scholarly journals Robust Single-cell Matching and Multi-modal Analysis Using Shared and Distinct Features Reveals Orchestrated Immune Responses

2021 ◽  
Author(s):  
Bokai Zhu ◽  
Shuxiao Chen ◽  
Yunhao Bai ◽  
Han Chen ◽  
Nilanjan Mukherjee ◽  
...  

The ability to align individual cellular information from multiple experimental sources, techniques and systems is fundamental for a true systems-level understanding of biological processes. While single-cell transcriptomic studies have transformed our appreciation for the complexities and contributions of diverse cell types to disease, they can be limited in their ability to assess protein-level phenotypic information and beyond. Therefore, matching and integrating single-cell datasets which utilize robust protein measurements across multiple modalities is critical for a deeper understanding of cell states, and signaling pathways particularly within their native tissue context. Current available tools are mainly designed for single-cell transcriptomics matching and integration, and generally rely upon a large number of shared features across datasets for mutual Nearest Neighbor (mNN) matching. This approach is unsuitable when applied to single-cell proteomic datasets, due to the limited number of parameters simultaneously accessed, and lack of shared markers across these experiments. Here, we introduce a novel cell matching algorithm, Matching with pARtIal Overlap (MARIO), that takes into account both shared and distinct features, while consisting of vital filtering steps to avoid sub-optimal matching. MARIO accurately matches and integrates data from different single-cell proteomic and multi-modal methods, including spatial techniques, and has cross-species capabilities. MARIO robustly matched tissue macrophages identified from COVID-19 lung autopsies via CODEX imaging to macrophages recovered from COVID-19 bronchoalveolar lavage fluid via CITE-seq. This cross-platform integrative analysis enabled the identification of unique orchestrated immune responses within the lung of complement-expressing macrophages and their impact on the local tissue microenvironment. MARIO thus provides an analytical framework for unified analysis of single-cell data for a comprehensive understanding of the underlying biological system.

2021 ◽  
Author(s):  
Yun Zhang ◽  
Brian Aevermann ◽  
Rohan Gala ◽  
Richard H. Scheuermann

Reference cell type atlases powered by single cell transcriptomic profiling technologies have become available to study cellular diversity at a granular level. We present FR-Match for matching query datasets to reference atlases with robust and accurate performance for identifying novel cell types and non-optimally clustered cell types in the query data. This approach shows excellent performance for cross-platform, cross-sample type, cross-tissue region, and cross-data modality cell type matching.


2020 ◽  
Author(s):  
Samantha M. Golomb ◽  
Ian H. Guldner ◽  
Anqi Zhao ◽  
Qingfei Wang ◽  
Bhavana Palakurthi ◽  
...  

ABSTRACTThe brain contains a diverse array of immune cell types. The phenotypic and functional plasticity of brain immune cells collectively contribute to brain tissue homeostasis and disease progression. Immune cell plasticity is profoundly influenced by local tissue microenvironment cues and systemic factors. Yet, the transcriptional mechanism by which systemic stimuli, such as aging and gut microbiota dysbiosis, reshape brain immune cell plasticity and homeostasis has not been fully delineated. Using Cellular Indexing of Transcriptomes and Epitopes by sequencing (CITE-seq), we analyzed compositional and transcriptional changes of the brain immune landscape in response to aging and gut dysbiosis. We first examined the discordance between canonical surface marker-defined immune cell types (Cell-ID) and their transcriptome signatures, which suggested transcriptional plasticity among immune cells despite sharing the same cell surface markers. Specifically, inflammatory and patrolling Ly6C+ monocytes were shifted predominantly to a pro-inflammatory transcriptional program in the aged brain, while brain ILCs shifted toward an ILC2 transcriptional profile. Finally, aging led to an increase of ILC-like cells expressing a T memory stemness (Tscm) signature in the brain. Antibiotics (ABX)-induced gut dysbiosis reduced the frequency of ILCs exhibiting Tscm-like properties in the aged mice, but not in the young mice. Enabled by high-resolution single-cell molecular phenotyping, our study revealed that systemic changes due to aging and gut dysbiosis prime the brain environment for an increased propensity for neuroinflammation, which provided insights into gut dysbiosis in age-related neurological diseases.Manuscript SummaryGolomb et al. performed Cellular Indexing of Transcriptomes and Epitopes by sequencing (CITE-seq) on immune cells from the brains of young and aged mice with and without antibiotics-induced gut dysbiosis. High resolution, single cell immunophenotyping enabled the dissection of extensive transcriptional plasticity of canonically identified monocytes and innate lymphoid cells (ILCs) in the aged brain. Through differential gene expression and trajectory inference analyses, the authors revealed tissue microenvironment-dependent cellular responses influenced by aging and gut dysbiosis that may potentiate neuroinflammatory diseases.Graphical Abstract


