Spatial Analysis of Tumor Microenvironment in Colon Cancer for Targeted Immunotherapy: Insights from Knowledge Graph and Tumor-Infiltrating Lymphocytes (TILs)
The figure shown below provides a comprehensive overview of the tumor microenvironment (TME) in colon cancer, highlighting the spatial expressions of various markers, the spatial distribution and co-expression of IL7R, MS4A1, and CXCL13, and a knowledge graph of key players in the TME. The image is divided into several sections:
Spatial Expressions of Markers: The top left section shows the spatial expressions of TME, T cells, B cells, TAM (tumor-associated macrophages), and pro-tumor markers in human colon cancer using the Xenium platform. The markers include tumor progression markers (e.g., PDPN, CA1), microenvironment markers (e.g., AQP8, TNC), TME markers (e.g., RGS5, MS4A1), T-cells and B-cells markers (e.g., CD3D, CD19), and TAM markers (e.g., CD68, CXCL13).
Spatial Distribution and Co-Expression: The bottom left section illustrates the spatial distribution and co-expression of IL7R, MS4A1, and CXCL13 in the TME. This includes detailed images showing the localization of these markers within the tissue.
Knowledge Graph of TME Key Players: The top right section presents a knowledge graph of key players in the TME of colon cancer. This graph categorizes various cell types and markers into groups such as angiogenic cell types, pro-angiogenic cells, anti-inflammatory cells, and more, showing their interactions and roles within the TME.
Segmented TILs: The bottom right section shows tumor-infiltrating lymphocytes (TILs) in the TME, with images of overlaid and segmented TILs, highlighting their distribution and density within the tissue.
This figure is significant for targeted immunotherapy as it provides detailed spatial and interaction data of various immune and tumor cells within the TME. Understanding these spatial relationships and marker expressions can help in identifying potential targets for immunotherapy, leading to more effective and personalized cancer treatments
Understanding the Tumor Microenvironment in Colorectal Cancer: A Knowledge Graph Approach
The bottom figures (shown avove) provide detailed information on specific gene-cell relationships within the TME of colon cancer. Each sub-figure focuses on a particular cell type and its associated genes. For instance, the first sub-figure shows the relationship between the enteroendocrine cell of the colon and genes like AGTR1, ANK2, and ABCC8. Similarly, other sub-figures highlight the relationships between tuft cells, stem cells, absorptive cells, progenitor cells, paneth cells, and their respective genes. These detailed mappings are essential for identifying potential biomarkers and therapeutic targets specific to different cell types within the TME, thereby aiding in the development of more precise and effective treatments for colon cancer.
For example, the predicted clustering of tuft cells (AFAP1L2 marker) and stem cells (ALDH1B1 marker), captured expression here on H&E image (xenium) with their corresponding marker genes and in the knowledge graph suggests a close interaction or shared pathways within the Tumor Microenvironment (TME) of colorectal cancer (CRC). This clustering can indicate important biological events such as cellular differentiation, immune response, or tumor progression. For example, tuft cells are known to play a role in sensing the environment and initiating immune responses, while stem cells are crucial for tissue regeneration and repair. Their close proximity in the graph may reflect their collaborative roles in maintaining the TME and responding to tumor-related changes.
Understanding these clusters can provide insights into the disease biology of CRC and help design targeted immunotherapy strategies. For instance, targeting the interactions between tuft cells and stem cells could potentially disrupt tumor growth and improve therapeutic outcomes. Additionally, integrating spatial omics data like Xenium can further enhance our understanding of these interactions and their spatial context within the TME, providing a more comprehensive view of the tumor landscape and aiding in the development of more effective treatments.
