Alzheimer's Research & Therapy最新文献

筛选
英文 中文
Prognostic model for predicting Alzheimer's disease conversion using functional connectome manifolds. 利用功能连接组流形预测阿尔茨海默病转归的预后模型。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-09 DOI: 10.1186/s13195-024-01589-3
Sunghun Kim, Mansu Kim, Jong-Eun Lee, Bo-Yong Park, Hyunjin Park
{"title":"Prognostic model for predicting Alzheimer's disease conversion using functional connectome manifolds.","authors":"Sunghun Kim, Mansu Kim, Jong-Eun Lee, Bo-Yong Park, Hyunjin Park","doi":"10.1186/s13195-024-01589-3","DOIUrl":"10.1186/s13195-024-01589-3","url":null,"abstract":"<p><strong>Background: </strong>Early detection of Alzheimer's disease (AD) is essential for timely management and consideration of therapeutic options; therefore, detecting the risk of conversion from mild cognitive impairment (MCI) to AD is crucial during neurodegenerative progression. Existing neuroimaging studies have mostly focused on group differences between individuals with MCI (or AD) and cognitively normal (CN), discarding the temporal information of conversion time. Here, we aimed to develop a prognostic model for AD conversion using functional connectivity (FC) and Cox regression suitable for conversion event modeling.</p><p><strong>Methods: </strong>We developed a prognostic model using a large-scale Alzheimer's Disease Neuroimaging Initiative dataset, and it was validated using external data obtained from the Open Access Series of Imaging Studies. We considered individuals who were initially CN or had MCI but progressed to AD and those with MCI with no progression to AD during the five-year follow-up period. As the exact conversion time to AD is unknown, we inferred this information using imputation approaches. We generated cortex-wide principal FC gradients using manifold learning techniques and computed subcortical-weighted manifold degrees from baseline functional magnetic resonance imaging data. A penalized Cox regression model with an elastic net penalty was adopted to define a risk score predicting the risk of conversion to AD, using FC gradients and clinical factors as regressors.</p><p><strong>Results: </strong>Our prognostic model predicted the conversion risk and confirmed the role of imaging-derived manifolds in the conversion risk. The brain regions that largely contributed to predicting AD conversion were the heteromodal association and visual cortices, as well as the caudate and hippocampus. Our risk score based on Cox regression was consistent with the expected disease trajectories and correlated with positron emission tomography tracer uptake and symptom severity, reinforcing its clinical usefulness. Our findings were validated using an independent dataset. The cross-sectional application of our model showed a higher risk for AD than that for MCI, which correlated with symptom severity scores in the validation dataset.</p><p><strong>Conclusion: </strong>We proposed a prognostic model predicting the risk of conversion to AD. The associated risk score may provide insights for early intervention in individuals at risk of AD conversion.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"217"},"PeriodicalIF":7.9,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465528/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142387251","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
Interactive effect of diabetes mellitus and subclinical MRI markers of cerebrovascular disease on cognitive decline and incident dementia: a memory-clinic study. 糖尿病和亚临床磁共振成像脑血管疾病标志物对认知能力下降和痴呆症的交互影响:一项记忆门诊研究。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-03 DOI: 10.1186/s13195-024-01577-7
Jiangbo Cui, Caroline Robert, Chia May Teh, Eddie Chong Jun Yi, Joyce R Chong, Boon Yeow Tan, Narayanaswamy Venketasubramanian, Mitchell K P Lai, Christopher Chen, Saima Hilal
{"title":"Interactive effect of diabetes mellitus and subclinical MRI markers of cerebrovascular disease on cognitive decline and incident dementia: a memory-clinic study.","authors":"Jiangbo Cui, Caroline Robert, Chia May Teh, Eddie Chong Jun Yi, Joyce R Chong, Boon Yeow Tan, Narayanaswamy Venketasubramanian, Mitchell K P Lai, Christopher Chen, Saima Hilal","doi":"10.1186/s13195-024-01577-7","DOIUrl":"10.1186/s13195-024-01577-7","url":null,"abstract":"<p><strong>Background: </strong>Cognitive impairment is an increasingly recognized comorbidity of diabetes, yet the mechanisms underlying this association remain poorly understood. This knowledge gap has contributed to conflicting findings regarding the impact of diabetes on long-term cognitive outcomes in older adults. The presence of cerebrovascular disease (CeVD) may potentially modify this relationship. However, interactive effect between diabetes and subclinical MRI markers of CeVD on cognitive trajectories and incident dementia remains unexplored.