2019 ◽  
Author(s):  
Yuchen Yang ◽  
Gang Li ◽  
Huijun Qian ◽  
Kirk C. Wilhelmsen ◽  
Yin Shen ◽  
...  

AbstractBatch effect correction has been recognized to be indispensable when integrating single-cell RNA sequencing (scRNA-seq) data from multiple batches. State-of-the-art methods ignore single-cell cluster label information, but such information can improve effectiveness of batch effect correction, particularly under realistic scenarios where biological differences are not orthogonal to batch effects. To address this issue, we propose SMNN for batch effect correction of scRNA-seq data via supervised mutual nearest neighbor detection. Our extensive evaluations in simulated and real datasets show that SMNN provides improved merging within the corresponding cell types across batches, leading to reduced differentiation across batches over MNN, Seurat v3, and LIGER. Furthermore, SMNN retains more cell type-specific features, partially manifested by differentially expressed genes identified between cell types after SMNN correction being biologically more relevant, with precision improving by up to 841%.Key PointsBatch effect correction has been recognized to be critical when integrating scRNA-seq data from multiple batches due to systematic differences in time points, generating laboratory and/or handling technician(s), experimental protocol, and/or sequencing platform.Existing batch effect correction methods that leverages information from mutual nearest neighbors across batches (for example, implemented in SC3 or Seurat) ignore cell type information and suffer from potentially mismatching single cells from different cell types across batches, which would lead to undesired correction results, especially under the scenario where variation from batch effects is non-negligible compared with biological effects.To address this critical issue, here we present SMNN, a supervised machine learning method that first takes cluster/cell-type label information from users or inferred from scRNA-seq clustering, and then searches mutual nearest neighbors within each cell type instead of global searching.Our SMNN method shows clear advantages over three state-of-the-art batch effect correction methods and can better mix cells of the same cell type across batches and more effectively recover cell-type specific features, in both simulations and real datasets.


2020 ◽  
Author(s):  
Jian Zhou ◽  
Olga G. Troyanskaya

AbstractScaling single-cell data exploratory analysis with the rapidly growing diversity and quantity of single-cell omics datasets demands more interpretable and robust data representation that is generalizable across datasets. To address this challenge, here we developed a novel ‘quasilinear’ framework that combines the interpretability and transferability of linear methods with the representational power of nonlinear methods. Within this framework, we introduce a data representation and visualization method, GraphDR, and a structure discovery method, StructDR, that unifies cluster, trajectory, and surface estimation and allows their confidence set inference. We applied both methods to diverse single-cell RNA-seq datasets from whole embryos and tissues. Unlike PCA and t-SNE, GraphDR and StructDR generated representations that both distinguished highly specific cell types and were comparable across datasets. In addition, GraphDR is at least an order of magnitude faster than commonly used nonlinear methods. Our visualizations of scRNA-seq data from developing zebrafish and Xenopus embryos revealed extruding branches of lineages from a continuum of cell states, suggesting that the current branch view of cell specification may be oversimplified. Moreover, StructDR identified a novel neuronal population using scRNA-seq data from mouse hippocampus. An open-source python library and a user-friendly graphical interface for 3D data visualization and analysis with these methods are available at https://github.com/jzthree/quasildr.


