Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.945
1.
BMC Geriatr ; 24(1): 501, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844858

BACKGROUND: Core biomarkers for Alzheimer's disease (AD), such as Aß42 and tau, have demonstrated high prognostic accuracy but do not fully capture the complex pathophysiology of AD. In this study, our objective was to identify novel cerebrospinal fluid (CSF) biomarkers using proteomics across the entire AD continuum to predict conversion to AD and explore their involvement in AD pathogenesis. METHODS: A cohort of 186 cognitively normal (CN), 127 subjective memory complaint (SMC), 79 early mild cognitive impairment (EMCI), 249 late MCI (LMCI), and 132 AD individuals was analyzed, with a follow-up period of over 3 years for non-AD participants. CSF 65 peptides, as well as hippocampal and entorhinal volumes were analyzed, and cognitive function was evaluated using the 13-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cog 13). Cox proportional hazards models and mediation analysis were performed to investigate associations and causal relationships. RESULTS: During the follow-up, approximately one-fourth (146/580) of the non-AD participants progressed to AD. After adjusting for baseline diagnosis (CN to LMCI) and other variables, multivariable Cox regression analysis identified three peptides (VAELEDEK, VSFELFADK, and VVSSIEQK) as significant predictors of conversion to AD. Incorporating these three peptides into the initial model significantly improved the C-statistic from 0.82 to 0.85 for predicting AD conversion, surpassing the predictive ability of Aß42 and P-tau. Moreover, hippocampal and entorhinal volumes mediated 30.3-53.8% of the association between the three peptides and ADAS-Cog 13 scores. CONCLUSIONS: These findings underscore the potential of these three peptides as robust prognostic biomarker candidates for AD conversion across the entire AD continuum, with a mechanism involving the mediation of hippocampal and entorhinal volumes.


Alzheimer Disease , Biomarkers , Proteomics , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Male , Female , Aged , Proteomics/methods , Prognosis , Biomarkers/cerebrospinal fluid , Follow-Up Studies , Cohort Studies , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/diagnosis , Aged, 80 and over , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Disease Progression , Middle Aged , Predictive Value of Tests , tau Proteins/cerebrospinal fluid
2.
Neurology ; 102(12): e209418, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38830138

BACKGROUND AND OBJECTIVES: Plasma ß-amyloid-1-42/1-40 (Aß42/40), phosphorylated-tau (P-tau), glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) have been widely examined in Alzheimer disease (AD), but little is known about their reflection of copathologies, clinical importance, and predictive value in dementia with Lewy bodies (DLB). We aimed to evaluate associations of these biomarkers with CSF amyloid, cognition, and core features in DLB. METHODS: This cross-sectional multicenter cohort study with prospective component included individuals with DLB, AD, and healthy controls (HCs), recruited from 2002 to 2020 with an annual follow-up of up to 5 years, from the European-Dementia With Lewy Bodies consortium. Plasma biomarkers were measured by single-molecule array (Neurology 4-Plex E kit). Amyloid status was determined by CSF Aß42 concentrations, and cognition was assessed by Mini-Mental State Examination (MMSE). Biomarker differences across groups, associations with amyloid status, and clinical core features were assessed by analysis of covariance. Associations with cognitive impairment and decline were assessed by linear regression and linear mixed-effects models. RESULTS: In our cohort consisting of 562 individuals (HC n = 89, DLB n = 342, AD n = 131; 250 women [44.5%], mean [SD] age of 71 [8] years), sex distribution did not differ between groups. Patients with DLB were significantly older, and had less years of education and worse baseline cognition than HC, but not AD. DLB participants stratified for amyloid status differed significantly in plasma Aß42/40 ratio (decreased in amyloid abnormal: ß = -0.008, 95% CI -0.016 to -0.0003, p = 0.01) and P-tau (increased in amyloid abnormal, P-tau181: ß = 0.246, 95% CI 0.011-0.481; P-tau231: ß = 0.227, 95% CI 0.035-0.419, both p < 0.05), but not in GFAP (ß = 0.068, 95% CI -0.018 to 0.153, p = 0.119), and NfL (ß = 0.004, 95% CI -0.087 to 0.096, p = 0.923) concentrations. Higher baseline GFAP, NfL, and P-tau concentrations were associated with lower MMSE scores in DLB, and GFAP and NfL were associated with a faster cognitive decline (GFAP: annual change of -2.11 MMSE points, 95% CI -2.88 to -1.35 MMSE points, p < 0.001; NfL: annual change of -2.13 MMSE points, 95% CI -2.97 to -1.29 MMSE points, p < 0.001). DLB participants with parkinsonism had higher concentrations of NfL (ß = 0.08, 95% CI 0.02-0.14, p = 0.006) than those without. DISCUSSION: Our study suggests a possible utility of plasma Aß42/40, P-tau181, and P-tau231 as a noninvasive biomarkers to assess amyloid copathology in DLB, and plasma GFAP and NfL as monitoring biomarkers for cognitive symptoms in DLB.


