Alzheimer's Research & Therapy最新文献

筛选
英文 中文
Plasma trimethylamine N-oxide (TMAO): associations with cognition, neuroimaging, and dementia. 血浆三甲胺 N-氧化物 (TMAO):与认知、神经影像学和痴呆症的关系。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-20 DOI: 10.1186/s13195-024-01480-1
Amber Yaqub, Dina Vojinovic, Meike W Vernooij, P Eline Slagboom, Mohsen Ghanbari, Marian Beekman, Jeroen van der Grond, Thomas Hankemeier, Cornelia M van Duijn, M Arfan Ikram, Shahzad Ahmad
{"title":"Plasma trimethylamine N-oxide (TMAO): associations with cognition, neuroimaging, and dementia.","authors":"Amber Yaqub, Dina Vojinovic, Meike W Vernooij, P Eline Slagboom, Mohsen Ghanbari, Marian Beekman, Jeroen van der Grond, Thomas Hankemeier, Cornelia M van Duijn, M Arfan Ikram, Shahzad Ahmad","doi":"10.1186/s13195-024-01480-1","DOIUrl":"10.1186/s13195-024-01480-1","url":null,"abstract":"<p><strong>Background: </strong>The gut-derived metabolite Trimethylamine N-oxide (TMAO) and its precursors - betaine, carnitine, choline, and deoxycarnitine - have been associated with an increased risk of cardiovascular disease, but their relation to cognition, neuroimaging markers, and dementia remains uncertain.</p><p><strong>Methods: </strong>In the population-based Rotterdam Study, we used multivariable regression models to study the associations between plasma TMAO, its precursors, and cognition in 3,143 participants. Subsequently, we examined their link to structural brain MRI markers in 2,047 participants, with a partial validation in the Leiden Longevity Study (n = 318). Among 2,517 participants, we assessed the risk of incident dementia using multivariable Cox proportional hazard models. Following this, we stratified the longitudinal associations by medication use and sex, after which we conducted a sensitivity analysis for individuals with impaired renal function.</p><p><strong>Results: </strong>Overall, plasma TMAO was not associated with cognition, neuroimaging markers or incident dementia. Instead, higher plasma choline was significantly associated with poor cognition (adjusted mean difference: -0.170 [95% confidence interval (CI) -0.297;-0.043]), brain atrophy and more markers of cerebral small vessel disease, such as white matter hyperintensity volume (0.237 [95% CI: 0.076;0.397]). By contrast, higher carnitine concurred with lower white matter hyperintensity volume (-0.177 [95% CI: -0.343;-0.010]). Only among individuals with impaired renal function, TMAO appeared to increase risk of dementia (hazard ratio (HR): 1.73 [95% CI: 1.16;2.60]). No notable differences were observed in stratified analyses.</p><p><strong>Conclusions: </strong>Plasma choline, as opposed to TMAO, was found to be associated with cognitive decline, brain atrophy, and markers of cerebral small vessel disease. These findings illustrate the complexity of relationships between TMAO and its precursors, and emphasize the need for concurrent study to elucidate gut-brain mechanisms.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11103865/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069939","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
Blood biomarkers of neurodegeneration associate differently with amyloid deposition, medial temporal atrophy, and cerebrovascular changes in APOE ε4-enriched cognitively unimpaired elderly. 在APOE ε4富集的认知功能未受损的老年人中,神经退行性变的血液生物标志物与淀粉样蛋白沉积、颞叶内侧萎缩和脑血管变化有不同的关联。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-18 DOI: 10.1186/s13195-024-01477-w
Mikko Koivumäki, Laura Ekblad, Juan Lantero-Rodriguez, Nicholas J Ashton, Thomas K Karikari, Semi Helin, Riitta Parkkola, Jyrki Lötjönen, Henrik Zetterberg, Kaj Blennow, Juha O Rinne, Anniina Snellman