2020 ◽  
Author(s):  
yan zheng ◽  
Yuanke Zhong ◽  
Jialu Hu ◽  
Xuequn Shang

Abstract Background: Single-cell RNA sequencing (scRNA-seq) enables the possibility of many in-depth transcriptomic analyses at a single-cell resolution, it’s already widely used for exploring the dynamic development process of life, studying the gene regulation mechanism, and discovering new cell types. However, the low RNA capture rate, which cause highly sparse expression with dropout, makes it difficult to do downstream analyses.Method: Most current methods use bimodal model to fit the gene expression with overwhelming zero. In this paper, we proposed scRNA-seq complementation (SCC) to solve the dropout problem in scRNA-seq data. Firstly, we find the nearest neighbor cells of every cell. Then we use a mixture model to impute the dropouts of scRNA-seq data. The model can identify the possibility of dropouts and estimates the reasonable gene expression value.Results: Experiment results show that SCC gives competitive results compared to two existing methods while showing superiority in reducing the intra-class distance of cells and improving the clustering accuracy in both simulation and real data.Conclusions: SCC is an effective tool to resolve the dropout noise in scRNA-seq data. The code is freely accessible at https://github.com/nwpuzhengyan/SCC.


2020 ◽  
Author(s):  
Wanqiu Chen ◽  
Yongmei Zhao ◽  
Xin Chen ◽  
Xiaojiang Xu ◽  
Zhaowei Yang ◽  
...  

AbstractSingle-cell RNA sequencing (scRNA-seq) has become a very powerful technology for biomedical research and is becoming much more affordable as methods continue to evolve, but it is unknown how reproducible different platforms are using different bioinformatics pipelines, particularly the recently developed scRNA-seq batch correction algorithms. We carried out a comprehensive multi-center cross-platform comparison on different scRNA-seq platforms using standard reference samples. We compared six pre-processing pipelines, seven bioinformatics normalization procedures, and seven batch effect correction methods including CCA, MNN, Scanorama, BBKNN, Harmony, limma and ComBat to evaluate the performance and reproducibility of 20 scRNA-seq data sets derived from four different platforms and centers. We benchmarked scRNA-seq performance across different platforms and testing sites using global gene expression profiles as well as some cell-type specific marker genes. We showed that there were large batch effects; and the reproducibility of scRNA-seq across platforms was dictated both by the expression level of genes selected and the batch correction methods used. We found that CCA, MNN, and BBKNN all corrected the batch variations fairly well for the scRNA-seq data derived from biologically similar samples across platforms/sites. However, for the scRNA-seq data derived from or consisting of biologically distinct samples, limma and ComBat failed to correct batch effects, whereas CCA over-corrected the batch effect and misclassified the cell types and samples. In contrast, MNN, Harmony and BBKNN separated biologically different samples/cell types into correspondingly distinct dimensional subspaces; however, consistent with this algorithm’s logic, MNN required that the samples evaluated each contain a shared portion of highly similar cells. In summary, we found a great cross-platform consistency in separating two distinct samples when an appropriate batch correction method was used. We hope this large cross-platform/site scRNA-seq data set will provide a valuable resource, and that our findings will offer useful advice for the single-cell sequencing community.


2022 ◽  
Author(s):  
Tony Pan ◽  
Guoshuai Cao ◽  
Erting Tang ◽  
Yu Zhao ◽  
Pablo Penaloza-MacMaster ◽  
...  

