首页 > 最新文献

Alzheimer's Research & Therapy最新文献

英文 中文
β-synuclein in cerebrospinal fluid as a potential biomarker for distinguishing human prion diseases from Alzheimer's and Parkinson's disease.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-07 DOI: 10.1186/s13195-025-01688-9
Bing Xu, Kang Xiao, Xiaoxi Jia, Rundong Cao, Donglin Liang, Ruhan A, Weiwei Zhang, Chunjie Li, Liping Gao, Cao Chen, Qi Shi, Xiaoping Dong

Background: β-synuclein (β-syn), mainly expressed in central nerve system, is one of the biomarkers in cerebrospinal fluid (CSF) and blood for synaptic damage, which has been reported to be elevated in CSF and blood of the patients of prion diseases (PrDs).

Methods: We analyzed 314 CSF samples from patients in China National Surveillance for CJD. The diagnostic groups of the 223 patients with PrDs included sporadic Creutzfeldt-Jacob disease (sCJD), genetic CJD (gCJD), fatal familial insomnia (FFI) and Gerstmann-Straussler-Scheinker (GSS). 91 patients with non-PrDs comprised Alzheimer's disease (AD), Parkinson's disease (PD), viral encephalitis (VE) or autoimmune encephalitis (AE) were enrolled in the control groups. The CSF β-syn levels were measured by a commercial microfluidic ELISA. The Mann-Whitney U test and Kruskal-Wallis H test were employed to analyze two or more sets of continuous variables. Multiple linear regression was also performed to evaluate the factors for CSF β-syn levels. Receiver operating characteristics (ROC) curves and area under the curve (AUC) values were used to assess the diagnostic performance of β-syn.

Results: The median of β-syn levels (2074 pg/ml; IQR: 691 to 4332) of all PrDs was significantly higher than that of non-PrDs group (504 pg/ml; IQR: 126 to 3374). The CSF β-syn values in the cohorts of sCJD, T188K-gCJD, E200K-gCJD and P102L-GSS were remarkably higher than that of the group of AD + PD, but similar as that of the group of VE + AE. The elevated CSF β-syn in sCJD and gCJD cases was statistically associated with CSF 14-3-3 positive and appearance of mutism. ROC curve analysis identified satisfied performance for distinguishing from AD + PD, with high AUC values in sCJD (0.7640), T188K-gCJD (0.8489), E200K-gCJD (0.8548), P102L-GSS (0.7689) and D178N-FFI (0.7210), respectively.

Conclusion: Our data here indicate that CSF β-syn is a potential biomarker for distinguishing PrDs (gCJD, sCJD and GSS) from AD and PD, but is much less efficient from VE and AE. These findings have critical implications for early diagnosis and monitoring of synaptic integrity in prion diseases.

{"title":"β-synuclein in cerebrospinal fluid as a potential biomarker for distinguishing human prion diseases from Alzheimer's and Parkinson's disease.","authors":"Bing Xu, Kang Xiao, Xiaoxi Jia, Rundong Cao, Donglin Liang, Ruhan A, Weiwei Zhang, Chunjie Li, Liping Gao, Cao Chen, Qi Shi, Xiaoping Dong","doi":"10.1186/s13195-025-01688-9","DOIUrl":"https://doi.org/10.1186/s13195-025-01688-9","url":null,"abstract":"<p><strong>Background: </strong>β-synuclein (β-syn), mainly expressed in central nerve system, is one of the biomarkers in cerebrospinal fluid (CSF) and blood for synaptic damage, which has been reported to be elevated in CSF and blood of the patients of prion diseases (PrDs).</p><p><strong>Methods: </strong>We analyzed 314 CSF samples from patients in China National Surveillance for CJD. The diagnostic groups of the 223 patients with PrDs included sporadic Creutzfeldt-Jacob disease (sCJD), genetic CJD (gCJD), fatal familial insomnia (FFI) and Gerstmann-Straussler-Scheinker (GSS). 91 patients with non-PrDs comprised Alzheimer's disease (AD), Parkinson's disease (PD), viral encephalitis (VE) or autoimmune encephalitis (AE) were enrolled in the control groups. The CSF β-syn levels were measured by a commercial microfluidic ELISA. The Mann-Whitney U test and Kruskal-Wallis H test were employed to analyze two or more sets of continuous variables. Multiple linear regression was also performed to evaluate the factors for CSF β-syn levels. Receiver operating characteristics (ROC) curves and area under the curve (AUC) values were used to assess the diagnostic performance of β-syn.</p><p><strong>Results: </strong>The median of β-syn levels (2074 pg/ml; IQR: 691 to 4332) of all PrDs was significantly higher than that of non-PrDs group (504 pg/ml; IQR: 126 to 3374). The CSF β-syn values in the cohorts of sCJD, T188K-gCJD, E200K-gCJD and P102L-GSS were remarkably higher than that of the group of AD + PD, but similar as that of the group of VE + AE. The elevated CSF β-syn in sCJD and gCJD cases was statistically associated with CSF 14-3-3 positive and appearance of mutism. ROC curve analysis identified satisfied performance for distinguishing from AD + PD, with high AUC values in sCJD (0.7640), T188K-gCJD (0.8489), E200K-gCJD (0.8548), P102L-GSS (0.7689) and D178N-FFI (0.7210), respectively.</p><p><strong>Conclusion: </strong>Our data here indicate that CSF β-syn is a potential biomarker for distinguishing PrDs (gCJD, sCJD and GSS) from AD and PD, but is much less efficient from VE and AE. These findings have critical implications for early diagnosis and monitoring of synaptic integrity in prion diseases.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"39"},"PeriodicalIF":7.9,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143370231","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Heterogeneous clinical phenotypes of sporadic early-onset Alzheimer's disease: a neuropsychological data-driven approach.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-06 DOI: 10.1186/s13195-025-01689-8
Deepti Putcha, Yuta Katsumi, Alexandra Touroutoglou, Ani Eloyan, Alexander Taurone, Maryanne Thangarajah, Paul Aisen, Jeffrey L Dage, Tatiana Foroud, Clifford R Jack, Joel H Kramer, Kelly N H Nudelman, Rema Raman, Prashanthi Vemuri, Alireza Atri, Gregory S Day, Ranjan Duara, Neill R Graff-Radford, Ian M Grant, Lawrence S Honig, Erik C B Johnson, David T Jones, Joseph C Masdeu, Mario F Mendez, Erik Musiek, Chiadi U Onyike, Meghan Riddle, Emily Rogalski, Stephen Salloway, Sharon Sha, R Scott Turner, Thomas S Wingo, David A Wolk, Kyle Womack, Maria C Carrillo, Gil D Rabinovici, Bradford C Dickerson, Liana G Apostolova, Dustin B Hammers

Background: The clinical presentations of early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease are distinct, with EOAD having a more aggressive disease course with greater heterogeneity. Recent publications from the Longitudinal Early-Onset Alzheimer's Disease Study (LEADS) described EOAD as predominantly amnestic, though this phenotypic description was based solely on clinical judgment. To better understand the phenotypic range of EOAD presentation, we applied a neuropsychological data-driven method to subtype the LEADS cohort.

