- Browse by Subject
Browsing by Subject "Spatial transcriptomics"
Now showing 1 - 10 of 12
Results Per Page
Sort Options
Item 302 Diagnostic Evidence Gauge of Spatial Transcriptomics (DEGAS-ST): Using transfer learning to map clinical data to spatial transcriptomics in prostate cancer(Cambridge University Press, 2024-04-03) Couetil, Justin; Alomari, Ahmed K.; Zhang, Jie; Huang, Kun; Johnson, Travis S.; Medical and Molecular Genetics, School of MedicineOBJECTIVES/GOALS: The 'field effect' is a concept in pathology that pre-malignant tissue changes forecast health. Spatial transcriptomics could detect these changes earlier than histopathology, suggesting new early cancer screening methods. Knowing how normal tissue damage relates to cancer’s origin and progression may improve long-term outcomes. METHODS/STUDY POPULATION: We trained DEGAS, our machine learning framework, with prostate cancer data, combining both general cancer patterns and in-depth genetic information from individual tumors. The Tumor Cancer Genome Atlas (TCGA) shows how gene patterns in tumors relate to patient outcomes, emphasizing the differences between tumors from different patients (intertumor). On the other hand, spatial transcriptomics (ST) shows the genetic variety within a single tumor (intratumor) but has limited samples, making it hard to know which genetic differences are important for treatment. DEGAS bridges these areas by finding tissue sections that resemble those in TCGA profiles and are key indicators of patient survival. DEGAS serves as a valuable tool for generating clinically-important hypotheses. RESULTS/ANTICIPATED RESULTS: DEGAS identified benign-appearing glands in a normal prostate as being highly associated with poor progression-free survival. These glands have transcriptional signatures similar to high-grade prostate cancer. We confirmed this finding in a separate prostate cancer ST dataset. By integrating single cell (SC) data we demonstrated that cells annotated as cancerous in the SC data map to regions of benign glands in the ST dataset. We pinpoint several genes, chiefly Microseminoprotein-β (MSMB, PSP94), where reduced expression is highly correlated with poor progression-free survival. Cell type specific differential expression analysis further revealed that loss of MSMB expression associated with poor outcomes occurs specifically in luminal epithelia, the putative progenitor of prostate cancer. DISCUSSION/SIGNIFICANCE: DEGAS reveals that normal-appearing tissue can be highly-associated with tumor progression and underscores the importance of the 'field effect' in cancer research. Traditional analysis may miss such nuance, hiding key transitional cell states. Validating gene markers could boost early cancer detection and understanding of metastasis.Item Deconvolution Tactics and Normalization in Renal Spatial Transcriptomics(Frontiers Media, 2022-01-13) Ferreira, Ricardo Melo; Freije, Benjamin J.; Eadon, Michael T.; Medicine, School of MedicineThe kidney is composed of heterogeneous groups of epithelial, endothelial, immune, and stromal cells, all in close anatomic proximity. Spatial transcriptomic technologies allow the interrogation of in situ expression signatures in health and disease, overlaid upon a histologic image. However, some spatial gene expression platforms have not yet reached single-cell resolution. As such, deconvolution of spatial transcriptomic spots is important to understand the proportion of cell signature arising from these varied cell types in each spot. This article reviews the various deconvolution strategies discussed in the 2021 Indiana O’Brien Center for Microscopy workshop. The unique features of Seurat transfer score methodology, SPOTlight, Robust Cell Type Decomposition, and BayesSpace are reviewed. The application of normalization and batch effect correction across spatial transcriptomic samples is also discussed.Item Define and visualize pathological architectures of human tissues from spatially resolved transcriptomics using deep learning(Elsevier, 2022-08-24) Chang, Yuzhou; He, Fei; Wang, Juexin; Chen, Shuo; Li, Jingyi; Liu, Jixin; Yu, Yang; Su, Li; Ma, Anjun; Allen, Carter; Lin, Yu; Sun, Shaoli; Liu, Bingqiang; Otero, José Javier; Chung, Dongjun; Fu, Hongjun; Li, Zihai; Xu, Dong; Ma, Qin; Medical and Molecular Genetics, School of MedicineSpatially resolved transcriptomics provides a new way to define spatial contexts and