Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.313
Filter
1.
Clin Chim Acta ; 564: 119941, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39181294

ABSTRACT

BACKGROUND: In Alzheimer's disease (AD) diagnosis, a cerebrospinal fluid (CSF) biomarker panel is commonly interpreted with binary cutoff values. However, these values are not generic and do not reflect the disease continuum. We explored the use of interval-specific likelihood ratios (LRs) and probability-based models for AD using a CSF biomarker panel. METHODS: CSF biomarker (Aß1-42, tTau and pTau181) data for both a clinical discovery cohort of 241 patients (measured with INNOTEST) and a clinical validation cohort of 129 patients (measured with EUROIMMUN), both including AD and non-AD dementia/cognitive complaints were retrospectively retrieved in a single-center study. Interval-specific LRs for AD were calculated and validated for univariate and combined (Aß1-42/tTau and pTau181) biomarkers, and a continuous bivariate probability-based model for AD, plotting Aß1-42/tTau versus pTau181 was constructed and validated. RESULTS: LR for AD increased as individual CSF biomarker values deviated from normal. Interval-specific LRs of a combined biomarker model showed that once one biomarker became abnormal, LRs increased even further when another biomarker largely deviated from normal, as replicated in the validation cohort. A bivariate probability-based model predicted AD with a validated accuracy of 88% on a continuous scale. CONCLUSIONS: Interval-specific LRs in a combined biomarker model and prediction of AD using a continuous bivariate biomarker probability-based model, offer a more meaningful interpretation of CSF AD biomarkers on a (semi-)continuous scale with respect to the post-test probability of AD across different assays and cohorts.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Probability , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Humans , Biomarkers/cerebrospinal fluid , Female , Male , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Likelihood Functions , Middle Aged , tau Proteins/cerebrospinal fluid , Retrospective Studies , Peptide Fragments/cerebrospinal fluid , Cohort Studies
2.
Croat Med J ; 65(4): 328-338, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39219196

ABSTRACT

AIM: To examine whether changes in biomarker concentrations in patients with idiopathic normal-pressure hydrocephalus (iNPH) during 72 h of external lumbar drainage (ELD) can differentiate between responders and non-responders. METHODS: Twenty patients with clinical and neuroradiological signs of iNPH underwent ELD over a period of 72 h. During this period, changes in cerebrospinal fluid (CSF) concentrations of biomarkers (amyloid-ß, total and phosphorylated tau proteins) and intracranial pressure were monitored, and the volume of drained CSF was measured. Changes in the concentrations of selected biomarkers at three time points (0, 36, and 72 h) during ELD were tested for association with changes in clinical condition. RESULTS: Ten patients showed significant clinical improvement after ELD, quantified as a difference of two or more points on the Mini-Mental State Examination and/or Japanese iNPH grading scale. The concentration of all tested biomarkers increased during the first 36 h. Respondents had higher Aß 1-42 at all time points, with a significant difference seen after 72 h. They also had a significantly higher Aß1-42/Aß1-40 ratio at all time points. CONCLUSION: A gradual increase in Aß 1-42 concentration during three-day ELD represents a possible positive prognostic factor for the placement of permanent CSF drainage in patients with iNPH.


Subject(s)
Amyloid beta-Peptides , Biomarkers , Drainage , Hydrocephalus, Normal Pressure , Humans , Hydrocephalus, Normal Pressure/cerebrospinal fluid , Hydrocephalus, Normal Pressure/diagnosis , Hydrocephalus, Normal Pressure/surgery , Male , Female , Pilot Projects , Biomarkers/cerebrospinal fluid , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Aged, 80 and over , tau Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Middle Aged , Intracranial Pressure , Spinal Puncture
3.
Alzheimers Res Ther ; 16(1): 199, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242539

ABSTRACT

BACKGROUND: There is initial evidence suggesting that biomarker neurogranin (Ng) may distinguish Alzheimer's disease (AD) from other neurodegenerative diseases. Therefore, we assessed (a) the discriminant ability of cerebrospinal fluid (CSF) Ng levels to distinguish between AD and frontotemporal lobar degeneration (FTLD) pathology and between different stages within the same disease, (b) the relationship between Ng levels and cognitive performance in both AD and FTLD pathology, and (c) whether CSF Ng levels vary by apolipoprotein E (APOE) polymorphism in the AD continuum. METHODS: Participants with subjective cognitive decline (SCD) (n = 33), amnestic mild cognitive impairment (aMCI) due to AD (n = 109), AD dementia (n = 67), MCI due to FTLD (n = 25), and FTLD dementia (n = 29) were recruited from the Czech Brain Aging Study. One-way analysis of covariance (ANCOVA) assessed Ng levels in diagnostic subgroups. Linear regressions evaluated the relationship between CSF Ng levels, memory scores, and APOE polymorphism. RESULTS: Ng levels were higher in aMCI-AD patients compared to MCI-FTLD (F[1, 134] = 15.16, p < .001), and in AD-dementia compared to FTLD-dementia (F[1, 96] = 4.60, p = .029). Additionally, Ng levels were higher in FTLD-dementia patients compared to MCI-FTLD (F[1, 54]= 4.35, p = .034), lower in SCD participants compared to aMCI-AD (F[1, 142] = 10.72, p = .001) and AD-dementia (F[1, 100] = 20.90, p < .001), and did not differ between SCD participants and MCI-FTLD (F[1, 58]= 1.02, p = .491) or FTLD-dementia (F[1, 62]= 2.27, p = .051). The main effect of diagnosis across the diagnostic subgroups on Aß1-42/Ng ratio was significant too (F[4, 263]=, p < .001). We found a non-significant association between Ng levels and memory scores overall (ß=-0.25, p = .154) or in AD diagnostic subgroups, and non-significant differences in this association between overall AD APOE ε4 carriers and non-carriers (ß=-0.32, p = .358). CONCLUSIONS: In this first study to-date to assess MCI and dementia due to AD or FTLD within one study, elevated CSF Ng appears to be an early biomarker of AD-related impairment, but its role as a biomarker appears to diminish after dementia diagnosis, whereby dementia-related underlying processes in AD and FTLD may begin to merge. The Aß1-42/Ng ratio discriminated AD from FTLD patients better than Ng alone. CSF Ng levels were not related to memory in AD or FTLD, suggesting that Ng may be a marker of the biological signs of disease state rather than cognitive deficits.