Methods: Neuropsychological test performance from 169 amyloid-positive EOAD participants were analyzed. Education-corrected normative comparisons were made using a sample of 98 cognitively normal participants. Comparing the relative levels of impairment between each cognitive domain, we applied a cut-off of 1 SD below all other domain scores to indicate a phenotype of "predominant" impairment in a given cognitive domain. Individuals were otherwise considered to have multidomain impairment. Whole-cortex general linear modeling of cortical atrophy was applied as an MRI-based validation of these distinct clinical phenotypes.

Results: We identified 6 phenotypic subtypes of EOAD: Dysexecutive Predominant (22% of sample), Amnestic Predominant (11%), Language Predominant (11%), Visuospatial Predominant (15%), Mixed Amnestic/Dysexecutive Predominant (11%), and Multidomain (30%). These phenotypes did not differ by age, sex, or years of education. The APOE ɛ4 genotype was enriched in the Amnestic Predominant group, who were also rated as least impaired. Cortical thickness analysis validated these clinical phenotypes with dissociations in atrophy patterns observed between the Dysexecutive and Amnestic Predominant groups. In contrast to the heterogeneity observed from our neuropsychological data-driven approach, diagnostic classifications for this same sample based solely on clinical judgment indicated that 82% of individuals were amnestic-predominant, 9% were non-amnestic, 4% met criteria for Posterior Cortical Atrophy, and 5% met criteria for Primary Progressive Aphasia.

Conclusion: A neuropsychological data-driven method to phenotype EOAD individuals uncovered a more detailed understanding of the presenting heterogeneity in this atypical AD sample compared to clinical judgment alone. Clinicians and patients may over-report memory dysfunction at the expense of non-memory symptoms. These findings have important implications for diagnostic accuracy and treatment considerations.

{"title":"Heterogeneous clinical phenotypes of sporadic early-onset Alzheimer's disease: a neuropsychological data-driven approach.","authors":"Deepti Putcha, Yuta Katsumi, Alexandra Touroutoglou, Ani Eloyan, Alexander Taurone, Maryanne Thangarajah, Paul Aisen, Jeffrey L Dage, Tatiana Foroud, Clifford R Jack, Joel H Kramer, Kelly N H Nudelman, Rema Raman, Prashanthi Vemuri, Alireza Atri, Gregory S Day, Ranjan Duara, Neill R Graff-Radford, Ian M Grant, Lawrence S Honig, Erik C B Johnson, David T Jones, Joseph C Masdeu, Mario F Mendez, Erik Musiek, Chiadi U Onyike, Meghan Riddle, Emily Rogalski, Stephen Salloway, Sharon Sha, R Scott Turner, Thomas S Wingo, David A Wolk, Kyle Womack, Maria C Carrillo, Gil D Rabinovici, Bradford C Dickerson, Liana G Apostolova, Dustin B Hammers","doi":"10.1186/s13195-025-01689-8","DOIUrl":"10.1186/s13195-025-01689-8","url":null,"abstract":"<p><strong>Background: </strong>The clinical presentations of early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease are distinct, with EOAD having a more aggressive disease course with greater heterogeneity. Recent publications from the Longitudinal Early-Onset Alzheimer's Disease Study (LEADS) described EOAD as predominantly amnestic, though this phenotypic description was based solely on clinical judgment. To better understand the phenotypic range of EOAD presentation, we applied a neuropsychological data-driven method to subtype the LEADS cohort.</p><p><strong>Methods: </strong>Neuropsychological test performance from 169 amyloid-positive EOAD participants were analyzed. Education-corrected normative comparisons were made using a sample of 98 cognitively normal participants. Comparing the relative levels of impairment between each cognitive domain, we applied a cut-off of 1 SD below all other domain scores to indicate a phenotype of \"predominant\" impairment in a given cognitive domain. Individuals were otherwise considered to have multidomain impairment. Whole-cortex general linear modeling of cortical atrophy was applied as an MRI-based validation of these distinct clinical phenotypes.</p><p><strong>Results: </strong>We identified 6 phenotypic subtypes of EOAD: Dysexecutive Predominant (22% of sample), Amnestic Predominant (11%), Language Predominant (11%), Visuospatial Predominant (15%), Mixed Amnestic/Dysexecutive Predominant (11%), and Multidomain (30%). These phenotypes did not differ by age, sex, or years of education. The APOE ɛ4 genotype was enriched in the Amnestic Predominant group, who were also rated as least impaired. Cortical thickness analysis validated these clinical phenotypes with dissociations in atrophy patterns observed between the Dysexecutive and Amnestic Predominant groups. In contrast to the heterogeneity observed from our neuropsychological data-driven approach, diagnostic classifications for this same sample based solely on clinical judgment indicated that 82% of individuals were amnestic-predominant, 9% were non-amnestic, 4% met criteria for Posterior Cortical Atrophy, and 5% met criteria for Primary Progressive Aphasia.</p><p><strong>Conclusion: </strong>A neuropsychological data-driven method to phenotype EOAD individuals uncovered a more detailed understanding of the presenting heterogeneity in this atypical AD sample compared to clinical judgment alone. Clinicians and patients may over-report memory dysfunction at the expense of non-memory symptoms. These findings have important implications for diagnostic accuracy and treatment considerations.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"38"},"PeriodicalIF":7.9,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11800584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143363391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A systematic review and meta-analysis of the impact of transcranial direct current stimulation on cognitive function in older adults with cognitive impairments: the influence of dosage parameters.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-04 DOI: 10.1186/s13195-025-01677-y
Thatchaya Prathum, Thanwarat Chantanachai, Oranich Vimolratana, Chotica Laksanaphuk, Irin Apiworajirawit, Benchaporn Aneksan, Kanthika Latthirun, Cheng-Ta Yang, Wanalee Klomjai

Introduction: Numerous studies have demonstrated the effects of transcranial direct current stimulation (tDCS) on cognitive function in the older people. This study further explores the impact of tDCS and its dosage parameters on cognitive enhancement in older people with cognitive impairments.