The knowledge graph (KG) provides a comprehensive understanding of the cellular interactions and regulatory mechanisms involved in liver injury and repair by visualizing the spatiotemporal dynamics and interactions of cell types, processes, and pathways. This visualization allows us to see how different cell types, such as cholangiocytes, liver progenitor-like cells (LPLCs), hepatocytes, immune cells, fibroblasts interact and contribute to liver repair. The KG highlights key processes like cholestatic injury, repair mechanisms, ductular reaction, and ECM remodeling, as well as important signaling pathways such as TGFβ, NOTCH, WNT, and inflammation pathways. These pathways orchestrate the activation and recruitment of immune cells and fibroblasts, contributing to extracellular matrix (ECM) remodeling. The research illustrated in the provided figures focuses on the cellular and molecular dynamics involved in liver injury, repair, and regeneration. The left panel depicts liver tissue samples over time (Days 0, 8, 17, R2, R7, R21) post-DSS treatment, a chemical inducing injury, showing changes in cellular proliferation as indicated by the marker Mki67. The timeline highlights a progression from injury to regeneration, providing insight into how liver tissue undergoes cycles of damage and repair. The right panel presents a knowledge graph (built from the data taken from this study PMID: 38627596) linking various biological entities and pathways.
Knowledge graphs offer significant benefits in understanding complex biological systems by systematically mapping the relationships among various entities involved in liver injury and repair. By visually structuring these interactions, knowledge graphs help us discern critical patterns and dependencies, enabling a comprehensive view of liver pathophysiology and the interplay of factors like inflammation, cellular regeneration, and ECM remodeling. Leveraging advanced tools like GraphRAG and Generative AI enhances this process by automating the integration of vast biomedical data, revealing latent connections that may be missed through traditional analysis. These insights can accelerate drug discovery by identifying novel therapeutic targets for conditions like Metabolic-Associated Steatohepatitis (MASH) and liver fibrosis, providing a robust framework for developing targeted therapies to halt or reverse liver damage. This model of building knowledge graphs in GenAI is currently under development for further analysis to understand liver injury and repair. It utilizes multimodal data, including spatial transcriptomics imaging and single-cell scRNA-seq data, to provide comprehensive insights into the cellular interactions and regulatory mechanisms involved in these processes
Mapping Mid-Lobular Gene Networks: Unraveling Liver Fibrosis Pathways with GenAI and Multimodal Data Integration
The knowledge graph illustrates the intricate relationships among genes associated with mid-lobular liver zonation, utilizing a combination of data from histopathology spatial transcriptomics (visium), spatial omics (CosMx), and single-cell RNA sequencing (scRNA-seq). Liver zonation refers to the functional specialization within different zones of liver lobules, influenced by gradients of oxygen, nutrients, and other signals. This zonation is essential for understanding how specific cellular responses vary across these regions under normal and disease conditions, particularly in the context of liver fibrosis.
In this network, genes such as VWF, TIE1, KDR, and APOC1 represent critical factors that play roles in endothelial function, immune response, and metabolism within liver lobules. The interconnections among these genes suggest a complex network of gene-gene interactions and co-expression patterns, highlighting how expression changes dynamically across liver zones in response to damage and fibrotic processes. These relationships are especially relevant because liver fibrosis often initiates in mid-lobular regions, where cells are subject to unique environmental conditions. Identifying these patterns provides insight into potential biomarkers of fibrosis severity and progression, as well as a foundation for targeted interventions.
The knowledge graph emphasizes pathways that could serve as therapeutic targets for fibrosis. Genes involved in angiogenesis (such as TIE1 and KDR) and immune response genes (such as C8A and C9) are particularly relevant. By targeting specific pathways in these networks, it may be possible to modulate the fibrotic process more effectively. Drugs designed to influence these pathways, or key regulatory genes could potentially slow or reverse fibrosis.
The integration of multimodal data from histopathology, spatial omics, and scRNA-seq allows for a comprehensive view of gene expression changes in distinct liver zones and cell types. This layered approach provides a detailed understanding of how fibrosis develops on a cellular level, thus aiding the identification of specific cell types or spatial regions within the liver that are most responsive to fibrotic stimuli.
