Pub Date : 2025-01-10eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcaf009
Hannah de Bruin, Colin Groot, Suzie Kamps, Everard G B Vijverberg, Anna Steward, Amir Dehsarvi, Yolande A L Pijnenburg, Rik Ossenkoppele, Nicolai Franzmeier
Traumatic brain injury is widely viewed as a risk factor for dementia, but the biological mechanisms underlying this association are still unclear. In previous studies, traumatic brain injury has been associated with the hallmark pathologies of Alzheimer's disease, i.e. amyloid-β plaques and neurofibrillary tangles comprised of hyperphosphorylated tau. Depending on the type and location of trauma, traumatic brain injury can induce spatially heterogeneous brain lesions that may pre-dispose for the development of Alzheimer's disease pathology in aging. Therefore, we hypothesized that a history of traumatic brain injury may be related to spatially heterogeneous amyloid-β and tau pathology patterns that deviate from the stereotypical temporo-parietal patterns in Alzheimer's disease. To test this, we included 103 Vietnam War veterans of whom 65 had experienced traumatic brain injury (n = 40, 38.8% mild; n = 25, 24.3% moderate/severe). Most individuals had a history of 1 (n = 35, 53.8%) or 2 (n = 15, 23.1%) traumatic brain injury events. We included the group without a history of traumatic brain injury (n = 38, 36.9%) as controls. The majority was cognitively normal (n = 80, 77.7%), while a subset had mild cognitive impairment (n = 23, 22.3%). All participants underwent [18F]florbetapir/Amyvid amyloid-β PET and [18F]flortaucipir/Tauvid tau-PET 39.63 ± 18.39 years after their last traumatic brain injury event. We found no differences in global amyloid-β and tau-PET levels between groups, suggesting that a history of traumatic brain injury does not pre-dispose to accumulate amyloid-β or tau pathology in general. However, we found that traumatic brain injury was associated with altered spatial patterns of amyloid-β and tau, with relatively greater deposition in fronto-parietal brain regions. These regions are prone to damage in traumatic brain injury, while they are typically only affected in later stages of Alzheimer's disease. Moreover, in our traumatic brain injury groups, the association between amyloid-β and tau was reduced in Alzheimer-typical temporal regions but increased in frontal regions that are commonly associated with traumatic brain injury. Altogether, while acknowledging the relatively small sample size and generally low levels of Alzheimer's disease pathology in this sample, our findings suggest that traumatic brain injury induces spatial patterns of amyloid-β and tau that differ from patterns observed in typical Alzheimer's disease. Furthermore, traumatic brain injury may be associated with a de-coupling of amyloid-β and tau in regions vulnerable in Alzheimer's disease. These findings indicate that focal brain damage in early/mid-life may change neurodegenerative trajectories in late-life.
{"title":"Amyloid-β and tau deposition in traumatic brain injury: a study of Vietnam War veterans.","authors":"Hannah de Bruin, Colin Groot, Suzie Kamps, Everard G B Vijverberg, Anna Steward, Amir Dehsarvi, Yolande A L Pijnenburg, Rik Ossenkoppele, Nicolai Franzmeier","doi":"10.1093/braincomms/fcaf009","DOIUrl":"10.1093/braincomms/fcaf009","url":null,"abstract":"<p><p>Traumatic brain injury is widely viewed as a risk factor for dementia, but the biological mechanisms underlying this association are still unclear. In previous studies, traumatic brain injury has been associated with the hallmark pathologies of Alzheimer's disease, i.e. amyloid-β plaques and neurofibrillary tangles comprised of hyperphosphorylated tau. Depending on the type and location of trauma, traumatic brain injury can induce spatially heterogeneous brain lesions that may pre-dispose for the development of Alzheimer's disease pathology in aging. Therefore, we hypothesized that a history of traumatic brain injury may be related to spatially heterogeneous amyloid-β and tau pathology patterns that deviate from the stereotypical temporo-parietal patterns in Alzheimer's disease. To test this, we included 103 Vietnam War veterans of whom 65 had experienced traumatic brain injury (<i>n</i> = 40, 38.8% mild; <i>n</i> = 25, 24.3% moderate/severe). Most individuals had a history of 1 (<i>n</i> = 35, 53.8%) or 2 (<i>n</i> = 15, 23.1%) traumatic brain injury events. We included the group without a history of traumatic brain injury (<i>n</i> = 38, 36.9%) as controls. The majority was cognitively normal (<i>n</i> = 80, 77.7%), while a subset had mild cognitive impairment (<i>n</i> = 23, 22.3%). All participants underwent [<sup>18</sup>F]florbetapir/Amyvid amyloid-β PET and [<sup>18</sup>F]flortaucipir/Tauvid tau-PET 39.63 ± 18.39 years after their last traumatic brain injury event. We found no differences in global amyloid-β and tau-PET levels between groups, suggesting that a history of traumatic brain injury does not pre-dispose to accumulate amyloid-β or tau pathology in general. However, we found that traumatic brain injury was associated with altered spatial patterns of amyloid-β and tau, with relatively greater deposition in fronto-parietal brain regions. These regions are prone to damage in traumatic brain injury, while they are typically only affected in later stages of Alzheimer's disease. Moreover, in our traumatic brain injury groups, the association between amyloid-β and tau was reduced in Alzheimer-typical temporal regions but increased in frontal regions that are commonly associated with traumatic brain injury. Altogether, while acknowledging the relatively small sample size and generally low levels of Alzheimer's disease pathology in this sample, our findings suggest that traumatic brain injury induces spatial patterns of amyloid-β and tau that differ from patterns observed in typical Alzheimer's disease. Furthermore, traumatic brain injury may be associated with a de-coupling of amyloid-β and tau in regions vulnerable in Alzheimer's disease. These findings indicate that focal brain damage in early/mid-life may change neurodegenerative trajectories in late-life.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcaf009"},"PeriodicalIF":4.1,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11752645/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143025382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alzheimer's disease may be associated with early dopamine dysfunction. However, its effects on neurofunctional alterations in the neurotransmission pathways remain elusive. In this study, positron emission tomography atlases and functional MRI data for 86 older adults with mild cognitive impairment Alzheimer's disease (MCI), 58 with mild Alzheimer's disease-dementia and 76 cognitively unimpaired were combined to investigate connectivity alterations associated with the dopaminergic and cholinergic systems. A cross-sectional design was used to compare neurotransmitter-related functional connectivity across groups and associations between functional connectivity and cognitive performance. The findings show that the Alzheimer's disease dementia group showed a decline in mesocorticolimbic dopamine-related connectivity in the precuneus but heightened connectivity in the thalamus, whereas the Alzheimer's disease-MCI group showed a decline in nigrostriatal connectivity in the left temporal areas. Acetylcholine-related connectivity decline was observed in both Alzheimer's disease-MCI and Alzheimer's disease-dementia primarily in the temporo-parietal areas. Episodic memory scores correlated positively with acetylcholine- and dopamine-related connectivity in the temporo-parietal cortex and negatively with dopamine-related functional connectivity in the fronto-thalamic areas. This study shows that connectivity alterations in acetylcholine and dopamine functional pathways parallel cognitive decline in Alzheimer's disease and might be a clinically relevant marker in early Alzheimer's disease.