Methods: Randomized controlled trials (RCTs) published through November 2023 were retrieved from databases including PubMed, Scopus, EMBASE, EBSCO, and the Cochrane Library. Participants were older adults with cognitive impairments, including Alzheimer's disease (AD), mild cognitive impairment (MCI), and dementia. AD was diagnosed based on the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), or the National Institute of Neurological and Communicative Disorders and Stroke - Alzheimer' Disease and Related Disorders Association (NINCDS-ADRDA) criteria. Dementia was diagnosed using the DSM-V or NINCDS-ADRDA criteria, while MCI was diagnosed using the DSM-V, the Petersen criteria, or assessments such as Montreal Cognitive Assessment (MoCA) and Clinical Dementia Rating (CDR). Standardized mean difference (SMD) values were analyzed to assess the effects.

Results: A total of 19 RCTs were included. tDCS significantly improved the Mini-Mental State Examination score both immediately post-intervention (SMD = 0.51, p = 0.005) and at follow-up (SMD = 2.29, p = 0.0003). Significant effects were observed when tDCS was used alone (SMD = 0.39, p = 0.04), at current densities 0.06 mA/cm2 (SMD = 0.25, p = 0.04), session durations exceeding 20 min (SMD = 0.89, p = 0.01), up to 15 sessions (SMD = 0.28, p = 0.009), and when an active electrode was placed over the temporal area (SMD = 0.33, p = 0.02). People with AD showed greater improvements compared to those with MCI or dementia (SMD = 0.91, p = 0.02). However, tDCS did not significantly improve memory or executive function.

Conclusion: tDCS demonstrated efficacy in enhancing global cognition in older people with cognitive impairments, providing insight into optimal parameters for clinical application. However, no improvement were observed in memory or executive function.

导言许多研究已经证明了经颅直流电刺激(tDCS)对老年人认知功能的影响。本研究进一步探讨了经颅直流电刺激(tDCS)及其剂量参数对认知障碍老年人认知能力增强的影响:从 PubMed、Scopus、EMBASE、EBSCO 和 Cochrane Library 等数据库中检索了截至 2023 年 11 月发表的随机对照试验(RCT)。参与者为有认知障碍的老年人,包括阿尔茨海默病(AD)、轻度认知障碍(MCI)和痴呆症。阿尔茨海默病的诊断依据是《精神疾病诊断与统计手册》第四版(DSM-IV)或美国国家神经与交流障碍和中风研究所-阿尔茨海默病及相关疾病协会(NINCDS-ADRDA)的标准。痴呆的诊断采用 DSM-V 或 NINCDS-ADRDA 标准,而 MCI 的诊断则采用 DSM-V、彼得森标准或蒙特利尔认知评估 (MoCA) 和临床痴呆评级 (CDR) 等评估方法。对标准化均差(SMD)值进行分析,以评估效果:tDCS 可显著改善干预后即刻(SMD = 0.51,p = 0.005)和随访时(SMD = 2.29,p = 0.0003)的迷你精神状态检查评分。当单独使用 tDCS 时(SMD = 0.39,p = 0.04)、电流密度≤ 0.06 mA/cm2 时(SMD = 0.25,p = 0.04)、疗程超过 20 分钟时(SMD = 0.89,p = 0.01)、最多 15 个疗程时(SMD = 0.28,p = 0.009)以及将有源电极置于颞部时(SMD = 0.33,p = 0.02),均观察到显著效果。与 MCI 或痴呆症患者相比,注意力缺失症患者的改善幅度更大(SMD = 0.91,p = 0.02)。结论:tDCS在提高认知障碍老年人的整体认知能力方面表现出疗效,为临床应用提供了最佳参数。但是,在记忆力和执行功能方面没有观察到任何改善。
{"title":"A systematic review and meta-analysis of the impact of transcranial direct current stimulation on cognitive function in older adults with cognitive impairments: the influence of dosage parameters.","authors":"Thatchaya Prathum, Thanwarat Chantanachai, Oranich Vimolratana, Chotica Laksanaphuk, Irin Apiworajirawit, Benchaporn Aneksan, Kanthika Latthirun, Cheng-Ta Yang, Wanalee Klomjai","doi":"10.1186/s13195-025-01677-y","DOIUrl":"10.1186/s13195-025-01677-y","url":null,"abstract":"<p><strong>Introduction: </strong>Numerous studies have demonstrated the effects of transcranial direct current stimulation (tDCS) on cognitive function in the older people. This study further explores the impact of tDCS and its dosage parameters on cognitive enhancement in older people with cognitive impairments.</p><p><strong>Methods: </strong>Randomized controlled trials (RCTs) published through November 2023 were retrieved from databases including PubMed, Scopus, EMBASE, EBSCO, and the Cochrane Library. Participants were older adults with cognitive impairments, including Alzheimer's disease (AD), mild cognitive impairment (MCI), and dementia. AD was diagnosed based on the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), or the National Institute of Neurological and Communicative Disorders and Stroke - Alzheimer' Disease and Related Disorders Association (NINCDS-ADRDA) criteria. Dementia was diagnosed using the DSM-V or NINCDS-ADRDA criteria, while MCI was diagnosed using the DSM-V, the Petersen criteria, or assessments such as Montreal Cognitive Assessment (MoCA) and Clinical Dementia Rating (CDR). Standardized mean difference (SMD) values were analyzed to assess the effects.</p><p><strong>Results: </strong>A total of 19 RCTs were included. tDCS significantly improved the Mini-Mental State Examination score both immediately post-intervention (SMD = 0.51, p = 0.005) and at follow-up (SMD = 2.29, p = 0.0003). Significant effects were observed when tDCS was used alone (SMD = 0.39, p = 0.04), at current densities <math><mo>≤</mo></math> 0.06 mA/cm<sup>2</sup> (SMD = 0.25, p = 0.04), session durations exceeding 20 min (SMD = 0.89, p = 0.01), up to 15 sessions (SMD = 0.28, p = 0.009), and when an active electrode was placed over the temporal area (SMD = 0.33, p = 0.02). People with AD showed greater improvements compared to those with MCI or dementia (SMD = 0.91, p = 0.02). However, tDCS did not significantly improve memory or executive function.</p><p><strong>Conclusion: </strong>tDCS demonstrated efficacy in enhancing global cognition in older people with cognitive impairments, providing insight into optimal parameters for clinical application. However, no improvement were observed in memory or executive function.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"37"},"PeriodicalIF":7.9,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11796231/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multi-omics analyses identify gut microbiota-fecal metabolites-brain-cognition pathways in the Alzheimer's disease continuum.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-01 DOI: 10.1186/s13195-025-01683-0
Han Zhao, Xia Zhou, Yu Song, Wenming Zhao, Zhongwu Sun, Jiajia Zhu, Yongqiang Yu