understand the pathogenesis of complex human diseases. Although some computational frameworks can characterize spatial context via various clustering methods, the detailed spatial architectures and functional zonation often cannot be revealed and localized due to the limited capacities of associating spatial information. We present RESEPT, a deep-learning framework for characterizing and visualizing tissue architecture from spatially resolved transcriptomics. Given inputs such as gene expression or RNA velocity, RESEPT learns a three-dimensional embedding with a spatial retained graph neural network from spatial transcriptomics. The embedding is then visualized by mapping into color channels in an RGB image and segmented with a supervised convolutional neural network model. Based on a benchmark of 10x Genomics Visium spatial transcriptomics datasets on the human and mouse cortex, RESEPT infers and visualizes the tissue architecture accurately. It is noteworthy that, for the in-house AD samples, RESEPT can localize cortex layers and cell types based on pre-defined region- or cell-type-enriched genes and furthermore provide critical insights into the identification of amyloid-beta plaques in Alzheimer's disease. Interestingly, in a glioblastoma sample analysis, RESEPT distinguishes tumor-enriched, non-tumor, and regions of neuropil with infiltrating tumor cells in support of clinical and prognostic cancer applications.Item DSTG: deconvoluting spatial transcriptomics data through graph-based artificial intelligence(Oxford University Press, 2021-09-02) Song, Qianqian; Su, Jing; Biostatistics, School of Public HealthRecent development of spatial transcriptomics (ST) is capable of associating spatial information at different spots in the tissue section with RNA abundance of cells within each spot, which is particularly important to understand tissue cytoarchitectures and functions. However, for such ST data, since a spot is usually larger than an individual cell, gene expressions measured at each spot are from a mixture of cells with heterogenous cell types. Therefore, ST data at each spot needs to be disentangled so as to reveal the cell compositions at that spatial spot. In this study, we propose a novel method, named deconvoluting spatial transcriptomics data through graph-based convolutional networks (DSTG), to accurately deconvolute the observed gene expressions at each spot and recover its cell constitutions, thus achieving high-level segmentation and revealing spatial architecture of cellular heterogeneity within tissues. DSTG not only demonstrates superior performance on synthetic spatial data generated from different protocols, but also effectively identifies spatial compositions of cells in mouse cortex layer, hippocampus slice and pancreatic tumor tissues. In conclusion, DSTG accurately uncovers the cell states and subpopulations based on spatial localization. DSTG is available as a ready-to-use open source software (https://github.com/Su-informatics-lab/DSTG) for precise interrogation of spatial organizations and functions in tissues.Item Editorial: Proceedings of the 2021 Indiana O'Brien Center Microscopy Workshop(Frontiers Media, 2022-05-02) Dunn, Kenneth W.; Hall, Andrew M.; Molitoris, Bruce A.; Medicine, School of MedicineItem Identification of Hypoxia-ALCAMhigh Macrophage- Exhausted T Cell Axis in Tumor Microenvironment Remodeling for Immunotherapy Resistance(Wiley, 2024) Xun, Zhenzhen; Zhou, Huanran; Shen, Mingyi; Liu, Yao; Sun, Chengcao; Du, Yanhua; Jiang, Zhou; Yang, Liuqing; Zhang, Qing; Lin, Chunru; Hu, Qingsong; Ye, Youqiong; Han, Leng; Biostatistics and Health Data Science, School of MedicineAlthough hypoxia is known to be associated with immune resistance, the adaptability to hypoxia by different cell populations in the tumor microenvironment and the underlying mechanisms remain elusive. This knowledge gap has hindered the development of therapeutic strategies to overcome tumor immune resistance induced by hypoxia. Here, bulk, single‐cell, and spatial transcriptomics are integrated to characterize hypoxia associated with immune escape during carcinogenesis and reveal a hypoxia‐based intercellular communication hub consisting of malignant cells, ALCAM high macrophages, and exhausted CD8+ T cells around the tumor boundary. A hypoxic microenvironment promotes binding of HIF‐1α complex is demonstrated to the ALCAM promoter therefore