{"title":"Changes in neurotransmitter-related functional connectivity along the Alzheimer's disease continuum.","authors":"Riccardo Manca, Matteo De Marco, Hilkka Soininen, Livia Ruffini, Annalena Venneri","doi":"10.1093/braincomms/fcaf008","DOIUrl":"10.1093/braincomms/fcaf008","url":null,"abstract":"<p><p>Alzheimer's disease may be associated with early dopamine dysfunction. However, its effects on neurofunctional alterations in the neurotransmission pathways remain elusive. In this study, positron emission tomography atlases and functional MRI data for 86 older adults with mild cognitive impairment Alzheimer's disease (MCI), 58 with mild Alzheimer's disease-dementia and 76 cognitively unimpaired were combined to investigate connectivity alterations associated with the dopaminergic and cholinergic systems. A cross-sectional design was used to compare neurotransmitter-related functional connectivity across groups and associations between functional connectivity and cognitive performance. The findings show that the Alzheimer's disease dementia group showed a decline in mesocorticolimbic dopamine-related connectivity in the precuneus but heightened connectivity in the thalamus, whereas the Alzheimer's disease-MCI group showed a decline in nigrostriatal connectivity in the left temporal areas. Acetylcholine-related connectivity decline was observed in both Alzheimer's disease-MCI and Alzheimer's disease-dementia primarily in the temporo-parietal areas. Episodic memory scores correlated positively with acetylcholine- and dopamine-related connectivity in the temporo-parietal cortex and negatively with dopamine-related functional connectivity in the fronto-thalamic areas. This study shows that connectivity alterations in acetylcholine and dopamine functional pathways parallel cognitive decline in Alzheimer's disease and might be a clinically relevant marker in early Alzheimer's disease.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcaf008"},"PeriodicalIF":4.1,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11840171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143470235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-09eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcaf005
Ming Ann Sim, Yingqi Liao, Siew Pang Chan, Eugene S J Tan, Cheuk Ni Kan, Joyce R Chong, Yuek Ling Chai, Narayanaswamy Venketasubramanian, Boon Yeow Tan, Saima Hilal, Xin Xu, Christopher L H Chen, Mitchell K P Lai
Brain serotonin dysregulation is associated with dementia and neuropsychiatric symptomology. However, the prognostic utility of circulating serotonin levels in detecting features of prodromal dementia including functional decline, cognitive impairment, mild behavioural impairment and brain atrophy remains unclear. In this prospective study of memory clinic subjects followed-up for ≤5 years, dementia-free subjects, classified as having no cognitive impairment or cognitive impairment, no dementia at baseline, underwent annual neuropsychological assessments including Montreal Cognitive Assessment, Global Cognition Z-scores and Clinical Dementia Rating Scale Global Scores (where a ≥ 0.5 increment from baseline denotes functional decline). Mild behavioural impairment was measured using baseline and annual Neuropsychiatric Inventory assessments, while brain atrophy was evaluated using cortical and medial temporal atrophy scores from baseline MRI scans. Baseline serum serotonin was then associated with the neuropsychological and neuroimaging measures cross-sectionally and longitudinally. Furthermore, associations of serum serotonin with cross-sectional brain atrophy scores were studied. Of the 191 elderly subjects included in the study, 63 (33.0%) had no cognitive impairment while 128 (67.0%) had cognitive impairment, no dementia. Fourteen subjects (9.0%) showed baseline mild behavioural impairment. Compared with the highest tertile, subjects within the lowest tertile of serotonin had greater Cortical Atrophy scores (adjusted odds ratio = 2.54, 95% confidence interval=1.22-5.30, P = 0.013). Serotonin levels were not significantly associated with cross-sectional neuropsychological or mild behavioural impairment scores (all P > 0.05). Of the 181 subjects with longitudinal cognitive follow-up (median duration 60.0 months), 56 (30.9%) developed functional decline, while incident mild behavioural impairment occurred in 26/119 (21.8%) subjects. Compared with the highest tertile, lower serotonin levels were associated with higher hazards of functional decline (lowest tertile: adjusted hazards ratio = 2.15, 95% confidence interval = 1.04-4.44, P = 0.039), and incident mild behavioural impairment (lowest tertile: adjusted hazards ratio = 3.82, 95% confidence interval = 1.13-12.87, P = 0.031, middle tertile: adjusted hazards ratio = 3.56, 95% confidence interval =1.05-12.15, P = 0.042). The association between the lowest serotonin tertile and functional decline was mediated via its effect on incident mild behavioural impairment (adjusted odds ratio = 3.96, 95% confidence interval = 1.15-13.61, P = 0.029). In conclusion, low circulating serotonin may be associated with cortical atrophy at baseline, as well as act as an early prognostic marker for functional decline and mild behavioural impairment in elderly, dementia-free subjects.
{"title":"Low serum serotonin is associated with functional decline, mild behavioural impairment and brain atrophy in dementia-free subjects.","authors":"Ming Ann Sim, Yingqi Liao, Siew Pang Chan, Eugene S J Tan, Cheuk Ni Kan, Joyce R Chong, Yuek Ling Chai, Narayanaswamy Venketasubramanian, Boon Yeow Tan, Saima Hilal, Xin Xu, Christopher L H Chen, Mitchell K P Lai","doi":"10.1093/braincomms/fcaf005","DOIUrl":"10.1093/braincomms/fcaf005","url":null,"abstract":"<p><p>Brain serotonin dysregulation is associated with dementia and neuropsychiatric symptomology. However, the prognostic utility of circulating serotonin levels in detecting features of prodromal dementia including functional decline, cognitive impairment, mild behavioural impairment and brain atrophy remains unclear. In this prospective study of memory clinic subjects followed-up for ≤5 years, dementia-free subjects, classified as having no cognitive impairment or cognitive impairment, no dementia at baseline, underwent annual neuropsychological assessments including Montreal Cognitive Assessment, Global Cognition <i>Z-</i>scores and Clinical Dementia Rating Scale Global Scores (where a ≥ 0.5 increment from baseline denotes functional decline). Mild behavioural impairment was measured using baseline and annual Neuropsychiatric Inventory assessments, while brain atrophy was evaluated using cortical and medial temporal atrophy scores from baseline MRI scans. Baseline serum serotonin was then associated with the neuropsychological and neuroimaging measures cross-sectionally and longitudinally. Furthermore, associations of serum serotonin with cross-sectional brain atrophy scores were studied. Of the 191 elderly subjects included in the study, 63 (33.0%) had no cognitive impairment while 128 (67.0%) had cognitive impairment, no dementia. Fourteen subjects (9.0%) showed baseline mild behavioural impairment. Compared with the highest tertile, subjects