Background: Gut microbiota dysbiosis is linked to Alzheimer's disease (AD), but our understanding of the molecular and neuropathological bases underlying such association remains fragmentary.

Methods: Using 16S rDNA amplicon sequencing, untargeted metabolomics, and multi-modal magnetic resonance imaging, we examined group differences in gut microbiome, fecal metabolome, neuroimaging measures, and cognitive variables across 30 patients with AD, 75 individuals with mild cognitive impairment (MCI), and 61 healthy controls (HC). Furthermore, we assessed the associations between these multi-omics changes using correlation and mediation analyses.

Results: There were significant group differences in gut microbial composition, which were driven by 8 microbial taxa (e.g., Staphylococcus and Bacillus) exhibiting a progressive increase in relative abundance from HC to MCI to AD, and 2 taxa (e.g., Anaerostipes) showing a gradual decrease. 26 fecal metabolites (e.g., Arachidonic, Adrenic, and Lithocholic acids) exhibited a progressive increase from HC to MCI to AD. We also observed progressive gray matter atrophy in broadly distributed gray matter regions and gradual micro-structural integrity damage in widespread white matter tracts along the AD continuum. Integration of these multi-omics changes revealed significant associations between microbiota, metabolites, neuroimaging, and cognition. More importantly, we identified two potential mediation pathways: (1) microbiota → metabolites → neuroimaging → cognition, and (2) microbiota → metabolites → cognition.

Conclusion: Aside from elucidating the underlying mechanism whereby gut microbiota dysbiosis is linked to AD, our findings may contribute to groundwork for future interventions targeting the microbiota-metabolites-brain-cognition pathways as a therapeutic strategy in the AD continuum.

{"title":"Multi-omics analyses identify gut microbiota-fecal metabolites-brain-cognition pathways in the Alzheimer's disease continuum.","authors":"Han Zhao, Xia Zhou, Yu Song, Wenming Zhao, Zhongwu Sun, Jiajia Zhu, Yongqiang Yu","doi":"10.1186/s13195-025-01683-0","DOIUrl":"10.1186/s13195-025-01683-0","url":null,"abstract":"<p><strong>Background: </strong>Gut microbiota dysbiosis is linked to Alzheimer's disease (AD), but our understanding of the molecular and neuropathological bases underlying such association remains fragmentary.</p><p><strong>Methods: </strong>Using 16S rDNA amplicon sequencing, untargeted metabolomics, and multi-modal magnetic resonance imaging, we examined group differences in gut microbiome, fecal metabolome, neuroimaging measures, and cognitive variables across 30 patients with AD, 75 individuals with mild cognitive impairment (MCI), and 61 healthy controls (HC). Furthermore, we assessed the associations between these multi-omics changes using correlation and mediation analyses.</p><p><strong>Results: </strong>There were significant group differences in gut microbial composition, which were driven by 8 microbial taxa (e.g., Staphylococcus and Bacillus) exhibiting a progressive increase in relative abundance from HC to MCI to AD, and 2 taxa (e.g., Anaerostipes) showing a gradual decrease. 26 fecal metabolites (e.g., Arachidonic, Adrenic, and Lithocholic acids) exhibited a progressive increase from HC to MCI to AD. We also observed progressive gray matter atrophy in broadly distributed gray matter regions and gradual micro-structural integrity damage in widespread white matter tracts along the AD continuum. Integration of these multi-omics changes revealed significant associations between microbiota, metabolites, neuroimaging, and cognition. More importantly, we identified two potential mediation pathways: (1) microbiota → metabolites → neuroimaging → cognition, and (2) microbiota → metabolites → cognition.</p><p><strong>Conclusion: </strong>Aside from elucidating the underlying mechanism whereby gut microbiota dysbiosis is linked to AD, our findings may contribute to groundwork for future interventions targeting the microbiota-metabolites-brain-cognition pathways as a therapeutic strategy in the AD continuum.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"36"},"PeriodicalIF":7.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786436/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073347","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of intensive lifestyle changes on the progression of mild cognitive impairment or early dementia due to Alzheimer's disease: the need for rigor.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-01 DOI: 10.1186/s13195-024-01621-6
Joshua D Grill, Daniel Gillen

We consider the recent publication by Ornish and colleagues and the rigor expected for interventional clinical trials. We contend that lifestyle intervention trials should strive for the same rigor as drug trials and highlight opportunities to improve rigor in this example, particularly in design, data analysis, and publication of results for this and other lifestyle intervention studies.