{"title":"Blood biomarkers of neurodegeneration associate differently with amyloid deposition, medial temporal atrophy, and cerebrovascular changes in APOE ε4-enriched cognitively unimpaired elderly.","authors":"Mikko Koivumäki, Laura Ekblad, Juan Lantero-Rodriguez, Nicholas J Ashton, Thomas K Karikari, Semi Helin, Riitta Parkkola, Jyrki Lötjönen, Henrik Zetterberg, Kaj Blennow, Juha O Rinne, Anniina Snellman","doi":"10.1186/s13195-024-01477-w","DOIUrl":"10.1186/s13195-024-01477-w","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) plaques, neurofibrillary tau tangles, and neurodegeneration in the brain parenchyma. Here, we aimed to (i) assess differences in blood and imaging biomarkers used to evaluate neurodegeneration among cognitively unimpaired APOE ε4 homozygotes, heterozygotes, and non-carriers with varying risk for sporadic AD, and (ii) to determine how different cerebral pathologies (i.e., Aβ deposition, medial temporal atrophy, and cerebrovascular pathology) contribute to blood biomarker concentrations in this sample.</p><p><strong>Methods: </strong>Sixty APOE ε4 homozygotes (n = 19), heterozygotes (n = 21), and non-carriers (n = 20) ranging from 60 to 75 years, were recruited in collaboration with Auria biobank (Turku, Finland). Participants underwent Aβ-PET ([<sup>11</sup>C]PiB), structural brain MRI including T1-weighted and T2-FLAIR sequences, and blood sampling for measuring serum neurofilament light chain (NfL), plasma total tau (t-tau), plasma N-terminal tau fragments (NTA-tau) and plasma glial fibrillary acidic protein (GFAP). [<sup>11</sup>C]PiB standardized uptake value ratio was calculated for regions typical for Aβ accumulation in AD. MRI images were analysed for regional volumes, atrophy scores, and volumes of white matter hyperintensities. Differences in biomarker levels and associations between blood and imaging biomarkers were tested using uni- and multivariable linear models (unadjusted and adjusted for age and sex).</p><p><strong>Results: </strong>Serum NfL concentration was increased in APOE ε4 homozygotes compared with non-carriers (mean 21.4 pg/ml (SD 9.5) vs. 15.5 pg/ml (3.8), p = 0.013), whereas other blood biomarkers did not differ between the groups (p > 0.077 for all). From imaging biomarkers, hippocampal volume was significantly decreased in APOE ε4 homozygotes compared with non-carriers (6.71 ml (0.86) vs. 7.2 ml (0.7), p = 0.029). In the whole sample, blood biomarker levels were differently predicted by the three measured cerebral pathologies; serum NfL concentration was associated with cerebrovascular pathology and medial temporal atrophy, while plasma NTA-tau associated with medial temporal atrophy. Plasma GFAP showed significant association with both medial temporal atrophy and Aβ pathology. Plasma t-tau concentration did not associate with any of the measured pathologies.</p><p><strong>Conclusions: </strong>Only increased serum NfL concentrations and decreased hippocampal volume was observed in cognitively unimpaired APOEε4 homozygotes compared to non-carriers. In the whole population the concentrations of blood biomarkers were affected in distinct ways by different pathologies.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11102270/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140955749","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
The impact of incident stroke on cognitive trajectories in later life. 中风对晚年认知轨迹的影响。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-18 DOI: 10.1186/s13195-024-01479-8
Swarna Vishwanath, Ingrid Hopper, Geoffrey C Cloud, Trevor T-J Chong, Raj C Shah, Geoffrey A Donnan, Jeff D Williamson, Charles B Eaton, Rory Wolfe, Christopher M Reid, Andrew M Tonkin, Suzanne G Orchard, Sharyn Fitzgerald, Anne M Murray, Robyn L Woods, Mark R Nelson, Ajay Sood, Claire J Steves, Joanne Ryan
{"title":"The impact of incident stroke on cognitive trajectories in later life.","authors":"Swarna Vishwanath, Ingrid Hopper, Geoffrey C Cloud, Trevor T-J Chong, Raj C Shah, Geoffrey A Donnan, Jeff D Williamson, Charles B Eaton, Rory Wolfe, Christopher M Reid, Andrew M Tonkin, Suzanne G Orchard, Sharyn Fitzgerald, Anne M Murray, Robyn L Woods, Mark R Nelson, Ajay Sood, Claire J Steves, Joanne Ryan","doi":"10.1186/s13195-024-01479-8","DOIUrl":"10.1186/s13195-024-01479-8","url":null,"abstract":"<p><strong>Background: </strong>Cognitive impairment is common after stroke, and a large proportion of stroke patients will develop dementia. However, there have been few large prospective studies which have assessed cognition both prior to and after stroke. This study aims to determine the extent to which incident stroke impacts different domains of cognitive function in a longitudinal cohort of older community-dwelling individuals.