GenAI (generative AI) plays a crucial role in analyzing this multimodal data, allowing for pattern recognition across vast datasets, hypothesis generation, and the suggestion of drug targets based on detailed molecular profiles. The application of GenAI accelerates the research process by automating data interpretation and identifying potential therapeutic targets, ultimately streamlining drug discovery for liver fibrosis and similar complex diseases. This approach not only facilitates a deeper understanding of disease mechanisms but also expedites the development of targeted treatments tailored to the specific pathology of liver fibrosis.
Leveraging Knowledge Graphs for Gene-LSEC-Disease Interactions in Liver Zone 2: Advancing GenAI Therapeutic Strategies
A knowledge graph specifically designed for the liver’s midlobular zone 2 offers a comprehensive framework to elucidate the complex interactions between zone-specific genes, liver sinusoidal endothelial cells (LSECs), and disease phenotypes. By mapping these relationships, it enhances our understanding of the molecular pathways that govern LSEC responses towards fibrosis or regeneration. This integrated approach facilitates the identification of key regulatory targets and mechanisms underlying liver pathology. Additionally, the knowledge graph serves as a robust foundation for GenAI-based therapeutic strategies, enabling the prediction and validation of novel interventions. The model is further under development to incorporate multimodal data, including spatial omics, single-cell RNA sequencing (scRNA-seq), multiomics, histopathology, and clinical data. Ultimately, this tool accelerates the discovery and development of targeted treatments for liver diseases by providing actionable insights into cellular and genetic drivers.
Leveraging Graph Neural Networks for Predicting Cell Types in Colorectal Cancer: A Spatial Transcriptomics Approach
The image presents below a study on modeling colorectal cancer (CRC) using a Graph Neural Network (GNN) to predict cell types, leveraging the Squidpy Visium CRC dataset. The top row shows histological images of CRC tissue sections labeled as "spatial1" and "spatial2," with different regions highlighted. Below these images are network graphs representing cell type interactions within these spatial contexts. The cell types include CK low HR low tumor cells, apoptotic tumor cells, macrophages, T cells, proliferative tumor cells, and small elongated stromal cells. The bottom section of the image shows additional histological images with different cell type annotations labeled as "ID1" and "SPP1."
Spatial Context: GNNs can capture the spatial relationships between different cell types within the tumor microenvironment, providing a more comprehensive understanding of cell interactions.
Predictive Accuracy: By modeling the complex interactions between cells, GNNs can improve the accuracy of cell type predictions, which is crucial for identifying specific targets for immunotherapy.
Personalized Treatment: This approach can help in identifying unique cellular compositions and interactions in individual patients, leading to more personalized and effective immunotherapy strategies.
Data Integration: Squidpy Visium CRC data integrates spatial transcriptomics with histological data, allowing for a multi-dimensional analysis that enhances the understanding of tumor biology and the immune landscape.
This approach demonstrates the application of advanced computational techniques like GNNs in cancer research, highlighting the potential for improved diagnostic and therapeutic strategies in colorectal cancer.
Modeling Cell Types Using Graph Neural Networks and Spatial Transcriptomics
The top row of the image shows tissue from a mouse brain, as indicated by the Squidpy Visium Fluorescent dataset. The bottom row features network graphs modeling cell types using Graph Neural Networks (GNN) at different neighbor variables (n_neighs=40, 20, 10).
Possible Cell Types in Mouse Brain:
T Cells: Present in the brain, especially in the context of neuroinflammation.
Macrophages: Microglia are the resident macrophages of the brain.
Endothelial Cells: Form the blood-brain barrier and are present in the brain vasculature.
Less Likely Cell Types in Mouse Brain:
Tumor Cells: Typically associated with cancerous tissues, not normal brain tissue.
Stromal Cells: More common in connective tissues and tumors, not typically found in the brain.
While some cell types like T cells, macrophages (microglia), and endothelial cells are indeed present in the mouse brain, others like tumor cells and stromal cells are more commonly associated with tumor environments. Therefore, the presence of these cell types in the bottom row suggests a pathological condition, such as a brain tumor, rather than normal brain tissue.