Leading Professional Society for Computational Biology and Bioinformatics
Connecting, Training, Empowering, Worldwide

banner

Posters - Schedules

Poster presentations at ISMB/ECCB 2021 will be presented virtually. Authors will pre-record their poster talk (5-7 minutes) and will upload it to the virtual conference platform site along with a PDF of their poster beginning July 19 and no later than July 23. All registered conference participants will have access to the poster and presentation through the conference and content until October 31, 2021. There are Q&A opportunities through a chat function and poster presenters can schedule small group discussions with up to 15 delegates during the conference.

Information on preparing your poster and poster talk are available at: https://www.iscb.org/ismbeccb2021-general/presenterinfo#posters

Ideally authors should be available for interactive chat during the times noted below:

View Posters By Category

Session A: Sunday, July 25 between 15:20 - 16:20 UTC
Session B: Monday, July 26 between 15:20 - 16:20 UTC
Session C: Tuesday, July 27 between 15:20 - 16:20 UTC
Session D: Wednesday, July 28 between 15:20 - 16:20 UTC
Session E: Thursday, July 29 between 15:20 - 16:20 UTC
Celloscope: a probabilistic model for cell type deconvolution in spatial transcriptomics data
COSI: Special Session 05
  • Agnieszka Geras, Warsaw University of Technology, University of Warsaw, Poland
  • Ewa Szczurek, University of Warsaw, Poland
  • Shadi Darvish Shafighi, University of Warsaw, Poland
  • Kacper Domżał, University of Warsaw, Poland
  • Igor Filipiuk, University of Warsaw, Poland
  • Łukasz Rączkowski, University of Warsaw, Poland
  • Hosein Toosi, Royal Institute of Technology, Sweden
  • Leszek Kaczmarek, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Poland
  • Łukasz Koperski, Medical University of Warsaw, Poland
  • Jens Lagergren, Royal Institute of Technology, Sweden
  • Dominika Nowis, Medical University of Warsaw, Poland

Short Abstract: The spatial transcriptomics procedure enables to gain an insight not only into the level of gene activity, but also enables to map this activity spatially. It is possible due to the fact that, unlike single cell RNA-seq experiments, spatial transcriptomics (ST) retains information on cells’ position within the tissue. However, ST spots contain multiple cells, therefore the observed signal inevitably includes information about mixtures of cells of different types. In order to deconvolute the aforementioned mixtures and infer the spatial cell types composition, various methods combining the two complementary technologies: ST and single cell RNA-seq have been proposed. Unfavourably, these methods require both types of data and may be prone to bias due to platform-specific effects, such as sequencing depth. To address those issues, we present an innovative approach that does not require single cell data, but instead needs additional prior knowledge on marker genes. Our novel probabilistic model for cell type deconvolution in ST data called Celloscope, was applied on mouse brain data and was able to successfully indicate brain structures and spatially distinguish between two main neuron types: inhibitory and excitatory.

CiteFuse enables multi-modal analysis of CITE-seq data
COSI: Special Session 05
  • Yingxin Lim, The University of Sydney, Australia
  • Jean Yee Hwa Yang, The University of Sydney, Australia
  • Hani Jieun Kim, The University of Sydney, Australia
  • Thomas Geddes, The University of Sydney, Australia
  • Pengyi Yang, The University of Sydney, Australia

Short Abstract: Multi-modal profiling of single cells represents one of the latest technological advancements in molecular biology. Among various single-cell multi-modal strategies, cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) allows simultaneous quantification of two distinct species: RNA and cell-surface proteins. Here, we introduce CiteFuse, a streamlined package consisting of a suite of tools for doublet detection, modality integration, clustering, differential RNA and protein expression analysis, antibody-derived tag evaluation, ligand–receptor interaction analysis and interactive web-based visualization of CITE-seq data.

We demonstrate the capacity of CiteFuse to integrate the two data modalities and its relative advantage against data generated from single-modality profiling using both simulations and real-world CITE-seq data. Furthermore, we illustrate a novel doublet detection method based on a combined index of cell hashing and transcriptome data. Finally, we demonstrate CiteFuse for predicting ligand–receptor interactions by using multi-modal CITE-seq data. Collectively, we demonstrate the utility and effectiveness of CiteFuse for the integrative analysis of transcriptome and epitope profiles from CITE-seq data.