</p><p><strong>Methods: </strong>19,114 older individuals without cardiovascular disease or major cognitive impairment were recruited and followed over a maximum 11 years. Stroke included ischaemic and haemorrhagic stroke and was adjudicated by experts. Cognitive function was assessed regularly using Modified Mini-Mental State Examination (3MS), Hopkins Verbal Learning Test-Revised (HVLT-R), Symbol Digit Modalities Test (SDMT), and Controlled Oral Word Association Test (COWAT). Linear mixed models were used to investigate the change in cognition at the time of stroke and decline in cognitive trajectories following incident stroke.</p><p><strong>Results: </strong>During a median follow-up period of 8.4 [IQR: 7.2, 9.6] years, 815 (4.3%) participants experienced a stroke. Over this time, there was a general decline observed in 3MS, HVLT-R delayed recall, and SDMT scores across participants. However, for individuals who experienced a stroke, there was a significantly greater decline across all cognitive domains immediately after the event immediately after the event (3MS: -1.03 [95%CI: -1.45, -0.60]; HVLT-R: -0.47 [-0.70, -0.24]; SDMT: -2.82 [-3.57, -2.08]; COWAT: -0.67 [-1.04, -0.29]) and a steeper long-term decline for three of these domains (3MS -0.62 [-0.88, -0.35]; COWAT: -0.30 [-0.46, -0.14]); HVLT-R: -0.12 [95%CI, -0.70, -0.24]). However individuals with stroke experienced no longer-term decline in SDMT compared to the rest of the participants.</p><p><strong>Conclusions: </strong>These findings highlight the need for comprehensive neuropsychology assessments for ongoing monitoring of cognition following incident stroke; and potential early intervention.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11102228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140955753","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
Comparison of plasma and neuroimaging biomarkers to predict cognitive decline in non-demented memory clinic patients. 比较血浆和神经影像生物标志物,预测非痴呆记忆门诊患者的认知能力衰退。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-16 DOI: 10.1186/s13195-024-01478-9
Augusto J Mendes, Federica Ribaldi, Aurelien Lathuiliere, Nicholas J Ashton, Henrik Zetterberg, Marc Abramowicz, Max Scheffler, Frédéric Assal, Valentina Garibotto, Kaj Blennow, Giovanni B Frisoni
{"title":"Comparison of plasma and neuroimaging biomarkers to predict cognitive decline in non-demented memory clinic patients.","authors":"Augusto J Mendes, Federica Ribaldi, Aurelien Lathuiliere, Nicholas J Ashton, Henrik Zetterberg, Marc Abramowicz, Max Scheffler, Frédéric Assal, Valentina Garibotto, Kaj Blennow, Giovanni B Frisoni","doi":"10.1186/s13195-024-01478-9","DOIUrl":"10.1186/s13195-024-01478-9","url":null,"abstract":"<p><strong>Background: </strong>Plasma biomarkers of Alzheimer's disease (AD) pathology, neurodegeneration, and neuroinflammation are ideally suited for secondary prevention programs in self-sufficient persons at-risk of dementia. Plasma biomarkers have been shown to be highly correlated with traditional imaging biomarkers. However, their comparative predictive value versus traditional AD biomarkers is still unclear in cognitively unimpaired (CU) subjects and with mild cognitive impairment (MCI).</p><p><strong>Methods: </strong>Plasma (Aβ42/40, p-tau181, p-tau231, NfL, and GFAP) and neuroimaging (hippocampal volume, centiloid of amyloid-PET, and tau-SUVR of tau-PET) biomarkers were assessed at baseline in 218 non-demented subjects (CU = 140; MCI = 78) from the Geneva Memory Center. Global cognition (MMSE) was evaluated at baseline and at follow-ups up to 5.7 years. We used linear mixed-effects models and Cox proportional-hazards regression to assess the association between biomarkers and cognitive decline. Lastly, sample size calculations using the linear mixed-effects models were performed on subjects positive for amyloid-PET combined with tau-PET and plasma biomarker positivity.