{"title":"Effects of intensive lifestyle changes on the progression of mild cognitive impairment or early dementia due to Alzheimer's disease: the need for rigor.","authors":"Joshua D Grill, Daniel Gillen","doi":"10.1186/s13195-024-01621-6","DOIUrl":"10.1186/s13195-024-01621-6","url":null,"abstract":"<p><p>We consider the recent publication by Ornish and colleagues and the rigor expected for interventional clinical trials. We contend that lifestyle intervention trials should strive for the same rigor as drug trials and highlight opportunities to improve rigor in this example, particularly in design, data analysis, and publication of results for this and other lifestyle intervention studies.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"32"},"PeriodicalIF":7.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073341","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Delta-opioid receptor signaling alleviates neuropathology and cognitive impairment in the mouse model of Alzheimer's disease by regulating microglia homeostasis and inhibiting HMGB1 pathway.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-01 DOI: 10.1186/s13195-025-01682-1
Yuan Xu, Naiyuan Shao, Feng Zhi, Ronghua Chen, Yilin Yang, Jiahui Li, Ying Xia, Ya Peng

Background: Recent studies suggest that opioid receptor signaling may differentially affect Alzheimer's disease (AD) pathology and the relevant behavioral dysfunctions. However, the precise roles and mechanisms of opioid receptor subtypes in AD pathologies are still unclear with major controversies.

Methods: We compared the delta-opioid receptor (DOR)- and mu-opioid receptor (MOR)-mediated effects on AD-associated cognitive deficits, pathologies, neuroinflammations, cell death using transgenic APP/PS1 mouse model and BV2 cell line at behavioral, molecular, and cellular levels. Unpaired t-test and one/two way analysis for variance (ANOVA) were used to analyze statistical significance of the data.

Results: We show a distinct role of DOR and its major difference with MOR in AD injury in an APP/PS1 mouse model. DOR activation by UFP-512, but not MOR activation by DAMGO, attenuated cognitive impairment, reduced beta-amyloid (Aβ) production and aggregation, as well as protected the neurons from apoptosis in APP/PS1 mice. DOR and MOR also differentially modulated microglia in APP/PS1 mice and in vitro AD cell model with a DOR-mediated inhibition on the excessive activation of microglia and the release of pro-inflammatory cytokines in AD pathologies. Gene expression profiling further revealed that the alternations in DOR/MOR are closely associated with microglial homeostatic signatures and high mobility group protein B1 (HMGB1) in AD. DOR activation inhibited HMGB1 secretion and its translocation from nuclear to cytoplasm. Our in-vitro studies further confirmed that DOR overexpression mitigated microglial inflammatory response and rescued neurons from AD injury via HMGB1-NF-κB signaling pathway.

Conclusions: These novel findings uncover previously unappreciated roles of DOR in neuroprotection against AD injury via modulating microglia-related inflammatory responses.

{"title":"Delta-opioid receptor signaling alleviates neuropathology and cognitive impairment in the mouse model of Alzheimer's disease by regulating microglia homeostasis and inhibiting HMGB1 pathway.","authors":"Yuan Xu, Naiyuan Shao, Feng Zhi, Ronghua Chen, Yilin Yang, Jiahui Li, Ying Xia, Ya Peng","doi":"10.1186/s13195-025-01682-1","DOIUrl":"10.1186/s13195-025-01682-1","url":null,"abstract":"<p><strong>Background: </strong>Recent studies suggest that opioid receptor signaling may differentially affect Alzheimer's disease (AD) pathology and the relevant behavioral dysfunctions. However, the precise roles and mechanisms of opioid receptor subtypes in AD pathologies are still unclear with major controversies.</p><p><strong>Methods: </strong>We compared the delta-opioid receptor (DOR)- and mu-opioid receptor (MOR)-mediated effects on AD-associated cognitive deficits, pathologies, neuroinflammations, cell death using transgenic APP/PS1 mouse model and BV2 cell line at behavioral, molecular, and cellular levels. Unpaired t-test and one/two way analysis for variance (ANOVA) were used to analyze statistical significance of the data.</p><p><strong>Results: </strong>We show a distinct role of DOR and its major difference with MOR in AD injury in an APP/PS1 mouse model. DOR activation by UFP-512, but not MOR activation by DAMGO, attenuated cognitive impairment, reduced beta-amyloid (Aβ) production and aggregation, as well as protected the neurons from apoptosis in APP/PS1 mice. DOR and MOR also differentially modulated microglia in APP/PS1 mice and in vitro AD cell model with a DOR-mediated inhibition on the excessive activation of microglia and the release of pro-inflammatory cytokines in AD pathologies. Gene expression profiling further revealed that the alternations in DOR/MOR are closely associated with microglial homeostatic signatures and high mobility group protein B1 (HMGB1) in AD. DOR activation inhibited HMGB1 secretion and its translocation from nuclear to cytoplasm. Our in-vitro studies further confirmed that DOR overexpression mitigated microglial inflammatory response and rescued neurons from AD injury via HMGB1-NF-κB signaling pathway.</p><p><strong>Conclusions: </strong>These novel findings uncover previously unappreciated roles of DOR in neuroprotection against AD injury via modulating microglia-related inflammatory responses.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"35"},"PeriodicalIF":7.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786345/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Retinal optical coherence tomography intensity spatial correlation features as new biomarkers for confirmed Alzheimer's disease.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-01 DOI: 10.1186/s13195-025-01676-z
Zi Jin, Xinmin Wang, Ying Lang, Yufeng Song, Huangxiong Zhan, Wuge Shama, Yingying Shen, Guihua Zeng, Faying Zhou, Hongjian Gao, Shuling Ye, Yanjiang Wang, Fan Lu, Meixiao Shen

Background: The nature and severity of Alzheimer's disease (AD) pathologies in the retina and brain correspond. However, retinal biomarkers need to be validated in clinical cohorts with confirmed AD biomarkers and optical coherence tomography (OCT). The main objective of this study was to investigate whether retinal metrics measured by OCT aid in the early screening and brain pathology monitoring for confirmed AD.

Methods: This was a case-control study. All participants underwent retinal OCT imaging, and neurological examinations, including amyloid-β (Aβ) positron emission tomography. Participants were subdivided into cognitively normal (CN), mild cognitive impairment (MCI), and AD-derived dementia (ADD). Except retinal thickness, we developed the grey level co-occurrence matrix algorithm to extract retinal OCT intensity spatial correlation features (OCT-ISCF), including angular second matrix (ASM), correlation (COR), and homogeneity (HOM), one-way analysis of variance was used to compare the differences in retinal parameters among the groups, and to analyze the correlation with brain Aβ plaques and cognitive scores. The repeatability and robustness of OCT-ISCF were evaluated using experimental and simulation methods.