within the lowest tertile of serotonin had greater Cortical Atrophy scores (adjusted odds ratio = 2.54, 95% confidence interval=1.22-5.30, <i>P</i> = 0.013). Serotonin levels were not significantly associated with cross-sectional neuropsychological or mild behavioural impairment scores (all <i>P</i> > 0.05). Of the 181 subjects with longitudinal cognitive follow-up (median duration 60.0 months), 56 (30.9%) developed functional decline, while incident mild behavioural impairment occurred in 26/119 (21.8%) subjects. Compared with the highest tertile, lower serotonin levels were associated with higher hazards of functional decline (lowest tertile: adjusted hazards ratio = 2.15, 95% confidence interval = 1.04-4.44, <i>P</i> = 0.039), and incident mild behavioural impairment (lowest tertile: adjusted hazards ratio = 3.82, 95% confidence interval = 1.13-12.87, <i>P</i> = 0.031, middle tertile: adjusted hazards ratio = 3.56, 95% confidence interval =1.05-12.15, <i>P</i> = 0.042). The association between the lowest serotonin tertile and functional decline was mediated via its effect on incident mild behavioural impairment (adjusted odds ratio = 3.96, 95% confidence interval = 1.15-13.61, <i>P</i> = 0.029). In conclusion, low circulating serotonin may be associated with cortical atrophy at baseline, as well as act as an early prognostic marker for functional decline and mild behavioural impairment in elderly, dementia-free subjects.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcaf005"},"PeriodicalIF":4.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11733688/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-08eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcae427
Michael Gurevich, Rina Zilkha-Falb, Jia Sherman, Maxime Usdin, Catarina Raposo, Licinio Craveiro, Polina Sonis, David Magalashvili, Shay Menascu, Mark Dolev, Anat Achiron
Primary progressive multiple sclerosis (PPMS) affects 10-15% of multiple sclerosis patients and presents significant variability in the rate of disability progression. Identifying key biological features and patients at higher risk for fast progression is crucial to develop and optimize treatment strategies. Peripheral blood cell transcriptome has the potential to provide valuable information to predict patients' outcomes. In this study, we utilized a machine learning framework applied to the baseline blood transcriptional profiles and brain MRI radiological enumerations to develop prognostic models. These models aim to identify PPMS patients likely to experience significant disease progression and who could benefit from early treatment intervention. RNA-sequence analysis was performed on total RNA extracted from peripheral blood mononuclear cells of PPMS patients in the placebo arm of the ORATORIO clinical trial (NCT01412333), using Illumina NovaSeq S2. Cross-validation algorithms from Partek Genome Suite (www.partek.com) were applied to predict disability progression and brain volume loss over 120 weeks. For disability progression prediction, we analysed blood RNA samples from 135 PPMS patients (61 females and 74 males) with a mean ± standard error age of 44.0 ± 0.7 years, disease duration of 5.9 ± 0.32 years and a median baseline Expanded Disability Status Scale (EDSS) score of 4.3 (range 3.5-6.5). Over the 120-week study, 39.3% (53/135) of patients reached the disability progression end-point, with an average EDSS score increase of 1.3 ± 0.16. For brain volume loss prediction, blood RNA samples from 94 PPMS patients (41 females and 53 males), mean ± standard error age of 43.7 ± 0.7 years and a median baseline EDSS of 4.0 (range 3.0-6.5) were used. Sixty-seven per cent (63/94) experienced significant brain volume loss. For the prediction of disability progression, we developed a two-level procedure. In the first level, a 10-gene predictor achieved a classification accuracy of 70.9 ± 4.5% in identifying patients reaching the disability end-point within 120 weeks. In the second level, a four-gene classifier distinguished between fast and slow disability progression with a 506-day cut-off, achieving 74.1 ± 5.2% accuracy. For brain volume loss prediction, a 12-gene classifier reached an accuracy of 70.2 ± 6.7%, which improved to 74.1 ± 5.2% when combined with baseline brain MRI measurements. In conclusion, our study demonstrates that blood transcriptome data, alone or combined with baseline brain MRI metrics, can effectively predict disability progression and brain volume loss in PPMS patients.
原发性进行性多发性硬化症(PPMS)影响10-15%的多发性硬化症患者,在残疾进展率方面表现出显著的可变性。确定关键的生物学特征和高风险的快速进展患者对于制定和优化治疗策略至关重要。外周血细胞转录组有可能为预测患者预后提供有价值的信息。在这项研究中,我们利用机器学习框架应用于基线血液转录谱和脑MRI放射计数来开发预后模型。这些模型旨在确定可能经历重大疾病进展的PPMS患者,以及谁可以从早期治疗干预中受益。使用Illumina NovaSeq S2对ORATORIO临床试验(NCT01412333)安慰剂组PPMS患者外周血单个核细胞中提取的总RNA进行RNA序列分析。来自Partek Genome Suite (www.partek.com)的交叉验证算法用于预测120周内的残疾进展和脑容量损失。为了预测残疾进展,我们分析了135名PPMS患者(61名女性和74名男性)的血液RNA样本,平均±标准误差年龄为44.0±0.7岁,疾病持续时间为5.9±0.32年,中位基线扩展残疾状态量表(EDSS)评分为4.3(范围为3.5-6.5)。在120周的研究中,39.3%(53/135)的患者达到残疾进展终点,平均EDSS评分增加1.3±0.16。为了预测脑容量损失,使用94名PPMS患者(41名女性,53名男性)的血液RNA样本,平均±标准误差年龄为43.7±0.7岁,基线EDSS中位数为4.0(范围为3.0-6.5)。67%(63/94)经历了显著的脑容量损失。为了预测残疾进展,我们制定了一个两级程序。在第一级,10个基因预测器在识别120周内达到残疾终点的患者时达到了70.9±4.5%的分类准确率。在第二级,四基因分类器区分快速和缓慢的残疾进展,截断时间为506天,准确率为74.1±5.2%。对于脑容量损失预测,12个基因分类器的准确率为70.2±6.7%,与基线脑MRI测量相结合时提高到74.1±5.2%。总之,我们的研究表明,血液转录组数据,单独或结合基线脑MRI指标,可以有效地预测PPMS患者的残疾进展和脑容量损失。
{"title":"Machine learning-based prediction of disease progression in primary progressive multiple sclerosis.","authors":"Michael Gurevich, Rina Zilkha-Falb, Jia Sherman, Maxime Usdin, Catarina Raposo, Licinio Craveiro, Polina Sonis, David Magalashvili, Shay Menascu, Mark Dolev, Anat Achiron","doi":"10.1093/braincomms/fcae427","DOIUrl":"https://doi.org/10.1093/braincomms/fcae427","url":null,"abstract":"<p><p>Primary progressive multiple sclerosis (PPMS) affects 10-15% of multiple sclerosis patients and presents significant variability in the rate of disability progression. Identifying key biological features and patients at higher risk for fast progression is crucial to develop and optimize treatment strategies. Peripheral blood cell transcriptome has the potential to provide valuable information to predict patients' outcomes. In this study, we utilized a machine learning framework applied to the baseline blood transcriptional profiles and brain MRI radiological enumerations to develop prognostic models. These models aim to identify PPMS patients likely to experience significant disease progression and who could benefit from early treatment intervention. RNA-sequence analysis was performed on total RNA extracted from peripheral blood mononuclear cells of PPMS patients in the placebo arm of the ORATORIO clinical trial (NCT01412333), using Illumina NovaSeq S2. Cross-validation algorithms from Partek Genome Suite (www.partek.com) were applied to predict disability progression and brain volume loss over 120 weeks. For disability progression prediction, we analysed blood RNA samples from 135 PPMS patients (61 females and 74 males) with a mean ± standard error age of 44.0 ± 0.7 years, disease duration of 5.9 ± 0.32 years and a median baseline Expanded Disability Status Scale (EDSS) score of 4.3 (range 3.5-6.5). Over the 120-week