Zexian Zeng’s research while affiliated with Peking University 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 (71)


Integrated computational analysis identifies therapeutic targets with dual action in cancer cells and T cells
  • Article

February 2025

·

19 Reads

·

2 Citations

Immunity

·

Rui Zhang

·

Rui Guo

·

[...]

·

Zexian Zeng

Figure 1. Silencing of ICAM-1 confers resistance to CTL and NK cell-mediated killing (A) The mRNA expression level of MHC and co-stimulatory molecules among 1,086 human cancer cell lines from the CCLE database. (B) Pan-cancer analysis of ICAM-1 expression level on different cancer types from TCGA database. ICAM-1 median expression value is show in red line. (C and D) FACS analysis of surface ICAM-1 level on indicated human (C) and murine (D) cancer cell lines. Murine cells were either untreated or treated with IFN-g (50 ng/mL) for 24 hours. (E) In vitro competition assay of tumor and CTL co-culture. Control (sgControl) SW480 cells were either mixed with tdTomato-labeled control (sgControl) cells or ICAM-1 KO cells. These mixture cells were then co-cultured with NY-ESO-1-specific T cells or control T cells without the expression of TCR against NY-ESO-1. Log 2 fold changes of the percentage of mixture SW480 cells upon co-culture with NY-ESO-1-specific CTLs as compared with that co-cultured with control T cells were shown (n = 3).
Figure 2. Tumor-intrinsic ICAM-1 is critical for immune evasion for both MHC-I-sufficient and deficient tumors (A and B) Vector-transduced or Icam1 OE B16F10 and 4T1 tumors were inoculated in wild-type mice (A) and NSG mice (B), respectively. Tumor growth curves were recorded and shown. n = 5-6 mice per group. (C) Control (sgControl) and Icam1 KO (sgIcam1) 4T1 tumors were inoculated in the wild-type and NSG mice, respectively. Tumor growth curves were recorded and shown. n = 4-5 mice per group. (D) Summary of FACS analysis comparing the number of indicated tumor-infiltrating immune cells between control and Icam1 KO 4T1 tumors on day 16 after tumor inoculation (n = 5-6). (E) B2m/Icam1 double KO (sgB2m + sgIcam1) or B2m single KO (sgB2m + sgControl) 4T1 tumors were inoculated in the wild-type and NSG mice, respectively. Tumor growth curves were recorded and shown. n = 4-6 mice per group. (F) Summary of FACS analysis comparing the number of indicated tumor-infiltrating immune cells between B2m/Icam1 double KO and B2m single KO 4T1 tumors on day 15 after tumor inoculation (n = 6). Data are presented as means ± SEM (A-F). *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 by two-way ANOVA (A-C and E) and one-way ANOVA (D and F). ns, not significant. Data are representative of at least two independent experiments (A-F).
Figure 3. ICAM-1 is co-expressed with a wide range of pro-inflammatory genes and is epigenetically regulated in tumor cells (A) Workflow of CRISPR screen to identify regulators of ICAM-1 expression. Cas9-expressing A549 cells were transduced with a genome-wide sgRNA library. CRISPR-edited A549 cells were then sorted into ICAM-1 high and ICAM-1 low fractions, followed by genomic DNA extraction and sequencing to determine the sgRNA abundance. (B) Volcano plot showing the log 2 fold change and p values of ICAM-1 regulators identified from CRISPR screen. The left graph shows the depleted hits (KO of the gene reduced ICAM-1 expression) and the right graph shows the enriched hits (KO of the gene enhanced ICAM-1 expression). Annotated genes represent the NFkB pathway (blue) and epigenetic regulators (red). (C) Log 2 fold change of sgRNAs against indicated genes in ICAM-1 high A549 cells as compared with control. Depleted sgRNA (KO leads to reduced ICAM-1) and enriched sgRNAs (KO leads to enhanced ICAM-1) are labeled in blue and red bars, respectively. The control sgRNAs are indicated by gray bars. (D) Gene ontology (GO) analysis in top 100 enriched hits from ICAM-1 high A549 cells of CRISPR screen. (E) FACS analysis of ICAM-1 level on A549-Cas9 cells expressing control sgRNA or sgRNAs targeting UHRF1, DNMT1, EED, BPTF, and STAG2. The same control sample was used for all comparisons shown in the panel. Data are representative of two independent experiments (E).
Figure 4. UHRF1-DNMT1-mediated methylation is a major ICAM-1 silencing mechanism in cancer cells (A) Illustration of functional domains in UHRF1. The indicated point mutations abolish the corresponding functions of the domains. (B) Western blot analysis of UHRF1 protein level in control and UHRF1 KO A549 cells expressing indicated UHRF1 mutants. (C) Mean fluorescence intensity (MFI) of surface ICAM-1 level determined by flow cytometry in cells expressing indicated UHRF1 mutants (n = 3). (D) RNA-seq and WGBS profiles of ICAM1 in UHRF1 KO and control A549 cells. CpG region is shaded in blue. One of representative biological replicates is shown for each sample. (E) Bisulfite sequencing of the ICAM1 CpG region in control (left) and UHRF1 KO (right) A549 cells. Each line represents a single clone (n = 20). Methylated CpG sites are shown in black circles and unmethylated sites in blank circles. The percentages of overall methylated CpGs are indicated. (F) Pearson's correlation of tumor ICAM-1 expression and ICAM1 promoter methylation score from the CCLE database. (G and H) Control or UHRF1 KO A549 cells co-cultured either with NY-ESO-1-specific CTLs (G) or NK-92MI cells (H) in the presence of isotype (mouse IgG1 kappa antibodies) or anti-ICAM-1-blocking antibodies (5 mg/mL). Specific lysis percentage was determined by FACS, counting the number of alive cells after co-culture with NY-ESO-1-specific CTLs or NK-92MI cells, as compared with control group (n = 3). Data are presented as means ± SEM (C and G and H). *p < 0.05 and ****p < 0.0001 by one-way ANOVA (C) and two-way ANOVA (G and H). ns, not significant. Data are representative of at least two independent experiments (B, C, G, and H).
Figure 5. Reconstitution of ICAM-1/LFA-1 signaling through fusion protein Cet3ICAM1-D1 (A) Schematic structure of Cet3ICAM1-D1 fusion protein (left) and working hypothesis (right). Cet3ICAM1-D1 is composed of Fab fragment of cetuximab and murine natural D1 domain of ICAM-1, fused to a ''LALA-PG'' human Fc fragment. Working hypothesis: in the absence of ICAM-1, the fusion protein could interact and activate with LFA-1 signaling through the ICAM-1 D1 domain. (B) Binding affinity of cetuximab and Cet3ICAM1-D1 to EGFR in MC38 cells (n = 3). (C and D) OT-1 T cells were co-cultured with MC38 (C) and B16F10 (D) tumor cells with serial dilutions of Cet3ICAM1-D1 or cetuximab. FACS analysis showing the percentage of intracellular IFN-g-producing OT-I T cells (n = 3). (E) OT-1 cells were co-cultured with SIINFEKL-pulsed or unpulsed MC38 tumor cells in the presence of 10 nM Cet3ICAM1-D1. FACS analysis showing the percentage of intracellular IFN-g-producing OT-I T cells (n = 3).
Potentiating anti-tumor immunity by re-engaging immune synapse molecules
  • Article
  • Full-text available