Subject(s)
Alzheimer Disease , Biomarkers , Cognitive Dysfunction , Frontotemporal Lobar Degeneration , Neurogranin , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoproteins E/genetics , Apolipoproteins E/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/diagnosis , Cross-Sectional Studies , Frontotemporal Lobar Degeneration/cerebrospinal fluid , Frontotemporal Lobar Degeneration/diagnosis , Neurogranin/cerebrospinal fluid , Neuropsychological Tests
4.
Neurology ; 103(8): e209828, 2024 Oct 22.
Article in English | MEDLINE | ID: mdl-39284112

ABSTRACT

OBJECTIVES: Recently, a subset of patients affected by cerebral amyloid angiopathy (CAA) distinguished by atypical juvenile onset and a hypothesized iatrogenic origin (iatrogenic CAA, iCAA) has emerged. ß-Amyloid (Aß) accumulation evidenced by amyloid PET positivity or CSF Aß decrease was included in the iCAA diagnostic criteria. Conversely, diagnostic criteria for sporadic CAA (sCAA) do not involve biomarker analysis. The aim of this study was to assess CSF and plasma levels of Aß and tau in iCAA and sCAA cohorts. METHODS: Patients affected by probable or possible CAA according to established criteria (Boston 2.0) were prospectively recruited at Fondazione IRCCS Carlo Besta and San Gerardo dei Tintori from May 2021 to January 2024. Patients with probable and possible iCAA or sCAA with available plasma and/or CSF samples were included. Clinical and neurologic data were collected, and levels of Aß40, Aß42, total tau, and phospho-tau (p-tau) were assessed in CSF and plasma by SiMoA and Lumipulse. RESULTS: 21 patients with iCAA (72% male, mean age at symptom onset 50 years [36-74]) and 32 patients with sCAA (44% male, mean age at symptom onset 68 years [52-80]) were identified. Cognitive impairment and cardiovascular risk factors in the sCAA cohort were more common compared with the iCAA cohort. Patients with sCAA and iCAA showed similar CSF levels for Aß40 (p = 0.5 [sCAA, 95% CI 2,604-4,228; iCAA, 95% CI 1,958-3,736]), Aß42 (p = 0.7 [sCAA, 95% CI 88-157; iCAA, 95% CI 83-155]), and total tau (p = 0.08 [sCAA, 95% CI 80-134; iCAA, 95% CI 37-99]). Plasma levels of Aß40 (p = 0.08, 95% CI 181-222), Aß42 (p = 0.3, 95% CI 6-8), and total tau (p = 0.4, 95% CI 3-6) were not statistically different in patients with sCAA compared with iCAA ones (Aß40, 95% CI 153-193; Aß42, 95% CI 6-7 and total tau, 95% CI 2-4). DISCUSSION: Despite presenting with a younger age at onset, fewer cardiovascular risk factors, and lower cognitive impairment, patients with iCAA demonstrated Aß and tau levels comparable with elderly patients with sCAA, supporting a common molecular paradigm between the 2 CAA forms.


Subject(s)
Amyloid beta-Peptides , Biomarkers , Cerebral Amyloid Angiopathy , Iatrogenic Disease , Peptide Fragments , tau Proteins , Humans , Male , Female , Cerebral Amyloid Angiopathy/blood , Cerebral Amyloid Angiopathy/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Biomarkers/blood , Biomarkers/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , tau Proteins/blood , Aged , Middle Aged , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Prospective Studies , Aged, 80 and over
5.
Nat Commun ; 15(1): 7998, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266542

ABSTRACT

Impaired clearance of amyloid ß (Aß) in late-onset Alzheimer's disease (AD) affects disease progression. The role of peripheral monocytes in Aß clearance from the central nervous system (CNS) is unclear. We use a flow cytometry assay to identify Aß-binding monocytes in blood, validated by confocal microscopy, Western blotting, and mass spectrometry. Flow cytometry immunophenotyping and correlation with AD biomarkers are studied in 150 participants from the AIBL study. We also examine monocytes in human cerebrospinal fluid (CSF) and their migration in an APP/PS1 mouse model. The assay reveals macrophage-like Aß-binding monocytes with high phagocytic potential in both the periphery and CNS. We find lower surface Aß levels in mild cognitive impairment (MCI) and AD-dementia patients compared to cognitively unimpaired individuals. Monocyte infiltration from blood to CSF and migration from CNS to peripheral lymph nodes and blood are observed. Here we show that Aß-binding monocytes may play a role in CNS Aß clearance, suggesting their potential as a biomarker for AD diagnosis and monitoring.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cognitive Dysfunction , Disease Progression , Mice, Transgenic , Monocytes , Alzheimer Disease/metabolism , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Alzheimer Disease/blood , Humans , Monocytes/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Female , Aged , Male , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/cerebrospinal fluid , Mice , Aged, 80 and over , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Biomarkers/metabolism , Flow Cytometry , Disease Models, Animal , Phagocytosis , Middle Aged
6.
Neurology ; 103(7): e209801, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39288341

