Aviv Regev’s research while affiliated with Genentech and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (400)


Deep cell atlas of the human lung reveals the presence of ionocytes in both human proximal and distal airways and proximal and distal ALI cultures
A Regional sampling for a deep lung cell atlas. Numbered circles represent sampled locations. B Lung cell atlas. Uniform manifold approximation and projection (UMAP) embedding of cell profiles (dots) from the large airways (left) and lung lobe regions (right) colored by cell type annotation. C–F Epithelial lung and ALI cell profiles. UMAP embeddings of epithelial cell profiles from the proximal airway (C), distal lung lobe (D), and ALI cultures generated from large airway basal cells isolated from primary bronchus (E) or from small airway basal cells isolated from microdissected small airway less than 2 mm in diameter (F). G–I Ionocyte abundance in the human proximal and distal airways and human proximal and distal ALI cultures. G Number of BSND+ mature ionocytes per ALI (y axis) in Large ALI and Small ALI cultures (x axis). n = 3 ALIs averaged from 3 separate donors, Two tailed unpaired T test. Error bars are standard deviation. H Whole mount images of dissected large (left) and small (right) airways stained for BSND (magenta) and acetylated Tubulin (green). Insets: Representative examples of BSND+ ionocytes (magenta). I Number of BSND+ mature ionocytes per mm2 (y axis) in microdissected large airways and small airways (x axis). n = 26 for large airways and 33 for small airways across three normal human lungs (Hu66, Hu67, and Hu68). One way ANOVA (Sidak’s multiple comparisons). Error bars are standard deviation. Elements of 1 A was created with BioRender https://BioRender.com/0ruztco.
Transcriptional and chromatin accessibility profiles reveal a replicative rare cell progenitor and a pre-ionocyte state
APOU2F3+ tuft-like cells are predicted to be progenitors of mature ionocytes. UMAP embedding of scRNA-seq profiles (dots) of rare epithelial cells in our deep lung cell atlas, colored by cell annotation (left) and showing RNA velocity vectors (right) directed from tuft-like cells to ionocytes. B Human large airways and human Large ALI cultures both contain POU2F3+ cells. Antibody staining of a section of the right primary bronchus for POU2F3 (red) and DAPI (blue) and LAE ALI for POU2F3 (green) and DAPI (blue). This staining has been repeated in 3 separate samples. C–EPOU2F3+ tuft-like cells include replicating and non-replicating cells. C Mean expression (dot color, relative expression) and percentage of cells (dot size) expressing selected cell identity and cell proliferation markers (columns) in different rare epithelial cell subsets (rows). D UMAP embedding of scRNA-seq profiles (dots) of rare cells, colored by cell cycle classification (left) or cell type annotation (right). E Large ALI cultures co-stained with POU2F3 (yellow, top) and MKI67 (purple, middle). The bottom panel shows cells expressing both markers (arrows). This staining was repeated in 3 samples. F Distinct chromatin state marks POU2F3+ progenitors. UMAP embedding of large airways epithelial cell scATAC-seq profiles (dots) colored by de novo cell type annotation. Zoom of boxed rare cells highlight chromatin accessibility at select gene loci associated with tuft cells, ionocytes, and progenitor cells.
Type 2 pathway cytokines induce mature tuft cell differentiation
A Neural and immune genes are induced in mature tuft cells. Significance (signed-log10 (q-value), x axis) of enrichment of the four functional gene sets (y axis) in the REACTOME database, most enriched in genes up-regulated in mature tuft cells (positive values) or ionocytes (negative values). B IL-13 treatment shifts the Large ALI cell composition. UMAP embedding of scRNA-seq profiles (dots) from control (left; same plot as in Fig. 1E, reproduced here for convenience) and IL-13-treated (right) LAE ALIs, colored by cell subset annotation. C–E Mature tuft cells are induced in IL-13-treated LAE ALIs. Zoom of a portion of the UMAP embedding in IL-13 treated LAE ALIs (from B, right) colored by scores for rare cell marker gene signatures (Supplementary Data 2) (C) or by expression of rare cell marker genes (D). E Number of antibody-stained cells (y axis) for GNAT3 expressing tuft cells (n = 2 ALIs (Hu19, Hu67)) and BSND expressing ionocytes n = 3 ALIs (Hu19, Hu62, Hu67) in LAE ALI treated with PBS or IL13 (10 ng/ml) (x axis). (All experimental treatments were done in parallel; Methods). F Mature SAE ALIs are treated for 96 h with PBS (control) or IL13 (20 ng/ml). IL-13 treatment shifts SAE ALI cell composition. UMAP embedding of scRNA-seq profiles (dots) from control (left; as in Fig. 1F) and IL-13-treated (right) SAE ALIs, colored by cell type annotation. G Mature tuft cells are induced in IL-13-treated SAE ALIs. Zoom of a portion of the UMAP embedding in IL-13 treated SAE ALIs (from G, right) colored by expression of rare cell marker genes.
Type 2 pathway cytokines redirect the lineage of bipotent Tuft-Ionocyte Progenitor (TIP) cells
A, B Sorting strategy to enrich rare cells. A. Mean expression (dot color, relative expression) and proportion of cells (dot size) expressing genes encoding the cell surface proteins NCAM1 and KIT in human in vivo scRNA-seq data from Fig. 1B. B Expression level (∂∂CT, qPCR, y axis) of key rare cell marker genes (marked on top left) in sorted cell populations from human ALI cultures (x axis, labeled by sorting marker). n = 3 technical replicates. Error bars are standard deviation. Based on this expression data, dissociated cells were stained for anti-human CD45–BV421 (1:100; BioLegend 368522), anti-human CD31–BV421 (1:100; BioLegend 303124), anti-human CD326(EPCAM)–APC (1:100 BioLegend 324208), CD117(KIT)–FITC (1:100; BioLegend 313231) and anti-human CD56(NCAM)–BV711 (1:100; BD Biosciences 563169). A negative sort was performed for CD45 (immune cells) and CD31 (endothelial cells), with positive selection for CD326 (epithelial cells), CD56 (NCAM – rare cell marker), and CD117 (KIT – rare cell marker). Please see Supplementary Fig. 9 for gating strategy. C, D Experimental strategy. C Left: Model of differentiating ALI. Right: POU2F3 mRNA expression (∂∂CT, qPCR, y axis) at different time points (x axis) during ALI differentiation. n = 3 technical replicates. Error bars are standard deviation. D Schematic of experimental time course, where starting at ALI D3 (top; the time point at which TIP cells are first present) cultures were treated with IL13 (10 ng/ml) or PBS control for 5 days and then CD45- CD31- EPCAM + NCAM1 + , CD45- CD31- EPCAM + KIT+ and CD45- CD31- EPCAM + KIT + NCAM1+ rare cells were collected, pooled, and profiled using scRNA-seq. E–H TIP cells give rise to ionocytes via defined transition states in control LAE ALI cultures. UMAP embedding of scRNA-seq profiles (dots) from PBS-treated (control) LAE ALI cultures colored by cell type annotation (E), overlaid RNA velocity vectors (F), cell cycle phase classification (G, left), G1/S (G, middle), and G2/M (G, right) gene signature scores. H Mean expression (dot color) and fraction of cells (dot size) expressing different lineage markers (columns) in each cell subset along the default lineage transition of TIP cells towards ionocytes (rows). I–L TIP cells are diverted towards mature tuft cell fate following IL-13 treatment. UMAP embedding of scRNA-seq profiles (dots) from IL-13-treated LAE ALI cultures colored by cell subsetannotation (I), overlaid RNA velocity vectors (J), cell cycle phase classification (K, left), G1/S (K, middle), and G2/M (K, right) gene signature scores. L Mean expression (dot color) and fraction of cells(dot size) expressing different lineage markers columns, same genes as in H) in each cell subset (rows) along the IL13-induced lineage transition of TIP cells towards mature tuft cells. M Left: Experimental setup schematic of differentiating ALI treated continuously with IL-13 for 25 days. Right: Number of BSND+ cells (ionocytes, y axis, n = 3 ALIs (Hu19, Hu60, Hu67)), error bars are standard deviation, and GNAT3+ cells (tuft cells, y axis, n = 2 ALIs (Hu60, Hu67)) in PBS and IL-13 conditions (x axis). N RNA velocity analysis of the pooled PBS and IL13-treated ALIs (clustering shown in Supplementary Fig. 10C, D) demonstrates that IL13 redirects TIP cell differentiation towards mature tuft cells and away from the default pathway of ionocyte differentiation. O Schematic depicting the observed expression of lineage-specifying TFs in TIP cell descendants that are differentiating towards either ionocyte or mature tuft cell fate. Orange arrows indicate default differentiation (PBS) and blue arrows indicate IL13-induced differentiation. P Proposed model of cytokine-mediated TIP cell lineage switching. Illustrations 4 C, M, O, P were created with BioRender: https://BioRender.com/dbkmcyx.
IL17A promotes tuft cell differentiation and asthmatic airway epithelium contains mature tuft cells
A–D Tuft cell abundance increases following IL17A treatment in LAE ALI. A, C Overview schematic of IL17A treatment experiments in mature LAE ALI treated with PBS (control) or IL17A (50 ng/ml; from D39 to D44) (A) and differentiating ALI at D3 (the first time point at which TIP cells are present) treated cells with IL17A (50 ng/ml) or PBS from D3 to D24 (C). B Number of GNAT3+ mature tuft cells (quantified by immunohistochemistry, y axis, n = 2 ALIs (Hu19, Hu67)), when mature LAE ALIs are treated with PBS or IL17A (50 ng/ul; from D39 to D44) (x axis) D Number of GNAT3+ mature tuft cells (quantified by immunohistochemistry, y axis, n = 2 ALIs, (Hu19, Hu67)), in LAE ALIs when treated with PBS or IL17A from D3 to D24. E, F Tuft cells are increased in ALIs and airways from asthmatic patients. E Number of GNAT3+ mature tuft cells (y axis) in LAE ALIs derived from two asthmatic individuals (Hu70 and Hu78) and in patients with no history of lung diseases (Hu66, Hu67) (x axis), n = 1 ALI per donor. F Whole mount staining of dissected airways from a patient with asthma exacerbation. Staining in the top panel with GNAT3 (green) shows a mature tuft cell, surrounded by ciliated cells (Atub, white). Staining with additional mature tuft cell marker ALOX5AP (magenta) reveals the characteristic bipolar morphology (arrows) associated with mature tuft cells (bottom panels). Due to the availability of tissue, immunostaining was performed on only 1 donor. Illustrations 4B and D were created with BioRender: https://BioRender.com/g37wh81.
Single cell profiling of human airway identifies tuft-ionocyte progenitor cells displaying cytokine-dependent differentiation bias in vitro
  • Article
  • Full-text available