</p><p><strong>Methods: </strong>A total of 654 participants underwent brain MRI at baseline, from whom 614 with at least one follow-up were selected for longitudinal analysis. Cognitive tests were performed annually up to 5 years. CeVD markers of interest were lacunes, white matter hyperintensities (WMHs), cerebral microbleeds (CMBs), cortical microinfarcts (CMIs), intracranial stenosis (ICS), and cortical infarcts. Blood-based Alzheimer biomarkers, including p-tau181 and p-tau181/Aβ42 ratio, were used as indicators of Alzheimer pathology.</p><p><strong>Results: </strong>At baseline, diabetes was associated with lower cognitive performance and higher burden of CeVD, but not p-tau181 or p-tau181/Aβ42 ratio. Longitudinally, we found an interactive effect of diabetes and WMHs, rather than an independent effect of diabetes, on cognitive decline and dementia risk. Subgroup analyses showed association of diabetes with cognitive outcomes was stronger in participants with high WMHs load but non-significant in those with low WMHs load. Moreover, these associations remained unchanged after adjusting for blood-based Alzheimer biomarkers.</p><p><strong>Conclusions: </strong>The effect of diabetes on cognitive decline is contingent upon the presence of WMHs and independent of Alzheimer's pathology. This finding raises the possibility of utilizing WMHs as an imaging biomarker to identify diabetic subgroup at greater risk of developing cognitive impairment. Furthermore, therapeutic interventions targeting WMHs may prevent cognitive deterioration in older adults with diabetes.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"214"},"PeriodicalIF":7.9,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370744","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
Public perceptions related to healthcare preparedness to anti-amyloid therapies for Alzheimer's Disease in Japan. 日本公众对阿尔茨海默病抗淀粉样蛋白疗法的医疗准备情况的看法。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-03 DOI: 10.1186/s13195-024-01568-8
Kenichiro Sato, Yoshiki Niimi, Ryoko Ihara, Atsushi Iwata, Kazushi Suzuki, Kiyotaka Nemoto, Tetsuaki Arai, Shinji Higashi, Ataru Igarashi, Kensaku Kasuga, Shuichi Awata, Takeshi Iwatsubo
{"title":"Public perceptions related to healthcare preparedness to anti-amyloid therapies for Alzheimer's Disease in Japan.","authors":"Kenichiro Sato, Yoshiki Niimi, Ryoko Ihara, Atsushi Iwata, Kazushi Suzuki, Kiyotaka Nemoto, Tetsuaki Arai, Shinji Higashi, Ataru Igarashi, Kensaku Kasuga, Shuichi Awata, Takeshi Iwatsubo","doi":"10.1186/s13195-024-01568-8","DOIUrl":"10.1186/s13195-024-01568-8","url":null,"abstract":"<p><strong>Background: </strong>The approval of lecanemab, an anti-amyloid therapy for Alzheimer's disease (AD), necessitates addressing healthcare preparedness for disease-modifying treatment (DMT) to ensure appropriate, safe, and sustainable drug administration. Understanding public perceptions on this matter is crucial. We aimed to assess discrepancies and similarities in the perceptions of Japanese trial-ready cohort study ('J-TRC webstudy') participants and clinical specialists in the fields of dementia treatment and radiology, concerning affairs related to challenges in DMT preparedness.</p><p><strong>Methods: </strong>This was a cross-sectional prospective observational study conducted in November-December 2023. The J-TRC webstudy participants were invited to participate in an online survey using Google Forms, and clinical specialists were invited to complete a mail-based survey. Main questionnaire items had been designed to be common in both surveys, and their responses were analyzed for participant attributes, interests, attitudes, expectations, and concerns about DMTs without specifying lecanemab.</p><p><strong>Results: </strong>Responses were obtained from n = 2,050 J-TRC webstudy participants and n = 1,518 clinical specialists. Compared to specialists, more J-TRC respondents perceived the eligible proportion for DMT as smaller (59.1% versus 30.7%), perceived the eligible severity for DMT as more limited (58.0% versus 24.5%), and perceived the efficacy of DMT as slightly more encouraging (29.3% versus 34.8%). In terms of treatment prioritization, both J-TRC respondents and specialist respondents exhibited similar levels of acceptance for prioritizing patients to treat: e.g., approximately two-thirds endorsed patient prioritization under hypothetical resource constraints or other reasons. A medical rationale emerged as the most compelling reason for acceptance of patient prioritization across the surveys. In contrast, the need to address vulnerable populations was the reason that led to the least acceptance of prioritization, followed by economic considerations.