ABSTRACT

BACKGROUND AND OBJECTIVES: Vascular risk factors (VRFs) and cerebral small vessel disease (cSVD) are common in patients with Alzheimer disease (AD). It remains unclear whether this coexistence reflects shared risk factors or a mechanistic relationship and whether vascular and amyloid pathologies have independent or synergistic influence on subsequent AD pathophysiology in preclinical stages. We investigated links between VRFs, cSVD, and amyloid levels (Aß1-42) and their combined effect on downstream AD biomarkers, that is, CSF hyperphosphorylated tau (P-tau181), atrophy, and cognition. METHODS: This retrospective study included nondemented participants (Clinical Dementia Rating < 1) from the European Prevention of Alzheimer's Dementia (EPAD) cohort and assessed VRFs with the Framingham risk score (FRS) and cSVD features on MRI using visual scales and white matter hyperintensity volumes. After preliminary linear analysis, we used structural equation modeling (SEM) to create a "cSVD severity" latent variable and assess the direct and indirect effects of FRS and cSVD severity on Aß1-42, P-tau181, gray matter volume (baseline and longitudinal), and cognitive performance (baseline and longitudinal). RESULTS: A total cohort of 1,592 participants were evaluated (mean age = 65.5 ± 7.4 years; 56.16% F). We observed positive associations between FRS and all cSVD features (all p < 0.05) and a negative association between FRS and Aß1-42 (ß = -0.04 ± 0.01). All cSVD features were negatively associated with CSF Aß1-42 (all p < 0.05). Using SEM, the cSVD severity fully mediated the association between FRS and CSF Aß1-42 (indirect effect: ß = -0.03 ± 0.01), also when omitting vascular amyloid-related markers. We observed a significant indirect effect of cSVD severity on P-tau181 (indirect effect: ß = 0.12 ± 0.03), baseline and longitudinal gray matter volume (indirect effect: ß = -0.10 ± 0.03; ß = -0.12 ± 0.05), and baseline cognitive performance (indirect effect: ß = -0.16 ± 0.03) through CSF Aß1-42. DISCUSSION: In a large nondemented population, our findings suggest that cSVD is a mediator of the relationship between VRFs and CSF Aß1-42 and affects downstream neurodegeneration and cognitive impairment. We provide evidence of VRFs indirectly affecting the pathogenesis of AD, highlighting the importance of considering cSVD burden in memory clinics for AD risk evaluation and as an early window for intervention. These results stress the role of VRFs and cerebrovascular pathology as key biomarkers for accurate design of anti-amyloid clinical trials and offer new perspectives for patient stratification.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cerebral Small Vessel Diseases , Peptide Fragments , tau Proteins , Humans , Alzheimer Disease/pathology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Male , Female , Aged , Risk Factors , Amyloid beta-Peptides/cerebrospinal fluid , Retrospective Studies , Cerebral Small Vessel Diseases/diagnostic imaging , Cerebral Small Vessel Diseases/complications , Cerebral Small Vessel Diseases/pathology , tau Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Middle Aged , Magnetic Resonance Imaging , Biomarkers/cerebrospinal fluid , Brain/pathology , Brain/diagnostic imaging , Atrophy/pathology
7.
Alzheimers Res Ther ; 16(1): 207, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39322953

ABSTRACT

BACKGROUND: Several blood-based biomarkers offer the opportunity of in vivo detection of brain pathology and neurodegeneration in Alzheimer disease with high specificity and sensitivity, but the performance of amyloid-ß (Aß) measurements remains under evaluation. Autosomal dominant Alzheimer disease (ADAD) with mutations in PSEN1, PSEN2 and APP can be studied as a model for sporadic Alzheimer disease. However, clarifying the genetic effects on the Aß-levels in different matrices such as cerebrospinal fluid or plasma is crucial for generalizability and utility of data. We aimed to explore plasma Aß concentrations over the Alzheimer disease continuum in a longitudinal cohort of genetic Alzheimer disease. METHODS: 92 plasma samples were collected from at-risk individuals (n = 47) in a Swedish cohort of ADAD, including 18 mutation carriers (13 APPswe (p.KM670/671NL) MC), 5 PSEN1 (p.H163Y) MC) and 29 non-carriers (NC) as the reference group. Concentrations of Aß1-38, Aß1-40 and Aß1-42 were analyzed in plasma using immunoprecipitation coupled to tandem liquid chromatography mass spectrometry (IP-LC-MS/MS). Cross-sectional and repeated-measures data analyses were investigated family-wise, applying non-parametric tests as well as mixed-effects models. RESULTS: Cross-sectional analysis at baseline showed more than a 3-fold increase in all plasma Aß peptides in APPswe MC, regardless of clinical status, compared to controls (p < 0.01). PSEN1 (p.H163Y) presymptomatic MC had a decrease of plasma Aß1-38 compared to controls (p < 0.05). There was no difference in Aß1-42/1-40 ratio between APPswe MC (PMC and SMC), PSEN1 MC (PMC) and controls at baseline. Notably, both cross-sectional data and repeated-measures analysis suggested that APPswe MC have a stable Aß1-42/1-40 ratio with increasing age, in contrast to the decrease seen with aging in both controls and PSEN1 (p.H163Y) MC. CONCLUSION: These data show very strong mutation-specific effects on Aß profiles in blood, most likely due to a ubiquitous production outside of the CNS. Hence, analyses in an unselected clinical setting might unintentionally disclose genetic status. Furthermore, our findings suggest that the Aß ratio might be a poor indicator of brain Aß pathology in selected genetic cases. The very small sample size is a limitation that needs to be considered but reflects the scarcity of longitudinal in vivo data from genetic cohorts.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Presenilin-1 , Presenilin-2 , Humans , Alzheimer Disease/blood , Alzheimer Disease/genetics , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Male , Female , Sweden , Middle Aged , Presenilin-1/genetics , Presenilin-2/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/blood , Aged , Mutation , Adult , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Longitudinal Studies , Cohort Studies , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/genetics
8.
Neurology ; 103(7): e209795, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39270153