increasing its expression in macrophages, and the ALCAM high macrophages co‐localize with exhausted CD8+ T cells in the tumor spatial microenvironment and promote T cell exhaustion. Preclinically, HIF‐1ɑ inhibition reduces ALCAM expression in macrophages and exhausted CD8+ T cells and potentiates T cell antitumor function to enhance immunotherapy efficacy. This study reveals the systematic landscape of hypoxia at single‐cell resolution and spatial architecture and highlights the effect of hypoxia on immunotherapy resistance through the ALCAM high macrophage‐exhausted T cell axis, providing a novel immunotherapeutic strategy to overcome hypoxia‐induced resistance in cancers.Item Integrating single-cell and spatial transcriptomics reveals endoplasmic reticulum stress-related CAF subpopulations associated with chordoma progression(Oxford University Press, 2024) Zhang, Tao-Lan; Xia, Chao; Zheng, Bo-Wen; Hu, Hai-Hong; Jiang, Ling-Xiang; Escobar, David; Zheng, Bo-Yv; Chen, Tian-Dong; Li, Jing; Lv, Guo-Hua; Huang, Wei; Yan, Yi-Guo; Zou, Ming-Xiang; Radiation Oncology, School of MedicineBackground: With cancer-associated fibroblasts (CAFs) as the main cell type, the rich myxoid stromal components in chordoma tissues may likely contribute to its development and progression. Methods: Single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, bulk RNA-seq, and multiplexed quantitative immunofluorescence (QIF) were used to dissect the heterogeneity, spatial distribution, and clinical implication of CAFs in chordoma. Results: We sequenced here 72 097 single cells from 3 primary and 3 recurrent tumor samples, as well as 3 nucleus pulposus samples as controls using scRNA-seq. We identified a unique cluster of CAF in recurrent tumors that highly expressed hypoxic genes and was functionally enriched in endoplasmic reticulum stress (ERS). Pseudotime trajectory and cell communication analyses showed that this ERS-CAF subpopulation originated from normal fibroblasts and widely interacted with tumoral and immune cells. Analyzing the bulk RNA-seq data from 126 patients, we found that the ERS-CAF signature score was associated with the invasion and poor prognosis of chordoma. By integrating the results of scRNA-seq with spatial transcriptomics, we demonstrated the existence of ERS-CAF in chordoma tissues and revealed that this CAF subtype displayed the most proximity to its surrounding tumor cells. In subsequent QIF validation involving 105 additional patients, we confirmed that ERS-CAF was abundant in the chordoma microenvironment and located close to tumor cells. Furthermore, both ERS-CAF density and its distance to tumor cells were correlated with tumor malignant phenotype and adverse patient outcomes. Conclusions: These findings depict the CAF landscape for chordoma and may provide insights into the development of novel treatment approaches.Item Sparse Latent-Space Learning for High-Dimensional Data: Extensions and Applications(2023-05) White, Alexander James; Cao, Sha; Tu, Wanzhu; Zhang, Chi; Zhao, YiThe successful treatment and potential eradication of many complex diseases, such as cancer, begins with elucidating the convoluted mapping of molecular profiles to phenotypical manifestation. Our observed molecular profiles (e.g., genomics, transcriptomics, epigenomics) are often high-dimensional and are collected from patient samples falling into heterogeneous disease subtypes. Interpretable learning from such data calls for sparsity-driven models. This dissertation addresses the high dimensionality, sparsity, and heterogeneity issues when analyzing multiple-omics data, where each method is implemented with a concomitant R package. First, we examine challenges in submatrix identification, which aims to find subgroups of samples that behave similarly across a subset of features. We resolve issues such as two-way sparsity, non-orthogonality, and parameter tuning with an adaptive thresholding procedure on the singular vectors computed via orthogonal iteration. We validate the method with simulation analysis and apply it to an Alzheimer’s disease dataset. The second project focuses on modeling relationships between large, matched datasets. Exploring regressional structures between large data sets can provide insights such as the effect of long-range epigenetic influences on gene expression. We present a high-dimensional version of mixture multivariate regression to detect patient