3 research outputs found

    Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer’s disease

    Get PDF
    Background Astrocytes and microglia react to Aβ plaques, neurofibrillary tangles, and neurodegeneration in the Alzheimer’s disease (AD) brain. Single-nuclei and single-cell RNA-seq have revealed multiple states or subpopulations of these glial cells but lack spatial information. We have developed a methodology of cyclic multiplex fluorescent immunohistochemistry on human postmortem brains and image analysis that enables a comprehensive morphological quantitative characterization of astrocytes and microglia in the context of their spatial relationships with plaques and tangles. Methods Single FFPE sections from the temporal association cortex of control and AD subjects were subjected to 8 cycles of multiplex fluorescent immunohistochemistry, including 7 astroglial, 6 microglial, 1 neuronal, Aβ, and phospho-tau markers. Our analysis pipeline consisted of: (1) image alignment across cycles; (2) background subtraction; (3) manual annotation of 5172 ALDH1L1+ astrocytic and 6226 IBA1+ microglial profiles; (4) local thresholding and segmentation of profiles; (5) machine learning on marker intensity data; and (6) deep learning on image features. Results Spectral clustering identified three phenotypes of astrocytes and microglia, which we termed “homeostatic,” “intermediate,” and “reactive.” Reactive and, to a lesser extent, intermediate astrocytes and microglia were closely associated with AD pathology (≤ 50 µm). Compared to homeostatic, reactive astrocytes contained substantially higher GFAP and YKL-40, modestly elevated vimentin and TSPO as well as EAAT1, and reduced GS. Intermediate astrocytes had markedly increased EAAT2, moderately increased GS, and intermediate GFAP and YKL-40 levels. Relative to homeostatic, reactive microglia showed increased expression of all markers (CD68, ferritin, MHC2, TMEM119, TSPO), whereas intermediate microglia exhibited increased ferritin and TMEM119 as well as intermediate CD68 levels. Machine learning models applied on either high-plex signal intensity data (gradient boosting machines) or directly on image features (convolutional neural networks) accurately discriminated control vs. AD diagnoses at the single-cell level. Conclusions Cyclic multiplex fluorescent immunohistochemistry combined with machine learning models holds promise to advance our understanding of the complexity and heterogeneity of glial responses as well as inform transcriptomics studies. Three distinct phenotypes emerged with our combination of markers, thus expanding the classic binary “homeostatic vs. reactive” classification to a third state, which could represent “transitional” or “resilient” glia.España Ministry of Science, Innovation, and Universities FPU fellowship to CM-CMassachusetts Alzheimer’s Disease Research Center grant P30AG062421 to BTH, and 1R56AG061196 to BTHAlzheimer’s Association (AACF17-524184 and AACF-17-524184-RAPID to AS-P

    Additional file 2 of Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer’s disease