Amyloid beta-Peptides , Biomarkers , Glial Fibrillary Acidic Protein , Lewy Body Disease , Neurofilament Proteins , tau Proteins , Humans , Female , Male , tau Proteins/cerebrospinal fluid , tau Proteins/blood , Aged , Lewy Body Disease/cerebrospinal fluid , Lewy Body Disease/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Glial Fibrillary Acidic Protein/blood , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Cross-Sectional Studies , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Middle Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/blood , Aged, 80 and over , Cohort Studies , Prospective Studies , Cognition/physiology , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/blood
3.
J Neuroimmune Pharmacol ; 19(1): 27, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38829507

Reverse transcriptase inhibitors (RTIs) are currently broadly prescribed for the treatment of HIV infection but are also thought to prevent Alzheimer's disease (AD) progression by protecting against amyloidosis. Our study evaluates the hypothesis that reverse transcriptase inhibitors protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection. We compiled a case series of participants from a prospective study of the neurological consequences of HIV infection at the HIV Neurobehavioral Research Program (HNRP) who had serial neuropsychological and neurological assessments and were on RTIs. Two participants had gross and microscopic examination and immunohistochemistry of the brain at autopsy; one was assessed clinically for Alzheimer's disease by cerebrospinal fluid (CSF) analysis of phosphorylated-Tau, Total-Tau and Aß42. Additionally, a larger cohort of 250 autopsied individuals was evaluated for presence of amyloid plaques, Tau, and related pathologies. Three older, virally suppressed individuals with HIV who had long-term treatment with RTIs were included in analyses. Two cases demonstrated substantial cerebral amyloid deposition at autopsy. The third case met clinical criteria for AD based on a typical clinical course and CSF biomarker profile. In the larger cohort of autopsied individuals, the prevalence of cerebral amyloidosis among people with HIV (PWH) was greater for those on RTIs. Our study showed that long-term RTI therapy did not protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection in these patients. Given the known toxicities of RTIs, it is premature to recommend them to individuals at risk or with Alzheimer's disease who do not have HIV infection.


Alzheimer Disease , Amyloidosis , HIV Infections , Humans , HIV Infections/complications , HIV Infections/drug therapy , Alzheimer Disease/pathology , Male , Middle Aged , Female , Aged , Prospective Studies , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism , Brain/pathology , Brain/metabolism , Brain/diagnostic imaging , Cohort Studies , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism
4.
Nat Commun ; 15(1): 4706, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38830849

The neuromodulatory subcortical nuclei within the isodendritic core (IdC) are the earliest sites of tauopathy in Alzheimer's disease (AD). They project broadly throughout the brain's white matter. We investigated the relationship between IdC microstructure and whole-brain white matter microstructure to better understand early neuropathological changes in AD. Using multiparametric quantitative magnetic resonance imaging we observed two covariance patterns between IdC and white matter microstructure in 133 cognitively unimpaired older adults (age 67.9 ± 5.3 years) with familial risk for AD. IdC integrity related to 1) whole-brain neurite density, and 2) neurite orientation dispersion in white matter tracts known to be affected early in AD. Pattern 2 was associated with CSF concentration of phosphorylated-tau, indicating AD specificity. Apolipoprotein-E4 carriers expressed both patterns more strongly than non-carriers. IdC microstructure variation is reflected in white matter, particularly in AD-affected tracts, highlighting an early mechanism of pathological development.


Alzheimer Disease , Magnetic Resonance Imaging , Tauopathies , White Matter , tau Proteins , Humans , White Matter/diagnostic imaging , White Matter/pathology , White Matter/metabolism , Female , Male , Aged , Middle Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/genetics , Tauopathies/cerebrospinal fluid , tau Proteins/metabolism , tau Proteins/cerebrospinal fluid , Brain/pathology , Brain/diagnostic imaging , Brain/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Neurites/metabolism , Neurites/pathology
5.
Alzheimers Res Ther ; 16(1): 103, 2024 May 10.
Article En | MEDLINE | ID: mdl-38725083

BACKGROUND: 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. METHODS: 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. RESULTS: 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. CONCLUSIONS: 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.


Biomarkers , Cognitive Dysfunction , alpha-Synuclein , tau Proteins , Humans , Female , Male , Cognitive Dysfunction/cerebrospinal fluid , alpha-Synuclein/cerebrospinal fluid , Aged , tau Proteins/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Aged, 80 and over , Neuropsychological Tests
6.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(4. Vyp. 2): 33-40, 2024.
Article Ru | MEDLINE | ID: mdl-38696149

OBJECTIVE: To study the severity and localization of dilated perivascular spaces (DPVS), the levels of protein markers of amyloidosis and neurodegeneration in the cerebrospinal fluid (CSF) at different daily blood pressure (BP) profiles in patients with Alzheimer's disease (AD) and other types of cognitive impairment. MATERIAL AND METHODS: A total of 119 people, aged 53 to 92 years, including 55 patients with AD, 27 patients with vascular cognitive disorders (VCD), 19 patients with frontotemporal degeneration (FTD). All patients underwent BP monitoring for 24 hours using a standard oscillometric measurement method, lumbar puncture to assess Aß-42 and Aß-40 amyloid protein, total and phosphorylated tau protein in the CSF, magnetic resonance imaging tomography of the brain with subsequent assessment of the severity of expansion and localization of DPVS according to the G.M. Potter scale. RESULTS: In 58.3% of patients with AD, there is no adequate reduction in BP at night in comparison with patients with VCD (p<0.05). A significant degree of expansion of the DPVS turned out to be most typical for patients with AD: grade 3 was detected in 45.7% of patients, and the maximum, grade 4, was detected in 13.4%. At the same time, DPVSs were significantly more often detected in the group of subjects with insufficient reduction in diastolic BP (DBP) at night. A strong inverse correlation was established between the level of Aß-42 in the CSF and the variability of DBP at night (r= -0.92; p<0.05). The decrease in the level of Aß-42 in AD, especially at the prodromal stage, is directly related to the low variability of DBP at night, which is more characteristic of an insufficient decrease or increase in BP during night sleep. CONCLUSION: Patients with AD were characterized by an insufficient decrease in BP at night, which is associated with the severity and degree of maximum expansion of the DPVS. A decrease in the level of Aß-42 amyloid protein in the CSF strongly correlates with the variability of DBP at night.