SARS-CoV-2 and HIV-1 are RNA viruses that have killed millions of people worldwide. Understanding the similarities and differences between these two infections is critical for understanding disease progression and for developing effective vaccines and therapies, particularly for 38 million HIV-1+ individuals who are vulnerable to SARS-CoV-2 co-infection. Here, we utilized single-cell transcriptomics to perform a systematic comparison of 94,442 PBMCs from 7 COVID-19 and 9 HIV-1+ patients in an integrated immune atlas, in which 27 different cell types were identified using an accurate consensus single-cell annotation method. While immune cells in both cohorts show shared inflammation and disrupted mitochondrial function, COVID-19 patients exhibit stronger humoral immunity, broader IFN-I signaling, elevated Rho GTPase and mTOR pathway activities, and downregulated mitophagy. Our results elucidate transcriptional signatures associated with COVID-19 and HIV-1 that may reveal insights into fundamental disease biology and potential therapeutic targets to treat these viral infections.


2017 ◽  
Author(s):  
Giovanni Iacono ◽  
Elisabetta Mereu ◽  
Amy Guillaumet-Adkins ◽  
Roser Corominas ◽  
Ivon Cuscó ◽  
...  

AbstractSingle-cell RNA sequencing significantly deepened our insights into complex tissues and latest techniques are capable processing ten-thousands of cells simultaneously. With bigSCale, we provide an analytical framework being scalable to analyze millions of cells, addressing challenges of future large datasets. Unlike previous methods, bigSCale does not constrain data to fit an a priori-defined distribution and instead uses an accurate numerical model of noise. We evaluated the performance of bigSCale using a biological model of aberrant gene expression in patient derived neuronal progenitor cells and simulated datasets, which underlined its speed and accuracy in differential expression analysis. We further applied bigSCale to analyze 1.3 million cells from the mouse developing forebrain. Herein, we identified rare populations, such as Reelin positive Cajal-Retzius neurons, for which we determined a previously not recognized heterogeneity associated to distinct differentiation stages, spatial organization and cellular function. Together, bigSCale presents a perfect solution to address future challenges of large single-cell datasets.Extended AbstractSingle-cell RNA sequencing (scRNAseq) significantly deepened our insights into complex tissues by providing high-resolution phenotypes for individual cells. Recent microfluidic-based methods are scalable to ten-thousands of cells, enabling an unbiased sampling and comprehensive characterization without prior knowledge. Increasing cell numbers, however, generates extremely big datasets, which extends processing time and challenges computing resources. Current scRNAseq analysis tools are not designed to analyze datasets larger than from thousands of cells and often lack sensitivity and specificity to identify marker genes for cell populations or experimental conditions. With bigSCale, we provide an analytical framework for the sensitive detection of population markers and differentially expressed genes, being scalable to analyze millions of single cells. Unlike other methods that use simple or mixture probabilistic models with negative binomial, gamma or Poisson distributions to handle the noise and sparsity of scRNAseq data, bigSCale does not constrain the data to fit an a priori-defined distribution. Instead, bigSCale uses large sample sizes to estimate a highly accurate and comprehensive numerical model of noise and gene expression. The framework further includes modules for differential expression (DE) analysis, cell clustering and population marker identification. Moreover, a directed convolution strategy allows processing of extremely large data sets, while preserving the transcript information from individual cells.We evaluate the performance of bigSCale using a biological model for reduced or elevated gene expression levels. Specifically, we perform scRNAseq of 1,920 patient derived neuronal progenitor cells from Williams-Beuren and 7q11.23 microduplication syndrome patients, harboring a deletion or duplication of 7q11.23, respectively. The affected region contains 28 genes whose transcriptional levels vary in line with their allele frequency. BigSCale detects expression changes with respect to cells from a healthy donor and outperforms other methods for single-cell DE analysis in sensitivity. Simulated data sets, underline the performance of bigSCale in DE analysis as it is faster and more sensitive and specific than other methods. The probabilistic model of cell-distances within bigSCale is further suitable for unsupervised clustering and the identification of cell types and subpopulations. Using bigSCale, we identify all major cell types of the somatosensory cortex and hippocampus analyzing 3,005 cells from adult mouse brains. Remarkably, we increase the number of cell population specific marker genes 4-6-fold compared to the original analysis and, moreover, define markers of higher order cell types. These include CD90 (Thy1), a neuronal surface receptor, potentially suitable for isolating intact neurons from complex brain samples.To test its applicability for large data sets, we apply bigSCale on scRNAseq data from 1.3 million cells derived from the pallium of the mouse developing forebrain (E18, 10x Genomics). Our directed down-sampling strategy accumulates transcript counts from cells with similar transcriptional profiles into index cell transcriptomes, thereby defining cellular clusters with improved resolution. Accordingly, index cell clusters provide a rich resource of marker genes for the main brain cell types and less frequent subpopulations. Our analysis of rare populations includes poorly characterized developmental cell types, such as neuron progenitors from the subventricular zone and neocortical Reelin positive neurons known as Cajal-Retzius (CR) cells. The latter represent a transient population which regulates the laminar formation of the developing neocortex and whose malfunctioning causes major neurodevelopmental disorders like autism or schizophrenia. Most importantly, index cell cluster can be deconvoluted to individual cell level for targeted analysis of populations of interest. Through decomposition of Reelin positive neurons, we determined a previously not recognized heterogeneity among CR cells, which we could associate to distinct differentiation stages as well as spatial and functional differences in the developing mouse brain. Specifically, subtypes of CR cells identified by bigSCale express different compositions of NMDA, AMPA and glycine receptor subunits, pointing to subpopulations with distinct membrane properties. Furthermore, we found Cxcl12, a chemokine secreted by the meninges and regulating the tangential migration of CR cells, to be also expressed in CR cells located in the marginal zone of the neocortex, indicating a self-regulated migration capacity.Together, bigSCale presents a perfect solution for the processing and analysis of scRNAseq data from millions of single cells. Its speed and sensitivity makes it suitable to the address future challenges of large single-cell data sets.