</p><p><strong>Results: </strong>Cognitive decline was significantly predicted in MCI by baseline plasma NfL (β=-0.55), GFAP (β=-0.36), hippocampal volume (β = 0.44), centiloid (β=-0.38), and tau-SUVR (β=-0.66) (all p < 0.05). Subgroup analysis with amyloid-positive MCI participants also showed that only NfL and GFAP were the only significant predictors of cognitive decline among plasma biomarkers. Overall, NfL and tau-SUVR showed the highest prognostic values (hazard ratios of 7.3 and 5.9). Lastly, we demonstrated that adding NfL to the inclusion criteria could reduce the sample sizes of future AD clinical trials by up to one-fourth in subjects with amyloid-PET positivity or by half in subjects with amyloid-PET and tau-PET positivity.</p><p><strong>Conclusions: </strong>Plasma NfL and GFAP predict cognitive decline in a similar manner to traditional imaging techniques in amyloid-positive MCI patients. Hence, even though they are non-specific biomarkers of AD, both can be implemented in memory clinic workups as important prognostic biomarkers. Likewise, future clinical trials might employ plasma biomarkers as additional inclusion criteria to stratify patients at higher risk of cognitive decline to reduce sample sizes and enhance effectiveness.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11097559/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140955751","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
Cholecystokinin B receptor agonists alleviates anterograde amnesia in cholecystokinin-deficient and aged Alzheimer's disease mice. 胆囊收缩素 B 受体激动剂可减轻胆囊收缩素缺陷和老年性阿尔茨海默病小鼠的逆行性遗忘。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-15 DOI: 10.1186/s13195-024-01472-1
Nan Zhang, Yixuan Sui, Peter Jendrichovsky, Hemin Feng, Heng Shi, Xu Zhang, Shenghui Xu, Wenjian Sun, Huatang Zhang, Xi Chen, Micky D Tortorella, Jufang He
{"title":"Cholecystokinin B receptor agonists alleviates anterograde amnesia in cholecystokinin-deficient and aged Alzheimer's disease mice.","authors":"Nan Zhang, Yixuan Sui, Peter Jendrichovsky, Hemin Feng, Heng Shi, Xu Zhang, Shenghui Xu, Wenjian Sun, Huatang Zhang, Xi Chen, Micky D Tortorella, Jufang He","doi":"10.1186/s13195-024-01472-1","DOIUrl":"10.1186/s13195-024-01472-1","url":null,"abstract":"<p><strong>Background: </strong>As one major symptom of Alzheimer's disease (AD), anterograde amnesia describes patients with an inability in new memory formation. The crucial role of the entorhinal cortex in forming new memories has been well established, and the neuropeptide cholecystokinin (CCK) is reported to be released from the entorhinal cortex to enable neocortical associated memory and long-term potentiation. Though several studies reveal that the entorhinal cortex and CCK are related to AD, it is less well studied. It is unclear whether CCK is a good biomarker or further a great drug candidate for AD.</p><p><strong>Methods: </strong>mRNA expressions of CCK and CCK-B receptor (CCKBR) were examined in two mouse models, 3xTg AD and CCK knock-out (CCK<sup>-/-</sup>) mice. Animals' cognition was investigated with Morris water maze, novel object recognition test and neuroplasticity with in-vitro electrophysiological recording. Drugs were given intraperitoneally to animals to investigate the rescue effects on cognitive deficits, or applied to brain slices directly to explore the influence in inducement of long-term potentiation.</p><p><strong>Results: </strong>Aged 3xTg AD mice exhibited reduced CCK mRNA expression in the entorhinal cortex but reduced CCKBR expression in the neocortex and hippocampus, and impaired cognition and neuroplasticity comparable with CCK<sup>-/-</sup> mice. Importantly, the animals displayed improved performance and enhanced long-term potentiation after the treatment of CCKBR agonists.</p><p><strong>Conclusions: </strong>Here we provide more evidence to support the role of CCK in learning and memory and its potential to treat AD. We elaborated on the rescue effect of a promising novel drug, HT-267, on aged 3xTg AD mice. Although the physiological etiology of CCK in AD still needs to be further investigated, this study sheds light on a potential pharmaceutical candidate for AD and dementia.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11094875/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140943713","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