June 2025

·

36 Reads

Viral S. Shah

·

·

·

[...]

·

Human airways contain specialized rare epithelial cells including CFTR-rich ionocytes that regulate airway surface physiology and chemosensory tuft cells that produce asthma-associated inflammatory mediators. Here, using a lung cell atlas of 311,748 single cell RNA-Seq profiles, we identify 687 ionocytes (0.45%). In contrast to prior reports claiming a lack of ionocytes in the small airways, we demonstrate that ionocytes are present in small and large airways in similar proportions. Surprisingly, we find only 3 mature tuft cells (0.002%), and demonstrate that previously annotated tuft-like cells are instead highly replicative progenitor cells. These tuft-ionocyte progenitor (TIP) cells produce ionocytes as a default lineage. However, Type 2 and Type 17 cytokines divert TIP cell lineage in vitro, resulting in the production of mature tuft cells at the expense of ionocyte differentiation. Our dataset thus provides an updated understanding of airway rare cell composition, and further suggests that clinically relevant cytokines may skew the composition of disease-relevant rare cells.

Download

CellFlow enables generative single-cell phenotype modeling with flow matching

April 2025

·

22 Reads

High-content phenotypic screens provide a powerful strategy for studying biological systems, but the scale of possible perturbations and cell states makes exhaustive experiments unfeasible. Computational models that are trained on existing data and extrapolate to correctly predict outcomes in unseen contexts have the potential to accelerate biological discovery. Here, we present CellFlow, a flexible framework based on flow matching that can model single cell phenotypes induced by complex perturbations. We apply CellFlow to various phenotypic screens, accurately predicting expression responses to a wide range of perturbations, including cytokine stimulation, drug treatments and gene knockouts. CellFlow successfully modeled developmental perturbations at the whole-embryo scale and guided cell fate and organoid engineering by predicting heterogeneous cell populations arising from combinatorial morphogen treatments and by performing a virtual organoid protocol screen. Taken together, CellFlow has the potential to accelerate discovery from phenotypic screens by learning from existing data and generating phenotypes induced by unseen conditions.