February 2025

·

1 Read

Cell Reports Medicine

The formation of immune synapses (ISs) between cytotoxic T cells and tumor cells is crucial for effective tumor elimination. However, the role of ISs in immune evasion and resistance to immune checkpoint blockades (ICBs) remains unclear. We demonstrate that ICAM-1, a key IS molecule activating LFA-1 signaling in T and natural killer (NK) cells, is often expressed at low levels in cancers. The absence of ICAM-1 leads to significant resistance to T and NK cell-mediated anti-tumor immunity. Using a CRISPR screen, we show that ICAM-1 is epigenetically regulated by the DNA methylation pathway involving UHRF1 and DNMT1. Furthermore, we engineer an antibody-based therapeutic agent, “LFA-1 engager,” to enhance T cell-mediated anti-tumor immunity by reconstituting LFA-1 signaling. Treatment with LFA-1 engagers substantially enhances immune-mediated cytotoxicity, potentiates anti-tumor immunity, and synergizes with ICB in mouse models of ICAM-1-deficient tumors. Our data provide promising therapeutic strategies for re-engaging immune stimulatory signals in cancer immunotherapy.

Download

Fig.1 Benchmarking gene detection sensitivity across spatial transcriptomics platforms a. Schematic overview of sample collection, processing, and data generation. Human tumour samples were divided into three sections: one was formalin-fixed and paraffinembedded (FFPE) for Visium HD FFPE, CosMx 6K, and Xenium 5K profiling; another was embedded in Optimal Cutting Temperature (OCT) for Stereo-seq v1.3 and Visium HD FF profiling; and the remaining tissue was dissociated for single-cell RNA sequencing (scRNA-seq). The spatial distributions of 16 proteins on adjacent sections were profiled with CODEX. b. H&E staining and spatial distribution of EPCAM transcripts in COAD sections. Color intensity represents the transcript counts within 8 × 8 μm bins. c. Average transcript counts for lineage marker genes across whole sections, calculated at the resolution of 8 μm. d. Pearson correlation of gene expression levels between different ST data and scRNA-seq data. For each gene, the total transcript counts across three cancer types were averaged and log10 transformed. The diagonal red line indicates a slope of 1, and color intensity corresponds to relative gene counts. R denotes the correlation coefficient, and n indicates the number of genes included in the analysis. e. Log2-transformed total transcript counts of each gene within the ten selected regions (400 × 400 μm) with similar morphology in HCC and OV.
Fig.2 Evaluation of false positives and transcript-protein correlation across spatial transcriptomics platforms a. Spatial distribution of detected negative controls and genes in shared regions between CosMx 6K and Xenium 5K data from COAD sections. Color intensity represents the normalized number of calls in each 8 × 8 μm bin. b. Number of calls and Moran's I for common genes (n = 2552), platform-specific genes (3623 for CosMx 6K and 2449 for Xenium 5K), negative control sequences (NegProbe, 20 for CosMx 6K and 40 for Xenium 5K), and negative control codes (NegCode, 324 for CosMx 6K and 609 for Xenium 5K) detected by CosMx 6K and Xenium 5K in COAD. c. Percentage of negative control signals for CosMx 6K and Xenium 5K across three cancer types. d. H&E staining and spatial distribution of transcripts detected within and outside the
Fig.3 Comparison of cell segmentation a. Cell boundaries generated using automatic cell segmentation algorithms implemented in each ST platform (left) and manual cell segmentation by human annotators (middle). The merged results are shown (right), with white polygons denoting automatic annotations and blue masks indicating manual annotations. b. Number of cells annotated by automatic cell segmentation algorithms and human annotators within 125 100 × 100 μm bins. c-d. Log2-transformed transcript and gene counts of each manually segmented cell for all detected genes (c) or common genes (d) detected by the two iST platforms. e. Density plot of CD3E/CD68 and EPCAM expressions in COAD. Only cells containing at least one transcript of either CD3E/CD68 or EPCAM were included. Color intensity indicates the number of single cells. f. Expression correlation of marker genes expected to be exclusively expressed in different major lineages (n = 36 gene pairs).
Fig.4 Comparative analysis of cell type annotations, immune cell detection, and spatial alignment with adjacent CODEX a. UMAP representation of ST and scRNA-seq data from COAD. Each point represents a single cell for scRNA-seq, Stereo-seq v1.3, CosMx 6K, and Xenium 5K data. For Visium HD FFPE, each point corresponds to an 8 × 8 μm bin. Distinct colors denote different clusters. b. Average silhouette width (ASW) of clustering results across platforms, with higher scores indicating better clustering quality. c. Proportion of cells consistently annotated as the same cell type by multiple automatic annotation tools. Colors represent the number of tools that have consistent annotations. d. Comparison of cell type annotations derived from ST data with those from adjacent CODEX. e. Correlation of cell counts between ST data and adjacent CODEX for each cell type over spatial grids. Pearson correlation coefficients are shown. Error bars represent SEM for correlations obtained under different grid sizes. f. Spatial distribution of major cell types within regions of high lymphocyte infiltration (500 × 500 μm). The H&E staining, cell type annotations of ST and CODEX data, and protein staining for CD8, CD4, and CD20 are shown together.
Fig.5 Comparison of spatial clustering and cell distributions a. Spatial clustering of ST and CODEX data from COAD sections, with distinct colors denoting different spatial clusters. b. Correlation of cluster proportions between ST and CODEX data across 100 × 100 μm spatial grids. Pearson correlation coefficients are reported. c. H&E staining and localization of malignant cells within the tumour core and at the tumour boundary. d. Spatial distribution of tumour-infiltrating CD8+ T cells and peripheral CD8+ T cells within the COAD sections.
Systematic Benchmarking of High-Throughput Subcellular Spatial Transcriptomics Platforms

December 2024

·

256 Reads

·

1 Citation

Recent advancements in spatial transcriptomics technologies have significantly enhanced resolution and throughput, underscoring an urgent need for systematic benchmarking. To address this, we collected clinical samples from three cancer types - colon adenocarcinoma, hepatocellular carcinoma, and ovarian cancer - and generated serial tissue sections for systematic evaluation. Using these uniformly processed samples, we generated spatial transcriptomics data across five high-throughput platforms with subcellular resolution: Stereo-seq v1.3, Visium HD FFPE, Visium HD FF, CosMx 6K, and Xenium 5K. To establish ground truth datasets, we profiled proteins from adjacent tissue sections corresponding to all five platforms using CODEX and performed single-cell RNA sequencing on the same samples. Leveraging manual cell segmentation and detailed annotations, we systematically assessed each platform's performance across key metrics, including capture sensitivity, specificity, diffusion control, cell segmentation, cell annotation, spatial clustering, and transcript-protein alignment with adjacent CODEX. The uniformly generated, processed, and annotated multi-omics dataset is valuable for advancing computational method development and biological discoveries. The dataset is accessible via SPATCH, a user-friendly web server for visualization and download (http://spatch.pku-genomics.org/).