Author(s):  
Yuchen Yang ◽  
Gang Li ◽  
Huijun Qian ◽  
Kirk C Wilhelmsen ◽  
Yin Shen ◽  
...  

Abstract Batch effect correction has been recognized to be indispensable when integrating single-cell RNA sequencing (scRNA-seq) data from multiple batches. State-of-the-art methods ignore single-cell cluster label information, but such information can improve the effectiveness of batch effect correction, particularly under realistic scenarios where biological differences are not orthogonal to batch effects. To address this issue, we propose SMNN for batch effect correction of scRNA-seq data via supervised mutual nearest neighbor detection. Our extensive evaluations in simulated and real datasets show that SMNN provides improved merging within the corresponding cell types across batches, leading to reduced differentiation across batches over MNN, Seurat v3 and LIGER. Furthermore, SMNN retains more cell-type-specific features, partially manifested by differentially expressed genes identified between cell types after SMNN correction being biologically more relevant, with precision improving by up to 841.0%.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Michael W. Dorrity ◽  
Cristina M. Alexandre ◽  
Morgan O. Hamm ◽  
Anna-Lena Vigil ◽  
Stanley Fields ◽  
...  

AbstractThe scarcity of accessible sites that are dynamic or cell type-specific in plants may be due in part to tissue heterogeneity in bulk studies. To assess the effects of tissue heterogeneity, we apply single-cell ATAC-seq to Arabidopsis thaliana roots and identify thousands of differentially accessible sites, sufficient to resolve all major cell types of the root. We find that the entirety of a cell’s regulatory landscape and its transcriptome independently capture cell type identity. We leverage this shared information on cell identity to integrate accessibility and transcriptome data to characterize developmental progression, endoreduplication and cell division. We further use the combined data to characterize cell type-specific motif enrichments of transcription factor families and link the expression of family members to changing accessibility at specific loci, resolving direct and indirect effects that shape expression. Our approach provides an analytical framework to infer the gene regulatory networks that execute plant development.


Sign in / Sign up

Export Citation Format

Share Document