Proximity extension assay in cerebrospinal fluid identifies neurofilament light chain as biomarker of neurodegeneration in sporadic cerebral amyloid angiopathy. 脑脊液中的邻近延伸测定确定神经丝蛋白轻链是散发性脑淀粉样血管病变中神经变性的生物标记。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-14 DOI: 10.1186/s13195-024-01473-0
Marc Vervuurt, H Bea Kuiperij, Anna M de Kort, Iris Kersten, Catharina J M Klijn, Floris H B M Schreuder, Marcel M Verbeek
{"title":"Proximity extension assay in cerebrospinal fluid identifies neurofilament light chain as biomarker of neurodegeneration in sporadic cerebral amyloid angiopathy.","authors":"Marc Vervuurt, H Bea Kuiperij, Anna M de Kort, Iris Kersten, Catharina J M Klijn, Floris H B M Schreuder, Marcel M Verbeek","doi":"10.1186/s13195-024-01473-0","DOIUrl":"10.1186/s13195-024-01473-0","url":null,"abstract":"<p><strong>Background: </strong>Sporadic cerebral amyloid angiopathy (sCAA) is a disease characterised by the progressive deposition of the amyloid beta (Aβ) in the cerebral vasculature, capable of causing a variety of symptoms, from (mild) cognitive impairment, to micro- and major haemorrhagic lesions. Modern diagnosis of sCAA relies on radiological detection of late-stage hallmarks of disease, complicating early diagnosis and potential interventions in disease progression. Our goal in this study was to identify and validate novel biomarkers for sCAA.</p><p><strong>Methods: </strong>We performed a proximity extension assay (PEA) on cerebrospinal fluid (CSF) samples of sCAA/control participants (n = 34/51). Additionally, we attempted to validate the top candidate biomarker in CSF and serum samples (n = 38/26) in a largely overlapping validation cohort, through analysis with a targeted immunoassay.</p><p><strong>Results: </strong>Thirteen proteins were differentially expressed through PEA, with top candidate NFL significantly increased in CSF of sCAA patients (p < 0.0001). Validation analyses using immunoassays revealed increased CSF and serum NFL levels in sCAA patients (both p < 0.0001) with good discrimination between sCAA and controls (AUC: 0.85; AUC: 0.79 respectively). Additionally, the CSF: serum NFL ratio was significantly elevated in sCAA (p = 0.002).</p><p><strong>Discussion: </strong>Large-scale targeted proteomics screening of CSF of sCAA patients and controls identified thirteen biomarker candidates for sCAA. Orthogonal validation of NFL identified NFL in CSF and serum as biomarker, capable of differentiating between sCAA patients and controls.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11092079/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140920487","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
Serum urate levels and neurodegenerative outcomes: a prospective cohort study and mendelian randomization analysis of the UK Biobank. 血清尿酸盐水平与神经退行性疾病的预后:英国生物库的前瞻性队列研究和孟德尔随机分析。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-11 DOI: 10.1186/s13195-024-01476-x
Tingjing Zhang, Yu An, Zhenfei Shen, Honghao Yang, Jinguo Jiang, Liangkai Chen, Yanhui Lu, Yang Xia
{"title":"Serum urate levels and neurodegenerative outcomes: a prospective cohort study and mendelian randomization analysis of the UK Biobank.","authors":"Tingjing Zhang, Yu An, Zhenfei Shen, Honghao Yang, Jinguo Jiang, Liangkai Chen, Yanhui Lu, Yang Xia","doi":"10.1186/s13195-024-01476-x","DOIUrl":"10.1186/s13195-024-01476-x","url":null,"abstract":"<p><strong>Background: </strong>Previous studies on the associations between serum urate levels and neurodegenerative outcomes have yielded inconclusive results, and the causality remains unclear. This study aimed to investigate whether urate levels are associated with the risks of Alzheimer's disease and related dementias (ADRD), Parkinson's disease (PD), and neurodegenerative deaths.