Figure 4. The immune infiltration and association with immunotherapy response of GRB7 in OC. (A) Immune cell enrichment in low and high expression levels of GRB7 in OC from CIBERSORT. (B) GRB7's expression levels in responders and non-responders of ICB in syngeneic mouse models.
Figure 5. GRB7's expression among different cell types and datasets. The cohorts highlighted in red are the ovarian cancer single-cell datasets.
Figure 6. Knockout GRB7 inhibits the proliferation of OVCAR3. (A) Western blot analysis of GRB7 knockout efficiency. (B) Colony formation capacity of GRB7 knockout and control. (C) CCK-8 assay of GRB7 knockout and control. Data are represented as mean ± standard deviation (SD) (A-C). The Shapiro-Wilk test confirmed normality, and Brown-Forsythe test confirmed homogeneity of variance (B,C). * p < 0.05; ** p < 0.01; *** p < 0.001 by one-way ANOVA (B) and two-way ANOVA (C). Data are representative of three independent experiments (B,C).
Figure 7. GRB7 knockout in OVCAR3 inhibits cell migration and sensitizes killing effect of CD8+ T cells. (A) GRB7 knockout in OVCAR3 reduces migrating cell numbers in transwell assay. (B) GRB7
GRB7 Plays a Vital Role in Promoting the Progression and Mediating Immune Evasion of Ovarian Cancer

August 2024

·

35 Reads

·

1 Citation

Background: Despite breakthroughs in treatment, ovarian cancer (OC) remains one of the most lethal gynecological malignancies, with an increasing age-standardized mortality rate. This underscores an urgent need for novel biomarkers and therapeutic targets. Although growth factor receptor-bound protein 7 (GRB7) is implicated in cell signaling and tumorigenesis, its expression pattern and clinical implications in OC remain poorly characterized. Methods: To systematically investigate GRB7’s expression in OC, our study utilized extensive datasets from TCGA, GTEx, CCLE, and GEO. The prognostic significance of GRB7 was evaluated by means of Kaplan–Meier and Cox regression analyses. Using a correlation analysis and gene set enrichment analysis, relationships between GRB7’s expression and gene networks, immune cell infiltration and immunotherapy response were investigated. In vitro experiments were conducted to confirm GRB7’s function in the biology of OC. Results: Compared to normal tissues, OC tissues exhibited a substantial upregulation of GRB7. Reduced overall survival, disease-specific survival, and disease-free interval were all connected with high GRB7 mRNA levels. The network study demonstrated that GRB7 is involved in pathways relevant to the course of OC and has a positive connection with several key driver genes. Notably, GRB7’s expression was linked to the infiltration of M2 macrophage and altered response to immunotherapy. Data from single-cell RNA sequencing data across multiple cancer types indicated GRB7’s predominant expression in malignant cells. Moreover, OC cells with GRB7 deletion showed decreased proliferation and migration, as well as increased susceptibility to T cell-mediated cytotoxicity. Conclusion: With respect to OC, our results validated GRB7 as a viable prognostic biomarker and a promising therapeutic target, providing information about its function in tumorigenesis and immune modulation. GRB7’s preferential expression in malignant cells highlights its significance in the biology of cancer and bolsters the possibility that it could be useful in enhancing the effectiveness of immunotherapy.


Challenge design. ICI immune checkpoint inhibitor, PD-L1 programmed death ligand 1, TCR T-cell receptor, TMB tumor mutational burden
Prediction of PFS with submitted models. A Bootstrapped estimates of model performance in CheckMate 026 (boxes are bound by the 25th and 75th percentiles). B Decision tree summarizing the Netphar model. C Netphar performance in the chemotherapy and nivolumab arms of CheckMate 026. D Netphar performance in the chemotherapy and nivolumab + ipilimumab arms of CheckMate 227. BL baseline, C-index concordance index, DSS BM difference in squared scaled basal metrics, PD-L1 programmed death ligand 1, PFS progression-free survival, TMB tumor mutational burden
Prediction of OS with submitted models. A Bootstrapped estimates of model performance in CheckMate 026 (Boxes are bound by the 25th and 75th percentile). B Classification principle of the I-MIRACLE model. C I-MIRACLE performance in the chemotherapy and nivolumab arms of CheckMate 026. D I-MIRACLE performance in the chemotherapy and nivolumab + ipilimumab arms of CheckMate 227. BL baseline, C-index concordance index, DSS BM difference in squared scaled basal metrics, ICR immunologic constant of rejection, OS overall survival, PD-L1 programmed death ligand 1, PFS progression-free survival, TMB tumor mutational burden
Prediction of BOR of PD with submitted models. A Bootstrapped estimates of model performance in CheckMate 026 (boxes are bound by the 25th and 75th percentiles). B Principle of the cSysImmunoOnco model. C cSysImmunoOnco model performance in CheckMate 026 and D CheckMate 227. The grey dotted line is the line of non-determination. AUC area under the curve, BL baseline, BOR best overall response, DSS BM difference in squared scaled basal metrics, EaSIeR estimate systems immune response, ICI immune checkpoint inhibitor, ICR immunologic constant of rejection, MSI microsatellite instability, NSCLC non-small cell lung cancer, OS overall survival, PD progressive disease, PD-L1 programmed death ligand 1, TMB tumor mutational burden
A community challenge to predict clinical outcomes after immune checkpoint blockade in non-small cell lung cancer