CONET: Copy number event tree model of evolutionary tumor history for single-cell data
COSI: Special Session 05
  • Magda Markowska, University of Warsaw, Poland
  • Tomasz Cąkała, University of Warsaw, Poland
  • Błażej Miasojedow, University of Warsaw, Poland
  • Dilafruz Juraeva, Merck Healthcare KGaA, Translational Medicine, Oncology Bioinformatics, Germany
  • Johanna Mazur, Merck Healthcare KGaA, Translational Medicine, Oncology Bioinformatics, Germany
  • Edith Ross, Merck Healthcare KGaA, Translational Medicine, Oncology Bioinformatics, Germany
  • Eike Staub, Merck Healthcare KGaA, Translational Medicine, Oncology Bioinformatics, Germany
  • Ewa Szczurek, University of Warsaw, Poland

Short Abstract: Copy number alterations constitute important events in tumor evolution. Whole genome single cell sequencing data provides unprecedented insights into copy number profiles of individual cells, but is highly noisy. Reconstructing copy number event histories from this data is further complicated by the fact that the events span potentially overlapping regions of the genome.
Here, we propose Copy Number Event Tree (CONET), a probabilistic model for joint inference of the evolutionary tree on copy number events and copy number calling. CONET fully exploits the signal in scDNA-seq, as it relies directly on both the per-breakpoint and per-bin data. The model jointly infers the structure of an evolutionary tree on copy number events and copy number profiles of the cells, gaining statistical power in both tasks. CONET employs an efficient MCMC procedure to search the space of possible model structures and parameters. We introduce a range of model priors and penalties for efficient regularization. On simulated data, we demonstrate excellent performance of CONET both in breakpoint identification and tree reconstruction. CONET outperforms compared approaches in modeling the evolution and copy number calling for 260 cells from xenograft breast cancer sample.
CONET implementation is available at github.com/tc360950/CONET.

DUBStepR: correlation-based feature selection for clustering single-cell RNA sequencing data
COSI: Special Session 05
  • Bobby Ranjan, Genome Institute of Singapore, Singapore
  • Florian Schmidt, Genome Insitute of Singapore, Singapore
  • Wenjie Sun, Genome Institute of Singapore, Singapore
  • Jinyu Park, Genome Institute of Singapore, Singapore
  • Nirmala Arul Rayan, Genome Institute of Singapore, Singapore
  • Kunal Mishra, Genome Institute of Singapore, Singapore
  • Ronald Xie, Genome Institute of Singapore, Singapore
  • Fatemeh Alipour, Genome Institute of Singapore, Singapore
  • Vipul Singhal, Genome Institute of Singapore, Singapore
  • Ignasius Joanito, Genome Institute of Singapore, Singapore
  • Michelle Gek Liang Lim, Genome Institute of Singapore, Singapore
  • Shyam Prabhakar, Genome Institute of Singapore, Singapore

Short Abstract: Feature selection (marker gene selection) is widely believed to improve clustering accuracy, and is thus a key component of single cell clustering pipelines. However, we found that the performance of existing feature selection methods was inconsistent across benchmark datasets, and occasionally even worse than without feature selection. Moreover, existing methods ignored information contained in gene-gene correlations. We therefore developed DUBStepR (Determining the Underlying Basis using Stepwise Regression), a feature selection algorithm that leverages gene-gene correlations with a novel measure of inhomogeneity in feature space, termed the Density Index (DI). Despite selecting a relatively small number of genes, DUBStepR substantially outperformed existing single-cell feature selection methods across diverse clustering benchmarks. DUBStepR also demonstrated a significant improvement in additional benchmarking analyses focused on detection of rare cell types. DUBStepR is scalable to over a million cells, and can be straightforwardly applied to other data types such as single-cell ATAC-seq. We propose DUBStepR as a general-purpose feature selection solution for accurately clustering single-cell data.