</p><p><strong>Methods: </strong>This prospective study included 382,182 participants (45.7% men) from the UK Biobank cohort. Cox proportional hazards models were used to assess the associations between urate levels and risk of neurodegenerative outcomes. In the Mendelian randomization (MR) analysis, urate-related single-nucleotide polymorphisms were identified through a genome-wide association study. Both linear and non-linear MR approaches were utilized to investigate the potential causal associations.</p><p><strong>Results: </strong>During a median follow-up period of 12 years, we documented 5,400 ADRD cases, 2,553 PD cases, and 1,531 neurodegenerative deaths. Observational data revealed that a higher urate level was associated with a decreased risk of ADRD (hazard ratio [HR]: 0.93, 95% confidence interval [CI]: 0.90, 0.96), PD (HR: 0.87, 95% CI: 0.82, 0.91), and neurodegenerative death (HR: 0.88, 95% CI: 0.83, 0.94). Negative linear associations between urate levels and neurodegenerative events were observed (all P-values for overall < 0.001 and all P-values for non-linearity > 0.05). However, MR analyses yielded no evidence of either linear or non-linear associations between genetically predicted urate levels and the risk of the aforementioned neurodegenerative events.</p><p><strong>Conclusion: </strong>Although the prospective cohort study demonstrated that elevated urate levels were associated with a reduced risk of neurodegenerative outcomes, MR analyses found no evidence of causality.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11088014/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903724","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
Plasma biomarkers increase diagnostic confidence in patients with Alzheimer's disease or frontotemporal lobar degeneration. 血浆生物标志物提高了阿尔茨海默氏症或额颞叶变性患者的诊断信心。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-11 DOI: 10.1186/s13195-024-01474-z
Daniele Altomare, Ilenia Libri, Antonella Alberici, Jasmine Rivolta, Alessandro Padovani, Nicholas J Ashton, Henrik Zetterberg, Kaj Blennow, Barbara Borroni
{"title":"Plasma biomarkers increase diagnostic confidence in patients with Alzheimer's disease or frontotemporal lobar degeneration.","authors":"Daniele Altomare, Ilenia Libri, Antonella Alberici, Jasmine Rivolta, Alessandro Padovani, Nicholas J Ashton, Henrik Zetterberg, Kaj Blennow, Barbara Borroni","doi":"10.1186/s13195-024-01474-z","DOIUrl":"10.1186/s13195-024-01474-z","url":null,"abstract":"<p><strong>Background: </strong>The recent development of techniques to assess plasma biomarkers has changed the way the research community envisions the future of diagnosis and management of Alzheimer's disease (AD) and other neurodegenerative disorders. This work aims to provide real world evidence on the clinical impact of plasma biomarkers in an academic tertiary care center.</p><p><strong>Methods: </strong>Anonymized clinical reports of patients diagnosed with AD or Frontotemporal Lobar Degeneration with available plasma biomarkers (Aβ<sub>42</sub>, Aβ<sub>42</sub>/Aβ<sub>40</sub>, p-tau<sub>181</sub>, p-tau<sub>231</sub>, NfL, GFAP) were independently assessed by two neurologists who expressed diagnosis and diagnostic confidence three times: (T0) at baseline based on the information collected during the first visit, (T1) after plasma biomarkers, and (T2) after traditional biomarkers (when available). Finally, we assessed whether clinicians' interpretation of plasma biomarkers and the consequent clinical impact are consistent with the final diagnosis, determined after the conclusion of the diagnostic clinical and instrumental work-up by the actual managing physicians who had complete access to all available information.