ABSTRACT

BACKGROUND AND OBJECTIVES: There is a clear need to characterize and validate molecular biomarkers of cerebral amyloid angiopathy (CAA), in an effort to improve diagnostics, especially in the context of patients with Alzheimer disease (AD) receiving immunotherapies (for whom underlying CAA is the driver of amyloid-related imaging abnormalities). We performed an updated meta-analysis of 5 core CSF biomarkers (Aß42, Aß40, Aß438, total tau [T-tau], and phosphorylated tau [P-tau]) to assess which of these are most altered in sporadic CAA. METHODS: We systematically searched PubMed for eligible studies reporting data on CSF biomarkers reflecting APP metabolism (Aß42, Aß40, Aß38), neurodegeneration (T-tau), and tangle pathology (P-tau), in symptomatic sporadic CAA cohorts (based on the Boston criteria) vs control groups and/or vs patients with AD. Biomarker performance was assessed in random-effects meta-analysis based on ratio of mean (RoM) biomarker concentrations in (1) patients with CAA to controls and (2) CAA to patients with AD. RoM >1 indicates higher biomarker concentration in CAA vs comparison population, and RoM <1 indicates higher concentration in comparison groups. RESULTS: 8 studies met inclusion criteria: a total of 11 CAA cohorts (n = 289), 9 control cohorts (n = 310), and 8 AD cohorts (n = 339). Overall included studies were of medium quality based on our assessment tools. CAA to controls had lower mean level of all amyloid markers with CSF Aß42, Aß40, and Aß38 RoMs of 0.46 (95% CI 0.38-0.55, p < 0.0001), 0.70 (95% CI 0.63-0.78, p < 0.0001), and 0.71 (95% CI 0.56-0.89, p = 0.003), respectively. CSF T-tau and P-tau RoMs of patients with CAA to controls were both greater than 1: 1.56 (95% CI 1.32-1.84, p < 0.0001) and 1.31 (95% CI 1.13-1.51, p < 0.0001), respectively. Differentiation between CAA and AD was strong for CSF Aß40 (RoM 0.76, 95% CI 0.69-0.83, p < 0.0001) and Aß38 (RoM 0.55, 95% CI 0.38-0.81, p < 0.0001), but not Aß42 (RoM 1.00; 95% CI 0.81-1.23, p = 0.970). For T-tau and P-tau, average CSF ratios in patients with CAA vs AD were 0.64 (95% CI 0.58-0.71, p < 0.0001) and 0.64 (95% CI 0.58-0.71, p < 0.0001), respectively. DISCUSSION: Specific CSF patterns of Aß42, Aß40, Aß38, T-tau, and P-tau might serve as molecular biomarkers of CAA, in research and clinical settings, offering the potential to improve the clinical diagnostic approach pathway in specific scenarios.


Subject(s)
Amyloid beta-Peptides , Biomarkers , Cerebral Amyloid Angiopathy , tau Proteins , Humans , Cerebral Amyloid Angiopathy/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis
9.
Talanta ; 280: 126699, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39142131

ABSTRACT

Accurate and sensitive detection of amyloid-ß 42 oligomer (Aß42O) is of great significance for early diagnosis of Alzheimer's disease (AD). Herein, a signal on-off ratiometric electrochemical immunosensor was developed for highly selective and quantitative determination of Aß42O by using novel covalent organic frameworks (COFs) composites as the sensing platform. This immunosensor produced two independent electrochemical signals from the [Fe(CN)6]3-/4- and methylene blue (MB) probes at different potentials based on the electrocatalytic activity of gold nanoparticle-functionalized porphyrinyl COFs nanocomposites toward [Fe(CN)6]3-/4- and the signal probe of MB encapsulated in the aptamer-modified alkynyl COFs. Because the two signals of [Fe(CN)6]3-/4- and MB changed in opposite directions, a signal on-off mode was generated which can correct the results by introducing a reference signal and effectively eliminate background interference. Under optimal experimental conditions, the current ratio (IMB/I[Fe(CN)6]3-/4-) was well linearly related to the logarithmic value of Aß42O concentrations in the range of 10 pM to 1 µM, and the detection limit was 5.1 pM (S/N = 3). Additionally, the immunosensor exhibited satisfactory performance in case of real cerebrospinal fluid samples. The designed ratiometric electrochemical immunosensor provides a valuable route for early diagnosis of AD and our results also pave the way for designing of sensing platforms using COF-based nanomaterials and extending their functions and applications to bioanalysis.


Subject(s)
Amyloid beta-Peptides , Electrochemical Techniques , Metal-Organic Frameworks , Peptide Fragments , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Electrochemical Techniques/methods , Metal-Organic Frameworks/chemistry , Humans , Peptide Fragments/analysis , Peptide Fragments/chemistry , Limit of Detection , Biosensing Techniques/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Methylene Blue/chemistry , Immunoassay/methods , Alzheimer Disease/diagnosis
10.
Alzheimers Res Ther ; 16(1): 179, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127661

ABSTRACT

BACKGROUND: Microglial activation has been suggested to be involved in the pathogenesis of depression and Alzheimer's disease (AD). Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) is a marker of microglial activation. The purpose of this study was to investigate the interrelationships of cerebrospinal fluid (CSF) sTREM2, AD pathology, as well as minimal depressive symptoms (MDSs), and cognition. METHODS: A total of 545 non-demented individuals from the Alzheimer's Disease Neuroimaging Initiative cohort were included in our study. The average age of the total population was 72.6 years and the percentage of females was 42.6%. Linear regression models were conducted to investigate the linear relationships of MDSs with CSF sTREM2, AD pathology, cognition, and brain structure. Mediation models and structural equation models (SEM) were conducted to examine whether CSF sTREM2 mediated the relationships of MDSs with AD pathology and cognition. RESULTS: Results revealed that individuals with MDSs had lower CSF sTREM2 levels than normal controls. Linear regression showed that MDSs were linearly associated with CSF sTREM2 (PFDR = 0.012) and amyloid biomarkers (PFDR < 0.05), as well as cognitive scores (PFDR < 0.05) and hippocampal volume (PFDR = 0.003). Mediation analyses revealed that CSF sTREM2 mediated the association between MDSs and amyloid pathology, with the mediating proportions ranging from 6.030 to 18.894%. However, SEM failed to reveal that MDS affected cognition through CSF amyloid pathology and CSF sTREM2. CONCLUSIONS: MDSs are associated with amyloid pathology and cognition. CSF sTREM2 may potentially be an intervenable target between depression and AD pathology.