February 2024

·

158 Reads

·

7 Citations

Journal of Translational Medicine

Background Predictive biomarkers of immune checkpoint inhibitor (ICI) efficacy are currently lacking for non-small cell lung cancer (NSCLC). Here, we describe the results from the Anti–PD-1 Response Prediction DREAM Challenge, a crowdsourced initiative that enabled the assessment of predictive models by using data from two randomized controlled clinical trials (RCTs) of ICIs in first-line metastatic NSCLC. Methods Participants developed and trained models using public resources. These were evaluated with data from the CheckMate 026 trial (NCT02041533), according to the model-to-data paradigm to maintain patient confidentiality. The generalizability of the models with the best predictive performance was assessed using data from the CheckMate 227 trial (NCT02477826). Both trials were phase III RCTs with a chemotherapy control arm, which supported the differentiation between predictive and prognostic models. Isolated model containers were evaluated using a bespoke strategy that considered the challenges of handling transcriptome data from clinical trials. Results A total of 59 teams participated, with 417 models submitted. Multiple predictive models, as opposed to a prognostic model, were generated for predicting overall survival, progression-free survival, and progressive disease status with ICIs. Variables within the models submitted by participants included tumor mutational burden (TMB), programmed death ligand 1 (PD-L1) expression, and gene-expression–based signatures. The best-performing models showed improved predictive power over reference variables, including TMB or PD-L1. Conclusions This DREAM Challenge is the first successful attempt to use protected phase III clinical data for a crowdsourced effort towards generating predictive models for ICI clinical outcomes and could serve as a blueprint for similar efforts in other tumor types and disease states, setting a benchmark for future studies aiming to identify biomarkers predictive of ICI efficacy. Trial registration: CheckMate 026; NCT02041533, registered January 22, 2014. CheckMate 227; NCT02477826, registered June 23, 2015.


ABHD6 suppresses colorectal cancer progression via AKT signaling pathway

January 2024

·

18 Reads

·

1 Citation

Molecular Carcinogenesis

Colorectal cancer (CRC) continues to be a prevalent malignancy, posing a significant risk to human health. The involvement of alpha/beta hydrolase domain 6 (ABHD6), a serine hydrolase family member, in CRC development was suggested by our analysis of clinical data. However, the role of ABHD6 in CRC remains unclear. This study seeks to elucidate the clinical relevance, biological function, and potential molecular mechanisms of ABHD6 in CRC. We investigated the role of ABHD6 in clinical settings, conducting proliferation, migration, and cell cycle assays. To determine the influence of ABHD6 expression levels on Oxaliplatin sensitivity, we also performed apoptosis assays. RNA sequencing and KEGG analysis were utilized to uncover the potential molecular mechanisms of ABHD6. Furthermore, we validated its expression levels using Western blot and reactive oxygen species (ROS) detection assays. Our results demonstrated that ABHD6 expression in CRC tissues was notably lower compared to adjacent normal tissues. This low expression correlated with a poorer prognosis for CRC patients. Moreover, ABHD6 overexpression impeded CRC cell proliferation and migration while inducing G0/G1 cell cycle arrest. In vivo experiments revealed that downregulation of ABHD6 resulted in an increase in tumor weight and volume. Mechanistically, ABHD6 overexpression inhibited the activation of the AKT signaling pathway and decreased ROS levels in CRC cells, suggesting the role of ABHD6 in CRC progression via the AKT signaling pathway. Our findings demonstrate that ABHD6 functions as a tumor suppressor, primarily by inhibiting the AKT signaling pathway. This role establishes ABHD6 as a promising prognostic biomarker and a potential therapeutic target for CRC patients.


Tumor aerobic glycolysis confers immune evasion through modulating sensitivity to T cell-mediated bystander killing via TNF-α

July 2023

·

97 Reads

·

104 Citations

Cell Metabolism

Metabolic reprogramming toward glycolysis is a hallmark of cancer malignancy. The molecular mechanisms by which the tumor glycolysis pathway promotes immune evasion remain to be elucidated. Here, by performing genome-wide CRISPR screens in murine tumor cells co-cultured with cytotoxic T cells (CTLs), we identified that deficiency of two important glycolysis enzymes, Glut1 (glucose transporter 1) and Gpi1 (glucose-6-phosphate isomerase 1), resulted in enhanced killing of tumor cells by CTLs. Mechanistically, Glut1 inactivation causes metabolic rewiring toward oxidative phosphorylation, which generates an excessive amount of reactive oxygen species (ROS). Accumulated ROS potentiate tumor cell death mediated by tumor necrosis factor alpha (TNF-α) in a caspase-8- and Fadd-dependent manner. Genetic and pharmacological inactivation of Glut1 sensitizes tumors to anti-tumor immunity and synergizes with anti-PD-1 therapy through the TNF-α pathway. The mechanistic interplay between tumor-intrinsic glycolysis and TNF-α-induced killing provides new therapeutic strategies to enhance anti-tumor immunity.