</p><p><strong>Results: </strong>Clinicians assessed 122 reports, and their concordance ranged from 81 to 91% at the three time points. At T1, the presentation of plasma biomarkers resulted in a change of diagnosis in 2% (2/122, p = 1.00) of cases, and in increased diagnostic confidence in 76% (91/120, p < 0.001) of cases with confirmed diagnosis. The change in diagnosis and the increase in diagnostic confidence after plasma biomarkers were consistent with the final diagnosis in 100% (2/2) and 81% (74/91) of cases, respectively. At T2, the presentation of traditional biomarkers resulted in a further change of diagnosis in 13% (12/94, p = 0.149) of cases, and in increased diagnostic confidence in 88% (72/82, p < 0.001) of cases with confirmed diagnosis.</p><p><strong>Conclusions: </strong>In an academic tertiary care center, plasma biomarkers supported clinicians by increasing their diagnostic confidence in most cases, despite a negligible impact on diagnosis. Future prospective studies are needed to assess the full potential of plasma biomarkers on clinical grounds.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11088144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140908286","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
Cerebrospinal fluid α-synuclein adds the risk of cognitive decline and is associated with tau pathology among non-demented older adults. 脑脊液中的α-突触核蛋白会增加非痴呆老年人认知能力下降的风险,并与tau病理学相关。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-10 DOI: 10.1186/s13195-024-01463-2
Wenying Liu, Wenwen Li, Zhaojun Liu, Yan Li, Xuechu Wang, Mengmeng Guo, Shiyuan Wang, Shuheng Wang, Yan Li, Jianping Jia
{"title":"Cerebrospinal fluid α-synuclein adds the risk of cognitive decline and is associated with tau pathology among non-demented older adults.","authors":"Wenying Liu, Wenwen Li, Zhaojun Liu, Yan Li, Xuechu Wang, Mengmeng Guo, Shiyuan Wang, Shuheng Wang, Yan Li, Jianping Jia","doi":"10.1186/s13195-024-01463-2","DOIUrl":"10.1186/s13195-024-01463-2","url":null,"abstract":"<p><strong>Background: </strong>The role of α-synuclein in dementia has been recognized, yet its exact influence on cognitive decline in non-demented older adults is still not fully understood.</p><p><strong>Methods: </strong>A total of 331 non-demented individuals were included in the study from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants were divided into two distinct groups based on their α-synuclein levels: one with lower levels (α-synuclein-L) and another with higher levels (α-synuclein-H). Measurements included neuropsychiatric scales, cerebrospinal fluid (CSF) biomarkers, and blood transcriptomics. The linear mixed-effects model investigated the longitudinal changes in cognition. Kaplan-Meier survival analysis and the Cox proportional hazards model were utilized to evaluate the effects of different levels of α-synuclein on dementia. Gene set enrichment analysis (GSEA) was utilized to investigate the biological pathways related to cognitive impairment. Pearson correlation, multiple linear regression models, and mediation analysis were employed to investigate the relationship between α-synuclein and neurodegenerative biomarkers, and their potential mechanisms affecting cognition.</p><p><strong>Results: </strong>Higher CSF α-synuclein levels were associated with increased risk of cognitive decline and progression to dementia. Enrichment analysis highlighted the activation of tau-associated and immune response pathways in the α-synuclein-H group. Further correlation and regression analysis indicated that the CSF α-synuclein levels were positively correlated with CSF total tau (t-tau), phosphorylated tau (p-tau) 181, tumor necrosis factor receptor 1 (TNFR1) and intercellular cell adhesion molecule-1 (ICAM-1). Mediation analysis further elucidated that the detrimental effects of CSF α-synuclein on cognition were primarily mediated through CSF t-tau and p-tau. Additionally, it was observed that CSF α-synuclein influenced CSF t-tau and p-tau181 levels via inflammatory pathways involving CSF TNFR1 and ICAM-1.</p><p><strong>Conclusions: </strong>These findings elucidate a significant connection between elevated levels of CSF α-synuclein and the progression of cognitive decline, highlighting the critical roles of activated inflammatory pathways and tau pathology in this association. They underscore the importance of monitoring CSF α-synuclein levels as a promising biomarker for identifying individuals at increased risk of cognitive deterioration and developing dementia.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11084056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140896451","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