Subject(s)
Alzheimer Disease , Depression , Membrane Glycoproteins , Receptors, Immunologic , Humans , Female , Male , Membrane Glycoproteins/cerebrospinal fluid , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Depression/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognition/physiology , Aged, 80 and over , Cohort Studies , Brain/pathology , Brain/diagnostic imaging , Amyloid beta-Peptides/cerebrospinal fluid , Middle Aged
11.
Alzheimers Res Ther ; 16(1): 186, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160628

ABSTRACT

BACKGROUND: Plasma p-tau217 has emerged as the most promising blood-based marker (BBM) for the detection of Alzheimer Disease (AD) pathology, yet few studies have evaluated plasma p-tau217 performance in memory clinic settings. We examined the performance of plasma p-tau217 for the detection of AD using a high-sensitivity immunoassay in individuals undergoing diagnostic lumbar puncture (LP). METHODS: Paired plasma and cerebrospinal fluid (CSF) samples were analysed from the TIMC-BRAiN cohort. Amyloid (Aß) and Tau (T) pathology were classified based on established cut-offs for CSF Aß42 and CSF p-tau181 respectively. High-sensitivity electrochemiluminescence (ECL) immunoassays were performed on paired plasma/CSF samples for p-tau217, p-tau181, Glial Fibrillary Acidic Protein (GFAP), Neurofilament Light (NfL) and total tau (t-tau). Biomarker performance was evaluated using Receiver-Operating Curve (ROC) and Area-Under-the-Curve (AUC) analysis. RESULTS: Of 108 participants (age: 69 ± 6.5 years; 54.6% female) with paired samples obtained at time of LP, 64.8% (n = 70/108) had Aß pathology detected (35 with Mild Cognitive Impairment and 35 with mild dementia). Plasma p-tau217 was over three-fold higher in Aß + (12.4 pg/mL; 7.3-19.2 pg/mL) vs. Aß- participants (3.7 pg/mL; 2.8-4.1 pg/mL; Mann-Whitney U = 230, p < 0.001). Plasma p-tau217 exhibited excellent performance for the detection of Aß pathology (AUC: 0.91; 95% Confidence Interval [95% CI]: 0.86-0.97)-greater than for T pathology (AUC: 0.83; 95% CI: 0.75-0.90; z = 1.75, p = 0.04). Plasma p-tau217 outperformed plasma p-tau181 for the detection of Aß pathology (z = 3.24, p < 0.001). Of the other BBMs, only plasma GFAP significantly differed by Aß status which significantly correlated with plasma p-tau217 in Aß + (but not in Aß-) individuals. Application of a two-point threshold at 95% and 97.5% sensitivities & specificities may have enabled avoidance of LP in 58-68% of cases. CONCLUSIONS: Plasma p-tau217 measured using a high-sensitivity ECL immunoassay demonstrated excellent performance for detection of Aß pathology in a real-world memory clinic cohort. Moving forward, clinical use of plasma p-tau217 to detect AD pathology may substantially reduce need for confirmatory diagnostic testing for AD pathology with diagnostic LP in specialist memory services.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , tau Proteins , Humans , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Female , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Male , Aged , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Alzheimer Disease/cerebrospinal fluid , Immunoassay/methods , Middle Aged , Cohort Studies , Luminescent Measurements/methods
12.
Commun Biol ; 7(1): 1037, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179782

ABSTRACT

Alterations in functional connectivity (FC) have been observed in individuals with Alzheimer's disease (AD) with elevated amyloid (Aß) and tau. However, it is not yet known whether directed FC is already influenced by Aß and tau load in cognitively healthy (CH) individuals. A 21-channel electroencephalogram (EEG) was used from 46 CHs classified based on cerebrospinal fluid (CSF) Aß tau ratio: pathological (CH-PAT) or normal (CH-NAT). Directed FC was estimated with Partial Directed Coherence in frontal, temporal, parietal, central, and occipital regions. We also examined the correlations between directed FC and various functional metrics, including neuropsychology, cognitive reserve, MRI volumetrics, and heart rate variability between both groups. Compared to CH-NATs, the CH-PATs showed decreased FC from the temporal regions, indicating a loss of relative functional importance of the temporal regions. In addition, frontal regions showed enhanced FC in the CH-PATs compared to CH-NATs, suggesting neural compensation for the damage caused by the pathology. Moreover, CH-PATs showed greater FC in the frontal and occipital regions than CH-NATs. Our findings provide a useful and non-invasive method for EEG-based analysis to identify alterations in brain connectivity in CHs with a pathological versus normal CSF Aß/tau.


