scholarly journals ImSig: A resource for the identification and quantification of immune signatures in blood and tissue transcriptomics data

2016 ◽  
Author(s):  
Ajit Johnson Nirmal ◽  
Tim Regan ◽  
Barbara Bo-Ju Shih ◽  
David Arthur Hume ◽  
Andrew Harvey Sims ◽  
...  

AbstractThe outcome of many diseases is commonly correlated with the immune response at the site of pathology. The ability to monitor the status of the immune system in situ provides a mechanistic understanding of disease progression, a prognostic assessment and a guide for therapeutic intervention. Global transcriptomic data can be deconvoluted to provide an indication of the cell types present and their activation state, but the gene signatures proposed to date are either disease-specific or have been derived from data generated from isolated cell populations. Here we describe an improved set of immune gene signatures, ImSig, derived based on their co-expression in blood and tissue datasets. ImSig includes validated lists of marker genes for the main immune cell types and a number of core pathways. When used in combination with network analysis, ImSig is an accurate and easy to use approach for monitoring immune phenotypes in transcriptomic data derived from clinical samples.

2021 ◽  
Author(s):  
Asif Zubair ◽  
Richard H. Chapple ◽  
Sivaraman Natarajan ◽  
William C. Wright ◽  
Min Pan ◽  
...  

The disorganization of cell types within tissues underlies many human diseases and has been studied for over a century using the conventional tools of pathology, including tissue-marking dyes such as the H&E stain. Recently, spatial transcriptomics technologies were developed that can measure spatially resolved gene expression directly in pathology-stained tissues sections, revealing cell types and their dysfunction in unprecedented detail. In parallel, artificial intelligence (AI) has approached pathologist-level performance in computationally annotating H&E images of tissue sections. However, spatial transcriptomics technologies are limited in their ability to separate transcriptionally similar cell types and AI-based pathology has performed less impressively outside their training datasets. Here, we describe a methodology that can computationally integrate AI-annotated pathology images with spatial transcriptomics data to markedly improve inferences of tissue cell type composition made over either class of data alone. We show that this methodology can identify regions of clinically relevant tumor immune cell infiltration, which is predictive of response to immunotherapy and was missed by an initial pathologist's manual annotation. Thus, combining spatial transcriptomics and AI-based image annotation has the potential to exceed pathologist-level performance in clinical diagnostic applications and to improve the many applications of spatial transcriptomics that rely on accurate cell type annotations.


Author(s):  
Xiaoyu Lu ◽  
Szu-Wei Tu ◽  
Wennan Chang ◽  
Changlin Wan ◽  
Jiashi Wang ◽  
...  

Abstract Deconvolution of mouse transcriptomic data is challenged by the fact that mouse models carry various genetic and physiological perturbations, making it questionable to assume fixed cell types and cell type marker genes for different data set scenarios. We developed a Semi-Supervised Mouse data Deconvolution (SSMD) method to study the mouse tissue microenvironment. SSMD is featured by (i) a novel nonparametric method to discover data set-specific cell type signature genes; (ii) a community detection approach for fixing cell types and their marker genes; (iii) a constrained matrix decomposition method to solve cell type relative proportions that is robust to diverse experimental platforms. In summary, SSMD addressed several key challenges in the deconvolution of mouse tissue data, including: (i) varied cell types and marker genes caused by highly divergent genotypic and phenotypic conditions of mouse experiment; (ii) diverse experimental platforms of mouse transcriptomics data; (iii) small sample size and limited training data source and (iv) capable to estimate the proportion of 35 cell types in blood, inflammatory, central nervous or hematopoietic systems. In silico and experimental validation of SSMD demonstrated its high sensitivity and accuracy in identifying (sub) cell types and predicting cell proportions comparing with state-of-the-arts methods. A user-friendly R package and a web server of SSMD are released via https://github.com/xiaoyulu95/SSMD.


2019 ◽  
Author(s):  
Chenling Xu ◽  
Romain Lopez ◽  
Edouard Mehlman ◽  
Jeffrey Regier ◽  
Michael I. Jordan ◽  
...  