study, 39.3% (53/135) of patients reached the disability progression end-point, with an average EDSS score increase of 1.3 ± 0.16. For brain volume loss prediction, blood RNA samples from 94 PPMS patients (41 females and 53 males), mean ± standard error age of 43.7 ± 0.7 years and a median baseline EDSS of 4.0 (range 3.0-6.5) were used. Sixty-seven per cent (63/94) experienced significant brain volume loss. For the prediction of disability progression, we developed a two-level procedure. In the first level, a 10-gene predictor achieved a classification accuracy of 70.9 ± 4.5% in identifying patients reaching the disability end-point within 120 weeks. In the second level, a four-gene classifier distinguished between fast and slow disability progression with a 506-day cut-off, achieving 74.1 ± 5.2% accuracy. For brain volume loss prediction, a 12-gene classifier reached an accuracy of 70.2 ± 6.7%, which improved to 74.1 ± 5.2% when combined with baseline brain MRI measurements. In conclusion, our study demonstrates that blood transcriptome data, alone or combined with baseline brain MRI metrics, can effectively predict disability progression and brain volume loss in PPMS patients.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcae427"},"PeriodicalIF":4.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11707605/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142960253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcaf004
Yi-Ting Wang, Nicholas J Ashton, Joseph Therriault, Andréa L Benedet, Arthur C Macedo, Ilaria Pola, Etienne Aumont, Guglielmo Di Molfetta, Jaime Fernandez-Arias, Kubra Tan, Nesrine Rahmouni, Stijn Johannes G Servaes, Richard Isaacson, Tevy Chan, Seyyed Ali Hosseini, Cécile Tissot, Sulantha Mathotaarachchi, Jenna Stevenson, Firoza Z Lussier, Tharick A Pascoal, Serge Gauthier, Kaj Blennow, Henrik Zetterberg, Pedro Rosa-Neto
Blood-based biomarkers have been revolutionizing the detection, diagnosis and screening of Alzheimer's disease. Specifically, phosphorylated-tau variants (p-tau181, p-tau217 and p-tau231) are promising biomarkers for identifying Alzheimer's disease pathology. Antibody-based assays such as single molecule arrays immunoassays are powerful tools to investigate pathological changes indicated by blood-based biomarkers and have been studied extensively in the Alzheimer's disease research field. A novel proteomic technology-NUcleic acid Linked Immuno-Sandwich Assay (NULISA)-was developed to improve the sensitivity of traditional proximity ligation assays and offer a comprehensive outlook for 120 protein biomarkers in neurodegenerative diseases. Due to the relative novelty of the NULISA technology in quantifying Alzheimer's disease biomarkers, validation through comparisons with more established methods is required. The main objective of the current study was to determine the capability of p-tau variants quantified using NULISA for identifying abnormal amyloid-β and tau pathology. We assessed 397 participants [mean (standard deviation) age, 64.8 (15.7) years; 244 females (61.5%) and 153 males (38.5%)] from the Translational Biomarkers in Aging and Dementia (TRIAD) cohort where participants had plasma measurements of p-tau181, p-tau217 and p-tau231 from NULISA and single molecule arrays immunoassays. Participants also underwent neuroimaging assessments, including structural MRI, amyloid-PET and tau-PET. Our findings suggest an excellent agreement between plasma p-tau variants quantified using NULISA and single molecule arrays immunoassays. Plasma p-tau217 measured with NULISA shows excellent discriminative accuracy for abnormal amyloid-PET (area under the receiver operating characteristic curve = 0.918, 95% confidence interval = 0.883 to 0.953, P < 0.0001) and tau-PET (area under the receiver operating characteristic curve = 0.939; 95% confidence interval = 0.909 to 0.969, P < 0.0001). It also presents the capability for differentiating tau-PET staging. Validation of the NULISA-measured plasma biomarkers adds to the current analytical methods for Alzheimer's disease diagnosis, screening and staging and could potentially expedite the development of a blood-based biomarker panel.
{"title":"Identify biological Alzheimer's disease using a novel nucleic acid-linked protein immunoassay.","authors":"Yi-Ting Wang, Nicholas J Ashton, Joseph Therriault, Andréa L Benedet, Arthur C Macedo, Ilaria Pola, Etienne Aumont, Guglielmo Di Molfetta, Jaime Fernandez-Arias, Kubra Tan, Nesrine Rahmouni, Stijn Johannes G Servaes, Richard Isaacson, Tevy Chan, Seyyed Ali Hosseini, Cécile Tissot, Sulantha Mathotaarachchi, Jenna Stevenson, Firoza Z Lussier, Tharick A Pascoal, Serge Gauthier, Kaj Blennow, Henrik Zetterberg, Pedro Rosa-Neto","doi":"10.1093/braincomms/fcaf004","DOIUrl":"10.1093/braincomms/fcaf004","url":null,"abstract":"<p><p>Blood-based biomarkers have been revolutionizing the detection, diagnosis and screening of Alzheimer's disease. Specifically, phosphorylated-tau variants (p-tau<sub>181</sub>, p-tau<sub>217</sub> and p-tau<sub>231</sub>) are promising biomarkers for identifying Alzheimer's disease pathology. Antibody-based assays such as single molecule arrays immunoassays are powerful tools to investigate pathological changes indicated by blood-based biomarkers and have been studied extensively in the Alzheimer's disease research field. A novel proteomic technology-NUcleic acid Linked Immuno-Sandwich Assay (NULISA)-was developed to improve the sensitivity of traditional proximity ligation assays and offer a comprehensive outlook for 120 protein biomarkers in neurodegenerative diseases. Due to the relative novelty of the NULISA technology in quantifying Alzheimer's disease biomarkers, validation through comparisons with more established methods is required. The main objective of the current study was to determine the capability of p-tau variants quantified using NULISA for identifying abnormal amyloid-β and tau pathology. We assessed 397 participants [mean (standard deviation) age, 64.8 (15.7) years; 244 females (61.5%) and 153 males (38.5%)] from the Translational Biomarkers in Aging and Dementia (TRIAD) cohort where participants had plasma measurements of p-tau<sub>181</sub>, p-tau<sub>217</sub> and p-tau<sub>231</sub> from NULISA and single molecule arrays immunoassays. Participants also underwent neuroimaging assessments, including structural MRI, amyloid-PET and tau-PET. Our findings suggest an excellent agreement between plasma p-tau variants quantified using NULISA and single molecule arrays immunoassays. Plasma p-tau<sub>217</sub> measured with NULISA shows excellent discriminative accuracy for abnormal amyloid-PET (area under the receiver operating characteristic curve = 0.918, 95% confidence interval = 0.883 to 0.953, <i>P</i> < 0.0001) and tau-PET (area under the receiver operating characteristic curve = 0.939; 95% confidence interval = 0.909 to 0.969, <i>P</i> < 0.0001). It also presents the capability for differentiating tau-PET staging. Validation of the NULISA-measured plasma biomarkers adds to the current analytical methods for Alzheimer's disease diagnosis, screening and staging and could potentially expedite the development of a blood-based biomarker panel.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcaf004"},"PeriodicalIF":4.1,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753389/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143025820","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcae448