Havcr2cKO microglia have a MGnD-like gene-expression profile
a,b, Gene expression in microglia during development. a, Heatmap visualization of the average expression across microarray probes mapping to the same gene (row), normalized across replicates (column) in progenitor, embryonic (E) and postnatal (P) adult stage (2 months old)²¹ microglia. YSM, yolk sac macrophage. b, Dot plot visualization of gene expression (rows) at P9 and P28 stages of development²⁴. The dot size indicates the proportion of cells expressing the gene, and the colour represents average expression. c,d, RT–qPCR analysis of co-inhibitory molecules (c) and TGFβ-pathway-related molecules (d) in microglia (live CD45intCD11b⁺) during the early postnatal period (n = 9 (P7), 3 (P13), 5 (P20), 4 (P26)). The mean expression value at P7 was set as 1 for each gene. a.u., arbitrary units. e, Flow cytometry analyses of primary microglia (live CD45intTMEM119⁺) cultured with TGFβ (10 ng ml–1) (n = 3 per condition). The mean fluorescence intensity (MFI) of samples without stimulation was set as 1 for each molecule. FMO, fluorescence minus one. f, RT–qPCR analysis of primary microglia (live CD45intCD11b⁺) cultured with TGFβ (10 ng ml–1) for 24 h (n = 3 per condition, independent samples). g, Fragments per kilobase of transcript per million mapped reads (FPKM) value of Havcr2 in Tgfbr2cKO microglia²⁶ (n = 3 per condition). h,i, RNA-seq analysis of microglia (live CD45intCD11b⁺) from 1-month-old Havcr2cKO mice (n = 4 (3 males, 1 female), independent mice) and Havcr2flox/flox (control) mice (n = 5 (4 males, 1 female), independent mice). h, Heatmap visualization of DEGs (FDR < 0.05). Rows represent genes and columns are biological replicates. Gene expression is row-normalized across replicates. i, Volcano plot of a DEG analysis performed using DESeq2. j–l, The scores of MGnD (j), homeostasis (k) and KEGG phagosome (l) signatures. The y axis represents log2-transformed average transcripts per million (TPM) (the same dataset as Fig. 1h, independent mice). m, RT–qPCR analysis of Havcr2cKO microglia (live CD45intCD11b⁺4D4⁺GR1⁻) (n = 8 control, 7 Havcr2cKO, all females). The mean expression value of controls was set as 1 for each gene. n, Flow cytometry analysis of AXL, LY9 and TIM-3 in Havcr2cKO microglia (n = 8 control, 8 Havcr2cKO, all females). Results are shown from one experiment, representing at least two independent experiments (e,f,n). Data are the mean ± s.e.m. Student’s two-tailed t-test.
Source data
Havcr2cKO and Tgfbr2cKO microglia share SMAD2-related gene-expression changes
a, Circos plot comparison of DEGs upregulated (yellow) and downregulated (green) in Havcr2cKO microglia (the same dataset as Fig. 1h) with those upregulated (pink) and downregulated (blue) in Tgfbr2cKO microglia²⁶. b, Scatter plot of genes based on expression differences represented by log2-transformed fold changes in Havcr2cKO (x axis) and Tgfbr2cKO microglia (y axis) compared with control microglia. Genes upregulated or downregulated in both comparisons are indicated in red and blue, respectively. c, Co-IP–MS screening for binding partners of TIM-3. d, Blots of HEK293 cells transfected with WT Havcr2 and co-immunoprecipitated for HA–TIM-3. The samples for the control β-actin were run on a separate gel as sample-processing controls. IP, immunoprecipitated. e, Blots of lysate from human iMG co-immunoprecipitated for TIM-3. The band with a lower molecular weight in the lanes of the co-immunoprecipitated samples for SMAD2 is from the heavy chain of the IgG (around 50 kDa) antibody used for Co-IP. f, Homer transcription factor motif-enrichment analysis of 82 upregulated and 79 downregulated DEGs in Havcr2cKO microglia described in Fig. 1h. g,h, Flow cytometry analysis of Havcr2cKO microglia for pSMAD2 (g) and SMAD2 (h) (n = 8 and 7 for g; 5 and 4 for h, independent samples). MFI, mean fluorescence intensity. i, RT–qPCR analysis of Havcr2cKO microglia (live CD45intCD11b⁺4D4⁺GR1⁻) (n = 8 control, 7 Havcr2cKO). j, Flow cytometry analysis of microglia (live CD45intCD11b⁺) for pSMAD2 cultured with M-CSF (10 ng ml–1), GM-CSF (10 ng ml–1) or TGFβ (10 ng ml–1) for 16 h (n = 3 per condition, independent samples). k, Flow cytometry analysis of pSMAd2 in HEK293 cells transfected with WT Havcr2 plasmid stimulated with TGFβ for 4 h (n = 3 per condition, independent samples). l, Flow cytometry analysis of Tgfbr2KO and Smad2KO BV2 cells transfected with WT Havcr2 plasmid (n = 4 per condition, independent samples). Results are shown from one experiment, representing at least two independent experiments (d,e,g,h,j–l). Data are the mean ± s.e.m. Statistical tests: permutation test (a), two-sided Spearman’s correlation test (b), two-sided moderated t-test with Benjamini–Hochberg correction (c) or Student’s two-tailed t-test (g,j–l). See Supplementary Fig. 1 for gel source data (d,e). NS, not significant.
Source data
Havcr2cKO microglia have increased phagocytic ability and share a gene-expression signature with phagocytosing microglia
a, WT and mutant Havcr2 structures. HA, haemagglutinin; TM, transmembrane. b, Blots of HEK293 cells transfected with Havcr2 constructs co-immunoprecipitated for HA–TIM-3. The samples for the control β-tubulin were run on a separate gel as sample-processing controls (see Supplementary Fig. 1 for gel source data). c, Flow cytometry analysis of pSMAD2 in Havcr2 construct-transfected HEK293 cells. A successfully transfected TIM-3-positive fraction was analysed and compared with all the live cells in the control group (Extended Data Fig. 1c) (n = 3 per condition, independent samples). d, Schematic of the regulation of SMAD2 phosphorylation by TIM-3. e, Havcr2cKO microglia were cultured with pH-sensitive dye-stained Aβ for 4 h (n = 8 control, 7 Havcr2cKO, 1 without Aβ, independent samples). f, Fluorescence-stained dead neurons were injected into the cortex and hippocampus of live male mice (3 months old, n = 8 control, 6 Havcr2cKO, independent mice). The fraction of phagocytosing microglia was analysed 16 h later. g, RNA-seq analysis of sorted microglia (live CD45intCD11b⁺4D4⁺Ly6C⁻) from the same experiment as f. The top DEGs shared by at least two of the following three comparisons are shown in the heatmap: (1) control phagocytosing (Phago(+)) versus control non-phagocytosing (Phago(–)); (2) Havcr2cKO phagocytosing versus control phagocytosing; (3) Havcr2cKO non-phagocytosing versus control non-phagocytosing. Selected genes are highlighted, including the KEGG phagosome