Alzheimer Disease , Amyloid beta-Peptides , Hypertension , tau Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Aged , Female , Male , Middle Aged , Amyloid beta-Peptides/cerebrospinal fluid , Hypertension/complications , Hypertension/cerebrospinal fluid , Aged, 80 and over , tau Proteins/cerebrospinal fluid , Magnetic Resonance Imaging , Glymphatic System/diagnostic imaging , Blood Pressure/physiology , Peptide Fragments/cerebrospinal fluid , Dementia, Vascular/cerebrospinal fluid , Dementia, Vascular/diagnostic imaging , Biomarkers/cerebrospinal fluid , Brain/diagnostic imaging , Brain/pathology
7.
Alzheimers Res Ther ; 16(1): 99, 2024 May 04.
Article En | MEDLINE | ID: mdl-38704569

BACKGROUND: Patients with sporadic cerebral amyloid angiopathy (sCAA) frequently report cognitive or neuropsychiatric symptoms. The aim of this study is to investigate whether in patients with sCAA, cognitive impairment and neuropsychiatric symptoms are associated with a cerebrospinal fluid (CSF) biomarker profile associated with Alzheimer's disease (AD). METHODS: In this cross-sectional study, we included participants with sCAA and dementia- and stroke-free, age- and sex-matched controls, who underwent a lumbar puncture, brain MRI, cognitive assessments, and self-administered and informant-based-questionnaires on neuropsychiatric symptoms. CSF phosphorylated tau, total tau and Aß42 levels were used to divide sCAA patients in two groups: CAA with (CAA-AD+) or without a CSF biomarker profile associated with AD (CAA-AD-). Performance on global cognition, specific cognitive domains (episodic memory, working memory, processing speed, verbal fluency, visuoconstruction, and executive functioning), presence and severity of neuropsychiatric symptoms, were compared between groups. RESULTS: sCAA-AD+ (n=31; mean age: 72 ± 6; 42%, 61% female) and sCAA-AD- (n=23; 70 ± 5; 42% female) participants did not differ with respect to global cognition or type of affected cognitive domain(s). The number or severity of neuropsychiatric symptoms also did not differ between sCAA-AD+ and sCAA-AD- participants. These results did not change after exclusion of patients without prior ICH. CONCLUSIONS: In participants with sCAA, a CSF biomarker profile associated with AD does not impact global cognition or specific cognitive domains, or the presence of neuropsychiatric symptoms.


Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Cerebral Amyloid Angiopathy , Neuropsychological Tests , tau Proteins , Humans , Female , Male , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Aged , Cross-Sectional Studies , Cerebral Amyloid Angiopathy/cerebrospinal fluid , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/diagnostic imaging , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/etiology , Peptide Fragments/cerebrospinal fluid , Cognition/physiology , Middle Aged , Magnetic Resonance Imaging
8.
J Prev Alzheimers Dis ; 11(3): 701-709, 2024.
Article En | MEDLINE | ID: mdl-38706286

BACKGROUND: The polygenic risk score (PRS) aggregates the effects of numerous genetic variants associated with a condition across the human genome and may help to predict late-onset Alzheimer's disease (LOAD). Most of the current PRS studies on Alzheimer's disease (AD) have been conducted in Caucasian ancestry populations, while it is less studied in Chinese. OBJECTIVE: To establish and examine the validity of Chinese PRS, and explore its racial heterogeneity. DESIGN: We constructed a PRS using both discovery (N = 2012) and independent validation samples (N = 1008) from Chinese population. The associations between PRS and age at onset of LOAD or cerebrospinal fluid (CSF) biomarkers were assessed. We also replicated the PRS in an independent replication cohort with CSF data and constructed an alternative PRS using European weights. SETTING: Multi-center genetics study. PARTICIPANTS: A total of 3020 subjects were included in the study. MEASUREMENTS: PRS was calculated using genome-wide association studies data and evaluated the performance alone (PRSnoAPOE) and with other predictors (full model: LOAD ~ PRSnoAPOE + APOE+ sex + age) by measuring the area under the receiver operating curve (AUC). RESULTS: PRS of the full model achieved the highest AUC of 84.0% (95% CI = 81.4-86.5) with pT< 0.5, compared with the model containing APOE alone (61.0%). The AUC of PRS with pT<5e-8 was 77.8% in the PRSnoAPOE model, 81.5% in the full model, and only ranged from 67.5% to 75.1% in the PRS with the European weights model. A higher PRS was significantly associated with an earlier age at onset (P <0.001). The PRS also performed well in the replication cohort of the full model (AUC=83.1%, 95% CI = 74.3-92.0). The CSF biomarkers of Aß42 and the ratio of Aß42/Aß40 were significantly inversely associated with the PRS, while p-Tau181 showed a positive association. CONCLUSIONS: This finding suggests that PRS reveal genetic heterogeneity and higher prediction accuracy of the PRS for AD can be achieved using a base dataset and validation within the same ethnicity. The effective PRS model has the clinical potential to predict individuals at risk of developing LOAD at a given age and with abnormal levels of CSF biomarkers in the Chinese population.