AbstractAs single-cell transcriptomics becomes a mainstream technology, the natural next step is to integrate the accumulating data in order to achieve a common ontology of cell types and states. However, owing to various nuisance factors of variation, it is not straightforward how to compare gene expression levels across data sets and how to automatically assign cell type labels in a new data set based on existing annotations. In this manuscript, we demonstrate that our previously developed method, scVI, provides an effective and fully probabilistic approach for joint representation and analysis of cohorts of single-cell RNA-seq data sets, while accounting for uncertainty caused by biological and measurement noise. We also introduce single-cell ANnotation using Variational Inference (scANVI), a semi-supervised variant of scVI designed to leverage any available cell state annotations — for instance when only one data set in a cohort is annotated, or when only a few cells in a single data set can be labeled using marker genes. We demonstrate that scVI and scANVI compare favorably to the existing methods for data integration and cell state annotation in terms of accuracy, scalability, and adaptability to challenging settings such as a hierarchical structure of cell state labels. We further show that different from existing methods, scVI and scANVI represent the integrated datasets with a single generative model that can be directly used for any probabilistic decision making task, using differential expression as our case study. scVI and scANVI are available as open source software and can be readily used to facilitate cell state annotation and help ensure consistency and reproducibility across studies.


2021 ◽  
Author(s):  
Lei Gao ◽  
Fu Li ◽  
Jiao Cai ◽  
Jia Liu ◽  
Xi Zhang ◽  
...  

Acute myeloid leukemia (AML) is a highly heterogeneous hematological malignancy. The bone marrow (BM) microenvironment in AML plays an important role in leukemogenesis, drug resistance and leukemia relapse. In this study, we aimed to identify reliable immune-related biomarkers for AML prognosis by multiomics analysis. We obtained expression profiles from The Cancer Genome Atlas (TCGA) database and constructed a LASSO-Cox regression model to predict the prognosis of AML using multiomics bioinformatic analysis data. This was followed by independent validation of the model in the GSE106291 (n=251), GSE12417 (n=163) and GSE37642 (n=137) datasets and mutated genes in clinical samples for predicting overall survival (OS). Molecular docking was performed to predict the most optimal ligands to these hub genes. The single-cell RNA sequence dataset GSE116256 was used to clarify the expression of the hub genes in different immune cell types. According to their significant differences in immune gene signatures and survival trends, we concluded that the immune infiltration-lacking subtype (IL type) is associated with better prognosis than the immune infiltration-rich subtype (IR type). Using the LASSO model, we built a classifier based on 5 hub genes to predict the prognosis of AML (risk score = -0.086×ADAMTS3 + 0.180×CD52 + 0.472×CLCN5 - 0.356×HAL + 0.368×ICAM3). In summary, we constructed a prognostic model of AML using integrated multiomics bioinformatic analysis that could serve as a therapeutic classifier.


2021 ◽  
Vol 11 ◽  
Author(s):  
Guanghui Zhao ◽  
Junhua An ◽  
Qian Pu ◽  
Wenwen Geng ◽  
Haiyun Song ◽  
...  

The N6-methyladenosine (m6A) has been considered as a new layer of epitranscriptomic regulation on mRNA processing, stability, and translation. However, potential roles of m6A RNA methylation modification in tumor immune microenvironment (TIME) of breast cancer are yet fully understood. In this study, we comprehensively evaluated the genetic variations and transcript expressions of 15 m6A regulators in 1,079 breast cancer samples from the Cancer Genome Atlas (TCGA) database. We validated major regulators had significantly differential mRNA and protein expression in tumor tissue compared to normal tissues from 39 pairs of clinical breast cancer samples with different molecular subtypes, and especially high expression of m6A readers YTHDF1 and YTHDF3 predicted poor survival. Two clusters of breast cancer patients identified by the 15 m6A regulators’ pattern showed distinct overall survival, immune activation status, and immune cell infiltration, and clinical samples confirmed the diversity of lymphocytic infiltration. The profiles of these two clusters accorded with that of two classical cancer-immune phenotypes, immune-excluded and immune-inflamed phenotypes, it suggested that m6A regulators-based patterns might serve as crucial mediators of TIME in breast cancer. Moreover, the m6A phenotype-related gene signatures could also be survival predictor in breast cancer. Therefore, comprehensive evaluation of tumor m6A modification pattern will contribute to enhance our understanding of the characterization of immune cell infiltration in the tumor microenvironment and promote the responsiveness of breast cancer to immunotherapy.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e20071-e20071
Author(s):  
Sarah Elizabeth Church ◽  
Carmen Ballesteros-Merino ◽  
Amy H Sullivan ◽  
Andrew M White ◽  
Michael D Bailey ◽  
...  