    No full text
    Additional File 2: Figure S1. A β pathology in the temporal pole cortex. Description: Immunohistochemistry for Aβ (mouse monoclonal antibody, clone 6F/3D, Agilent, #M0872, 1:600) with peroxidase/DAB was performed in nearly-adjacent sections to those used for cyclic multiplex fluorescent immunohistochemistry in a Leica BOND-III automated stainer. Sections were counterstained with hematoxylin. Scale bars: 5 mm, insets 200 μm. Figure S2. Phospho-tau pathology in the temporal pole cortex. Description: Immunohistochemsitry for phospho-tauSer202/Thr205(mouse monoclonal antibody, clone AT8, Thermo-Scientific, #MN1020, 1:10,000) with peroxidase/DAB was performed in nearly-adjacent sections to those used for cyclic multiplex fluorescent immunohistochemistry in a Leica BOND-III automated stainer. Sections were counterstained with hematoxylin. Scale bars: 5 mm, insets 200 μm. Figure S3. Expression levels of selected markers across astrocytic and microglial subclusters from public single-nuclei RNA-seq studies. Description: Bubble plots illustrate the percent of nuclei (bubble size) and the gene expression levels (z-scores, color bar) of the astrocytic and microglial markers used in our cyclic multiplex fluorescent immunohistochemistry protocol across the astrocytic and microglial subclusters rendered by several published single-nuclei RNA-seq data sets. Note that our set of markers discriminates some of these transcriptomic subclusters. Figure S4. Characterization of astrocytes and microglia in AD vs. CTRL by cortical layer. Description: Box and whisker plots illustrate the distribution (box: median and interquartile range [IQR]; whiskers: 1.5 × IQR) of mean gray intensity (MGI) z-scores for (a) each astrocytic marker and (b) each microglial marker across the CTRL and AD groups by cortical layer. Only layers II to VI were included in this study. Figure S5. Characterization of astrocytic and microglial states by cortical layer. Description: Box and whisker plots show the distribution (box: median and interquartile range [IQR]; whiskers: 1.5 × IQR) of mean gray intensity (MGI) z-scores for each astrocytic (a) or microglial (b) marker across the three phenotypes by cortical layer. Only layers II to VI were included in this study. Figure S6. Effects of proximity to AD neuropathological changes on astrocytic and microglial phenotypes from two CTRL subjects with abundant Aβ plaques. Description: (a) Representative high-plex image of astrocytes from a CTRL subject with abundant Aβ plaques; note the differences with AD astrocytes in Fig. 5a. For clarity, only ALDH1L1, EAAT2, and GFAP markers are shown together with Aβ. Scale bar: 100 µm, insets a1–a3: 10 µm. (b) Histograms show the proportion of each astrocyte phenotype in n=2 CTRL subjects with abundant Aβ plaques relative to all their astrocytes as a function of their distance (µm, x axis) to the nearest Aβ plaque. Note that there are equal numbers of astrocytes within 25 µm from the nearest Aβ plaque classified as homeostatic, intermediate, or reactive. (c) Representative high-plex image of microglia from the same field of the same CTRL with abundant Aβ plaques; note the differences when compared to AD microglia in Fig. 5c. For clarity, only IBA1, TMEM119, and CD68 markers are shown together with Aβ. Scale bar: 100 µm, insets c1–c3: 10 µm. (d) Histograms indicate the proportion of each microglial phenotype in n=2 CTRL subjects with abundant Aβ plaques relative to all their microglial profiles as a function of their distance (µm, x axis) to the nearest Aβ plaque. Note that most microglia in the vicinity of Aβ plaques were classified as homeostatic, suggesting that their phenotypic transition to intermediate and reactive had not yet occurred. Figure S7. Differences in neuritic component of Aβ plaques from CTRL and AD subjects. Description: Representative images of Aβ and phospho-tau (PHF1) immunohistochemistry corresponding to the same fields of the AD and CTRL subjects shown in Fig. 5 and Fig. S6, respectively. Note the differences in the PHF1+ neuritic changes between CTRL and AD Aβ plaques. Scale bar: 100 µm, insets a1 and b1: 10 µm. Figure S8. Gradient boosting machine models accurately discriminate between glial phenotypes. Description: Receiver operating characteristic (ROC) curves demonstrate the high discriminative power of the gradient boosting machine (GBM) models to discern between states (i.e., homeostatic vs. intermediate vs. reactive) of (a) astrocytes and (b) microglia based on mean gray intensity (MGI) data from thousands of high-plex single-cell profiles. Rankings of the variable importance scores shown in the horizontal bar plots reveal the most relevant markers for each classification task, respectively. Figure S9. Application of deep learning model interpretability functions to astrocytes with extreme classification probabilities. Description: Examples of the convolutional neural network (CNN) model interpretability functions applied to astrocytes with extreme classification probabilities (i.e., confident and correct predictions). Columns 1 and 5 show DAPI and all astrocyte markers of the high-plex image of a single astrocyte cell body from a CTRL and an AD subject, respectively, after performing the CNN normalization steps described (i.e., segmentation, interpolation, channel-level z-score). Hence, the signal intensity is represented by dynamic range rather than by pixel intensity. Columns 2–4 and 6–8 show the saliency (2 and 6), integrated gradient (3 and 7), and GradCAM (4 and 8) maps, which illustrate the pixels of each marker that the CNN considered most important for the classification of these two astrocytes as CTRL or AD. Figure S10. Application of deep learning model interpretability functions to microglia with extreme classification probabilities. Description: Examples of the convolutional neural network (CNN) model interpretability functions applied to microglia with extreme classification probabilities (i.e., confident and correct predictions). Columns 1 and 5 show DAPI and all microglial markers of the high-plex image of a single microglial cell from a CTRL and an AD subject, respectively, after performing the CNN normalization steps described (i.e., segmentation, interpolation, channel-level z-score). Hence, the signal intensity is represented by dynamic range rather than by pixel intensity. Columns 2–4 and 6–8 show the saliency (2 and 6), integrated gradient (3 and 7), and GradCAM (4 and 8) maps, which illustrate the pixels of each marker that the CNN considered most important for the classification of these two microglia as CTRL or AD.Ministerio de Ciencia, Innovación y Universidades Real Colegio Complutense National Institute on Aging Alzheimer's Association.Peer reviewe

    Susceptibility-weighted imaging reveals cerebral microvascular injury in severe COVID-19

    No full text
    We evaluated the incidence, distribution, and histopathologic correlates of microvascular brain lesions in patients with severe COVID-19. Sixteen consecutive patients admitted to the intensive care unit with severe COVID-19 undergoing brain MRI for evaluation of coma or neurologic deficits were retrospectively identified. Eleven patients had punctate susceptibility-weighted imaging (SWI) lesions in the subcortical and deep white matter, eight patients had >10 SWI lesions, and four patients had lesions involving the corpus callosum. The distribution of SWI lesions was similar to that seen in patients with hypoxic respiratory failure, sepsis, and disseminated intravascular coagulation. Brain autopsy in one patient revealed that SWI lesions corresponded to widespread microvascular injury, characterized by perivascular and parenchymal petechial hemorrhages and microscopic ischemic lesions. Collectively, these radiologic and histopathologic findings add to growing evidence that patients with severe COVID-19 are at risk for multifocal microvascular hemorrhagic and ischemic lesions in the subcortical and deep white matter.National Institute of Neurological Disorders and Stroke (Grants R21NS109627, R21AG067562, RF1NS115268)NIH Director’s Office (Grant DP2HD101400)NIH National Institute of Mental Health (Grant K23MH115812)NIH National Institutes of Allergy and Infectious Diseases (Grant 2U19AI110818
    corecore