</p><p><strong>Conclusions: </strong>Our findings offer valuable insights into the discrepancies in knowledge and perception between patients and healthcare providers. This could enhance the delivery of patient information in clinical settings and inform the discussion surrounding patient prioritization strategies.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"205"},"PeriodicalIF":7.9,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448067/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363967","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
High-affinity antibodies specific to the core region of the tau protein exhibit diagnostic and therapeutic potential for Alzheimer's disease. 针对 tau 蛋白核心区域的特异性高亲和力抗体具有诊断和治疗阿尔茨海默病的潜力。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-02 DOI: 10.1186/s13195-024-01561-1
Mohammad Arastoo, Lewis K Penny, Richard Lofthouse, Aya Abdallah, Anna Abrahamsson, Pietro Marini, Valeria Melis, Gernot Riedel, Charles R Harrington, Claude M Wischik, Andrew Porter, Soumya Palliyil
{"title":"High-affinity antibodies specific to the core region of the tau protein exhibit diagnostic and therapeutic potential for Alzheimer's disease.","authors":"Mohammad Arastoo, Lewis K Penny, Richard Lofthouse, Aya Abdallah, Anna Abrahamsson, Pietro Marini, Valeria Melis, Gernot Riedel, Charles R Harrington, Claude M Wischik, Andrew Porter, Soumya Palliyil","doi":"10.1186/s13195-024-01561-1","DOIUrl":"10.1186/s13195-024-01561-1","url":null,"abstract":"<p><strong>Background: </strong>Recent advances in blood-based biomarker discovery are paving the way for simpler, more accessible diagnostic tools that can detect early signs of Alzheimer's disease (AD). Recent successes in the development of amyloid-targeting immunotherapy approaches mark an important advancement in providing new options for the treatment of AD. We have developed a set of high-affinity monoclonal antibodies (mAbs) to tau protein that have the potential as tools for diagnosis and treatment of AD.</p><p><strong>Methods: </strong>Sheep were immunised with either full-length tau (1-441) or truncated paired helical filament (PHF)-core tau (297-391). A stringent bio-panning and epitope selection strategy, with a particular focus directed to epitopes within the disease-relevant PHF-core tau, was used to identify single-chain antibodies (scAbs). These scAbs were ranked by affinity for each epitope class, with leads converted to high-affinity mAbs. These antibodies and their potential utility were assessed by their performance in tau immunoassays, as well as their ability to prevent tau aggregation and propagation. Further characterisation of these antibodies was performed by immunohistochemical staining of brain sections and immuno-gold electronmicroscopy of isolated PHFs.</p><p><strong>Results: </strong>Our work resulted in a set of high-affinity antibodies reacting with multiple epitopes spanning the entire tau protein molecule. The tau antibodies directed against the core tau unit of the PHF inhibited pathological aggregation and seeding using several biochemical and cell assay systems. Through staining of brain sections and PHFs, the panel of antibodies revealed which tau epitopes were available, truncated, or occluded. In addition, highly sensitive immunoassays were developed with the ability to distinguish between and quantify various tau fragments.</p><p><strong>Conclusion: </strong>This article introduces an alternative immunodiagnostic approach based on the concept of a \"tauosome\" - the diverse set of tau fragments present within biological fluids. The development of an antibody panel that can distinguish a range of different tau fragments provides the basis for a novel approach to potential diagnosis and monitoring of disease progression. Our results further support the notion that tau immunotherapy targeting the PHF-core needs to combine appropriate selection of both the target epitope and antibody affinity to optimise therapeutic potential.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"209"},"PeriodicalIF":7.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448309/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363964","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 analysis reveals the key factors involved in the severity of the Alzheimer's disease. 多组学分析揭示了阿尔茨海默病严重程度的关键因素。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-02 DOI: 10.1186/s13195-024-01578-6