Entropy-based Clustering for Single-Cell Transcriptomics
COSI: Special Session 05
  • Michael Casey, University of Southampton, United Kingdom
  • Joerg Fliege, University of Southampton, United Kingdom
  • Ruben Sanchez-Garcia, University of Southampton, United Kingdom
  • Ben MacArthur, University of Southampton, United Kingdom

Short Abstract: Single-cell transcriptomics lacks a cohesive mathematics of gene expression heterogeneity and cell type, limiting the interpretability of associated computational methods. We here provide such mathematics, developing an information-theoretic framework for quantifying the deviation of observed gene expression from a minimal ideal of cell type: that two cells of the same type should be statistically interchangeable. We showcase a novel unsupervised clustering algorithm based on our information-theoretic framework, which has no free parameters (beyond cluster number choice) and a natural correspondence to existing notions of cell type.

Evaluating Data Integration Tools on Clustering Spatial Transcriptomics Data
COSI: Special Session 05
  • Yijun Li, University of Michigan Ann Arbor, United States
  • Lana Garmire, University of Michigan Ann Arbor, United States
  • Bing He, University of Michigan Ann Arbor, United States
  • Stefan Stanojevic, University of Michigan Ann Arbor, United States
  • Zheng Jing, University of Michigan Ann Arbor, United States
  • Qianhui Huang, University of Michigan Ann Arbor, United States
  • Jian Kang, University of Michigan Ann Arbor, United States

Short Abstract: The increasing popularity of spatial transcriptomics has allowed researchers to analyze transcriptome data in its tissue sample's spatial context. There have been many methods developed for detecting genes with distinct spatial expression patterns. However, the application of such genes in clustering cell types has not been thoroughly studied. In the typical analysis pipeline of current RNA-seq data, clustering analysis is done on highly variable genes. Spatially variable genes can be novel markers to identify cell type clusters. Therefore, combining highly variable genes and spatially variable genes could potentially improve clustering performance. We applied six different integration methods commonly used in sequencing data, including CIMLR, MOFA+, scVI, WNN, SNF, and naive concatenation to combine spatially variable and highly variable genes and extract useful features on a lower dimension. We applied shared Nearest Neighbor clustering to the reduced extracted features and evaluated their performance. We performed the first comprehensive benchmark study that evaluates integration tools for handling both genetic and spatial data, using both real datasets from different spatial transcriptomics technologies and simulated datasets. Our results show WNN is recommended for integrating spatially variable genes and highly variable genes and extracting useful information to improve clustering.

Giotto suite, a flexible and high-performing framework for spatial multi-modal analysis
COSI: Special Session 05
  • Natalie Del Rossi, Icahn School of Medicine at Mount Sinai, United States
  • Jiaji George Chen, Boston University School of Medicine, United States
  • Guo-Cheng Yuan, Icahn School of Medicine at Mount Sinai, United States
  • Ruben Dries, Boston University School of Medicine, United States

Short Abstract: Previously our team introduced Giotto, an R package for the analysis and visualization of single-cell spatial data. Giotto represents an easy-to-use toolbox agnostic of the spatial platform used and offers a range of innovative algorithms to characterize tissue composition, identify spatial expression patterns, and find cellular interactions. To further support our overarching philosophy to create a multi-functional tool we built Giotto suite, which is backwards compatible and incorporates a number of extensions and enhancements. It provides an improved framework to represent current and future spatial datasets. First, we designed data structures that capture cell morphology features (e.g. cell boundary) and that enable incorporation of individual transcript information at the subcellular level for one or more modalities. To further handle such large datasets we integrated an HDF5 representation and optimized our computations. Finally, to improve (inter)operability we created i) object converters between Giotto and other popular tools (e.g. Seurat, SpatialExperiment, etc.) ii) changed to a modular package format to promote contributions from external developers, and iii) developed a reactive object for interactive selection and analysis using the R/Shiny platform. Altogether, Giotto Suite represents a powerful toolbox ready to tackle the next generation of challenges in spatial data analysis and visualization.