Subject(s)
Amyloid beta-Peptides , Brain , Electroencephalography , tau Proteins , Humans , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism , Female , Male , Brain/diagnostic imaging , Brain/physiopathology , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Aged , Cognition , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/physiopathology , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Magnetic Resonance Imaging , Middle Aged
13.
Alzheimers Res Ther ; 16(1): 192, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187891

ABSTRACT

BACKGROUND: Protein biomarkers have been broadly investigated in cerebrospinal fluid and blood for the detection of neurodegenerative diseases, yet a clinically useful diagnostic test to detect early, pre-symptomatic Alzheimer's disease (AD) remains elusive. We conducted this study to quantify Aß40, Aß42, total Tau (t-Tau), hyperphosphorylated Tau (ptau181), glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) in eye fluids relative to blood. METHODS: In this cross-sectional study we collected vitreous humor, aqueous humor, tear fluid and plasma in patients undergoing surgery for eye disease. All six biomarkers were quantitatively measured by digital immunoassay. Spearman and Bland-Altman correlation analyses were performed to assess the agreement of levels between ocular fluids and plasma. RESULTS: Seventy-nine adults underwent pars-plana vitrectomy in at least one eye. Of the 79, there were 77 vitreous, 67 blood, 56 tear fluid, and 51 aqueous samples. All six biomarkers were quantified in each bio-sample, except GFAP and NfL in tear fluid due to low sample volume. All six biomarkers were elevated in vitreous humor compared to plasma samples. T-Tau, ptau181, GFAP and NfL were higher in aqueous than in plasma, and t-Tau and ptau181 concentrations were higher in tear fluid than in plasma. Significant correlations were found between Aß40 in plasma and tears (r = 0.5; p = 0.019), t-Tau in plasma and vitreous (r = 0.4; p = 0.004), NfL in plasma and vitreous (r = 0.3; p = 0.006) and plasma and aqueous (r = 0.5; p = 0.004). No significant associations were found for Aß42, ptau181 and GFAP among ocular fluids relative to plasma. Bland-Altman analysis showed aqueous humor had the closest agreement to plasma across all biomarkers. Biomarker levels in ocular fluids revealed statistically significant associations between vitreous and aqueous for t-Tau (r = 0.5; p = 0.001), GFAP (r = 0.6; p < 0.001) and NfL (r = 0.7; p < 0.001). CONCLUSION: AD biomarkers are detectable in greater quantities in eye fluids than in plasma and show correlations with levels in plasma. Future studies are needed to assess the utility of ocular fluid biomarkers as diagnostic and prognostic markers for AD, especially in those at risk with eye disease.


Subject(s)
Amyloid beta-Peptides , Aqueous Humor , Biomarkers , Glial Fibrillary Acidic Protein , Neurofilament Proteins , Tears , Vitreous Body , tau Proteins , Humans , Female , Male , Biomarkers/blood , tau Proteins/blood , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism , Aged , Cross-Sectional Studies , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Middle Aged , Aqueous Humor/metabolism , Aqueous Humor/chemistry , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Tears/chemistry , Tears/metabolism , Vitreous Body/metabolism , Glial Fibrillary Acidic Protein/blood , Glial Fibrillary Acidic Protein/metabolism , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Aged, 80 and over , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/metabolism , Adult
14.
Alzheimers Res Ther ; 16(1): 182, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135067

ABSTRACT

BACKGROUND: Precisely defining the delay in onset of dementia is a particular challenge for early diagnosis. Brain [18F] fluoro-2-deoxy-2-D-glucose (18F-FDG) Positron Emission Tomography (PET) is a particularly interesting tool for the early diagnosis of neurodegenerative diseases, through the measurement of the cerebral glucose metabolic rate. There is currently a lack of longitudinal studies under real-life conditions, with sufficient patients, to accurately evaluate the predictive values of brain 18F-FDG PET scans. Here, we aimed to estimate the value of brain 18F-FDG PET for predicting the risk of dementia conversion and the risk of occurrence of a neurodegenerative pathology. METHODS: Longitudinal data for a cohort of patients with no diagnosis of dementia at the time of recruitment referred by a tertiary memory clinic for brain 18F-FDG PET were matched with (Prince M, Wimo A, Guerchet Maëlenn, Ali G-C, Wu Y-T et al. World Alzheimer Report 2015. The Global Impact of Dementia: An analysis of prevalence, incidence, cost and trends. [Research Report] Alzheimer's Disease International. 2015. 2015.) data from the French National Health Data System (NHDS), (Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14(4):535-62.) data from the National Alzheimer Bank (NAB), and (Davis M, O`Connell T, Johnson S, Cline S, Merikle E, Martenyi F, et al. Estimating Alzheimer's Disease Progression Rates from Normal Cognition Through Mild Cognitive Impairment and Stages of Dementia. CAR. 2018;15(8):777-88.) lumbar puncture (LP) biomarker data. The criteria for dementia conversion were the designation, within the three years after the brain 18F-FDG PET scan, of a long-term condition for dementia in the NHDS and a dementia stage of cognitive impairment in the NAB. The criterion for the identification of a neurodegenerative disease in the medical records was the determination of LP biomarker levels. RESULTS: Among the 403 patients (69.9 ± 11.4 years old, 177 women) from the initial cohort with data matched with the NHDS data, 137 were matched with the NAB data, and 61 were matched with LP biomarker data. Within three years of the scan, a 18F-FDG PET had negative predictive values of 85% for dementia conversion (according to the NHDS and NAB datasets) and 95% for the presence of LP neurodegeneration biomarkers. CONCLUSION: A normal brain 18F-FDG PET scan can help rule out the risk of dementia conversion and the presence of cerebrospinal fluid (CSF) biomarker of neurodegeneration early with high certainty, allowing modifications to patient management regimens in the short term. TRIAL REGISTRATION: Clinical Trials database (NCT04804722). March 18, 2021. Retrospectively registered.