pathway gene set (mmu04145), the top 100 DEGs of Clec7Aa⁺ versus Clec7a⁻ microglia¹⁰ and selected genes (Cd9, Cd33, Cst7, Cstb, Cxcl16, Ly9, Lyz2 and H2). h–j, The expression of Marco, Cd33 and Havcr2 in microglia from the same experiment as g (n = 8, 5, 5 and 4, from the left, independent mice in each genotype). k, Pathway analysis of the top DEGs comparing non-phagocytosing Havcr2cKO to non-phagocytosing control microglia (FDR < 0.1). Disease pathways (pathways under sections 6.1–6.10 from https://www.kegg.jp/kegg/pathway.html) and ribosomal genes were excluded from the analysis. l, Gene-expression signatures of Havcr2cKO microglia (DEGs between Havcr2cKO non-phagocytosing and control non-phagocytosing microglia) and phagocytosing microglia (DEGs between control phagocytosing and control non-phagocytosing microglia) were compared using the same RNA-seq data as in g. The same set of genes is highlighted as in g. m, The number of overlapping genes (n) between each pair of DEGs upregulated and downregulated in Havcr2cKO, phagocytosing, Tgfbr2cKO and Clec7a⁺ microglia compared with control microglia. N is the total number of up- or downregulated genes in the condition (row). Results are shown from one experiment, representing at least two independent experiments (b,c,e,f). Data are the mean ± s.e.m. Statistical tests: one-way analysis of variance (ANOVA) with Dunnett’s multiple comparisons test (c) or Student’s two-tailed t-test (e,f,h–j). One-sided permutation test is described in the section ‘Circos plot and permutation test’ in the Methods for l and m. The schematic in d was created using BioRender (https://biorender.com).
Source data
Havcr2icKO microglia improve cognitive function and mitigate pathological changes in the brain of a mouse model of AD
a,b, Spatial memory function was assessed using spontaneous alternation Y-mazes tests in mice aged 7–8 months (n = 37, 33, 30 and 35, from the left, independent mice) (a). Multivariate linear regression analysis confirmed that memory function was restored in Havcr2icKO:5×FAD mice compared with 5×FAD mice (b). c,d, Cognitive function was assessed using forced alternation Y-maze tests in mice aged 7–8 months (n = 53, 52, 50 and 55, from the left) (c). Multivariate linear regression analysis (d). e–g, Anxiety-like behaviour was longitudinally assessed using open-field tests in mice aged 4, 6, 8 and 10 months (groups from the left: n = 41, 49, 39 and 37 (4 months old); 44, 49, 39 and 41 (6 months old); 44, 47, 38 and 40 (8 months old); and 39, 40, 30 and 35 (10 months old)) (e,f). Multivariate linear regression analysis for mice aged 10 months (g). h, Images of immunostaining for Aβ (HJ3.4b antibody) and CLEC7A in 6 month-old female mice. i, Confocal images of CLEC7A, P2RY12 and HJ3.4b in 6-month-old female mice. j,k, Quantification of HJ3.4b-positive (j) and CLEC7A-positive (k) area and fluorescence intensity in cortical regions at 6 months (n = 4 and 7 females in 5 × FAD and Havcr2icKO:5 × FAD, respectively. n = 6 males per group). l, Images of plaques in the cortices of mice aged 7 months were classified into filamentous, compact and inert areas by double staining with methoxy-X04 and HJ3.4b (n = 336 and 251 plaques). The percentage of each area was calculated per plaque >100 µm² in size. As plaque groups, plaques with ≥50% area of any phenotype were classified into the category of that phenotype. The difference in filamentous and inert areas was not significant when assessed only for male mice. Results are shown from one experiment, representing at least two independent experiments (h–k). The results were reproduced with an alternative method in Extended Data Fig. 5d,e, for l. Data are the mean ± s.e.m. Statistical tests: Kruskal–Wallis test adjusted by the FDR (a,f), unpaired two-sided Wilcoxon test (c) or Student’s two-tailed t-test (j–l). Scale bars, 1 mm (h), 50 µm (i) or 10 µm (l).
Source data
Havcr2icKO mice have distinct microglial states in an AD model
a, UMAP visualization of snRNA-seq data of microglia prepared from mice aged 5 months (colour, number on legend). b, Probability density curves of the signature score for the hallmark TGFβ pathway in C2 from 5×FAD (purple) and Havcr2icKO:5×FAD (yellow) mice. Individual cells of each genotype are represented by the bars on top. c–e, Projection of P1 (magenta) and P2 (green) marker genes on the volcano plot of DEGs in different microglial perturbations: phagocytosis assay (c), MGnD (d) and Tgfbr2cKO (e). Only marker genes that are also significant in each perturbation are indicated. Spearman’s correlation was analysed for log2 fold changes of all expressed genes in each perturbation condition (c–e) with those in P1 versus P2. f, Dot plot representation of P1 and P2 marker genes that are significant DEGs in perturbations (c–e) and known anti-inflammatory, pro-inflammatory and phagocytic properties. Size of the dot is the percentage of cells in the genotype (row) expressing the marker gene. Colour represents the average scaled expression of the gene. g,h, Violin plots of alternative macrophage (g) and phagocytic (h) signature genes among genotypes in C2 (MGnD). i, Flow cytometry analysis of microglia prepared from mice aged 4 months (n = 3, 3 female; 4, 3 male, independent mice). Each data point in the bar plots represent the MFI for each sample only from Clec7a⁺ MGnD (the phenograph C12 of the UMAP, as shown in Extended Data Fig. 10j). j, Gating of microglia based on the expressions of NRP1 and IL-10RA in Clec7a⁺ MGnD. The frequencies of each population are shown (n = 3 5xFAD female mice, 3 Havcr2icKO:5×FAD female mice, 4 5xFAD male mice and 3 Havcr2icKO:5xFAD male mice). k, UMAP visualization of transcriptomic profiles of Havcr2icKO: 5×FAD and 5×FAD microglia coloured by clusters. Each dot is a microglial cell. l, KEGG, GO and MSigDB hallmark signatures and the cGAS–STING signature from a previous study⁶⁴ were compared between Havcr2icKO:5×FAD and 5×FAD mice in each microglial cluster defined in k. Colour is Hedges’ g effect size, and asterisks are the significance level of the Benjamini–Hochberg-adjusted t-test (Havcr2icKO:5×FAD against 5×FAD). Data are the mean ± s.e.m. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001 and ****P ≤ 0.0001. Statistical tests: Student’s two-tailed t-test (i,j) or one-way ANOVA with Tukey’s honestly significant difference correction (g,h). CC, cell cycling; HMG, homeostatic microglia.
Source data
Immune checkpoint TIM-3 regulates microglia and Alzheimer’s disease