Lingqi Meng, Han Jin, Burak Yulug, Ozlem Altay, Xiangyu Li, Lutfu Hanoglu, Seyda Cankaya, Ebru Coskun, Ezgi Idil, Rahim Nogaylar, Ahmet Ozsimsek, Saeed Shoaie, Hasan Turkez, Jens Nielsen, Cheng Zhang, Jan Borén, Mathias Uhlén, Adil Mardinoglu
{"title":"Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease.","authors":"Lingqi Meng, Han Jin, Burak Yulug, Ozlem Altay, Xiangyu Li, Lutfu Hanoglu, Seyda Cankaya, Ebru Coskun, Ezgi Idil, Rahim Nogaylar, Ahmet Ozsimsek, Saeed Shoaie, Hasan Turkez, Jens Nielsen, Cheng Zhang, Jan Borén, Mathias Uhlén, Adil Mardinoglu","doi":"10.1186/s13195-024-01578-6","DOIUrl":"10.1186/s13195-024-01578-6","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a debilitating neurodegenerative disorder with a global impact, yet its pathogenesis remains poorly understood. While age, metabolic abnormalities, and accumulation of neurotoxic substances are potential risk factors for AD, their effects are confounded by other factors. To address this challenge, we first utilized multi-omics data from 87 well phenotyped AD patients and generated plasma proteomics and metabolomics data, as well as gut and saliva metagenomics data to investigate the molecular-level alterations accounting the host-microbiome interactions. Second, we analyzed individual omics data and identified the key parameters involved in the severity of the dementia in AD patients. Next, we employed Artificial Intelligence (AI) based models to predict AD severity based on the significantly altered features identified in each omics analysis. Based on our integrative analysis, we found the clinical relevance of plasma proteins, including SKAP1 and NEFL, plasma metabolites including homovanillate and glutamate, and Paraprevotella clara in gut microbiome in predicting the AD severity. Finally, we validated the predictive power of our AI based models by generating additional multi-omics data from the same group of AD patients by following up for 3 months. Hence, we observed that these results may have important implications for the development of potential diagnostic and therapeutic approaches for AD patients.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"213"},"PeriodicalIF":7.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448018/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363966","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
Brain Iron in signature regions relating to cognitive aging in older adults: the Taizhou Imaging Study. 与老年人认知老化有关的特征区域的脑铁:台州影像研究。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-02 DOI: 10.1186/s13195-024-01575-9
Rui Li, Yi-Ren Fan, Ying-Zhe Wang, He-Yang Lu, Pei-Xi Li, Qiang Dong, Yan-Feng Jiang, Xing-Dong Chen, Mei Cui
{"title":"Brain Iron in signature regions relating to cognitive aging in older adults: the Taizhou Imaging Study.","authors":"Rui Li, Yi-Ren Fan, Ying-Zhe Wang, He-Yang Lu, Pei-Xi Li, Qiang Dong, Yan-Feng Jiang, Xing-Dong Chen, Mei Cui","doi":"10.1186/s13195-024-01575-9","DOIUrl":"10.1186/s13195-024-01575-9","url":null,"abstract":"<p><strong>Background: </strong>Recent magnetic resonance imaging (MRI) studies have established that brain iron accumulation might accelerate cognitive decline in Alzheimer's disease (AD) patients. Both normal aging and AD are associated with cerebral atrophy in specific regions. However, no studies have investigated aging- and AD-selective iron deposition-related cognitive changes during normal aging. Here, we applied quantitative susceptibility mapping (QSM) to detect iron levels in cortical signature regions and assessed the relationships among iron, atrophy, and cognitive changes in older adults.</p><p><strong>Methods: </strong>In this Taizhou Imaging Study, 770 older adults (mean age 62.0 ± 4.93 years, 57.5% women) underwent brain MRI to measure brain iron and atrophy, of whom 219 underwent neuropsychological tests nearly every 12 months for up to a mean follow-up of 2.68 years. Global cognition was assessed using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). Domain-specific cognitive scores were obtained from MoCA subscore components. Regional analyses were performed for cortical regions and 2 signature regions where atrophy affected by aging and AD only: Aging (AG) -specific and AD signature meta-ROIs. The QSM and cortical morphometry means of the above ROIs were also computed.