Subject(s)
Biomarkers , Dementia , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Humans , Positron-Emission Tomography/methods , Female , Male , Aged , Dementia/diagnostic imaging , Dementia/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Longitudinal Studies , Brain/diagnostic imaging , Brain/metabolism , Middle Aged , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Disease Progression , Aged, 80 and over , Early Diagnosis , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Cohort Studies
15.
J Alzheimers Dis ; 100(4): 1333-1343, 2024.
Article in English | MEDLINE | ID: mdl-39093070

ABSTRACT

Background: The relationship between Alzheimer's disease (AD)-related pathology and cognition was not exactly consistent. Objective: To explore whether the association between AD pathology and cognition can be moderated by frailty. Methods: We included 1711 participants from the Alzheimer's Disease Neuroimaging Initiative database. Levels of cerebrospinal fluid amyloid-ß, p-tau, and t-tau were identified for AD-related pathology based on the amyloid-ß/tau/neurodegeneration (AT[N]) framework. Frailty was measured using a modified Frailty Index-11 (mFI-11). Regression and interaction models were utilized to assess the relationship among frailty, AT(N) profiles, and cognition. Moderation models analyzed the correlation between AT(N) profiles and cognition across three frailty levels. All analyses were corrected for age, sex, education, and APOEɛ4 status. Results: In this study, frailty (odds ratio [OR] = 1.71, p < 0.001) and AT(N) profiles (OR = 2.00, p < 0.001) were independently associated with cognitive status. The model fit was improved when frailty was added to the model examining the relationship between AT(N) profiles and cognition (p < 0.001). There was a significant interaction between frailty and AT(N) profiles in relation to cognitive status (OR = 1.12, pinteraction = 0.028). Comparable results were obtained when Mini-Mental State Examination scores were utilized as the measure of cognitive performance. The association between AT(N) profiles and cognition was stronger with the levels of frailty. Conclusions: Frailty may diminish patients' resilience to AD pathology and accelerate cognitive decline resulting from abnormal AD-related pathology. In summary, frailty contributes to elucidating the relationship between AD-related pathology and cognitive impairment.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cognition , Frailty , tau Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/psychology , Alzheimer Disease/complications , Male , Female , Aged , Frailty/complications , Frailty/psychology , Amyloid beta-Peptides/cerebrospinal fluid , Cognition/physiology , tau Proteins/cerebrospinal fluid , Aged, 80 and over , Neuropsychological Tests , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/psychology
17.
J Alzheimers Dis ; 100(s1): S103-S114, 2024.
Article in English | MEDLINE | ID: mdl-39121126

ABSTRACT

Background: Synaptic dysfunction is closely associated with cognitive function in Alzheimer's disease (AD), and is present already in an early stage of the disease. Objective: Using serial cerebrospinal fluid (CSF) sampling, we aimed to investigate slopes of CSF synaptic proteins, and their relation with cognition along the AD continuum. Methods: We included subjects with subjective cognitive decline (SCD) or mild cognitive impairment (MCI) (n = 50 amyloid-ß+ [A +], n = 50 A-) and 50 patients with AD dementia from the Amsterdam dementia cohort, with CSF at two time points (median[IQR] 2.1[1.4-2.7] years). We analyzed 17 synaptic proteins and neurofilament light (NfL). Using linear mixed models we assessed trajectories of protein levels, and associations with cognitive decline (repeated Mini-Mental State Examination). We used Cox regression models to assess predictive value of protein levels for progression to AD dementia. Results: At baseline most proteins showed increased levels in AD dementia compared to the other groups. In contrast NPTX2 levels were lower in AD dementia. Higher baseline levels of SNAP25, ß-syn, and 14-3-3 proteins were associated with faster cognitive decline (St.B[SE] -0.27[0.12] to -0.61[0.12]). Longitudinal analyses showed that SYT1 and NPTX levels decreased over time in AD dementia (st.B[SE] -0.10[0.04] to -0.15[0.05]) and SCD/MCI-A+ (St.B[SE] -0.07[0.03] to -0.12[0.03]), but not in SCD/MCI-A- (pinteraction < 0.05). Increase over time in NfL levels was associated with faster cognitive decline in AD dementia (St.B[SE] -1.75[0.58]), but not in the other groups (pinteraction < 0.05). Conclusions: CSF synaptic proteins showed different slopes over time, suggesting complex synaptic dynamics. High levels of especially SNAP-25 may have value for prediction of cognitive decline in early AD stages, while increase in NfL over time correlates better with cognitive decline in later stages.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Cognitive Dysfunction , Disease Progression , Neurofilament Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Male , Female , Aged , Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Neurofilament Proteins/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Middle Aged , Nerve Tissue Proteins/cerebrospinal fluid , Synapses , Synaptosomal-Associated Protein 25/cerebrospinal fluid , Cohort Studies , C-Reactive Protein
18.
J Alzheimers Dis ; 100(s1): S93-S101, 2024.
Article in English | MEDLINE | ID: mdl-39121127

ABSTRACT

Background: The N-glycan structure bisecting N-acetylglucosamine (bisecting GlcNAc) is present on several N-glycans that are elevated in Alzheimer's disease (AD), and previous studies have shown that bisecting GlcNAc levels correlate with total tau and phospho-tau181 in cerebrospinal fluid at early stages of AD. A recent population-based study showed that bisecting GlcNAc correlates with total tau also in blood and that this correlation could predict conversion to dementia. Objective: In this study, we have further investigated how bisecting GlcNAc relates to total tau and phospho-tau 181 in cerebrospinal fluid samples from controls and cases with early cognitive deficits, stratified by amyloid/tau status and gender. Methods: Relative levels of bisecting GlcNAc in cerebrospinal fluid were measured by an enzyme-linked lectin assay in individuals with subjective cognitive decline, mild cognitive impairment and controls from the Norwegian Dementia Disease Initiation cohort. Results: As in our previous study, the correlation between bisecting GlcNAc and total tau or phospho-tau181 was particularly strong in the subjective cognitive decline group. The correlation was observed in amyloid negative and tau negative as well as amyloid positive and tau positive individuals, both in females and in males. Interestingly, among the amyloid negative and tau negative individuals, the correlation was observed in individuals with subjective cognitive decline but not in the controls. Conclusions: Thus, bisecting GlcNAc could be a biomarker for early cognitive decline.