HSCRM: a comprehensive reference map for human Single-cell RNA-Seq data
COSI: Special Session 05
  • Jinmiao Chen, Singapore Immunology Network (SIgN), Singapore
  • Mengwei Li, Singapore Immunology Network (SIgN), Singapore
  • Xiaomeng Zhang, Singapore Immunology Network (SIgN), Singapore
  • Jingjing Ling, Singapore Immunology Network (SIgN), Singapore
  • Nicole Yee Shin Lee, Singapore Immunology Network (SIgN), Singapore
  • Florent Ginhoux, Singapore Immunology Network (SIgN), Singapore

Short Abstract: The application of single-cell omics technologies has resolved cell heterogeneity at increasing scale and resolution, leading to the identification of new subpopulations in both healthy and diseased states and across various tissues. Most recently, these technologies, have been exploited to generate several human atlases, in addition to organ- or disease-specific sub-atlases. However, comparison between these various atlases and extraction of fundamental commonalties is difficult owing to the plethora of sequencing protocols and analytical pipelines employed. Currently, due to the limitation of integration methods, most studies involved only a few dozen of samples, which may have inadequate power to identify rare cell types. Moreover, different studies adopted different metadata standards and cell type ontologies which makes it difficult to integrate and compare them. As a result, there is an unmet need for unified analysis, integration and annotation of public datasets in order to reveal synergy between studies and to avoid duplication of effort and irregularities of nomenclature. Here, we re-analysed all available datasets with unified pipeline and integrate them to build a comprehensive Human Single-Cell Reference Map(HSCRM), in an attempt to elucidate system-wide characteristics of human cells in different biological contexts.

Inferring the origin of cells at the maternal and fetal interface
COSI: Special Session 05
  • Thomas Varley, Case Western Reserve University, United States
  • Sam Mesiano, Case Western Reserve University, United States
  • Mehmet Koyutürk, Case Western Reserve University, United States

Short Abstract: The placenta consists of cells of maternal and fetal origin. Single cell RNA-seq (sc-RNAseq) presents unprecedented opportunities to characterize interactions between maternal and fetal cells in driving human labor. An important first step in analyzing this data is to identify the origins of each cell. We present plaSCenta, an algorithm that integrates genomic and transcriptomic data in a supervised learning framework to infer the maternal or fetal origin of cells. plaSCenta uses Y-chromosomal reads in samples from male pregnancies to identify fetal cells and assign labels (maternal/fetal) to cells. It then uses these labeled samples to train machine learning algorithms to predict the origin based on gene expression. Utilizing single-cell data from samples representing different tissues and states of labor, we observe that plaSCenta accurately predicts the maternal/fetal origin of cells with an AUC score greater than 0.9. We find that plaSCenta outperforms freemuxlet, a general purpose origin inference algorithm that uses genomic variants. We investigate plaSCenta’s predictions across cell types and placental locations and find the maternal/fetal breakdown concordant with established biological knowledge. plaSCenta is implemented as a dashboard to visualize the breakdown of origin across placental locations, cell types, and maternal fetal identification methods.

Met-Flow: Single-cell protein analysis reveals metabolic remodelling in immune cell populations
COSI: Special Session 05
  • Patricia Ahl, Institute of Molecular and Cell Biology, A*STAR, Singapore
  • Richard Hopkins, Institute of Molecular and Cell Biology, A*STAR, Singapore
  • Wen Wei Xiang Singapore
  • Bijin Au, Institute of Molecular and Cell Biology, A*STAR, Singapore
  • Nivashini Kaliaperumal, Institute of Molecular and Cell Biology, A*STAR, Singapore
  • Anna-Marie Fairhurst, Institute of Molecular and Cell Biology, A*STAR, Singapore
  • John Connolly, Institute of Molecular and Cell Biology, A*STAR, Singapore

Short Abstract: Metabolism plays a central role in shaping immune cell function. Within the tumour microenvironment, immune cells are in dynamic interaction with multiple different cell types, shifting the balance of tumour progression and anti-tumour immunity. This interplay is influenced by resource availability as well as cell-type specific metabolic states. We have developed a high-throughput flow cytometry method named Met-Flow, that measures key metabolic protein levels in a heterogeneous population, and at the single-cell level. This integrative proteomic analysis combines immune cell surface markers, cellular signalling by phosphorylation and cytokine production, which is directly linked to divergent metabolic states. Using Fast Fourier Transform-accelerated interpolation-based t-distributed stochastic neighbour embedding (FitSNE), we demonstrated metabolic remodelling of immune populations at single-cell resolution. By multiplexing metabolism to immune function, we identified unique metabolic states of memory T cell subsets under glycolytic perturbation. Measuring the capacity for flux through metabolic pathways using Met-Flow can be applied to any given cell type, and opens up the opportunity to target specific pathways across diseases.