Alzheimer Disease , East Asian People , Genome-Wide Association Study , Multifactorial Inheritance , White People , Aged , Female , Humans , Male , Middle Aged , Age of Onset , Alzheimer Disease/genetics , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , China/epidemiology , East Asian People/genetics , Genetic Heterogeneity , Genetic Predisposition to Disease , Genetic Risk Score , Risk Factors , tau Proteins/cerebrospinal fluid , tau Proteins/genetics , White People/genetics
9.
J Prev Alzheimers Dis ; 11(3): 672-683, 2024.
Article En | MEDLINE | ID: mdl-38706283

BACKGROUND: We aimed to explore whether the relationships of blood pressures (BPs) with Alzheimer's disease (AD) endophenotypes varied by usage of antihypertensive drugs (AHDs). METHODS: A total of 765 non-demented older adults (mean age: 74.4 years; female: 43.1%) with a self-reported history of hypertension were followed for 6 years. Multiple linear regression and linear-mixed effect models were used to investigate the interaction effects of five categories of AHDs (angiotensin-converting enzyme inhibitors [ACEI], angiotensin II receptor blockers [ARBs], ß-blocker, calcium channel blockers [CCB], diuretic) with BPs (systolic blood pressure [SBP], diastolic blood pressure [DBP], and pulse pressure [PP]) on AD core pathology and neurodegenerative markers. RESULTS: After Bonferroni correction, significant interaction effects of BPs with AHDs were observed. Elevated SBP or PP in late-life was associated with higher levels of cerebral Aß burden (diuretic alone/ß-blocker × SBP), higher levels of CSF tau proteins (diuretic × SBP/PP, ARBs/CCB × SBP), and lower volume of entorhinal region (ß-blocker × SBP, diuretic × PP) only among hypertensive patients who received no anti-hypertensive treatments, while these associations became compromised or null for users of specific AHDs except for ACEI. Compared to taking other classes of AHDs, elevated SBP in late-life was associated with lower cerebral Aß burden in diuretic users (padjusted = 0.08) and was associated with higher CSF tau proteins in ACEI alone users (padjusted = 0.03). Longitudinal data validated the above-mentioned interaction effects on changes of cerebral Aß burden (padjusted < 0.05), CSF tau proteins (padjusted < 0.10), and brain atrophy (padjusted < 0.05). CONCLUSIONS: The relationships of late-life BP with AD pathology and neurodegeneration could be modified by anti-hypertensive treatments and varied by AHD classification. These findings provide preliminary evidence for tailored BP management strategy for preventing AD among late-life hypertensive adults.


Alzheimer Disease , Antihypertensive Agents , Blood Pressure , Hypertension , Humans , Aged , Female , Male , Hypertension/drug therapy , Alzheimer Disease/drug therapy , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Blood Pressure/physiology , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Aged, 80 and over , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Calcium Channel Blockers/therapeutic use
10.
Alzheimer Dis Assoc Disord ; 38(2): 107-111, 2024.
Article En | MEDLINE | ID: mdl-38752577

BACKGROUND: Blood-brain barrier (BBB) dysfunction is emerging as an important pathophysiologic factor in Alzheimer disease (AD). Cerebrospinal fluid (CSF) platelet-derived growth factor receptor-ß (PDGFRß) is a biomarker of BBB pericyte injury and has been implicated in cognitive impairment and AD. METHODS: We aimed to study CSF PDGFRß protein levels, along with CSF biomarkers of brain amyloidosis and tau pathology in a well-characterized population of cognitively unimpaired individuals and correlated CSF findings with amyloid-PET positivity. We performed an institutional review board (IRB)-approved cross-sectional analysis of a prospectively enrolled cohort of 36 cognitively normal volunteers with available CSF, Pittsburgh compound B PET/CT, Mini-Mental State Exam score, Global Deterioration Scale, and known apolipoprotein E ( APOE ) ε4 status. RESULTS: Thirty-six subjects were included. Mean age was 63.3 years; 31 of 36 were female, 6 of 36 were amyloid-PET-positive and 12 of 36 were APOE ε4 carriers. We found a moderate positive correlation between CSF PDGFRß and both total Tau (r=0.45, P =0.006) and phosphorylated Tau 181 (r=0.51, P =0.002). CSF PDGFRß levels were not associated with either the CSF Aß42 or the amyloid-PET. CONCLUSIONS: We demonstrated a moderate positive correlation between PDGFRß and both total Tau and phosphorylated Tau 181 in cognitively normal individuals. Our data support the hypothesis that BBB dysfunction represents an important early pathophysiologic step in AD, warranting larger prospective studies. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT00094939.