April 2025

·

309 Reads

·

2 Citations

Nature

Microglia are the resident immune cells in the brain and have pivotal roles in neurodevelopment and neuroinflammation1,2. This study investigates the function of the immune-checkpoint molecule TIM-3 (encoded by HAVCR2) in microglia. TIM-3 was recently identified as a genetic risk factor for late-onset Alzheimer’s disease³, and it can induce T cell exhaustion⁴. However, its specific function in brain microglia remains unclear. We demonstrate in mouse models that TGFβ signalling induces TIM-3 expression in microglia. In turn, TIM-3 interacts with SMAD2 and TGFBR2 through its carboxy-terminal tail, which enhances TGFβ signalling by promoting TGFBR-mediated SMAD2 phosphorylation, and this process maintains microglial homeostasis. Genetic deletion of Havcr2 in microglia leads to increased phagocytic activity and a gene-expression profile consistent with the neurodegenerative microglial phenotype (MGnD), also referred to as disease-associated microglia (DAM). Furthermore, microglia-targeted deletion of Havcr2 ameliorates cognitive impairment and reduces amyloid-β pathology in 5×FAD mice (a transgenic model of Alzheimer’s disease). Single-nucleus RNA sequencing revealed a subpopulation of MGnD microglia in Havcr2-deficient 5×FAD mice characterized by increased pro-phagocytic and anti-inflammatory gene expression alongside reduced pro-inflammatory gene expression. These transcriptomic changes were corroborated by single-cell RNA sequencing data across most microglial clusters in Havcr2-deficient 5×FAD mice. Our findings reveal that TIM-3 mediates microglia homeostasis through TGFβ signalling and highlight the therapeutic potential of targeting microglial TIM-3 in Alzheimer’s disease.




A Sample processing pipeline depicting sample acquisition, preparation for snRNAseq and spatial transcriptomic profiling, data generation, integration, and in silico functionalization. B Uniform manifold approximation and projection (UMAP) for all cells passing quality control (n = 80,808, Hepatocytes, n = 51,605; Immune / blood cells, n = 12,346; Endothelial cells: n = 9278, Mesenchymal cells (n = 4647); Biliary epithelial cells / Cholangiocytes, n = 2932). C Heatmap capturing the expression of marker genes across the 5 major compartments. D UMAP plots depicting gene marker expression for each compartment
Overview of the digital spatial profiling. A Regions of interest (ROIs), corresponding to the liver lobule and the portal area. Gene expression in each region was profiled using the NanoString GeoMx Digital Spatial Profiling (DSP) Whole Transcriptome Atlas (WTA) platform. B Diagram of the spatial arrangement of cellular subpopulations in the liver lobule and interactions in the context of COVID-19 (HA, hepatic artery; PV, portal vein; CV, central vein; BD, bile duct). C Principal component analysis (PCA) embeddings based on batch-corrected probe counts of all ROIs (right) and for the liver lobule ROIs (left) reveal that the DSP WTA platform correctly separates the lobular region from the portal, and reveals significant expression differences between the 3 zones. D Normalized pathway activity scores (PAS) between lobule regions. The DSP WTA is able to capture known zone-specific pathways as well as reveal perturbed pathways related to liver pathology and viral infection
A Uniform manifold approximation and projection (UMAP) for Hepatocytes (HEP1 n = 13,951, HEP2 n = 11,187, HEP3 n = 9956, HEP4 n = 9241, HEP5 n = 4056, HEP6 n = 1612, HEP7 n = 1602). B Heatmap capturing the expression of marker genes across the hepatocyte and the biliary epithelial cell compartments. C Slingshot pseudotime values (left) projected on the 2 primary harmony embeddings across 5 lineages for hepatocyte and biliary epithelial cells from COVID-19 and healthy liver nuclei. The starting and ending lineage points are represented with green and red, respectively. Slingshot-derived lineages (right), coupled with cell composition fold-change differences between healthy and COVID-19 liver samples on a log2 scale. D Cell proportion differences between COVID-19 and healthy liver samples. Significantly different proportions are marked in red (higher in COVID-19), in blue (higher in Controls), and denoted with * (* FDR < 0.05, ** FDR < 0.01; Binomial Generalized Linear Mixed Model). COVID-19-specific clusters are denoted with dark red. E Abundance of SARS-CoV-2 RNA+ nuclei in the snRNAseq clusters. The bars are colored by the scaled viral enrichment score estimated per cluster. Significantly enriched clusters are marked in red and denoted with * (* FDR < 0.05, ** FDR < 0.01; Viral enrichment test). F Uniform manifold approximation and projection (UMAP) plots depicting the average expression of different heat shock proteins (HSPA1A, HSPA1B, HSPA5, HSPA6, HSPA9, HSPB1, HSPD1) in hepatocytes (upper left), pathway activity scores for GO term “regulation of type I interferon-mediated signaling pathway” (GO:0060338, bottom left), the viral load in all the cellular compartments (upper right), in hepatocytes (lower middle), and the average expression on NFKB1 in hepatocytes (lower right)
A Abundance of SARS-CoV-2 RNA+ nuclei in the snRNAseq data for each donor. The bars are colored by the scaled viral enrichment score estimated per donor. Only donor L1 has a significant viral enrichment score (* FDR < 0.01; viral enrichment test). B Distribution of the NanoString GeoMx DSP SARS-CoV-2 probe enrichment score across donors. Donor L1 has a significantly higher enrichment score (FDR = 0.037, t-test) compared to the rest of the donors (L2-4). C Uniform manifold approximation and projection (UMAP) for biliary epithelial cells (BEC1 n = 736; BEC2 n = 687; BEC3 n = 457; BEC4 n = 373; BEC5 n = 371; BEC6 n = 281; BEC7 n = 27)
A Uniform manifold approximation and projection (UMAP) for the A immune / blood, B endothelial cell, and C mesenchymal cell compartments (AImmune: MAC1 n = 2798, MAC2 n = 2601, TC1 n = 1522, TC2 n = 388, TC3 n = 327, TC4 n = 29, DBL1 n = 1331, MAC3 n = 1038, NK n = 857, PC1 n = 397, PC2 n = 124, BC n = 124, ERY-P n = 359 MAC4 n = 222, MAST n = 36 DBL2 n = 193; BEndothelial: EC1 n = 2338, EC2 n = 2247, EC3 n = 1563, EC4 n = 1117, EC5, n = 795, EC6 n = 379, EC7 n = 328, EC8 n = 166, EC9 n = 116, DBL3 n = 91, EC11 n = 73, EC12 n = 65; CMesenchymal: MES1 n = 1223, MES2 n = 1065, MES3 n = 1040, MES4 n = 374, MES5 n = 328, MES6 n = 312, MES7 n = 275, MES8 n = 30). Heatmaps capturing the expression of marker genes across the 3 distinct major compartments are displayed. D Heatmap portraying the cell–cell communications between the cell populations. The color gradient indicates the strength of interaction between any two cell groups. Recipient/Donor cell-type color is portrayed in a blue (healthy) to red (COVID-19) gradient, relevant to the cell composition fold-change differences between healthy and COVID-19 liver samples. E Circle plot portraying the aggregated cell–cell communication network in the TGFb pathway. This analysis includes the enriched hepatocytes in SARS-CoV-2 reads as a separate population (HEP Inf). Thicker edge lines indicate a stronger signal, while circle sizes are proportional to the number of cells in each cellular compartment. Donor edge-line and circle color are portrayed in blue (significantly increased in healthy liver samples), red (significantly increased in COVID-19 liver samples), and black (no significant difference between COVID-19 and controls in cell proportions), concordantly with the cell composition fold-change differences between healthy and COVID-19 liver samples
A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients

March 2025

·

44 Reads

·

3 Citations

Genome Biology

Background The molecular underpinnings of organ dysfunction in severe COVID-19 and its potential long-term sequelae are under intense investigation. To shed light on these in the context of liver function, we perform single-nucleus RNA-seq and spatial transcriptomic profiling of livers from 17 COVID-19 decedents. Results We identify hepatocytes positive for SARS-CoV-2 RNA with an expression phenotype resembling infected lung epithelial cells, and a central role in a pro-fibrotic TGFβ signaling cell–cell communications network. Integrated analysis and comparisons with healthy controls reveal extensive changes in the cellular composition and expression states in COVID-19 liver, providing the underpinning of hepatocellular injury, ductular reaction, pathologic vascular expansion, and fibrogenesis characteristic of COVID-19 cholangiopathy. We also observe Kupffer cell proliferation and erythrocyte progenitors for the first time in a human liver single-cell atlas. Despite the absence of a clinical acute liver injury phenotype, endothelial cell composition is dramatically impacted in COVID-19, concomitantly with extensive alterations and profibrogenic activation of reactive cholangiocytes and mesenchymal cells. Conclusions Our atlas provides novel insights into liver physiology and pathology in COVID-19 and forms a foundational resource for its investigation and understanding.



Figure 1: Method overview. CellUntangler takes as input scRNA-sequencing measurements and one or more batch effects, and embeds cells into decomposed latent subspaces with geometries tailored to separate distinct biological signals into individual representations. (a) The CellUntangler model with two latent subspaces: one to capture the cell cycle, z 1 , and the other to capture non-cell cycle-specific signals, z 2 . The parameters of q(z | x; ϕ) and p(x | z, y; θ) are represented by ϕ and θ, respectively. (b) The cell cycle (cell cycle stage indicated by color) obscures the control and disease signal (indicated by shape). CellUntangler disentangles these signals by embedding the cell cycle signal in z 1 , revealing the control and disease signal in z 2 . z 2 is projected to 2D using UMAP for visualization.
Figure 7: Probabilistic graphical models for CellUntangler. The probabilistic graphical model for CellUntangler when the latent representation is decomposed into z 1 and z 2 , (a) when stop gradient is not used, and (b) when it is used.
CellUntangler: separating distinct biological signals in single-cell data with deep generative models

January 2025

·

36 Reads

Single-cell RNA-seq data have provided new insights into intracellular and intercellular processes. Because multiple processes are active in each cell simultaneously, such as its cell type program, differentiation, the cell cycle, and environmental responses, their respective signals can confound one another, requiring methods that can separate and filter different complex biological signals. Each such signal is based on different gene activities and can define different relationships between cells. However, existing methods often focus on a single process or rely on overly restrictive assumptions, thus removing, rather than disentangling biological signals. Here, we develop CellUntangler, a deep generative model that embeds cells into a flexible latent space composed of multiple subspaces, each designed with an appropriate geometry to capture a distinct signal. We apply CellUntangler to datasets containing only cycling cells and both cycling and non-cycling cells, generating embeddings in which the cell cycle signal is disentangled from non-cell cycle specific signals, such as cell type or differentiation trajectory. We demonstrate CellUntangler's extensibility by using it to capture and separate spatial from non-spatial signals. With CellUntangler, we can obtain latent embeddings that capture various biological signals and perform enhancement or filtering at the gene expression level for downstream analyses.