e20071 Background: Malignant mesothelioma has been an incurable disease with few effective therapies. While PD-1 targeted therapies have elicited some patient responses, the overall response rate for mesothelioma is low. Since mesothelioma is derived from the mesothelium of the lung, we hypothesize that immune cells in the tumor microenvironment (TME) may behave differently than other solid tumors that are responsive to immunotherapy. Here we characterize prognostic immune gene signatures and spatial protein expression in the mesothelioma TME. Methods: 50 FFPE mesothelioma samples were analyzed using the NanoString PanCancer IO360 assay which measures expression of 770 genes, including the abundance of 14 immune cell types and of 32 IO signatures. All genes and signatures were correlated with overall survival (OS). GeoMx digital spatial profiling (DSP) was performed on 40 samples assessing the protein expression along 12 geometric circular regions-of-interest (ROI). Tissue slides were stained with a combination of fluorescent-labeled antibodies (pan-cytokeratin, CD3, CD68) and a panel of 38-antibodies each conjugated to a unique UV-photocleavable DNA barcode. UV light was applied to the defined ROI, which releases the DNA barcodes for quantitation on the nCounter platform. Results: Unsupervised clustering of samples based on gene signatures showed two distinct groups; one, with low expression of lymphocyte activation/exhaustion signatures and the second, with moderate expression of immune signatures. Two samples had high expression of all immune gene signatures, also confirmed by DSP had increased expression of T cell markers. Tumor proliferation (p < 0.001), hypoxia (p = 0.04), glycolysis (p < 0.001), B7-H3 (p = 0.007) and TGF-β (p = 0.001) signatures were significantly associated with shorter OS. Additional DSP profiling of these mesothelioma samples showed both T cell excluded and desert TMEs. Conclusions: We show that the mesothelioma TME has distinct immune biology associated with OS. Tumors from patients with poor survival had expression profiles previously described to be associated with immune excluded and desert phenotypes. We show that gene expression and DSP identifies unique targets for immunotherapy and we hypothesize that these findings may guide the development of combination trials that will be effective against mesothelioma.


2020 ◽  
Author(s):  
JINZHUANG DOU ◽  
Shaoheng Liang ◽  
Vakul Mohanty ◽  
Xuesen Cheng ◽  
Sangbae Kim ◽  
...  

Acquiring accurate single-cell multiomics profiles often requires performing unbiased in silico integration of data matrices generated by different single-cell technologies from the same biological sample. However, both the rows and the columns can represent different entities in different data matrices, making such integration a computational challenge that has only been solved approximately by existing approaches. Here, we present bindSC, a single-cell data integration tool that realizes simultaneous alignment of the rows and the columns between data matrices without making approximations. Using datasets produced by multiomics technologies as gold standard, we show that bindSC generates accurate multimodal co-embeddings that are substantially more accurate than those generated by existing approaches. Particularly, bindSC effectively integrated single cell RNA sequencing (scRNA-seq) and single cell chromatin accessibility sequencing (scATAC-seq) data towards discovering key regulatory elements in cancer cell-lines and mouse cells. It achieved accurate integration of both common and rare cell types (<0.25% abundance) in a novel mouse retina cell atlas generated using the 10x Genomics Multiome ATAC+RNA kit. Further, it achieves unbiased integration of scRNA-seq and 10x Visium spatial transcriptomics data derived from mouse brain cortex samples. Lastly, it demonstrated efficacy in delineating immune cell types via integrating single-cell RNA and protein data. Thus, bindSC, available at https://github.com/KChen-lab/bindSC, can be applied in a broad variety of context to accelerate discovery of complex cellular and biological identities and associated molecular underpinnings in diseases and developing organisms.