Shreya Mukherjee, Tracey Singer, Aditi Venkatesh, Natasha A Choudhury, Gina S Perez Giraldo, Millenia Jimenez, Janet Miller, Melissa Lopez, Barbara A Hanson, Aasheeta P Bawa, Ayush Batra, Eric M Liotta, Igor J Koralnik
<p><p>Persistent symptoms after COVID-19 constitute the long COVID syndrome, also called post-acute sequelae of SARS-CoV-2 infection (PASC). COVID-19 vaccines reduce the gravity of ensuing SARS-CoV-2 infections. However, whether vaccines also have an impact on PASC remain unknown. We investigated whether vaccination prior to infection alters the subsequent neurologic post-acute sequelae of SARS-CoV-2 infection (Neuro-PASC). We studied prospectively the first consecutive 200 post-hospitalization Neuro-PASC (PNP) and 1100 non-hospitalized Neuro-PASC (NNP) patients evaluated at our neuro-COVID-19 clinic between May 2020 and January 2023. Among PNP patients, 87% had a pre-vaccination infection and 13% had a breakthrough infection post-vaccination. Among the NNP patients, 70.7% had a pre-vaccination infection and 29.3% had a breakthrough infection. Both PNP and NNP breakthrough infection patients had more frequent pre-existing depression/anxiety than their respective pre-vaccination infection groups, and NNP breakthrough infection patients also had more frequent comorbidities of headache, lung and gastrointestinal diseases than the NNP pre-vaccination infection group. An average of 10 months after symptom onset, the three most common neurological symptoms for PNP patients were brain fog (86.5%), numbness/tingling (56.5%) and headache (56.5%). Of all Neuro-PASC symptoms, PNP breakthrough infection more frequently reported anosmia compared to PNP pre-vaccination infection patients (69.2 versus 37.9%; <i>P</i> = 0.005). For NNP patients, the three most common neurological symptoms were brain fog (83.9%), headache (70.9%) and dizziness (53.8%). NNP pre-vaccination infection reported anosmia (56.6 versus 39.1%; <i>P</i> < 0.0001) and dysgeusia (53.3 versus 37.3%; <i>P</i> < 0.0001) more frequently than breakthrough infection patients. NNP breakthrough infection more frequently reported dizziness compared to NNP pre-vaccination infection patients (61.5 versus 50.6%; <i>P</i> = 0.001). Both PNP and NNP patients had impaired quality-of-life in cognitive, fatigue, sleep, anxiety and depression domains with no differences between pre-vaccination infection and breakthrough infection groups. PNP patients performed worse on National Institutes of Health Toolbox tests of processing speed, attention, executive function and working memory than a US normative population whereas NNP patients had lower results in processing, speed, attention and working memory, without differences between pre-vaccination infection and breakthrough infection groups. These results indicate that vaccination prior to SARS-CoV-2 infection does not affect the neurologic manifestations of long COVID in either PNP or NNP patients. Minor differences in neurologic symptoms between pre-vaccination infection and breakthrough infection groups may be caused by SARS-CoV-2 strains evolution. Patients developing Neuro-PASC after breakthrough infection have a higher burden of comorbidities, highlighting
COVID-19后的持续症状构成长COVID综合征,也称为SARS-CoV-2感染急性后后遗症(PASC)。COVID-19疫苗降低了随后的SARS-CoV-2感染的严重性。然而,疫苗是否对PASC也有影响尚不清楚。我们调查了感染前接种疫苗是否会改变SARS-CoV-2感染后的神经系统急性后后遗症(neuropasc)。我们前瞻性研究了2020年5月至2023年1月在我们的神经- covid -19诊所评估的首批连续200例住院后神经- pasc (PNP)和1100例非住院神经- pasc (NNP)患者。在PNP患者中,87%的人在接种前感染,13%的人在接种后出现突破性感染。在NNP患者中,接种前感染占70.7%,突破感染占29.3%。PNP和NNP突破感染患者的既往抑郁/焦虑发生率均高于接种前感染组,NNP突破感染患者的头痛、肺部和胃肠道疾病合并症发生率均高于接种前感染组。症状出现后平均10个月,PNP患者最常见的3种神经系统症状为脑雾(86.5%)、麻木/刺痛(56.5%)和头痛(56.5%)。在所有神经- pasc症状中,与接种前感染PNP的患者相比,PNP突破感染更频繁地报告嗅觉缺失(69.2 vs 37.9%;P = 0.005)。NNP患者最常见的3种神经系统症状为脑雾(83.9%)、头痛(70.9%)和头晕(53.8%)。NNP疫苗接种前感染报告嗅觉缺失(56.6%对39.1%;P < 0.0001)和发音困难(53.3 vs 37.3%;P < 0.0001)高于突破感染患者。与NNP疫苗接种前感染患者相比,NNP突破感染更频繁地报告头晕(61.5%对50.6%;P = 0.001)。PNP和NNP患者在认知、疲劳、睡眠、焦虑和抑郁领域的生活质量受损,接种前感染组和突破感染组之间没有差异。PNP患者在美国国立卫生研究院工具箱测试的处理速度、注意力、执行功能和工作记忆方面的表现低于美国正常人群,而NNP患者在处理、速度、注意力和工作记忆方面的结果低于接种前感染组和突破感染组。这些结果表明,在感染SARS-CoV-2之前接种疫苗对PNP或NNP患者的长冠状病毒神经系统表现没有影响。疫苗接种前感染组与突破感染组神经系统症状的微小差异可能是由SARS-CoV-2毒株进化引起的。突破性感染后发生神经pasc的患者有更高的合并症负担,突出了不同的危险因素,需要有针对性的管理。
{"title":"Vaccination prior to SARS-CoV-2 infection does not affect the neurologic manifestations of long COVID.","authors":"Shreya Mukherjee, Tracey Singer, Aditi Venkatesh, Natasha A Choudhury, Gina S Perez Giraldo, Millenia Jimenez, Janet Miller, Melissa Lopez, Barbara A Hanson, Aasheeta P Bawa, Ayush Batra, Eric M Liotta, Igor J Koralnik","doi":"10.1093/braincomms/fcae448","DOIUrl":"https://doi.org/10.1093/braincomms/fcae448","url":null,"abstract":"<p><p>Persistent symptoms after COVID-19 constitute the long COVID syndrome, also called post-acute sequelae of SARS-CoV-2 infection (PASC). COVID-19 vaccines reduce the gravity of ensuing SARS-CoV-2 infections. However, whether vaccines also have an impact on PASC remain unknown. We investigated whether vaccination prior to infection alters the subsequent neurologic post-acute sequelae of SARS-CoV-2 infection (Neuro-PASC). We studied prospectively the first consecutive 200 post-hospitalization Neuro-PASC (PNP) and 1100 non-hospitalized Neuro-PASC (NNP) patients evaluated at our neuro-COVID-19 clinic between May 2020 and January 2023. Among PNP patients, 87% had a pre-vaccination infection and 13% had a breakthrough infection post-vaccination. Among the NNP patients, 70.7% had a pre-vaccination infection and 29.3% had a breakthrough infection. Both PNP and NNP breakthrough infection patients had more frequent pre-existing depression/anxiety than their respective pre-vaccination infection groups, and NNP breakthrough infection patients also had more frequent comorbidities of headache, lung and gastrointestinal diseases than the NNP pre-vaccination infection group. An average of 10 months after symptom onset, the three most common neurological symptoms for PNP patients were brain fog (86.5%), numbness/tingling (56.5%) and headache (56.5%). Of all Neuro-PASC symptoms, PNP breakthrough infection more frequently reported anosmia compared to PNP pre-vaccination infection patients (69.2 versus 37.9%; <i>P</i> = 0.005). For NNP patients, the three most common neurological symptoms were brain fog (83.9%), headache (70.9%) and dizziness (53.8%). NNP pre-vaccination infection reported anosmia (56.6 versus 39.1%; <i>P</i> < 0.0001) and dysgeusia (53.3 versus 37.3%; <i>P</i> < 0.0001) more frequently than breakthrough infection patients. NNP breakthrough infection more frequently reported dizziness compared to NNP pre-vaccination infection patients (61.5 versus 50.6%; <i>P</i> = 0.001). Both PNP and NNP patients had impaired quality-of-life in cognitive, fatigue, sleep, anxiety and depression domains with no differences between pre-vaccination infection and breakthrough infection groups. PNP patients performed worse on National Institutes of Health Toolbox tests of processing speed, attention, executive function and working memory than a US normative population whereas NNP patients had lower results in processing, speed, attention and working memory, without differences between pre-vaccination infection and breakthrough infection groups. These results indicate that vaccination prior to SARS-CoV-2 infection does not affect the neurologic manifestations of long COVID in either PNP or NNP patients. Minor differences in neurologic symptoms between pre-vaccination infection and breakthrough infection groups may be caused by SARS-CoV-2 strains evolution. Patients developing Neuro-PASC after breakthrough infection have a higher burden of comorbidities, highlighting ","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcae448"},"PeriodicalIF":4.1,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11703551/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142960176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-07eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcae477