Alzheimer Disease , Biomarkers , Pericytes , tau Proteins , Humans , Female , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Male , Biomarkers/cerebrospinal fluid , Middle Aged , Cross-Sectional Studies , Aged , tau Proteins/cerebrospinal fluid , Pericytes/pathology , Positron-Emission Tomography , Amyloid beta-Peptides/cerebrospinal fluid , Blood-Brain Barrier , Receptor, Platelet-Derived Growth Factor beta/cerebrospinal fluid , Prospective Studies , Cohort Studies
11.
J Alzheimers Dis ; 99(3): 883-885, 2024.
Article En | MEDLINE | ID: mdl-38759014

With the advent of therapeutics with potential to slow Alzheimer's disease progression the necessity of understanding the diagnostic value of plasma biomarkers is critical, not only for understanding the etiology and progression of Alzheimer's disease, but also for access and response to potentially disease modifying therapeutic agents. Multiple studies are currently assessing the sensitivity and specificity of plasma biomarkers in large cohorts such as the Alzheimer's Disease Neuroimaging Initiative. This study uses machine learning to predict the progression from mild cognitive impairment using plasma biomarkers in conjunction with well-established cerebrospinal fluid and imaging biomarkers of disease progression.


Alzheimer Disease , Biomarkers , Disease Progression , Machine Learning , Neurofilament Proteins , tau Proteins , Humans , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Alzheimer Disease/cerebrospinal fluid , Biomarkers/blood , Biomarkers/cerebrospinal fluid , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Aged , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Female , Male , Prognosis , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnosis , Cohort Studies , Aged, 80 and over
12.
Int J Mol Sci ; 25(10)2024 May 08.
Article En | MEDLINE | ID: mdl-38791145

The diagnostic and prognostic value of plasma glial fibrillary acidic protein (pl-GFAP) in sporadic Creutzfeldt-Jakob disease (sCJD) has never been assessed in the clinical setting of rapidly progressive dementia (RPD). Using commercially available immunoassays, we assayed the plasma levels of GFAP, tau (pl-tau), and neurofilament light chain (pl-NfL) and the CSF total tau (t-tau), 14-3-3, NfL, phospho-tau181 (p-tau), and amyloid-beta isoforms 42 (Aß42) and 40 (Aß40) in sCJD (n = 132) and non-prion RPD (np-RPD) (n = 94) patients, and healthy controls (HC) (n = 54). We also measured the CSF GFAP in 67 sCJD patients. Pl-GFAP was significantly elevated in the sCJD compared to the np-RPD and HC groups and affected by the sCJD subtype. Its diagnostic accuracy (area under the curve (AUC) 0.760) in discriminating sCJD from np-RPD was higher than the plasma and CSF NfL (AUCs of 0.596 and 0.663) but inferior to the 14-3-3, t-tau, and pl-tau (AUCs of 0.875, 0.918, and 0.805). Pl-GFAP showed no association with sCJD survival after adjusting for known prognostic factors. Additionally, pl-GFAP levels were associated with 14-3-3, pl-tau, and pl-NfL but not with CSF GFAP, Aß42/Aß40, and p-tau. The diagnostic and prognostic value of pl-GFAP is inferior to established neurodegeneration biomarkers. Nonetheless, pl-GFAP noninvasively detects neuroinflammation and neurodegeneration in sCJD, warranting potential applications in disease monitoring.


Amyloid beta-Peptides , Biomarkers , Creutzfeldt-Jakob Syndrome , Dementia , Glial Fibrillary Acidic Protein , tau Proteins , Humans , Creutzfeldt-Jakob Syndrome/blood , Creutzfeldt-Jakob Syndrome/diagnosis , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Female , Male , Glial Fibrillary Acidic Protein/blood , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Aged , Middle Aged , Prognosis , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Dementia/blood , Dementia/diagnosis , Dementia/cerebrospinal fluid , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Disease Progression , 14-3-3 Proteins/cerebrospinal fluid , 14-3-3 Proteins/blood
13.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article En | MEDLINE | ID: mdl-38731812

We compared the clinical and analytical performance of Alzheimer's disease (AD) plasma biomarkers measured using the single-molecule array (Simoa) and Lumipulse platforms. We quantified the plasma levels of amyloid beta 42 (Aß42), Aß40, phosphorylated tau (Ptau181), and total tau biomarkers in 81 patients with mild cognitive impairment (MCI), 30 with AD, and 16 with non-AD dementia. We found a strong correlation between the Simoa and Lumipulse methods. Concerning the clinical diagnosis, Simoa Ptau181/Aß42 (AUC 0.739, 95% CI 0.592-0.887) and Lumipulse Aß42 and Ptau181/Aß42 (AUC 0.735, 95% CI 0.589-0.882 and AUC 0.733, 95% CI 0.567-0.900) had the highest discriminating power. However, their power was significantly lower than that of CSF Aß42/Aß40, as measured by Lumipulse (AUC 0.879, 95% CI 0.766-0.992). Simoa Ptau181 and Lumipulse Ptau181/Aß42 were the markers most consistent with the CSF Aß42/Aß40 status (AUC 0.801, 95% CI 0.712-0.890 vs. AUC 0.870, 95% CI 0.806-0.934, respectively) at the ≥2.127 and ≥0.084 cut-offs, respectively. The performance of the Simoa and Lumipulse plasma AD assays is weaker than that of CSF AD biomarkers. At present, the analysed AD plasma biomarkers may be useful for screening to reduce the number of lumbar punctures in the clinical setting.


Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Cognitive Dysfunction , tau Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Male , Female , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Aged , tau Proteins/cerebrospinal fluid , tau Proteins/blood , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/blood , Middle Aged , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Aged, 80 and over , Phosphorylation
14.
Nat Med ; 30(5): 1284-1291, 2024 May.
Article En | MEDLINE | ID: mdl-38710950

This study aimed to evaluate the impact of APOE4 homozygosity on Alzheimer's disease (AD) by examining its clinical, pathological and biomarker changes to see whether APOE4 homozygotes constitute a distinct, genetically determined form of AD. Data from the National Alzheimer's Coordinating Center and five large cohorts with AD biomarkers were analyzed. The analysis included 3,297 individuals for the pathological study and 10,039 for the clinical study. Findings revealed that almost all APOE4 homozygotes exhibited AD pathology and had significantly higher levels of AD biomarkers from age 55 compared to APOE3 homozygotes. By age 65, nearly all had abnormal amyloid levels in cerebrospinal fluid, and 75% had positive amyloid scans, with the prevalence of these markers increasing with age, indicating near-full penetrance of AD biology in APOE4 homozygotes. The age of symptom onset was earlier in APOE4 homozygotes at 65.1, with a narrower 95% prediction interval than APOE3 homozygotes. The predictability of symptom onset and the sequence of biomarker changes in APOE4 homozygotes mirrored those in autosomal dominant AD and Down syndrome. However, in the dementia stage, there were no differences in amyloid or tau positron emission tomography across haplotypes, despite earlier clinical and biomarker changes. The study concludes that APOE4 homozygotes represent a genetic form of AD, suggesting the need for individualized prevention strategies, clinical trials and treatments.


Alzheimer Disease , Apolipoprotein E4 , Biomarkers , Homozygote , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Age of Onset , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/cerebrospinal fluid , Amyloid/metabolism , Amyloid/genetics , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Biomarkers/cerebrospinal fluid , Cohort Studies , Positron-Emission Tomography , tau Proteins/genetics , tau Proteins/cerebrospinal fluid
15.
J Alzheimers Dis ; 99(3): 1077-1092, 2024.
Article En | MEDLINE | ID: mdl-38759016

Background: Although Lumipulse assays and conventional ELISA are strongly correlated, the precise relationship between their measured values remains undetermined. Objective: To determine the relationship between Lumipulse and ELISA measurement values. Methods: Patients who underwent cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarker measurements and consented to biobanking between December 2021 and June 2023 were included. The relationship between values measured via Lumipulse assays and conventional ELISA were evaluated by Passing-Bablok analyses for amyloid-ß 1-42 (Aß42), total tau (t-tau), and phospho-tau 181 (p-tau 181). Studies using both assays were systematically searched for in PubMed and summarized after quality assessment. Results: Regression line slopes and intercepts were 1.41 (1.23 to 1.60) and -77.8 (-198.4 to 44.5) for Aß42, 0.94 (0.88 to 1.01) and 98.2 (76.9 to 114.4) for t-tau, and 1.60 (1.43 to 1.75) and -21.1 (-26.9 to -15.6) for p-tau181. Spearman's correlation coefficients were 0.90, 0.95, and 0.95 for Aß42, t-tau, and p-tau181, respectively. We identified 13 other studies that included 2,117 patients in total. Aß42 slope varied among studies, suggesting inter-lab difference of ELISA. The slope and intercept of t-tau were approximately 1 and 0, respectively, suggesting small proportional and systematic differences. Conversely, the p-tau181 slope was significantly higher than 1, distributed between 1.5-2 in most studies, with intercepts significantly lower than 0, suggesting proportional and systematic differences. Conclusions: We characterized different relationship between measurement values for each biomarker, which may be useful for understanding the differences in CSF biomarker measurement values on different platforms and for future global harmonization.


Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Enzyme-Linked Immunosorbent Assay , Peptide Fragments , tau Proteins , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Humans , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay/methods , tau Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid
16.
Lakartidningen ; 1212024 May 31.
Article Sv | MEDLINE | ID: mdl-38818759

Technical developments have paved the way for the development of ultrasensitive analytical methods that allow for precise quantification of brain-specific proteins in blood samples. Plasma levels of amyloid ß, specifically the Aß42/40 ratio, are reduced in Alzheimer's disease (AD) and show concordance with brain amyloidosis assessed by PET, but the overlap with normal elderly may be too large for reliable use in clinical applications. Plasma phosphorylated tau (P-tau), especially a variant called P-tau217, is markedly increased in the early symptomatic stages of AD but remains normal in other neurodegenerative disorders. Total tau (T-tau) is measurable in blood and shows most promise as a biomarker for acute neuronal injury (e.g. acute traumatic or hypoxic brain injury), where T-tau shows a fast and dramatic increase but does not work well as an AD biomarker due to contributions to blood levels from peripheral tissues. Instead, a novel method for tau protein produced only in the CNS called brain-derived tau (BD-tau) shows promise as a biomarker for AD-type neurodegeneration. Neurofilament light (NFL) levels in blood correlate tightly with levels in CSF and reflect axonal injury irrespective of the underlying cause. Increased blood NFL concentration is found in several neurodegenerative disorders, including AD, but even more so in disorders such as motor neuron disease and frontotemporal dementia. Glial fibrillary acidic protein (GFAP) is expressed with activation of astrocytes, and is mildly increased in AD, but is also very high also in acute brain disorders. These blood tests show promise as tools to identify AD pathophysiology in the first assessment of patients with early cognitive symptoms, also in primary care, to guide clinical management and possible admission to the specialist clinic. A two-step model will result in a very high accuracy to either predict or exclude brain amyloidosis of the Alzheimer type.


Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Brain , Neurofilament Proteins , tau Proteins , Humans , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Alzheimer Disease/physiopathology , Biomarkers/blood , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/diagnostic imaging , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Glial Fibrillary Acidic Protein/blood
17.
Neurology ; 102(12): e209426, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38776513

BACKGROUND AND OBJECTIVES: With the aging US population and increasing incidence of Alzheimer disease (AD), understanding factors contributing to driving cessation among older adults is crucial for clinicians. Driving is integral for maintaining independence and functional mobility, but the risk factors for driving cessation, particularly in the context of normal aging and preclinical AD, are not well understood. We studied a well-characterized community cohort to examine factors associated with driving cessation. METHODS: This prospective, longitudinal observation study enrolled participants from the Knight Alzheimer Disease Research Center and The DRIVES Project. Participants were enrolled if they were aged 65 years or older, drove weekly, and were cognitively normal (Clinical Dementia Rating [CDR] = 0) at baseline. Participants underwent annual clinical, neurologic, and neuropsychological assessments, including ß-amyloid PET imaging and CSF (Aß42, total tau [t-Tau], and phosphorylated tau [p-Tau]) collection every 2-3 years. The primary outcome was time from baseline visit to driving cessation, accounting for death as a competing risk. The cumulative incidence function of driving cessation was estimated for each biomarker. The Fine and Gray subdistribution hazard model was used to examine the association between time to driving cessation and biomarkers adjusting for clinical and demographic covariates. RESULTS: Among the 283 participants included in this study, there was a mean follow-up of 5.62 years. Driving cessation (8%) was associated with older age, female sex, progression to symptomatic AD (CDR ≥0.5), and poorer performance on a preclinical Alzheimer cognitive composite (PACC) score. Aß PET imaging did not independently predict driving cessation, whereas CSF biomarkers, specifically t-Tau/Aß42 (hazard ratio [HR] 2.82, 95% CI 1.23-6.44, p = 0.014) and p-Tau/Aß42 (HR 2.91, 95% CI 1.28-6.59, p = 0.012) ratios, were independent predictors in the simple model adjusting for age, education, and sex. However, in the full model, progression to cognitive impairment based on the CDR and PACC score across each model was associated with a higher risk of driving cessation, whereas AD biomarkers were not statistically significant. DISCUSSION: Female sex, CDR progression, and neuropsychological measures of cognitive functioning obtained in the clinic were strongly associated with future driving cessation. The results emphasize the need for early planning and conversations about driving retirement in the context of cognitive decline and the immense value of clinical measures in determining functional outcomes.


Alzheimer Disease , Amyloid beta-Peptides , Automobile Driving , Biomarkers , tau Proteins , Humans , Female , Male , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/epidemiology , Alzheimer Disease/diagnosis , Aged , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , tau Proteins/cerebrospinal fluid , Aged, 80 and over , Longitudinal Studies , Prospective Studies , Positron-Emission Tomography , Neuropsychological Tests , Cognition/physiology , Peptide Fragments/cerebrospinal fluid
18.
Cephalalgia ; 44(5): 3331024241248203, 2024 May.
Article En | MEDLINE | ID: mdl-38690635

BACKGROUND: Idiopathic intracranial hypertension is a secondary headache disorder potentially causing visual loss. Neurofilament light chain is a candidate, prognostic biomarker, but further studies of neuronal biomarkers are needed. Our objective was to investigate neurofilament light chain in cerebrospinal fluid (cNfL) and plasma (pNfL), amyloid-beta 42 (Aß-42), total-tau and phosphorylated-tau in cerebrospinal fluid in new-onset idiopathic intracranial hypertension. METHODS: Prospective case-control study including new-onset idiopathic intracranial hypertension and age, sex and BMI matched controls. Biomarkers were compared between patients and controls and related to papilledema, visual fields and opening pressure. RESULTS: We included 37 patients and 35 controls. Patients had higher age-adjusted cNfL (1.4 vs. 0.6 pg/mL, p-adjusted < 0.001), pNfL (0.5 vs. 0.3 pg/mL, p-adjusted < 0.001) and total-tau/Aß-42 (0.12 vs. 0.11, p-adjusted = 0.039). Significant, positive linear correlations were found between cNfL, pNfL, total-tau/Aß-42 and opening pressure. Patients with severe papilledema had elevated cNfL compared to mild-moderate papilledema (median cNfL: 4.3 pg/mL (3.7) versus 1.0 pg/mL (1.4), p-adjusted = 0.009). cNFL was inversely associated with perimetric mean deviation (r = -0.47, p-adjusted < 0.001). CONCLUSIONS: cNfL, pNfL and total-tau/Aß-42 were elevated in new-onset idiopathic intracranial hypertension. cNfL was associated with severity of papilledema and visual field defects at diagnosis. This indicates early axonal damage. Neurofilament light chain is a candidate biomarker for disease severity.


