7 research outputs found

    Cerebrospinal Fluid Inflammatory Biomarkers Reflect Clinical Severity in Huntington’s Disease

    Get PDF
    <div><p>Introduction</p><p>Immune system activation is involved in Huntington’s disease (HD) pathogenesis and biomarkers for this process could be relevant to study the disease and characterise the therapeutic response to specific interventions. We aimed to study inflammatory cytokines and microglial markers in the CSF of HD patients.</p><p>Methods</p><p>CSF TNF-α, IL-1β, IL-6, IL-8, YKL-40, chitotriosidase, total tau and neurofilament light chain (NFL) from 23 mutation carriers and 14 healthy controls were assayed.</p><p>Results</p><p>CSF TNF-α and IL-1β were below the limit of detection. Mutation carriers had higher YKL-40 (p = 0.003), chitotriosidase (p = 0.015) and IL-6 (p = 0.041) than controls. YKL-40 significantly correlated with disease stage (p = 0.007), UHDRS total functional capacity score (r = -0.46, p = 0.016), and UHDRS total motor score (r = 0.59, p = 4.5*10<sup>−4</sup>) after adjustment for age.</p><p>Conclusion</p><p>YKL-40 levels in CSF may, after further study, come to have a role as biomarkers for some aspects of HD. Further investigation is needed to support our exploratory findings.</p></div

    Relationship between CSF concentration of YKL-40 and measures of disease progression and clinical severity.

    No full text
    <p>a, disease stage; b, UHDRS total functional score; c, UHDRS total motor score. The p-values for partial Pearson’s correlation coefficient adjusted for age are shown.</p

    Additional file 2 of A proteomics analysis of 5xFAD mouse brain regions reveals the lysosome-associated protein Arl8b as a candidate biomarker for Alzheimer’s disease

    No full text
    Additional file 2: Fig. S1. Aβ peptide levels and APP and PSEN1 expression in hippocampus and cortex of 5xFAD mice. Fig. S2. Analysis of Aβ aggregate formation using membrane filter assays and sucrose gradient centrifugations. Fig. S3. Analysis of wild-type expression profiles to assess whether the protein abundance changes detected in 5xFAD brains are more frequent among highly expressed mouse proteins. Fig. S4. Functional analysis of dysregulated proteins defined with a pairwise model in brains of 5xFAD mice. Fig. S5. Enrichment analysis of cell-type-specific marker proteins among dysregulated proteins in brains of 5xFAD mice. Fig. S6. IPA and gene ontology enrichment analysis of differentially expressed proteins defined with the full model in cortical and hippocampal tissues of 5xFAD mice. Fig. S7. Ingenuity pathway analysis of Aβ-correlated and anticorrelated DEPs defined by the pairwise model in brains of 5xFAD mice. Fig. S8. Numbers of pairwise common DEPs in the mouse datasets and datasets from human studies. Fig. S9. Strategy to define mouse protein signatures that are concordantly altered also in AD patient brains. Fig. S10. Investigation of the overlap of DEPs in brains of 5xFAD mice with DEPs in asymptomatic AD brains. Fig. S11. Analysis of the correlation in protein effect sizes between 5xFAD mouse and AD patient brains for proteins present in all studies. Fig. S12. Selection of the neuronal lysosome-associated protein Arl8b by step-by-step data filtering. Fig. S13. Immunofluorescence analysis of 5xFAD brain slices. Fig. S14. Analysis of Arl8b protein aggregates using human brain homogenates derived from AD patients and control individuals

    Additional file 3 of A proteomics analysis of 5xFAD mouse brain regions reveals the lysosome-associated protein Arl8b as a candidate biomarker for Alzheimer’s disease

    No full text
    Additional file 3: Supplementary Excel File 1a. DEPs from 5xFAD versus wild-type tissue comparisons in hippocampus and cortex; DEPs were defined using the “pairwise model”; Supplementary Excel File 1b. Summary of the statistical calculations from Supplementary Excel File 1a; Supplementary Excel File 1c. DEPs in both hippocampal and cortical tissues defined through the “full model”; Supplementary Excel File 2. DEPs that correlate or anticorrelate to Aβ aggregate load in both hippocampal and cortical tissues defined through the “pairwise model”; Supplementary Excel File 3. Identified genes differentially down- or upregulatedin cortex or hippocampus of 5xFAD mice; Supplementary Excel File 4. Results of the Ingenuity Pathway Analyses performed in Figures 3g and 4e; Supplementary Excel File 5a. Proteins with significant abundance changes from Johnson et al. 2020; Supplementary Excel File 5b. Proteins with significant abundance changes from Johnson et al. 2022; Supplementary Excel File 5c. Proteins with significant abundance changes from Johnson et al. 2022; Supplementary Excel File 5d. Proteins with significant abundance changes from Drummond et al. 2022; Supplementary Excel File 6. Numbers of common DEPs in mouse and human datasets; Supplementary Excel File 7. DEPs defined using a “pairwise model” and concomitantly altered both in mouse and human brain tissues from J20, J22 and D22. Supplementary Excel File 8a. The step-by-step selection process to identify the potential AD biomarker Arl8b. Supplementary Excel File 8b. The top 25 Aβ-correlated proteins in mouse hippocampal tissues
    corecore