SCimilarity metric learning enables cell search in large human scale atlases
a, Cell querying with SCimilarity. Left, a query cell profile is compared to a searchable reference foundation model of 23.4 million profiles from 412 studies. Middle, samples with similar cells are identified and returned with information about the original sample conditions, including tissue, in vitro or diseases contexts. Right, a SCimilarity score is computed between the query cell and each cell within a tissue sample. b, Triplet loss training. Left, 56 training and 15 test datasets with Cell Ontology annotations from across the body are used as input. Middle, cell triplets are sampled, each consisting of an anchor cell (A), a positive cell (P, anchor-similar) and a negative cell (N, anchor-dissimilar), based on Cell Ontology annotations. Only non-ambiguous relationships are allowed. Right, triplets are used to train a neural network that embeds similar cells closer than dissimilar ones, forming a foundation model. Treg, regulatory T cells. The loss function is computed using a cell triplet, a reconstructed anchor cell profile (Â), and a weighting parameter (β) to balance the triplet loss (Ltriplet) and the mean squared error loss (LMSE).
SCimilarity learns a universal representation that generalizes to new datasets
a, A large-scale reference database of public gene expression datasets across tissues and diseases. The number of cells (circle size) across tissues (outermost light blue circles) and disease states (middle green circles) across individual studies (innermost circles) in the training (gold), test (pink) or unannotated (purple) datasets is shown. SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; NAFLD, non-alcoholic fatty liver disease; MS, multiple sclerosis; LCH, Langerhans cell histiocytosis; LAM, lymphangioleiomyomatosis; IBD, inflammatory bowel disease. b, Benchmarking SCimilarity against established data integration models. Ontology-aware ARI (study ARI, y axis, top left), NMI (study NMI, y axis, top middle), batch ASW (y axis, top right), cell type ASW (y axis, bottom left) and graph connectivity (y axis, bottom right) for different integration methods and SCimilarity (coloured bars), each applied to integrate two kidney datasets, two lung datasets, two PBMC datasets and all 15 held out (test) datasets (x axis) are shown. c, SCimilarity generalizes new datasets and flags outlier cells across different tissues and conditions. The fraction (x axis) of cells with low similarity to training data (SCimilarity score of <50) in each study (points) from different diseases (y axis, top) or healthy tissues (y axis, bottom callout) is shown.
SCimilarity accurately annotates cell types across the human body
a, SCimilarity cell annotation. A new unannotated cell (grey, bottom left) is embedded in SCimilarity’s common low-dimensional space and compared against the precomputed reference for cell type annotation (0.02 s per cell). b–d, SCimilarity annotation of a kidney scRNA-seq dataset. b,c, Uniform manifold approximation and projection (UMAP) embedding of cell profiles (dots) from SCimilarity’s latent representation of a held-out kidney dataset²⁵ coloured by author-provided (b) or SCimilarity-predicted (c) cell type annotations. LoH TAL, loop of Henle thick ascending limb; LoH tDL, loop of Henle thin descending limb. d, The percentage (colour bar and number) of author-annotated cells (columns) with each SCimilarity annotation (rows). e, Cell type annotation performance. Left, the accuracy (percentile F1 scores, higher is better; y axis) of SCimilarity and each of three annotation methods (colour bars) in matching author annotations in each of 15 test datasets (x axis) withheld from SCimilarity training. Right, the distribution of percentile F1 scores for each method (colour) across all 15 datasets. The box plots show the upper/lower quartiles (box limits), minimum/maximum values (whiskers) and median (centre line). F1 scores are calculated using a random sample of n = 10,000 cells per study. Data from refs. 1,25, 26, 27, 28–29,56, 57, 58, 59, 60, 61, 62, 63–64. Epi., epithelial; prox., proximal; pDC, plasmacytoid DC.
SCimilarity cell search reveals FMs across ILD and other diseases
a, SCimilarity cell search. A query cell profile (bottom left) is embedded into the SCimilarity representation with 23.4 million reference cells. Its nearest neighbours by distance are tabulated by study, tissue and disease. b–e, Identification of FMs across tissues. b, SCimilarity scores (y axis, log10 scale and colour bar) against an FM query profile for all monocytes and macrophages (dots) from 1,041 in vivo tissue samples from 143 studies (x axis), ordered by the mean SCimilarity score. c, The number of cells (circle size) across tissues (outermost light blue circles), disease states (middle green circles) and individual studies (innermost circles, coloured by the fraction of monocytes and macrophages with SCimilarity scores >99th percentile of all FM SCimilarity scores (log-scaled colour bar)). Circle sizes for disease and individual study are scaled relative to other diseases in the same tissue or studies in the same disease. d,e, UMAP of all single-cell profiles (macrophages and otherwise, dots) from the SCimilarity representation for ILD⁴⁰ (d) and PDAC⁴⁷ (e) studies, coloured by FM query SCimilarity scores (colour bar). f, SCimilarity’s explainability framework scores FM-associated genes by importance. The distribution of Integrated Gradients attribution scores (y axis, top; horizontal bars show the mean) for genes (x axis, top; columns, bottom) with the top 50 scores for FMs versus lung macrophages and their membership (red, presence; grey, absence) in published macrophage signatures (bottom, rows). The left colour bar represents the AUC for the attribute score match to published signatures. The signature publication source and P value (two-sided Mann–Whitney U-tests; in signature > not in signature) across the top 3,000 genes by mean attribution score are shown on the right. Attribution scores, AUC values and P values were calculated using the n = 500 cells most similar to FMs against n = 500 randomly sampled cells from the full n = 2,578,221 cell monocyte and macrophage query set.
SCimilarity cell search identifies in vitro cells matching an in vivo FM state and a novel in vitro disease model
a, Identification of FM-like cells across in vitro samples with a SCimilarity cell search. SCimilarity scores (y axis, log10 scale, colour bar) against a FM query profile for each annotated myeloid cell (dot) from n = 40 in vitro samples (x axis) from n = 17 studies, ordered by the mean SCimilarity score. The grey boxes show day 0 and day 5 samples from a 3D-hydrogel culture system². b–f, 3D conditions yield FM-like cells in vitro in validation experiments. b, SCimilarity scores (y axis, log10 scale, colour bar) against an FM query profile for each annotated myeloid cell (dot) in the original 3D-hydrogel culture system dataset² from n = 2 independent donors at day 0 and day 5 and from n = 3 independent donors in the day 8 validation experiment (x axis). c, The mean expression (dot colour) and percentage of cells (dot size) expressing genes (rows) with a high SCimilarity attribution score for distinguishing FMs in vivo (as in f) in myeloid cells in the original 3D-hydrogel culture system² and in the validation experiment (columns). d–f, UMAP embedding from SCimilarity’s query model latent space of cell profiles (dots) from day 0 (d) or day 5 (e) of the original 3D-hydrogel culture system², or from day 8 (f) of the replication experiment, coloured by FM SCimilarity score (colour bar). g, Replication of original finding of HSC expansion. The proportion of HSCs in n = 2 donors from ref. ² at day 0 and day 5 and n = 3 donors from the day 8 validation experiment.
A cell atlas foundation model for scalable search of similar human cells

November 2024

·

55 Reads

·

31 Citations

Nature

Single-cell RNA sequencing has profiled hundreds of millions of human cells across organs, diseases, development and perturbations to date. Mining these growing atlases could reveal cell–disease associations, identify cell states in unexpected tissue contexts and relate in vivo biology to in vitro models. These require a common measure of cell similarity across the body and an efficient way to search. Here we develop SCimilarity, a metric-learning framework to learn a unified and interpretable representation that enables rapid queries of tens of millions of cell profiles from diverse studies for cells that are transcriptionally similar to an input cell profile or state. We use SCimilarity to query a 23.4-million-cell atlas of 412 single-cell RNA-sequencing studies for macrophage and fibroblast profiles from interstitial lung disease¹ and reveal similar cell profiles across other fibrotic diseases and tissues. The top scoring in vitro hit for the macrophage query was a 3D hydrogel system², which we experimentally demonstrated reproduces this cell state. SCimilarity serves as a foundation model for single-cell profiles that enables researchers to query for similar cellular states across the human body, providing a powerful tool for generating biological insights from the Human Cell Atlas.