Results: This study enrolled 82 participants, subdivided into 20 CN, 22 MCI, and 40 ADD. Compared with the CN, the thickness of retinal nerve fiber layer and myoid and ellipsoid zone were significantly thinner (P < 0.05), and ASM, COR, and HOM in several retinal sublayers changed significantly in the ADD (P < 0.05). Notably, the MCI showed significant differences in ASM and COR in the outer segment of photoreceptor compared with the CN (P < 0.05). The changing pattern of OCT-ISCF with interclass correlation coefficients above 0.8 differed from that caused by speckle noise, and was affected by OCT image quality index. Moreover, the retinal OCT-ISCF were more strongly correlated with brain Aβ plaque burden and MoCA scores than retinal thickness. The accuracy using retinal OCT-ISCF (AUC = 0.935, 0.830) was better than that using retinal thickness (AUC = 0.795, 0.705) in detecting ADD and MCI.

Conclusions: The study demonstrates that retinal OCT-ISCF enhance the association and detection efficacy of AD pathology compared to retinal thickness, suggesting retinal OCT-ISCF have the potential to be new biomarkers for AD.

{"title":"Retinal optical coherence tomography intensity spatial correlation features as new biomarkers for confirmed Alzheimer's disease.","authors":"Zi Jin, Xinmin Wang, Ying Lang, Yufeng Song, Huangxiong Zhan, Wuge Shama, Yingying Shen, Guihua Zeng, Faying Zhou, Hongjian Gao, Shuling Ye, Yanjiang Wang, Fan Lu, Meixiao Shen","doi":"10.1186/s13195-025-01676-z","DOIUrl":"10.1186/s13195-025-01676-z","url":null,"abstract":"<p><strong>Background: </strong>The nature and severity of Alzheimer's disease (AD) pathologies in the retina and brain correspond. However, retinal biomarkers need to be validated in clinical cohorts with confirmed AD biomarkers and optical coherence tomography (OCT). The main objective of this study was to investigate whether retinal metrics measured by OCT aid in the early screening and brain pathology monitoring for confirmed AD.</p><p><strong>Methods: </strong>This was a case-control study. All participants underwent retinal OCT imaging, and neurological examinations, including amyloid-β (Aβ) positron emission tomography. Participants were subdivided into cognitively normal (CN), mild cognitive impairment (MCI), and AD-derived dementia (ADD). Except retinal thickness, we developed the grey level co-occurrence matrix algorithm to extract retinal OCT intensity spatial correlation features (OCT-ISCF), including angular second matrix (ASM), correlation (COR), and homogeneity (HOM), one-way analysis of variance was used to compare the differences in retinal parameters among the groups, and to analyze the correlation with brain Aβ plaques and cognitive scores. The repeatability and robustness of OCT-ISCF were evaluated using experimental and simulation methods.</p><p><strong>Results: </strong>This study enrolled 82 participants, subdivided into 20 CN, 22 MCI, and 40 ADD. Compared with the CN, the thickness of retinal nerve fiber layer and myoid and ellipsoid zone were significantly thinner (P < 0.05), and ASM, COR, and HOM in several retinal sublayers changed significantly in the ADD (P < 0.05). Notably, the MCI showed significant differences in ASM and COR in the outer segment of photoreceptor compared with the CN (P < 0.05). The changing pattern of OCT-ISCF with interclass correlation coefficients above 0.8 differed from that caused by speckle noise, and was affected by OCT image quality index. Moreover, the retinal OCT-ISCF were more strongly correlated with brain Aβ plaque burden and MoCA scores than retinal thickness. The accuracy using retinal OCT-ISCF (AUC = 0.935, 0.830) was better than that using retinal thickness (AUC = 0.795, 0.705) in detecting ADD and MCI.</p><p><strong>Conclusions: </strong>The study demonstrates that retinal OCT-ISCF enhance the association and detection efficacy of AD pathology compared to retinal thickness, suggesting retinal OCT-ISCF have the potential to be new biomarkers for AD.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"33"},"PeriodicalIF":7.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786474/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rotation errors in path integration are associated with Alzheimer's disease tau pathology: a cross-sectional study.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-02-01 DOI: 10.1186/s13195-025-01679-w
Lise Colmant, Lisa Quenon, Lara Huyghe, Adrian Ivanoiu, Thomas Gérard, Renaud Lhommel, Pauline Coppens, Yasmine Salman, Vincent Malotaux, Laurence Dricot, Lukas Kunz, Nikolai Axmacher, Philippe Lefèvre, Bernard Hanseeuw

Background: Early Alzheimer's disease diagnosis is crucial for preventive therapy development. Standard neuropsychological evaluation does not identify clinically normal individuals with brain amyloidosis, the first stage of the pathology, defined as preclinical Alzheimer's disease. Spatial navigation assessment, in particular path integration, appears promising to detect preclinical symptoms, as the medial temporal lobe plays a key role in navigation and is the first cortical region affected by tau pathology.

Methods: We have conducted a cross-sectional study. We related the path integration performance of 102 individuals without dementia, aged over 50, to amyloid and tau pathologies, measured using positron emission tomography. We included 75 clinically normal individuals (19 with brain amyloidosis, 56 without) and 27 individuals with mild cognitive impairment (18 with brain amyloidosis, 9 without). We fitted linear mixed models to predict the path integration performances according to amyloid status or tau pathology in the medial temporal lobal, adjusting for age, gender, cognitive status, education, and video game experience. We decomposed the error into rotation and distance errors.

Results: We observed that clinically normal adults with brain amyloidosis (preclinical Alzheimer's disease) had spatial navigation deficits when relying only on self-motion cues. However, they were able to use a landmark to reduce their errors. Individuals with mild cognitive impairment had deficits in path integration that did not improve when a landmark was added in the environment. The amyloid status did not influence performance among individuals with mild cognitive impairment. Among all individuals, rotation, but not distance, errors increased with the level of tau pathology in the medial temporal lobe.

Conclusion: Our results suggest that path integration performance in an environment without external cues allows identifying individuals with preclinical Alzheimer's disease, before overt episodic memory impairment is noticeable. Specifically, we demonstrated that poor angular estimation is an early cognitive marker of tau pathology, whereas distance estimation relates to older ages, not to Alzheimer's disease.

Trial registration: Eudra-CT 2018-003473-94.