Lina Rydén, Nazib M Seidu, Hanna Wetterberg, Jenna Najar, Margda Waern, Silke Kern, Kaj Blennow, Henrik Zetterberg, Ingmar Skoog, Anna Zettergren
Atrial fibrillation and heart failure have both been suggested to increase stroke and dementia risk. However, in observational studies, reversed causation and unmeasured confounding may occur. To mitigate these issues, this study aims to investigate if higher genetic risk for atrial fibrillation and heart failure increases dementia and stroke risk. Data were obtained from the population-based Gothenburg H70 Birth Cohort Studies in Sweden. Participants (N = 984) were born in 1930 with baseline examinations at age 70, 75, 79 or 85 and follow-ups until age 88-89. Polygenic risk scores at the 5 × 10-8, 1 × 10-5, 1 × 10-3 and 1 × 10-1 thresholds were generated for atrial fibrillation and heart failure. Stroke was diagnosed based on self-reports, close-informant interviews, and the National Patient Register. Dementia was diagnosed based on neuropsychiatric examinations, close-informant interviews, and the National Patient Register. Cox regression analyses were performed, adjusted for sex, age at baseline and the first five principal components to correct for population stratification. Those within the highest atrial fibrillation-polygenic risk score tertile had a 1.5 (95% CI 1.09-2.03) increased risk of dementia (at the 1 × 10-5 threshold) and a 1.5 (95% CI 1.07-2.03) increased risk of stroke (at the 1 × 10-3 threshold) compared to the lowest tertile. Those within the highest heart failure-polygenic risk score tertile had a 1.6 (95% CI 1.19-2.27) increased risk of dementia (at the 5 × 10-8 threshold), but no increased risk of stroke (HR 1.2; 95% CI 0.83-1.60 at the 1 × 10-5 threshold), compared to the lowest tertile. When analysing the polygenic risk scores as a continuous variable, the associations were in the same direction, although weaker. This study, investigating genetic risk of atrial fibrillation and heart failure in relation to stroke and dementia, supports the increasing body of evidence suggesting that atrial fibrillation is associated with both stroke and dementia risk. Whether heart failure increases dementia risk is less established, but the present study found that genetic risk of heart failure increased dementia risk. The finding that genetic risk for heart failure did not increase stroke risk needs to be interpreted with caution, as it may be due to a lack of statistical power. There are guidelines on how to best treat atrial fibrillation to prevent stroke, but more knowledge is needed on how to treat atrial fibrillation and heart failure to prevent dementia.
心房颤动和心力衰竭都被认为会增加中风和痴呆的风险。然而,在观察性研究中,可能会出现反向因果关系和未测量的混淆。为了缓解这些问题,本研究旨在调查心房颤动和心力衰竭的较高遗传风险是否会增加痴呆和中风的风险。数据来自瑞典以人群为基础的哥德堡H70出生队列研究。参与者(N = 984)出生于1930年,在70岁、75岁、79岁或85岁时进行基线检查,并随访至88-89岁。心房颤动和心力衰竭的多基因风险评分分别为5 × 10-8、1 × 10-5、1 × 10-3和1 × 10-1阈值。中风的诊断是基于自我报告、近距离访谈和国家患者登记。痴呆的诊断是基于神经精神病学检查、密切的知情访谈和国家患者登记册。进行Cox回归分析,调整性别、基线年龄和前五个主要成分以校正人口分层。与最低分位数相比,房颤-多基因风险评分最高分位数的患者痴呆风险增加1.5 (95% CI 1.09-2.03) (1 × 10-5阈值),卒中风险增加1.5 (95% CI 1.07-2.03) (1 × 10-3阈值)。心力衰竭-多基因风险评分最高的患者痴呆风险增加1.6 (95% CI 1.19-2.27) (5 × 10-8阈值),但卒中风险未增加(HR 1.2;95% CI 0.83-1.60 (1 × 10-5阈值),与最低分位数相比。当将多基因风险评分作为一个连续变量进行分析时,相关性虽然较弱,但方向相同。这项研究调查了心房颤动和心力衰竭与中风和痴呆的遗传风险,支持越来越多的证据表明心房颤动与中风和痴呆风险相关。心力衰竭是否会增加患痴呆症的风险尚不确定,但目前的研究发现,心力衰竭的遗传风险会增加患痴呆症的风险。心力衰竭的遗传风险不会增加中风的风险,这一发现需要谨慎解释,因为这可能是由于缺乏统计能力。有关于如何最好地治疗房颤以预防中风的指南,但如何治疗房颤和心力衰竭以预防痴呆需要更多的知识。
{"title":"Polygenic risk scores for atrial fibrillation and heart failure and the risk of stroke and dementia.","authors":"Lina Rydén, Nazib M Seidu, Hanna Wetterberg, Jenna Najar, Margda Waern, Silke Kern, Kaj Blennow, Henrik Zetterberg, Ingmar Skoog, Anna Zettergren","doi":"10.1093/braincomms/fcae477","DOIUrl":"10.1093/braincomms/fcae477","url":null,"abstract":"<p><p>Atrial fibrillation and heart failure have both been suggested to increase stroke and dementia risk. However, in observational studies, reversed causation and unmeasured confounding may occur. To mitigate these issues, this study aims to investigate if higher genetic risk for atrial fibrillation and heart failure increases dementia and stroke risk. Data were obtained from the population-based Gothenburg H70 Birth Cohort Studies in Sweden. Participants (<i>N</i> = 984) were born in 1930 with baseline examinations at age 70, 75, 79 or 85 and follow-ups until age 88-89. Polygenic risk scores at the 5 × 10<sup>-8</sup>, 1 × 10<sup>-5</sup>, 1 × 10<sup>-3</sup> and 1 × 10<sup>-1</sup> thresholds were generated for atrial fibrillation and heart failure. Stroke was diagnosed based on self-reports, close-informant interviews, and the National Patient Register. Dementia was diagnosed based on neuropsychiatric examinations, close-informant interviews, and the National Patient Register. Cox regression analyses were performed, adjusted for sex, age at baseline and the first five principal components to correct for population stratification. Those within the highest atrial fibrillation-polygenic risk score tertile had a 1.5 (95% CI 1.09-2.03) increased risk of dementia (at the 1 × 10<sup>-5</sup> threshold) and a 1.5 (95% CI 1.07-2.03) increased risk of stroke (at the 1 × 10<sup>-3</sup> threshold) compared to the lowest tertile. Those within the highest heart failure-polygenic risk score tertile had a 1.6 (95% CI 1.19-2.27) increased risk of dementia (at the 5 × 10<sup>-8</sup> threshold), but no increased risk of stroke (HR 1.2; 95% CI 0.83-1.60 at the 1 × 10<sup>-5</sup> threshold), compared to the lowest tertile. When analysing the polygenic risk scores as a continuous variable, the associations were in the same direction, although weaker. This study, investigating genetic risk of atrial fibrillation and heart failure in relation to stroke and dementia, supports the increasing body of evidence suggesting that atrial fibrillation is associated with both stroke and dementia risk. Whether heart failure increases dementia risk is less established, but the present study found that genetic risk of heart failure increased dementia risk. The finding that genetic risk for heart failure did not increase stroke risk needs to be interpreted with caution, as it may be due to a lack of statistical power. There are guidelines on how to best treat atrial fibrillation to prevent stroke, but more knowledge is needed on how to treat atrial fibrillation and heart failure to prevent dementia.</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcae477"},"PeriodicalIF":4.1,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11748287/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018297","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcae478