clusters, each with different correlation structures of matched-omics datasets. Results are validated via simulation and applied to matched-omics data sets. In the third project, we introduce a novel approach to modeling spatial transcriptomics (ST) data with a spatially penalized multinomial model of the expression counts. This method solves the low-rank structures of zero-inflated ST data with spatial smoothness constraints. We validate the model using manual cell structure annotations of human brain samples. We then applied this technique to additional ST datasets.Item Spatial Transcriptomic Analysis Reveals Associations between Genes and Cellular Topology in Breast and Prostate Cancers(MDPI, 2022-10-04) Alsaleh, Lujain; Li, Chen; Couetil, Justin L.; Ye, Ze; Huang, Kun; Zhang, Jie; Chen, Chao; Johnson, Travis S.; Biostatistics and Health Data Science, Richard M. Fairbanks School of Public HealthBackground: Cancer is the leading cause of death worldwide with breast and prostate cancer the most common among women and men, respectively. Gene expression and image features are independently prognostic of patient survival; but until the advent of spatial transcriptomics (ST), it was not possible to determine how gene expression of cells was tied to their spatial relationships (i.e., topology). Methods: We identify topology-associated genes (TAGs) that correlate with 700 image topological features (ITFs) in breast and prostate cancer ST samples. Genes and image topological features are independently clustered and correlated with each other. Themes among genes correlated with ITFs are investigated by functional enrichment analysis. Results: Overall, topology-associated genes (TAG) corresponding to extracellular matrix (ECM) and Collagen Type I Trimer gene ontology terms are common to both prostate and breast cancer. In breast cancer specifically, we identify the ZAG-PIP Complex as a TAG. In prostate cancer, we identify distinct TAGs that are enriched for GI dysmotility and the IgA immunoglobulin complex. We identified TAGs in every ST slide regardless of cancer type. Conclusions: These TAGs are enriched for ontology terms, illustrating the biological relevance to our image topology features and their potential utility in diagnostic and prognostic models.Item Spatial Transcriptomics Analysis Reveals Transcriptomic and Cellular Topology Associations in Breast and Prostate Cancers(2022-05) Alsaleh, Lujain; Johnson, Travis S.; Fadel, William; Tu, WanzhuBackground: Cancer is the leading cause of death worldwide and as a result is one of the most studied topics in public health. Breast cancer and prostate cancer are the most common cancers among women and men respectively. Gene expression and image features are independently prognostic of patient survival. However, it is sometimes difficult to discern how the molecular profile, e.g., gene expression, of given cells relate to their spatial layout, i.e., topology, in the tumor microenvironment (TME). However, with the advent of spatial transcriptomics (ST) and integrative bioinformatics analysis techniques, we are now able to better understand the TME of common cancers. Method: In this paper, we aim to determine the genes that are correlated with image topology features (ITFs) in common cancers which we denote topology associated genes (TAGs). To achieve this objective, we generate the correlation coefficient between genes and image features after identifying the optimal number of clusters for each of them. Applying this correlation matrix to heatmap using R package pheatmap to visualize the correlation between the two sets. The objective of this study is to identify common themes for the genes correlated with ITFs and we can pursue this using functional enrichment analysis. Moreover, we also find the similarity between gene clusters and some image features clusters using the ranking of correlation coefficient in order to identify, compare and contrast the TAGs across breast and prostate cancer ST slides. Result: The analysis shows that there are groups of gene ontology terms that are common within breast cancer, prostate cancer, and across both cancers. Notably, extracellular matrix (ECM) related terms appeared regularly in all ST slides. Conclusion: We identified TAGs in every ST slide regardless of cancer type. These TAGs were enriched for ontology terms that add context to the ITFs generated from ST cancer slides.