Department of Psychiatry, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (Escamilla)
| |
Collapse
|