Burak Yulug, Ozlem Altay, Xiangyu Li, Lutfu Hanoglu, Seyda Cankaya, Halil A Velioglu, Simon Lam, Hong Yang, Ebru Coskun, Ezgi Idil, Zubeyir Bayraktaroglu, Rahim Nogaylar, Ahmet Ozsimsek, Serkan Yildirim, Ismail Bolat, Metin Kiliclioglu, Cemil Bayram, Nursena Yuksel, Ozlem O Tozlu, Muhammad Arif, Saeed Shoaie, Ahmet Hacimuftuoglu, Cheng Zhang, Jens Nielsen, Hasan Turkez, Jan Borén, Mathias Uhlén, Adil Mardinoglu
Parkinson's disease is primarily marked by mitochondrial dysfunction and metabolic abnormalities. We recently reported that the combined metabolic activators improved the immunohistochemical parameters and behavioural functions in Parkinson's disease and Alzheimer's disease animal models and the cognitive functions in Alzheimer's disease patients. These metabolic activators serve as the precursors of nicotinamide adenine dinucleotide and glutathione, and they can be used to activate mitochondrial metabolism and eventually treat mitochondrial dysfunction. Here, we designed a randomized, double-blinded, placebo-controlled phase II study in Parkinson's disease patients with 84 days combined metabolic activator administration. A single dose of combined metabolic activator contains L-serine (12.35 g), N-acetyl-L-cysteine (2.55 g), nicotinamide riboside (1 g) and L-carnitine tartrate (3.73 g). Patients were administered either one dose of combined metabolic activator or a placebo daily for the initial 28 days, followed by twice-daily dosing for the next 56 days. The main goal of the study was to evaluate the clinical impact on motor functions using the Unified Parkinson's Disease Rating Scale and to determine the safety and tolerability of combined metabolic activator. A secondary objective was to assess cognitive functions utilizing the Montreal Cognitive Assessment and to analyse brain activity through functional MRI. We also performed comprehensive plasma metabolomics and proteomics analysis for detailed characterization of Parkinson's disease patients who participated in the study. Although no improvement in motor functions was observed, cognitive function was shown to be significantly improved (P < 0.0000) in Parkinson's disease patients treated with the combined metabolic activator group over 84 days, whereas no such improvement was noted in the placebo group (P > 0.05). Moreover, a significant reduction (P = 0.001) in Montreal Cognitive Assessment scores was observed in the combined metabolic activator group, with no decline (P > 0.05) in the placebo group among severe Parkinson's disease patients with lower baseline Montreal Cognitive Assessment scores. We showed that improvement in cognition was associated with critical brain network alterations based on functional MRI analysis, especially relevant to areas with cognitive functions in the brain. Finally, through a comprehensive multi-omics analysis, we elucidated the molecular mechanisms underlying cognitive improvements observed in Parkinson's disease patients. Our results show that combined metabolic activator administration leads to enhanced cognitive function and improved metabolic health in Parkinson's disease patients as recently shown in Alzheimer's disease patients. The trial was registered in ClinicalTrials.gov NCT04044131 (17 July 2019, https://clinicaltrials.gov/ct2/show/NCT04044131).
{"title":"Multi-omics characterization of improved cognitive functions in Parkinson's disease patients after the combined metabolic activator treatment: a randomized, double-blinded, placebo-controlled phase II trial.","authors":"Burak Yulug, Ozlem Altay, Xiangyu Li, Lutfu Hanoglu, Seyda Cankaya, Halil A Velioglu, Simon Lam, Hong Yang, Ebru Coskun, Ezgi Idil, Zubeyir Bayraktaroglu, Rahim Nogaylar, Ahmet Ozsimsek, Serkan Yildirim, Ismail Bolat, Metin Kiliclioglu, Cemil Bayram, Nursena Yuksel, Ozlem O Tozlu, Muhammad Arif, Saeed Shoaie, Ahmet Hacimuftuoglu, Cheng Zhang, Jens Nielsen, Hasan Turkez, Jan Borén, Mathias Uhlén, Adil Mardinoglu","doi":"10.1093/braincomms/fcae478","DOIUrl":"10.1093/braincomms/fcae478","url":null,"abstract":"<p><p>Parkinson's disease is primarily marked by mitochondrial dysfunction and metabolic abnormalities. We recently reported that the combined metabolic activators improved the immunohistochemical parameters and behavioural functions in Parkinson's disease and Alzheimer's disease animal models and the cognitive functions in Alzheimer's disease patients. These metabolic activators serve as the precursors of nicotinamide adenine dinucleotide and glutathione, and they can be used to activate mitochondrial metabolism and eventually treat mitochondrial dysfunction. Here, we designed a randomized, double-blinded, placebo-controlled phase II study in Parkinson's disease patients with 84 days combined metabolic activator administration. A single dose of combined metabolic activator contains L-serine (12.35 g), <i>N</i>-acetyl-L-cysteine (2.55 g), nicotinamide riboside (1 g) and L-carnitine tartrate (3.73 g). Patients were administered either one dose of combined metabolic activator or a placebo daily for the initial 28 days, followed by twice-daily dosing for the next 56 days. The main goal of the study was to evaluate the clinical impact on motor functions using the Unified Parkinson's Disease Rating Scale and to determine the safety and tolerability of combined metabolic activator. A secondary objective was to assess cognitive functions utilizing the Montreal Cognitive Assessment and to analyse brain activity through functional MRI. We also performed comprehensive plasma metabolomics and proteomics analysis for detailed characterization of Parkinson's disease patients who participated in the study. Although no improvement in motor functions was observed, cognitive function was shown to be significantly improved (<i>P</i> < 0.0000) in Parkinson's disease patients treated with the combined metabolic activator group over 84 days, whereas no such improvement was noted in the placebo group (<i>P</i> > 0.05). Moreover, a significant reduction (<i>P</i> = 0.001) in Montreal Cognitive Assessment scores was observed in the combined metabolic activator group, with no decline (<i>P</i> > 0.05) in the placebo group among severe Parkinson's disease patients with lower baseline Montreal Cognitive Assessment scores. We showed that improvement in cognition was associated with critical brain network alterations based on functional MRI analysis, especially relevant to areas with cognitive functions in the brain. Finally, through a comprehensive multi-omics analysis, we elucidated the molecular mechanisms underlying cognitive improvements observed in Parkinson's disease patients. Our results show that combined metabolic activator administration leads to enhanced cognitive function and improved metabolic health in Parkinson's disease patients as recently shown in Alzheimer's disease patients. The trial was registered in ClinicalTrials.gov NCT04044131 (17 July 2019, https://clinicaltrials.gov/ct2/show/NCT04044131).</p>","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcae478"},"PeriodicalIF":4.1,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11733689/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143018231","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-06eCollection Date: 2025-01-01DOI: 10.1093/braincomms/fcaf002