Fig. 1 | Geographical distribution of locations of members of the Human Cell Atlas. Geographical distribution of locations of members of the Human Cell Atlas, most of whom are scientists engaged in HCA activities or have expressed an interest to participate in the HCA. (see https://www.humancellatlas.org/learnmore/hca-metrics/ for details).
Meetings, roadshows and workshops organized by the Human Cell Atlas for researchers in various global regions
The commitment of the human cell atlas to humanity

November 2024

·

151 Reads

·

6 Citations

The Human Cell Atlas (HCA) is a global partnership "to create comprehensive reference maps of all human cells-the fundamental units of life - as a basis for both understanding human health and diagnosing, monitoring, and treating disease." ( https://www.humancellatlas.org/ ) The atlas shall characterize cells from diverse individuals across the globe to better understand human biology. HCA proactively considers the priorities of, and benefits accrued to, contributing communities. Here, we lay out principles and action items that have been adopted to affirm HCA's commitment to equity so that the atlas is beneficial to all of humanity.


Citations (82)


... Significant differences in cell type abundances between the distinct clinical groups at the different timepoints were detected by using a binomial generalized linear model (GLM) 83 . A model taking into account Cluster and Condition was fitted, i.e.,~Cluster*Condition, by using Lme4 version 1.1-27.1. ...

Reference:

Durable response to CAR T is associated with elevated activation and clonotypic expansion of the cytotoxic native T cell repertoire
A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients

Genome Biology

... With the myriad of possible applications of scRNA-seq technologies in precision medicine, 39 scRNA-seq reference atlases should be diverse from their inception to maximize the global benefit to all populations. 90 Limitations of the study One caveat for our analyses is that human demographics such as age, sex, self-reported ethnicity, and genetic ancestry can be confounded by correlated sociocultural, environmental (e.g., exposure to infectious agents), and lifestyle (e.g., diet) factors, all of which can contribute to phenotypic variation. Furthermore, technical variation across study sites can introduce biases that are challenging to disentangle from biological variation between population groups. ...

The commitment of the human cell atlas to humanity

... Embeddings of single-cell profiles are now routinely used as a research tool in biological investigation to characterize cell types and states, their changes over time and their distinction between conditions, including diseases, organs or drug treatments 1,2 . With a dramatic growth in single-cell data, including the Human Cell Atlas 3,4 , multiple efforts have focused on learning universal embeddings for diverse single-cell data, with different integration methods or foundation models [5][6][7][8][9][10] . Given their broad use, it is crucial to scrutinize the quality of embeddings to evaluate the performance of the underlying integration methods [11][12][13] and zero-shot capabilities of the resulting foundation models 14,15 . ...

A cell atlas foundation model for scalable search of similar human cells

Nature

... app/ publi catio ns/ htapp_ mbc_ klugh ammer_ 2023? tab= overv iew) [39]. Raw sequencing data are available on dbGAP (accession number: phs002371, https:// www. ...

A multi-modal single-cell and spatial expression map of metastatic breast cancer biopsies across clinicopathological features

Nature Medicine

... In melanoma, axonogenesis and the sensitivity of nociceptor sensory neurons are both enhanced.Nociceptor-produced neuropeptide calcitonin gene-related peptide (CGRP) exacerbates the exhaustion of CD8 + T cells via the CGRP/RAMP1 pathway and attenuates anti-tumor immunity against melanoma 6 . Conversely, the CGRP/RAMP3 axis reportedly promotes IFN-γ-producing TH1 and CD8 + T cell responses by activating cAMP signaling133 . It would be intriguing to explore whether the cAMP signaling elicited by sensory neurotransmitters could induce ChAT expression in T cells. ...

Neuropeptide signalling orchestrates T cell differentiation

Nature

... The growing number of biobanks alike illustrates its value in advancing precision therapy for GBM, especially with the rapid rise and availability of multi-omics capabilities and wealth of data that require rigorous validation using reliable preclinical models and well-curated molecular data. 14,15,[17][18][19][20][78][79][80] Here, we tapped into Glioportal to identify Health Research awarded to S.Y. The organizations that provided the grants had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. ...

Glioblastoma Cortical Organoids Recapitulate Cell-State Heterogeneity and Intercellular Transfer

... Traditional methods, using bulk tissue or broad populations of sorted cells, fail to fully capture the intricate, often highly cell type specific, molecular changes associated with these diseases. Recent advances in single-cell expression profiling address these limitations and have facilitated the generation of larger datasets, most notably for AD where the combined data now nears 1,000 cases 3,[8][9][10][11] . However, single cell resolution datasets for other diseases are considerably smaller. ...

Cellular communities reveal trajectories of brain ageing and Alzheimer’s disease

Nature

... To evaluate the levels of Asn metabolism proteins present in CD4 + T cells, we used a publicly available bulk RNA-seq dataset consisting of naive CD4 + T cells differentiated in vitro under T helper 1 (TH1), non-pathogenic T helper 17 (npTH17), and pathogenic T helper 17 (pTH17) polarizing conditions for 1, 6, 12, 20 or 48 hours (Figure 2B-C). 19 These data suggest that the Asn synthesizing enzyme, asparagine synthase (Asns), increases in expression upon activation under all polarizing conditions, whereas the asparagine catabolizing enzyme, Asrgl1, expression demonstrates little change upon activation. We validated these findings in CD4 + T cells activated under non polarizing conditions (Figure 2D-E) using quantitative PCR (qPCR). ...

BACH2 regulates diversification of regulatory and proinflammatory chromatin states in TH17 cells

Nature Immunology

... Chemokines classically related to IFN response such as CXCL9, CXCL10, CXCL11 and IFN are also more abundant post-ICB 27 . CXCL16, which was one of the three pre-ICB proteins, was recently shown to play important roles in tumor immunity and resistance 28,29 . Strikingly, the canonical immune-related proteins that were most uniformly increased in abundance post-ICB in both responders and non-responders were T cell-related proteins such as GZMA, GZMB, CXCL13 and PDCD1. ...

Pan-cancer mapping of single CD8+ T cell profiles reveals a TCF1:CXCR6 axis regulating CD28 co-stimulation and anti-tumor immunity

Cell Reports Medicine

... Interestingly, a recent study of mosaic, human iPSC-derived brain organoids encountered related challenges, with EB-derived, multi-donor human brain cortical organoids exhibiting extensive skewing 56 . The authors solved this by initiating brain organoid derivation on single-donor-derived stem cell aggregates, but then disaggregating and pooling these several weeks into the protocol. ...

Brain Chimeroids reveal individual susceptibility to neurotoxic triggers

Nature