{"title":"Rotation errors in path integration are associated with Alzheimer's disease tau pathology: a cross-sectional study.","authors":"Lise Colmant, Lisa Quenon, Lara Huyghe, Adrian Ivanoiu, Thomas Gérard, Renaud Lhommel, Pauline Coppens, Yasmine Salman, Vincent Malotaux, Laurence Dricot, Lukas Kunz, Nikolai Axmacher, Philippe Lefèvre, Bernard Hanseeuw","doi":"10.1186/s13195-025-01679-w","DOIUrl":"10.1186/s13195-025-01679-w","url":null,"abstract":"<p><strong>Background: </strong>Early Alzheimer's disease diagnosis is crucial for preventive therapy development. Standard neuropsychological evaluation does not identify clinically normal individuals with brain amyloidosis, the first stage of the pathology, defined as preclinical Alzheimer's disease. Spatial navigation assessment, in particular path integration, appears promising to detect preclinical symptoms, as the medial temporal lobe plays a key role in navigation and is the first cortical region affected by tau pathology.</p><p><strong>Methods: </strong>We have conducted a cross-sectional study. We related the path integration performance of 102 individuals without dementia, aged over 50, to amyloid and tau pathologies, measured using positron emission tomography. We included 75 clinically normal individuals (19 with brain amyloidosis, 56 without) and 27 individuals with mild cognitive impairment (18 with brain amyloidosis, 9 without). We fitted linear mixed models to predict the path integration performances according to amyloid status or tau pathology in the medial temporal lobal, adjusting for age, gender, cognitive status, education, and video game experience. We decomposed the error into rotation and distance errors.</p><p><strong>Results: </strong>We observed that clinically normal adults with brain amyloidosis (preclinical Alzheimer's disease) had spatial navigation deficits when relying only on self-motion cues. However, they were able to use a landmark to reduce their errors. Individuals with mild cognitive impairment had deficits in path integration that did not improve when a landmark was added in the environment. The amyloid status did not influence performance among individuals with mild cognitive impairment. Among all individuals, rotation, but not distance, errors increased with the level of tau pathology in the medial temporal lobe.</p><p><strong>Conclusion: </strong>Our results suggest that path integration performance in an environment without external cues allows identifying individuals with preclinical Alzheimer's disease, before overt episodic memory impairment is noticeable. Specifically, we demonstrated that poor angular estimation is an early cognitive marker of tau pathology, whereas distance estimation relates to older ages, not to Alzheimer's disease.</p><p><strong>Trial registration: </strong>Eudra-CT 2018-003473-94.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"34"},"PeriodicalIF":7.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786419/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073427","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Glymphatic system clearance and Alzheimer's disease risk: a CSF proteome-wide study.
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-01-31 DOI: 10.1186/s13195-024-01612-7
Natalia Cullell, Giovanni Caruana, Andrea Elias-Mas, Ariane Delgado-Sanchez, Cristina Artero, Maria Teresa Buongiorno, Marta Almería, Nicola J Ray, Sonia A L Correa, Jerzy Krupinski

Background: The emerging evidence of the role of the glymphatic system (GS) in Alzheimer's disease (AD) provides new opportunities for intervention from the earliest stages of the disease. The aim of the study is to evaluate the efficacy of GS in AD to identify new disease biomarkers.

Methods: We performed a two-stage proteomic study to evaluate the GS health using intravenous gadolinium-based contrast agent (GBCA) with serial T1 3T magnetic resonance imaging (MRI) in individuals with amnestic mild cognitive impairment (aMCI). In Stage 1 (evaluated in the Cohort 1 of aMCI participants (n = 11)), we correlated the levels of 7K cerebrospinal fluid (CSF) proteins (estimated by SOMAscan) with GS health in 78 Freesurfer-segmented brain regions of interest (ROIs).

Results: A total of seven different proteins were significantly associated with GS health (p-value < 6.4 × 10-4). The stronger correlations were identified for NSUN6, GRAAK, OLFML3, ACTN2, RUXF, SHPS1 and TIM-4. A pathway enrichment analysis revealed that the proteins associated with GS health were mainly implicated in neurodegenerative processes, immunity and inflammation. In Stage 2, we validated these proteomic results in a new cohort of aMCI participants (with and without evidence of AD pathology in CSF (aMCI(-) and aMCI/AD( +); n = 22 and 7, respectively) and healthy controls (n = 10). Proteomic prediction models were generated in each ROI. These were compared with demographic-only models for identifying participants with aMCI(-) and aMCI/AD( +) vs controls. This analysis was repeated to determine if the models could identify those with aMCI/AD( +) from both aMCI(-) and controls. The proteomic models were found to outperform the demographic-only models.

Conclusions: Our study identifies proteins linked with GS health and involved the immune system in aMCI participants.