</p><p><strong>Results: </strong>Significant associations were found between QSM levels and cognitive scores. In particular, after adjusting for cortical thickness of regions of interest (ROIs), participants in the upper tertile of the cortical and AG-specific signature QSM exhibited worse ZMMSE than did those in the lower tertile [ <math><mrow><mspace></mspace> <mi>β</mi> <mspace></mspace></mrow> </math> = -0.104, p = 0.026; <math><mrow><mspace></mspace> <mi>β</mi> <mspace></mspace></mrow> </math> = -0.118, p = 0.021, respectively]. Longitudinal analysis suggested that QSM values in all ROIs might predict decline in ZMoCA and key domains such as attention and visuospatial function (all p < 0.05). Furthermore, iron levels were negatively correlated with classic MRI markers of cortical atrophy (cortical thickness, gray matter volume, and local gyrification index) in total, AG-specific signature and AD signature regions (all p < 0.05).</p><p><strong>Conclusion: </strong>AG- and AD-selective iron deposition was associated with atrophy and cognitive decline in elderly people, highlighting its potential as a neuroimaging marker for cognitive aging.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"211"},"PeriodicalIF":7.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448274/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363962","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
Cholinesterase inhibitor use in amyloid PET-negative mild cognitive impairment and cognitive changes. 在淀粉样蛋白 PET 阴性的轻度认知障碍患者中使用胆碱酯酶抑制剂以及认知变化。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-02 DOI: 10.1186/s13195-024-01580-y
Jung-Min Pyun, Young Ho Park, Min Ju Kang, SangYun Kim
{"title":"Cholinesterase inhibitor use in amyloid PET-negative mild cognitive impairment and cognitive changes.","authors":"Jung-Min Pyun, Young Ho Park, Min Ju Kang, SangYun Kim","doi":"10.1186/s13195-024-01580-y","DOIUrl":"10.1186/s13195-024-01580-y","url":null,"abstract":"<p><strong>Background: </strong>Cholinesterase inhibitors (ChEIs) are prescribed for Alzheimer's disease (AD) and sometimes for mild cognitive impairment (MCI) without knowing underlying pathologies and its effect on cognition. We investigated the frequency of ChEI prescriptions in amyloid-negative MCI and their association with cognitive changes in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort.</p><p><strong>Methods: </strong>We included participants with amyloid positron emission tomography (PET)-negative MCI from the ADNI. We analyzed the associations of ChEI use with cognitive changes, brain volume, and cerebrospinal fluid (CSF) total tau (t-tau), hyperphosphorylated tau<sub>181</sub> (p-tau<sub>181</sub>), and p-tau<sub>181</sub>/t-tau ratio.</p><p><strong>Results: </strong>ChEIs were prescribed in 27.4% of amyloid PET-negative MCI and were associated with faster cognitive decline, reduced baseline hippocampal volume and entorhinal cortical thickness, and a longitudinal decrease in the frontal lobe cortical thickness.</p><p><strong>Conclusions: </strong>The association between ChEI use and accelerated cognitive decline may stem from underlying pathologies involving reduced hippocampal volume, entorhinal cortical thickness and faster frontal lobe atrophy. We suggest that ChEI use in amyloid PET-negative MCI patients might need further consideration, and studies investigating the causality between ChEI use and cognitive decline are warranted in the future.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"210"},"PeriodicalIF":7.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448210/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363963","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
Inflammatory proteins associated with Alzheimer's disease reduced by a GLP1 receptor agonist: a post hoc analysis of the EXSCEL randomized placebo controlled trial. GLP1 受体激动剂可减少与阿尔茨海默病相关的炎症蛋白:EXSCEL 随机安慰剂对照试验的事后分析。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-02 DOI: 10.1186/s13195-024-01573-x
Ivan Koychev, Graham Reid, Maggie Nguyen, Robert J Mentz, Dan Joyce, Svati H Shah, Rury R Holman