Subject(s)
Acetylglucosamine , Cognitive Dysfunction , tau Proteins , Humans , Male , Female , Acetylglucosamine/metabolism , tau Proteins/cerebrospinal fluid , Aged , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/blood , Middle Aged , Phosphorylation , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Aged, 80 and over , Amyloid beta-Peptides/cerebrospinal fluid
19.
Neurology ; 103(6): e209818, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39208367

ABSTRACT

BACKGROUND AND OBJECTIVES: There is no disease-modifying treatment of corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP), 2 disorders characterized by their striking phenotypic, and, in CBS, pathologic heterogeneity. Seed amplification assays (SAAs) could enable the detection of neuropathologic processes, such as α-synuclein (αSyn) copathology, that affect the success of future disease-modifying treatment strategies. The primary objective was to assess possible αSyn copathology in CBS and PSP, as detected in CSF using an αSyn SAA (αSyn-SAA). Secondary objectives were to evaluate the association of αSyn-SAA positivity with other biomarkers including of Alzheimer disease (AD), and with clinical presentation. We hypothesized that αSyn-SAA positivity would be detectable in CBS and PSP and that it would be associated with AD biomarker positivity and ß-amyloid (Aß) 42 levels, neurodegeneration as assessed by neurofilament light chain (NfL) levels, and symptoms associated with synucleinopathies. METHODS: This cross-sectional observational study included patients clinically diagnosed with CBS and PSP who underwent a lumbar puncture between 2012 and 2021 (Toronto Western Hospital, Canada). CSF was tested for αSyn-SAA positivity, AD biomarkers, and NfL levels. Clinical data were derived from medical records. RESULTS: We tested the CSF of 40 patients with CBS (19 female patients, 65.9 ± 8.6 years) and 28 with PSP (13 female patients, 72.5 ± 8.7 years old), mostly White (n = 50) or Asian (n = 14). αSyn-SAA positivity was observed in 35.9% patients with CBS and 28.6% with PSP. In young-onset, but not late-onset patients, αSyn-SAA positivity and AD positivity were associated (odds ratio [OR] 8.8, 95% CI 1.2-82.6, p < 0.05). A multivariable linear regression analysis showed a significant interaction of αSyn-SAA status by age at onset on CSF Aß42 levels (ß = 0.3 ± 0.1, p < 0.05). Indeed, age at onset was positively related to Aß42 levels only in αSyn-SAA-positive patients, as shown by slope comparison. A logistic regression analysis also suggested that REM sleep behavior disorder was associated with αSyn-SAA positivity (OR 60.2, 95% CI 5.2-1,965.8; p < 0.01). DISCUSSION: We detected a frequency of αSyn-SAA positivity in CBS and PSP in line with pathologic studies, highlighting the usefulness of SAAs for in vivo detection of otherwise undetectable neuropathologic processes. Our results also suggest that AD status (specifically low Aß42) and older age at onset may contribute to αSyn-SAA positivity. This opens new perspectives for the stratification of patients in clinical trials.


Subject(s)
Amyloid beta-Peptides , Biomarkers , alpha-Synuclein , Humans , alpha-Synuclein/cerebrospinal fluid , Female , Male , Aged , Middle Aged , Biomarkers/cerebrospinal fluid , Cross-Sectional Studies , Amyloid beta-Peptides/cerebrospinal fluid , Parkinsonian Disorders/cerebrospinal fluid , Parkinsonian Disorders/diagnosis , Supranuclear Palsy, Progressive/cerebrospinal fluid , Supranuclear Palsy, Progressive/diagnosis , Peptide Fragments/cerebrospinal fluid , Neurofilament Proteins/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Aged, 80 and over
20.
J Alzheimers Dis ; 100(s1): S223-S241, 2024.
Article in English | MEDLINE | ID: mdl-39177605

ABSTRACT

Background: While drainage/removal of fluid and toxins from the brain by cerebrospinal fluid (CSF) directly into venous blood is well-known, a second drainage route has recently been (re)discovered-meningeal lymphatic vessels (mLVs)-which are responsible for up to half of total brain fluid/toxin drainage. The cytokine vascular endothelial growth factor (VEGF) increases mLV diameter and numbers to increase mLV drainage, resulting in increased mLV drainage. Alzheimer's disease (AD) is characterized by low plasma and CSF levels of VEGF. Objective: To determine if non-invasive transcranial radiofrequency wave treatment (TRFT), through modulation of VEGF levels in blood and CSF, can affect removal of toxins tau and amyloid-ß (Aß) from the brain. Methods: Eight mild/moderate AD subjects were given twice-daily 1-hour TRFT sessions at home by their caregivers. Blood and CSF samples were taken at baseline and following completion of 2 months of TRFT. Results: In plasma and/or CSF, strong baseline correlations between VEGF levels and AD markers (t-tau, p-tau, Aß1-40, Aß1-42) were eliminated by TRFT. This effect was primarily due to TRFT-induced increases in VEGF levels in AD subjects with low or unmeasurable "baseline" VEGF levels. These increased VEGF levels were associated with increased clearance/drainage of tau and Aß from the brain, likely through VEGF's actions on mLVs. Conclusions: A new mechanism of TRFT is identified (facilitation of brain tau and Aß clearance via VEGF) that is likely contributory to TRFT's reversal of cognitive impairment in AD subjects. TRFT may be particularly effective for cognitive benefit in AD subjects who have low VEGF levels.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Vascular Endothelial Growth Factor A , tau Proteins , Humans , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/cerebrospinal fluid , Male , Female , Aged , Brain/metabolism , Aged, 80 and over , Middle Aged , Peptide Fragments/cerebrospinal fluid , Radio Waves
SELECTION OF CITATIONS
SEARCH DETAIL