Tutorial: integrative computational analysis of bulk RNA-sequencing data to characterize tumor immunity using RIMA

June 2023

·

150 Reads

·

17 Citations

Nature Protocols

RNA-sequencing (RNA-seq) has become an increasingly cost-effective technique for molecular profiling and immune characterization of tumors. In the past decade, many computational tools have been developed to characterize tumor immunity from gene expression data. However, the analysis of large-scale RNA-seq data requires bioinformatics proficiency, large computational resources and cancer genomics and immunology knowledge. In this tutorial, we provide an overview of computational analysis of bulk RNA-seq data for immune characterization of tumors and introduce commonly used computational tools with relevance to cancer immunology and immunotherapy. These tools have diverse functions such as evaluation of expression signatures, estimation of immune infiltration, inference of the immune repertoire, prediction of immunotherapy response, neoantigen detection and microbiome quantification. We describe the RNA-seq IMmune Analysis (RIMA) pipeline integrating many of these tools to streamline RNA-seq analysis. We also developed a comprehensive and user-friendly guide in the form of a GitBook with text and video demos to assist users in analyzing bulk RNA-seq data for immune characterization at both individual sample and cohort levels by using RIMA.


Transfer learning enables predictions in network biology

May 2023

·

1,868 Reads

·

441 Citations

Nature

Mapping gene networks requires large amounts of transcriptomic data to learn the connections between genes, which impedes discoveries in settings with limited data, including rare diseases and diseases affecting clinically inaccessible tissues. Recently, transfer learning has revolutionized fields such as natural language understanding1,2 and computer vision³ by leveraging deep learning models pretrained on large-scale general datasets that can then be fine-tuned towards a vast array of downstream tasks with limited task-specific data. Here, we developed a context-aware, attention-based deep learning model, Geneformer, pretrained on a large-scale corpus of about 30 million single-cell transcriptomes to enable context-specific predictions in settings with limited data in network biology. During pretraining, Geneformer gained a fundamental understanding of network dynamics, encoding network hierarchy in the attention weights of the model in a completely self-supervised manner. Fine-tuning towards a diverse panel of downstream tasks relevant to chromatin and network dynamics using limited task-specific data demonstrated that Geneformer consistently boosted predictive accuracy. Applied to disease modelling with limited patient data, Geneformer identified candidate therapeutic targets for cardiomyopathy. Overall, Geneformer represents a pretrained deep learning model from which fine-tuning towards a broad range of downstream applications can be pursued to accelerate discovery of key network regulators and candidate therapeutic targets.