{"title":"Glymphatic system clearance and Alzheimer's disease risk: a CSF proteome-wide study.","authors":"Natalia Cullell, Giovanni Caruana, Andrea Elias-Mas, Ariane Delgado-Sanchez, Cristina Artero, Maria Teresa Buongiorno, Marta Almería, Nicola J Ray, Sonia A L Correa, Jerzy Krupinski","doi":"10.1186/s13195-024-01612-7","DOIUrl":"10.1186/s13195-024-01612-7","url":null,"abstract":"<p><strong>Background: </strong>The emerging evidence of the role of the glymphatic system (GS) in Alzheimer's disease (AD) provides new opportunities for intervention from the earliest stages of the disease. The aim of the study is to evaluate the efficacy of GS in AD to identify new disease biomarkers.</p><p><strong>Methods: </strong>We performed a two-stage proteomic study to evaluate the GS health using intravenous gadolinium-based contrast agent (GBCA) with serial T1 3T magnetic resonance imaging (MRI) in individuals with amnestic mild cognitive impairment (aMCI). In Stage 1 (evaluated in the Cohort 1 of aMCI participants (n = 11)), we correlated the levels of 7K cerebrospinal fluid (CSF) proteins (estimated by SOMAscan) with GS health in 78 Freesurfer-segmented brain regions of interest (ROIs).</p><p><strong>Results: </strong>A total of seven different proteins were significantly associated with GS health (p-value < 6.4 × 10<sup>-4</sup>). The stronger correlations were identified for NSUN6, GRAAK, OLFML3, ACTN2, RUXF, SHPS1 and TIM-4. A pathway enrichment analysis revealed that the proteins associated with GS health were mainly implicated in neurodegenerative processes, immunity and inflammation. In Stage 2, we validated these proteomic results in a new cohort of aMCI participants (with and without evidence of AD pathology in CSF (aMCI(-) and aMCI/AD( +); n = 22 and 7, respectively) and healthy controls (n = 10). Proteomic prediction models were generated in each ROI. These were compared with demographic-only models for identifying participants with aMCI(-) and aMCI/AD( +) vs controls. This analysis was repeated to determine if the models could identify those with aMCI/AD( +) from both aMCI(-) and controls. The proteomic models were found to outperform the demographic-only models.</p><p><strong>Conclusions: </strong>Our study identifies proteins linked with GS health and involved the immune system in aMCI participants.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"31"},"PeriodicalIF":7.9,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11786353/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073346","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
RADAR-AD: assessment of multiple remote monitoring technologies for early detection of Alzheimer's disease. RADAR-AD:评估用于早期检测阿尔茨海默病的多种远程监控技术。
IF 7.9 1区 医学 Q1 CLINICAL NEUROLOGY Pub Date : 2025-01-27 DOI: 10.1186/s13195-025-01675-0
Manuel Lentzen, Srinivasan Vairavan, Marijn Muurling, Vasilis Alepopoulos, Alankar Atreya, Merce Boada, Casper de Boer, Pauline Conde, Jelena Curcic, Giovanni Frisoni, Samantha Galluzzi, Martha Therese Gjestsen, Mara Gkioka, Margarita Grammatikopoulou, Lucrezia Hausner, Chris Hinds, Ioulietta Lazarou, Alexandre de Mendonça, Spiros Nikolopoulos, Dorota Religa, Gaetano Scebba, Pieter Jelle Visser, Gayle Wittenberg, Vaibhav A Narayan, Neva Coello, Anna-Katharine Brem, Dag Aarsland, Holger Fröhlich

Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting millions worldwide, leading to cognitive and functional decline. Early detection and intervention are crucial for enhancing the quality of life of patients and their families. Remote Monitoring Technologies (RMTs) offer a promising solution for early detection by tracking changes in behavioral and cognitive functions, such as memory, language, and problem-solving skills. Timely detection of these symptoms can facilitate early intervention, potentially slowing disease progression and enabling appropriate treatment and care.

Methods: The RADAR-AD study was designed to evaluate the accuracy and validity of multiple RMTs in detecting functional decline across various stages of AD in a real-world setting, compared to standard clinical rating scales. Our approach involved a univariate analysis using Analysis of Covariance (ANCOVA) to analyze individual features of six RMTs while adjusting for variables such as age, sex, years of education, clinical site, BMI and season. Additionally, we employed four machine learning classifiers - Logistic Regression, Decision Tree, Random Forest, and XGBoost - using a nested cross-validation approach to assess the discriminatory capabilities of the RMTs.

Results: The ANCOVA results indicated significant differences between healthy and AD subjects regarding reduced physical activity, less REM sleep, altered gait patterns, and decreased cognitive functioning. The machine-learning-based analysis demonstrated that RMT-based models could identify subjects in the prodromal stage with an Area Under the ROC Curve of 73.0 %. In addition, our findings show that the Amsterdam iADL questionnaire has high discriminatory abilities.

Conclusions: RMTs show promise in AD detection already in the prodromal stage. Using them could allow for earlier detection and intervention, thereby improving patients' quality of life. Furthermore, the Amsterdam iADL questionnaire holds high potential when employed remotely.

{"title":"RADAR-AD: assessment of multiple remote monitoring technologies for early detection of Alzheimer's disease.","authors":"Manuel Lentzen, Srinivasan Vairavan, Marijn Muurling, Vasilis Alepopoulos, Alankar Atreya, Merce Boada, Casper de Boer, Pauline Conde, Jelena Curcic, Giovanni Frisoni, Samantha Galluzzi, Martha Therese Gjestsen, Mara Gkioka, Margarita Grammatikopoulou, Lucrezia Hausner, Chris Hinds, Ioulietta Lazarou, Alexandre de Mendonça, Spiros Nikolopoulos, Dorota Religa, Gaetano Scebba, Pieter Jelle Visser, Gayle Wittenberg, Vaibhav A Narayan, Neva Coello, Anna-Katharine Brem, Dag Aarsland, Holger Fröhlich","doi":"10.1186/s13195-025-01675-0","DOIUrl":"10.1186/s13195-025-01675-0","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting millions worldwide, leading to cognitive and functional decline. Early detection and intervention are crucial for enhancing the quality of life of patients and their families. Remote Monitoring Technologies (RMTs) offer a promising solution for early detection by tracking changes in behavioral and cognitive functions, such as memory, language, and problem-solving skills. Timely detection of these symptoms can facilitate early intervention, potentially slowing disease progression and enabling appropriate treatment and care.</p><p><strong>Methods: </strong>The RADAR-AD study was designed to evaluate the accuracy and validity of multiple RMTs in detecting functional decline across various stages of AD in a real-world setting, compared to standard clinical rating scales. Our approach involved a univariate analysis using Analysis of Covariance (ANCOVA) to analyze individual features of six RMTs while adjusting for variables such as age, sex, years of education, clinical site, BMI and season. Additionally, we employed four machine learning classifiers - Logistic Regression, Decision Tree, Random Forest, and XGBoost - using a nested cross-validation approach to assess the discriminatory capabilities of the RMTs.</p><p><strong>Results: </strong>The ANCOVA results indicated significant differences between healthy and AD subjects regarding reduced physical activity, less REM sleep, altered gait patterns, and decreased cognitive functioning. The machine-learning-based analysis demonstrated that RMT-based models could identify subjects in the prodromal stage with an Area Under the ROC Curve of 73.0 %. In addition, our findings show that the Amsterdam iADL questionnaire has high discriminatory abilities.</p><p><strong>Conclusions: </strong>RMTs show promise in AD detection already in the prodromal stage. Using them could allow for earlier detection and intervention, thereby improving patients' quality of life. Furthermore, the Amsterdam iADL questionnaire holds high potential when employed remotely.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"29"},"PeriodicalIF":7.9,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11771057/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Alzheimer's Research & Therapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1