2019 ◽  
Author(s):  
Feiyang Ma ◽  
Matteo Pellegrini

AbstractCell type identification is one of the major goals in single cell RNA sequencing (scRNA-seq). Current methods for assigning cell types typically involve the use of unsupervised clustering, the identification of signature genes in each cluster, followed by a manual lookup of these genes in the literature and databases to assign cell types. However, there are several limitations associated with these approaches, such as unwanted sources of variation that influence clustering and a lack of canonical markers for certain cell types. Here, we present ACTINN (Automated Cell Type Identification using Neural Networks), which employs a neural network with 3 hidden layers, trains on datasets with predefined cell types, and predicts cell types for other datasets based on the trained parameters. We trained the neural network on a mouse cell type atlas (Tabula Muris Atlas) and a human immune cell dataset, and used it to predict cell types for mouse leukocytes, human PBMCs and human T cell sub types. The results showed that our neural network is fast and accurate, and should therefore be a useful tool to complement existing scRNA-seq pipelines.Author SummarySingle cell RNA sequencing (scRNA-seq) provides high resolution profiling of the transcriptomes of individual cells, which inevitably results in high volumes of data that require complex data processing pipelines. Usually, one of the first steps in the analysis of scRNA-seq is to assign individual cells to known cell types. To accomplish this, traditional methods first group the cells into different clusters, then find marker genes, and finally use these to manually assign cell types for each cluster. Thus these methods require prior knowledge of cell type canonical markers, and some level of subjectivity to make the cell type assignments. As a result, the process is often laborious and requires domain specific expertise, which is a barrier for inexperienced users. By contrast, our neural network ACTINN automatically learns the features for each predefined cell type and uses these features to predict cell types for individual cells. This approach is computationally efficient and requires no domain expertise of the tissues being studied. We believe ACTINN allows users to rapidly identify cell types in their datasets, thus rendering the analysis of their scRNA-seq datasets more efficient.


2019 ◽  
Author(s):  
Arezo Torang ◽  
Paraag Gupta ◽  
David J. Klinke

AbstractBackgroundHost immune response is coordinated by a variety of different specialized cell types that vary in time and location. While host immune response can be studied using conventional low-dimensional approaches, advances in transcriptomics analysis may provide a less biased view. Yet, leveraging transcriptomics data to identify immune cell subtypes presents challenges for extracting informative gene signatures hidden within a high dimensional transcriptomics space characterized by low sample numbers with noisy and missing values. To address these challenges, we explore using machine learning methods to select gene subsets and estimate gene coefficients simultaneously.ResultsElastic-net logistic regression, a type of machine learning, was used to construct separate classifiers for ten different types of immune cell and for five T helper cell subsets. The resulting classifiers were then used to develop gene signatures that best discriminate among immune cell types and T helper cell subsets using RNA-seq datasets. We validated the approach using single-cell RNA-seq (scRNA-seq) datasets, which gave consistent results. In addition, we classified cell types that were previously unannotated. Finally, we benchmarked the proposed gene signatures against other existing gene signatures.ConclusionsDeveloped classifiers can be used as priors in predicting the extent and functional orientation of the host immune response in diseases, such as cancer, where transcriptomic profiling of bulk tissue samples and single cells are routinely employed. Information that can provide insight into the mechanistic basis of disease and therapeutic response. The source code and documentation are available through GitHub: https://github.com/KlinkeLab/ImmClass2019.


2020 ◽  
Author(s):  
Jinzhuang Dou ◽  
Shaoheng Liang ◽  
Vakul Mohanty ◽  
Xuesen Cheng ◽  
Sangbae Kim ◽  
...  

Abstract Acquiring accurate single-cell multiomics profiles often requires performing unbiased in silico integration of data matrices generated by different single-cell technologies from the same biological sample. However, both the rows and the columns can represent different entities in different data matrices, making such integration a computational challenge that has only been solved approximately by existing approaches. Here, we present bindSC, a single-cell data integration tool that realizes simultaneous alignment of the rows and the columns between data matrices without making approximations. Using datasets produced by multiomics technologies as gold standard, we show that bindSC generates accurate multimodal co-embeddings that are substantially more accurate than those generated by existing approaches. Particularly, bindSC effectively integrated single cell RNA sequencing (scRNA-seq) and single cell chromatin accessibility sequencing (scATAC-seq) data towards discovering key regulatory elements in cancer cell-lines and mouse cells. It achieved accurate integration of both common and rare cell types (<0.25% abundance) in a novel mouse retina cell atlas generated using the 10x Genomics Multiome ATAC+RNA kit. Further, it achieves unbiased integration of scRNA-seq and 10x Visium spatial transcriptomics data derived from mouse brain cortex samples. Lastly, it demonstrated efficacy in delineating immune cell types via integrating single-cell RNA and protein data. Thus, bindSC, available at https://github.com/KChen-lab/bindSC, can be applied in a broad variety of context to accelerate discovery of complex cellular and biological identities and associated molecular underpinnings in diseases and developing organisms. 


Sign in / Sign up

Export Citation Format

Share Document