Overview of MetaTiME
MetaTiME integrates 1.7 million single cells to learn common transcriptional programs in the tumor microenvironment (TME). a Steps for Meta-components (MeCs) discovery. For each scRNA dataset, the expression matrix of TME cells is decomposed into a loading matrix (red) and an independent component (IC) matrix through independent component analysis (ICA). The ICs represent mutually independent sources of transcriptional variation. ICs from each dataset are concatenated and clustered into groups of ICs with high similarity, representing transcriptional programs shared across TME. MeCs are then calculated as averaged profiles of ICs from each cluster. Each MeC is interpretable, representing gene signatures of cell type, cell states, or signaling pathway activities. b Left: MetaTiME provides 86 functionally annotated MeCs that depict the TME transcriptional landscape. They are grouped into six lineage-related categories and one category reflecting signaling activities, each using a background color corresponding to the lineage. Middle: the MetaTiME annotation tool facilitates automatic annotation of cell states for new tumor scRNA data. Right: candidate regulators of each MeC are prioritized by combining MeC gene weights with epigenetics data. MeC: meta-components, TME: tumor microenvironment, ICA: independent component analysis, MeC: meta-component, TF: transcription factor.
MetaTiME meta-components are biologically interpretable with top genes
a Heatmap of top ten most recurrent clusters of MeCs showing normalized gene weights. b Biological characterization of each MeC with top genes. To facilitate biological interpretation, MeCs are categorized into six lineage-associated classes (B cells, T cells for CD4T, CD8T and NK cells, dendritic cells, monocyte and macrophages, other myeloid cell types, and stroma cells) and one signaling pathway-associated class. c Examples of T cell related MeCs with top 20 genes with largest weights. d Gene contribution of known lineage-related biomarkers for each MeC, and correlation with known immune markers from Azimuth. In the top dot plot, size and color represents log-scaled MeC z-weights of each gene in each MeC. In the bottom dot plot, size and color represents the maximum correlation coefficient between MeC and Azimuth defined marker genes per cell type. MeC meta-component, DC dendritic cell, Mono/Mac monocytes and macrophages. Each MeC and gene is shaded with a background color corresponding to the lineage category. Source data are provided as a Source Data file.
MetaTiME annotates cell states with high resolution on tumor microenvironment single-cell data
a MetaTiME cell state annotation of cell clusters in a basal cell carcinoma scRNA dataset based on top enriched MeCs. b Manual annotation labels by experts from the original study shown on the same UMAP space. c Signature continuums of four MeCs representing the mature dendritic cell state, the CXCL13-secreting exhausted T cell state, the CXCL13-secreting T follicular helper cell state, and the IL1B pathway-activated macrophage state. d Marker gene expression for each annotated cell cluster as in (a). e Bar plot showing cell state composition of tumor microenvironment for tumor scRNA dataset cell states. The proportion of cell states from the same MeC category are aggregated. Source data are provided as a Source Data file.
Differential signature analysis and delineated macrophage states in TME
a Differential MeC signature testing for enriched cell states comparing pre- and post- immunotherapy conditions in basal cell carcinoma (BCC) with two-sided t-test. X-axis: Difference of mean signature scores between post- and pre-immunotherapy conditions. Y-axis: -log(p-value) from two-sided t-test. The significant cluster-wise differential signature is marked as “EnrichedMeC@ClusterName”; when the enriched MeC is the same as cluster name, the signature is marked “ClusterName”. Red dots, differentially increased signature; size of the dots is proportionally to the mean signature score of cells from the post-immunotherapy condition in the cluster. Blue dots, differentially repressed signature; size of the dots is proportionally to the mean signature score of cells from the pre-immunotherapy condition in the cluster. b Differential signature testing for enriched cell states comparing non-responders and responders from pre-treatment condition in bladder carcinoma (BLCA) with two-sided t-test. Red dots, differentially increased signature; size of the dots is proportionally to the mean signature score of cells from the responder condition in the cluster. Blue dots, differentially repressed signature; size of the dots is proportionally to the mean signature score of cells from the non-responder condition in the cluster. c Model of monocytes and macrophage states in tumor and their metabolic differences. d Top pathways enriched in different macrophage MeCs, with adjusted hypergeometric tested p-value from Enrichr. BCC basal cell carcinoma, BLCA bladder carcinoma. Source data are provided as a Source Data file.
MetaTiME prioritizes tumor immunity transcriptional regulators
For selected MeCs, TFs are prioritized by their MeC expression representation and Lisa, ChIP-seq based, regulatory potentials. X-axis: gene z-weight of the TF for the current MeC. Y-axis: Lisa-based regulatory potential significance for top genes in the current MeC. Orange factors: MeC regulators prioritized based on both MeC gene weights and Lisa regulatory potential significance. Green factors: TFs highly weighted in MeCs and not in Lisa analysis. Blue factors: TFs with high Lisa regulatory potential and not highly weighted in MeCs. a TFs prioritized for three MeCs in the signaling category. b TFs prioritized for three MeCs in the dendritic cell category. c TFs prioritized for three MeCs representing different macrophage states. d TFs prioritized for three MeCs representing different T cell states. TF transcription factor, MeC meta-component. Source data are provided as a Source Data file.
MetaTiME integrates single-cell gene expression to characterize the meta-components of the tumor immune microenvironment

May 2023

·

206 Reads

·

12 Citations

Recent advances in single-cell RNA sequencing have shown heterogeneous cell types and gene expression states in the non-cancerous cells in tumors. The integration of multiple scRNA-seq datasets across tumors can indicate common cell types and states in the tumor microenvironment (TME). We develop a data driven framework, MetaTiME, to overcome the limitations in resolution and consistency that result from manual labelling using known gene markers. Using millions of TME single cells, MetaTiME learns meta-components that encode independent components of gene expression observed across cancer types. The meta-components are biologically interpretable as cell types, cell states, and signaling activities. By projecting onto the MetaTiME space, we provide a tool to annotate cell states and signature continuums for TME scRNA-seq data. Leveraging epigenetics data, MetaTiME reveals critical transcriptional regulators for the cell states. Overall, MetaTiME learns data-driven meta-components that depict cellular states and gene regulators for tumor immunity and cancer immunotherapy.


Citations (54)


... Spatially resolved transcriptome analysis is achieved by capturing mRNA in tissue sections using a barcoded solid matrix, enabling unbiased whole transcriptome analysis using poly(dT) oligonucleotides to capture target sequences on spatial barcode arrays with poly(A) tails and correlating transcripts with their spatial locations. 20 10x Genomics recently launched a new technology called Visium, which enables large-scale tissue detection without pre-selecting the region of interest (ROI). It analyses the entire transcriptome from tissue sections by capturing polyadenylated RNA on spatially barcoded microarray slides (Figure 2A). ...