Frederik O Hansen, Karoline Knudsen, Malene F Damholdt, Toke Bek, Per Borghammer, Niels Okkels
<p><p>Asymmetric dopaminergic degeneration of the striatum is a characteristic feature of Parkinson's disease, associated with right-left asymmetry in motor function. As such, studying asymmetry provides insights into progressive neurodegeneration between cerebral hemispheres. Given the impact of Lewy pathology on various neurotransmitter systems beyond the dopaminergic, it may be that other neuronal systems in the predominantly affected hemisphere are similarly affected. According to this hypothesis, asymmetry in dopaminergic degeneration would be expected to coincide with asymmetry in other neurotransmitter systems. Consequently, asymmetry in functions primarily dependent on dopaminergic integrity, such as motor function, should correlate with asymmetry in bilateral non-motor functions that rely on other cerebral systems, such as pupillary function. Therefore, this study tested whether right-left asymmetry in bilateral non-motor measures correlates with asymmetry in dopaminergic striatal integrity. We also tested whether asymmetric striatal degeneration is associated with greater asymmetry in non-motor measures overall. Using a comparative cross-sectional design, we recruited newly diagnosed patients with Parkinson's disease with predominantly right-sided (<i>n</i> = 18), left-sided (<i>n</i> = 15) or symmetric nigrostriatal denervation (<i>n</i> = 15) assessed on dopamine PET. Detailed examinations of lateralized non-motor function included lacrimation, hand skin wrinkling, salivation, olfaction and pupillary function. Healthy controls were recruited for comparison. We observed a moderate-to-strong correlation between right-left asymmetry of putamen dopamine binding and asymmetry in pupillary redilation speed [Spearman's rank correlation coefficient (<i>r<sub>s</sub></i> ) = -0.53, 95% confidence interval (-0.77; -0.14), <i>P</i> = 0.0084]. We also observed moderate correlations between non-negative putaminal asymmetry and lacrimation [<i>r<sub>s</sub></i> = 0.35, (-0.00; 0.62), <i>P</i> = 0.0464] and word recognition [<i>r<sub>s</sub></i> = 0.36, (0.01; 0.63), <i>P</i> = 0.0410]. However, none were significant after false discovery rate correction. We observed significant group differences in non-negative asymmetry in salivation (<i>P</i> = 0.0390, ANOVA) and a trend towards greater asymmetric lacrimation in participants with asymmetric striatal dopamine loss compared with healthy controls (<i>P</i> = 0.0330, unadjusted). Additionally, participants with asymmetric striatal dopaminergic binding showed greater, though non-significant, asymmetry in all pupillary measures compared with those with symmetric dopaminergic binding. In conclusion, this study contributes to our understanding of neurodegeneration progression in Parkinson's disease and suggests a link between dopaminergic degeneration and non-motor measures related to autonomic function, particularly salivation, lacrimation and pupillary function. While our findings do not support a stri
{"title":"Non-motor asymmetry and dopamine degeneration in Parkinson's disease.","authors":"Frederik O Hansen, Karoline Knudsen, Malene F Damholdt, Toke Bek, Per Borghammer, Niels Okkels","doi":"10.1093/braincomms/fcaf002","DOIUrl":"10.1093/braincomms/fcaf002","url":null,"abstract":"<p><p>Asymmetric dopaminergic degeneration of the striatum is a characteristic feature of Parkinson's disease, associated with right-left asymmetry in motor function. As such, studying asymmetry provides insights into progressive neurodegeneration between cerebral hemispheres. Given the impact of Lewy pathology on various neurotransmitter systems beyond the dopaminergic, it may be that other neuronal systems in the predominantly affected hemisphere are similarly affected. According to this hypothesis, asymmetry in dopaminergic degeneration would be expected to coincide with asymmetry in other neurotransmitter systems. Consequently, asymmetry in functions primarily dependent on dopaminergic integrity, such as motor function, should correlate with asymmetry in bilateral non-motor functions that rely on other cerebral systems, such as pupillary function. Therefore, this study tested whether right-left asymmetry in bilateral non-motor measures correlates with asymmetry in dopaminergic striatal integrity. We also tested whether asymmetric striatal degeneration is associated with greater asymmetry in non-motor measures overall. Using a comparative cross-sectional design, we recruited newly diagnosed patients with Parkinson's disease with predominantly right-sided (<i>n</i> = 18), left-sided (<i>n</i> = 15) or symmetric nigrostriatal denervation (<i>n</i> = 15) assessed on dopamine PET. Detailed examinations of lateralized non-motor function included lacrimation, hand skin wrinkling, salivation, olfaction and pupillary function. Healthy controls were recruited for comparison. We observed a moderate-to-strong correlation between right-left asymmetry of putamen dopamine binding and asymmetry in pupillary redilation speed [Spearman's rank correlation coefficient (<i>r<sub>s</sub></i> ) = -0.53, 95% confidence interval (-0.77; -0.14), <i>P</i> = 0.0084]. We also observed moderate correlations between non-negative putaminal asymmetry and lacrimation [<i>r<sub>s</sub></i> = 0.35, (-0.00; 0.62), <i>P</i> = 0.0464] and word recognition [<i>r<sub>s</sub></i> = 0.36, (0.01; 0.63), <i>P</i> = 0.0410]. However, none were significant after false discovery rate correction. We observed significant group differences in non-negative asymmetry in salivation (<i>P</i> = 0.0390, ANOVA) and a trend towards greater asymmetric lacrimation in participants with asymmetric striatal dopamine loss compared with healthy controls (<i>P</i> = 0.0330, unadjusted). Additionally, participants with asymmetric striatal dopaminergic binding showed greater, though non-significant, asymmetry in all pupillary measures compared with those with symmetric dopaminergic binding. In conclusion, this study contributes to our understanding of neurodegeneration progression in Parkinson's disease and suggests a link between dopaminergic degeneration and non-motor measures related to autonomic function, particularly salivation, lacrimation and pupillary function. While our findings do not support a stri","PeriodicalId":93915,"journal":{"name":"Brain communications","volume":"7 1","pages":"fcaf002"},"PeriodicalIF":4.1,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11752486/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143025830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}