{"title":"Inflammatory proteins associated with Alzheimer's disease reduced by a GLP1 receptor agonist: a post hoc analysis of the EXSCEL randomized placebo controlled trial.","authors":"Ivan Koychev, Graham Reid, Maggie Nguyen, Robert J Mentz, Dan Joyce, Svati H Shah, Rury R Holman","doi":"10.1186/s13195-024-01573-x","DOIUrl":"10.1186/s13195-024-01573-x","url":null,"abstract":"<p><strong>Background: </strong>Glucagon-like peptide-1 receptor agonists are a viable option for the prevention of Alzheimer's disease (AD) but the mechanisms of this potential disease modifying action are unclear. We investigated the effects of once-weekly exenatide (EQW) on AD associated proteomic clusters.</p><p><strong>Methods: </strong>The Exenatide Study of Cardiovascular Event Lowering study compared the cardiovascular effects of EQW 2 mg with placebo in 13,752 people with type 2 diabetes mellitus. 4,979 proteins were measured (Somascan V0.4) on baseline and 1-year plasma samples of 3,973 participants. C-reactive protein (CRP), ficolin-2 (FCN2), plasminogen activator inhibitor 1 (PAI-1), soluble vascular cell adhesion protein 1 (sVCAM1) and 4 protein clusters were tested in multivariable mixed models.</p><p><strong>Results: </strong>EQW affected FCN2 (Cohen's d -0.019), PAI-1 (Cohen's d -0.033), sVCAM-1 (Cohen's d 0.035) and a cytokine-cytokine cluster (Cohen's d 0.037) significantly compared with placebo. These effects were sustained in individuals over the age of 65 but not in those under 65.</p><p><strong>Conclusions: </strong>EQW treatment was associated with significant change in inflammatory proteins associated with AD.</p><p><strong>Trial registration: </strong>EXSCEL is registered on ClinicalTrials.gov: NCT01144338 on 10th of June 2010.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"212"},"PeriodicalIF":7.9,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448378/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363965","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
Impact of APOE ε4 and ε2 on plasma neurofilament light chain and cognition in autosomal dominant Alzheimer's disease. APOE ε4和ε2对常染色体显性阿尔茨海默病患者血浆神经丝蛋白轻链和认知能力的影响
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-10-01 DOI: 10.1186/s13195-024-01572-y
Stephanie Langella, Kyra Bonta, Yinghua Chen, Yi Su, Daniel Vasquez, David Aguillon, Natalia Acosta-Baena, Ana Y Baena, Gloria Garcia-Ospina, Margarita Giraldo-Chica, Victoria Tirado, Claudia Muñoz, Silvia Ríos-Romenets, Claudia Guzman-Martínez, Jeremy J Pruzin, Valentina Ghisays, Joseph F Arboleda-Velasquez, Kenneth S Kosik, Pierre N Tariot, Eric M Reiman, Francisco Lopera, Yakeel T Quiroz
{"title":"Impact of APOE ε4 and ε2 on plasma neurofilament light chain and cognition in autosomal dominant Alzheimer's disease.","authors":"Stephanie Langella, Kyra Bonta, Yinghua Chen, Yi Su, Daniel Vasquez, David Aguillon, Natalia Acosta-Baena, Ana Y Baena, Gloria Garcia-Ospina, Margarita Giraldo-Chica, Victoria Tirado, Claudia Muñoz, Silvia Ríos-Romenets, Claudia Guzman-Martínez, Jeremy J Pruzin, Valentina Ghisays, Joseph F Arboleda-Velasquez, Kenneth S Kosik, Pierre N Tariot, Eric M Reiman, Francisco Lopera, Yakeel T Quiroz","doi":"10.1186/s13195-024-01572-y","DOIUrl":"10.1186/s13195-024-01572-y","url":null,"abstract":"<p><strong>Background: </strong>Apolipoprotein E (APOE) genotypes have been suggested to influence cognitive impairment and clinical onset in presenilin-1 (PSEN1) E280A carriers for autosomal dominant Alzheimer's disease (ADAD). Less is known about their impact on the trajectory of biomarker changes. Neurofilament light chain (NfL), a marker of neurodegeneration, begins to accumulate in plasma about 20 years prior to the clinical onset of ADAD. In this study we investigated the impact of APOE ε4 and ε2 variants on age-related plasma NfL increases and cognition in PSEN1 E280A mutation carriers.</p><p><strong>Methods: </strong>We analyzed cross-sectional data from PSEN1 E280A mutation carriers and non-carriers recruited from the Alzheimer's Prevention Initiative Registry of ADAD. All participants over 18 years with available APOE genotype, plasma NfL, and neuropsychological evaluation were included in this study. APOE genotypes and plasma NfL concentrations were characterized for each participant. Cubic spline models using a Hamiltonian Markov chain Monte Carlo method were used to characterize the respective impact of at least one APOE ε4 or ε2 allele on age-related log-transformed plasma NfL increases. Linear regression models were estimated to explore the impact of APOE ε4 and ε2 variants and plasma NfL on a composite cognitive test score in the ADAD mutation carrier and non-carrier groups.