Reference:

Spatial omics strategies for investigating human carotid atherosclerotic disease
Systematic Benchmarking of High-Throughput Subcellular Spatial Transcriptomics Platforms

... The tumor microenvironment (TME) plays a vital role in both tumor progression and the effectiveness of immunotherapy. It was found that the knockout of GRB7 was associated with increased T cell-mediated cytotoxicity, suggesting the possibility of GRB7 being a potential target for immunotherapy [25]. In this study, the relationship between GRB7 expression and immune cell infiltration in KICH, KIRC, and PAAD was examined using the TIMER database. ...

GRB7 Plays a Vital Role in Promoting the Progression and Mediating Immune Evasion of Ovarian Cancer

... The combination of ICR and TMB/proliferation resulted in the top performer in terms of overall survival prediction to ICI, corroborating the importance to assess these parameters simultaneously. 90 In summary, while the phenomenology determining cancer immune responsiveness is increasingly being understood, future challenges remain about how to take advantage of the insights gained to develop better therapeutics. 40 Surrogate markers for transcriptional patterns: artificial intelligence in histopathology Measuring the abundance of RNA transcripts is the gold standard to assess transcriptional patterns in cancer tissue. ...

A community challenge to predict clinical outcomes after immune checkpoint blockade in non-small cell lung cancer

Journal of Translational Medicine

... Several members of the ABHD family have been implicated in the development and progression of malignancies. For example, ABHD2 has been associated with the invasiveness of breast cancer cells (7), ABHD3 was identified in a pro-apoptotic gene screen (8), ABHD6 promotes colorectal cancer progression (9), but the tumor-suppressive effects have also been reported on non-small cell lung cancer (10), and ABHD9 is linked to prostate cancer recurrence (11,12). ...

ABHD6 suppresses colorectal cancer progression via AKT signaling pathway
  • Citing Article
  • January 2024

Molecular Carcinogenesis

... On the one hand, when the level of aerobic glycolysis in cancer cells decreases, mitochondrial oxidative phosphorylation significantly increases, leading to the production of a large amount of ROS. Increased ROS levels result in the downregulation of antiapoptotic proteins [80]. On the other hand, increased aerobic glycolysis directly affects the levels of antiapoptotic and proapoptotic proteins [81]. ...

Tumor aerobic glycolysis confers immune evasion through modulating sensitivity to T cell-mediated bystander killing via TNF-α
  • Citing Article
  • July 2023

Cell Metabolism

... Immune repertoire analysis was performed using the TRUST4 [29] algorithm, implemented in the RNA-seq tumor Immune Analysis (RIMA) pipeline [30]. All TCR metrics shown here are explained in details in the pipeline description (https://liulab-dfci.github.io/RIMA/). ...

Tutorial: integrative computational analysis of bulk RNA-sequencing data to characterize tumor immunity using RIMA
  • Citing Article
  • June 2023

Nature Protocols

... Advances in pretraining and scaling foundation models mean that researchers are now seeking to understand how large unlabeled datasets can be used to initialize models with a general understanding of biology, and initiatives like CELLx-GENE [4] are rising to this data demand. These developments have spurred a variety of proposed foundation models [5][6][7][8][9][10][11][12], all of which pretrain on large cell datasets. ...

Transfer learning enables predictions in network biology

Nature

... com/ yizhang/ MetaT iME). This tool provided each cell with a MeC signature score and identified the enriched MeC status for each cluster, using the top-enriched MeC for each to facilitate uniform manifold approximation and projection (UMAP) visualization [15]. ...

MetaTiME integrates single-cell gene expression to characterize the meta-components of the tumor immune microenvironment

... An algorithm 37 was used to search for regulatory factors that explain the differentially expressed genes (DEGs) specific to the subgroup of identified transcription factors, including E2F1 and TFDP1 ( Supplementary Fig. 6a). These factors form heterodimeric complexes, which negatively impact immune activity 37 and are directly involved in cell cycle progression; thus, E2F1 is a potential dual-action regulatory target in this subgroup. ...

Discovery of Targets for Immune-Metabolic Antitumor Drugs Identifies Estrogen-Related Receptor Alpha

Cancer Discovery

... We subsequently optimized and condensed it into a fixed 20-gene panel, showing prognostic significance in different cancer types (for example, melanoma 10 , bladder cancer 10 , breast cancer 20,21 , neuroblastoma 22 and soft-tissue sarcoma 23 ). The ICR also correlates with response to immunotherapy across multiple cancer types, including breast 24 , melanoma 10 and non-small-cell lung cancer 25 . The ICR signature includes gene modules that reflect the activation of type 1 T (T H 1) cell signaling, expression of CXCR3/CCR5 chemokine ligands, cytotoxicity and counter-activation of immunoregulatory mechanisms 21 (Fig. 1b). ...

A Community Challenge to Predict Clinical Outcomes After Immune Checkpoint Blockade in Non-Small Cell Lung Cancer