Updated safety results from phase 3 lecanemab study in early Alzheimer's disease. 早期阿尔茨海默病 Lecanemab 3 期研究的最新安全性结果。
IF 7.9 1区 医学
Alzheimer's Research & Therapy Pub Date : 2024-05-10 DOI: 10.1186/s13195-024-01441-8
Lawrence S Honig, Marwan N Sabbagh, Christopher H van Dyck, Reisa A Sperling, Steven Hersch, Andre Matta, Luigi Giorgi, Michelle Gee, Michio Kanekiyo, David Li, Derk Purcell, Shobha Dhadda, Michael Irizarry, Lynn Kramer
{"title":"Updated safety results from phase 3 lecanemab study in early Alzheimer's disease.","authors":"Lawrence S Honig, Marwan N Sabbagh, Christopher H van Dyck, Reisa A Sperling, Steven Hersch, Andre Matta, Luigi Giorgi, Michelle Gee, Michio Kanekiyo, David Li, Derk Purcell, Shobha Dhadda, Michael Irizarry, Lynn Kramer","doi":"10.1186/s13195-024-01441-8","DOIUrl":"10.1186/s13195-024-01441-8","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer disease (AD) is a major health problem of aging, with tremendous burden on healthcare systems, patients, and families globally. Lecanemab, an FDA-approved amyloid beta (Aβ)-directed antibody indicated for the treatment of early AD, binds with high affinity to soluble Aβ protofibrils, which have been shown to be more toxic to neurons than monomers or insoluble fibrils. Lecanemab has been shown to be well tolerated in multiple clinical trials, although risks include an increased rate of amyloid-related imaging abnormalities (ARIA) and infusion reactions relative to placebo.</p><p><strong>Methods: </strong>Clarity AD was an 18-month treatment (Core study), multicenter, double-blind, placebo-controlled, parallel-group study with open-label extension (OLE) in participants with early AD. Eligible participants were randomized 1:1 across 2 treatment groups (placebo and lecanemab 10 mg/kg biweekly). Safety evaluations included monitoring of vital signs, physical examinations, adverse events, clinical laboratory parameters, and 12-lead electrocardiograms. ARIA occurrence was monitored throughout the study by magnetic resonance imaging, read both locally and centrally.</p><p><strong>Results: </strong>Overall, 1795 participants from Core and 1612 participants with at least one dose of lecanemab (Core + OLE) were included. Lecanemab was generally well-tolerated in Clarity AD, with no deaths related to lecanemab in the Core study. There were 9 deaths during the OLE, with 4 deemed possibly related to study treatment. Of the 24 deaths in Core + OLE, 3 were due to intracerebral hemorrhage (ICH): 1 placebo in the Core due to ICH, and 2 lecanemab in OLE with concurrent ICH (1 on tissue plasminogen activator and 1 on anticoagulant therapy). In the Core + OLE, the most common adverse events in the lecanemab group (> 10%) were infusion-related reactions (24.5%), ARIA with hemosiderin deposits (ARIA-H) microhemorrhages (16.0%), COVID-19 (14.7%), ARIA with edema (ARIA-E; 13.6%), and headache (10.3%). ARIA-E and ARIA-H were largely radiographically mild-to-moderate. ARIA-E generally occurred within 3-6 months of treatment, was more common in ApoE e4 carriers (16.8%) and most common in ApoE ε4 homozygous participants (34.5%).</p><p><strong>Conclusions: </strong>Lecanemab was generally well-tolerated, with the most common adverse events being infusion-related reactions, ARIA-H, ARIA-E. Clinicians, participants, and caregivers should understand the incidence, monitoring, and management of these events for optimal patient care.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov numbers: Clarity AD NCT03887455).</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11084061/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903728","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