</p><p><strong>Results: </strong>Analyses included 788 PSEN1 E280A mutation carriers (169 APOE ε4 + , 114 ε2 +) and 650 mutation non-carriers (165 APOE ε4 + , 80 ε2 +), aged 18-75 years. APOE ε4 allele carriers were distinguished from ε4 non-carriers by greater age-related NfL elevations in the ADAD mutation carrier group, beginning about three years after the mutation carriers' estimated median age at mild cognitive impairment onset. APOE ε2 allele carriers had lower plasma NfL concentrations than ε2 non-carriers in both the ADAD mutation carrier and non-carrier groups, unrelated to age, and an attenuated relationship between higher NfL levels on cognitive decline in the ADAD mutation carrier group.</p><p><strong>Conclusions: </strong>APOE ε4 accelerates age-related plasma NfL increases and APOE ε2 attenuates the relationship between higher plasma NfL levels and cognitive decline in ADAD. NfL may be a useful biomarker to assess clinical efficacy of APOE-modifying drugs with the potential to help in the treatment and prevention of ADAD.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"208"},"PeriodicalIF":7.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443799/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142360939","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
Gene-variant specific effects of plasma amyloid-β levels in Swedish autosomal dominant Alzheimer disease. 瑞典常染色体显性阿尔茨海默病血浆淀粉样蛋白-β水平的基因变异特异性影响。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-09-25 DOI: 10.1186/s13195-024-01574-w
Charlotte Johansson, Steinunn Thordardottir, José Laffita-Mesa, Josef Pannee, Elena Rodriguez-Vieitez, Henrik Zetterberg, Kaj Blennow, Caroline Graff
{"title":"Gene-variant specific effects of plasma amyloid-β levels in Swedish autosomal dominant Alzheimer disease.","authors":"Charlotte Johansson, Steinunn Thordardottir, José Laffita-Mesa, Josef Pannee, Elena Rodriguez-Vieitez, Henrik Zetterberg, Kaj Blennow, Caroline Graff","doi":"10.1186/s13195-024-01574-w","DOIUrl":"https://doi.org/10.1186/s13195-024-01574-w","url":null,"abstract":"<p><strong>Background: </strong>Several blood-based biomarkers offer the opportunity of in vivo detection of brain pathology and neurodegeneration in Alzheimer disease with high specificity and sensitivity, but the performance of amyloid-β (Aβ) measurements remains under evaluation. Autosomal dominant Alzheimer disease (ADAD) with mutations in PSEN1, PSEN2 and APP can be studied as a model for sporadic Alzheimer disease. However, clarifying the genetic effects on the Aβ-levels in different matrices such as cerebrospinal fluid or plasma is crucial for generalizability and utility of data. We aimed to explore plasma Aβ concentrations over the Alzheimer disease continuum in a longitudinal cohort of genetic Alzheimer disease.</p><p><strong>Methods: </strong>92 plasma samples were collected from at-risk individuals (n = 47) in a Swedish cohort of ADAD, including 18 mutation carriers (13 APPswe (p.KM670/671NL) MC), 5 PSEN1 (p.H163Y) MC) and 29 non-carriers (NC) as the reference group. Concentrations of Aβ1-38, Aβ1-40 and Aβ1-42 were analyzed in plasma using immunoprecipitation coupled to tandem liquid chromatography mass spectrometry (IP-LC-MS/MS). Cross-sectional and repeated-measures data analyses were investigated family-wise, applying non-parametric tests as well as mixed-effects models.</p><p><strong>Results: </strong>Cross-sectional analysis at baseline showed more than a 3-fold increase in all plasma Aβ peptides in APPswe MC, regardless of clinical status, compared to controls (p < 0.01). PSEN1 (p.H163Y) presymptomatic MC had a decrease of plasma Aβ1-38 compared to controls (p < 0.05). There was no difference in Aβ1-42/1-40 ratio between APPswe MC (PMC and SMC), PSEN1 MC (PMC) and controls at baseline. Notably, both cross-sectional data and repeated-measures analysis suggested that APPswe MC have a stable Aβ1-42/1-40 ratio with increasing age, in contrast to the decrease seen with aging in both controls and PSEN1 (p.H163Y) MC.</p><p><strong>Conclusion: </strong>These data show very strong mutation-specific effects on Aβ profiles in blood, most likely due to a ubiquitous production outside of the CNS. Hence, analyses in an unselected clinical setting might unintentionally disclose genetic status. Furthermore, our findings suggest that the Aβ ratio might be a poor indicator of brain Aβ pathology in selected genetic cases. The very small sample size is a limitation that needs to be considered but reflects the scarcity of longitudinal in vivo data from genetic cohorts.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"16 1","pages":"207"},"PeriodicalIF":7.9,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11423518/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142339241","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信