Biomarkers , Neurofilament Proteins , Pseudotumor Cerebri , Humans , Female , Male , Neurofilament Proteins/cerebrospinal fluid , Neurofilament Proteins/blood , Adult , Pseudotumor Cerebri/diagnosis , Pseudotumor Cerebri/cerebrospinal fluid , Pseudotumor Cerebri/blood , Pseudotumor Cerebri/complications , Prospective Studies , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Case-Control Studies , Middle Aged , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , tau Proteins/blood
19.
J Prev Alzheimers Dis ; 11(3): 730-738, 2024.
Article En | MEDLINE | ID: mdl-38706289

BACKGROUND: Serum-measured fragments of Tau cleaved by ADAM-10 (Tau-A) and Caspase-3 (Tau-C) have been found linked to change in cognitive function and risk of dementia. OBJECTIVES: 1) To determine the discriminatory abilities of Tau-A, and Tau-C in subjects with either mild cognitive impairment (MCI) due to Alzheimer's disease (AD) or AD dementia compared to a control group. 2) To determine if there is a relation between Tau-A, and Tau-C and established cerebrospinal fluid (CSF) markers of AD- ß-Amyloid1-42 (AB42), Phosphorylated-tau-181 (p-tau), and total-tau. 3) To determine if Tau-A and Tau-C are associated with progression rate from MCI due to AD to AD dementia. DESIGN: Cross-sectional and a substudy using a retrospective cohort design. SETTING: Memory clinic derived subjects contributing to the Danish Dementia Biobank. PARTICIPANTS: Cognitively unimpaired subjects (n=49), patients with mild cognitive impairment (MCI) due to AD (n=45), and Alzheimer's dementia (n=52). MEASUREMENTS: Competitive enzyme-linked immunosorbent assay (ELISA)-measured serum levels of Tau-A, and Tau-C. RESULTS: The ratio between Tau-A and Tau-C differed between the three groups (p=0.015). Age- and sex-adjusted Tau-A differed between groups with lower ratios being associated with more severe disease (p=0.023). Tau-C was trending towards significant correlation to CSF-levels of AB42 (Pearson correlation coefficient 0.164, p=0.051). Those with Tau-C-levels in the 2nd quartile had a hazard ratio (HR) of 2.91 (95% CI 1.01 - 8.44, p=0.04) of progression compared to those in the 1st quartile. Those in the 3rd quartile was found to have a borderline significant (p=0.055) HR of 2.63 (95% CI 0.98 - 7.05) when compared to those in the lowest quartile. CONCLUSIONS: Tau-A and the ratio between Tau-A and Tau-C showed significant differences between groups and were correlated to CSF-AB42. Tau-C values in the middle range were associated with faster progression from MCI to dementia. This pilot study adds to the mounting data suggesting serum-measured Tau-A and Tau-C as biomarkers useful in relation to diagnosis and progression rate in AD but need further validation.


Alzheimer Disease , Biomarkers , Cognitive Dysfunction , Disease Progression , tau Proteins , Humans , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnosis , Male , Female , Aged , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Cross-Sectional Studies , Retrospective Studies , Middle Aged , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Dementia/blood , Cohort Studies , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid
20.
Sci Rep ; 14(1): 11307, 2024 05 17.
Article En | MEDLINE | ID: mdl-38760423

We aimed to assess diagnostic accuracy of plasma p-tau181 and NfL separately and in combination in discriminating Subjective Cognitive Decline (SCD) and Mild Cognitive Impairment (MCI) patients carrying Alzheimer's Disease (AD) pathology from non-carriers; to propose a flowchart for the interpretation of the results of plasma p-tau181 and NfL. We included 43 SCD, 41 MCI and 21 AD-demented (AD-d) patients, who underwent plasma p-tau181 and NfL analysis. Twenty-eight SCD, 41 MCI and 21 AD-d patients underwent CSF biomarkers analysis (Aß1-42, Aß1-42/1-40, p-tau, t-tau) and were classified as carriers of AD pathology (AP+) it they were A+/T+ , or non-carriers (AP-) when they were A-, A+/T-/N-, or A+/T-/N+ according to the A/T(N) system. Plasma p-tau181 and NfL separately showed a good accuracy (AUC = 0.88), while the combined model (NfL + p-tau181) showed an excellent accuracy (AUC = 0.92) in discriminating AP+ from AP- patients. Plasma p-tau181 and NfL results were moderately concordant (Coehn's k = 0.50, p < 0.001). Based on a logistic regression model, we estimated the risk of AD pathology considering the two biomarkers: 10.91% if both p-tau181 and NfL were negative; 41.10 and 76.49% if only one biomarker was positive (respectively p-tau18 and NfL); 94.88% if both p-tau181 and NfL were positive. Considering the moderate concordance and the risk of presenting an underlying AD pathology according to the positivity of plasma p-tau181 and NfL, we proposed a flow chart to guide the combined use of plasma p-tau181 and NfL and the interpretation of biomarker results to detect AD pathology.


Alzheimer Disease , Biomarkers , Cognitive Dysfunction , Neurofilament Proteins , tau Proteins , Humans , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnosis , Male , Female , Neurofilament Proteins/blood , Aged , Biomarkers/blood , Phosphorylation , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Middle Aged , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid
...