Niche-specific gene expression using PIC-seq analysis during mouse embryonic development
COSI: Special Session 05
  • Junil Kim, Soongsil University, South Korea
  • Michaela Rothova, University of Copenahgen, Denmark
  • Joshua Brickman, University of Copenahgen, Denmark
  • Kyoung Jae Won, U of Copenhagen, Denmark

Short Abstract: Cells continuously interact with each other to provide signaling cues to nearby cells during development. However, our understanding about the role of cell contact is still very limited. Approaches to study cell-cell interaction from single cell RNAseq (scRNAseq) relies heavily on co-expression of ligand-receptor pairs.

To understand cell-cell interactions beyond ligand-receptor can explain, we used RNA sequencing from physically interacting cells (PIC-seq) as well scRNAseq from developing mouse embryo (at embryonic day E7.5, E8.5, and E9.5). PIC-seq was generated by mildly dissociating mouse embryo. Our computational framework based on support vector machine (SVM) found that cells exhibit specific gene expression when they are contacting with other types of cells. For instance, Lhx5 is expressed in neural progenitor (NP) cells neighboring with definitive endoderm (DE), while Gsc is expressed in the DE cells interacting with NP. Based on it, we were able to predict the neighboring cell type by reading the transcriptome of a cell. We further developed spatial-tSNE to show the spatial organization of tissue in the 2 dimensional space based on the cell neighbor prediction. Our study reveals niche-specific gene expression and explains the cell heterogeneity in the scRNAseq.

PseudotimeDE: inference of differential gene expression along cell pseudotime with well-calibrated p-values from single-cell RNA sequencing data
COSI: Special Session 05
  • Dongyuan Song, University of California, Los Angeles, United States
  • Jingyi Li, University of California, Los Angeles, United States

Short Abstract: To investigate molecular mechanisms underlying cell state changes, a crucial analysis is to identify differentially expressed (DE) genes along the pseudotime inferred from single-cell RNA-sequencing data. However, existing methods do not account for pseudotime inference uncertainty, and they have either ill-posed p-values or restrictive models. Here we propose PseudotimeDE, a DE gene identification method that adapts to various pseudotime inference methods, accounts for pseudotime inference uncertainty, and outputs well-calibrated p-values. Comprehensive simulations and real-data applications verify that PseudotimeDE outperforms existing methods in false discovery rate control and power.

RESEPT: a computational framework for REconstructing and Segmenting Expression mapped pseudo-image based on sPatially resolved Transcriptomics
COSI: Special Session 05
  • Juexin Wang, University of Missouri, United States
  • Qin Ma, The Ohio State University, United States
  • Dong Xu, Univ. of Missouri-Columbia, United States
  • Yuzhou Chang, The Ohio State University, United States
  • Fei He, Northeast Normal University, China

Short Abstract: Recent advances in spatially resolved transcriptomics provide spatial context for tissue domains, cell types, and their underlying functions. However, intrinsic tissue architectures often cannot be fully revealed using existing cell clustering methods due to a lack of strong representation for the biological context in tissues. We introduced RESEPT (REconstructing and Segmenting Expression mapped pseudo-RGB images based on sPatially resolved Transcriptomics), a novel framework to accurately characterize spatial heterogeneity from spatially resolved transcriptomics. Learning a spatial constrained graph neural network, RESEPT first converts spatial gene expression to a pseudo-RGB image for single-cell data visualization and interpretation. Then a CNN-based supervised segmentation model is trained on these pseudo images to reveal spatial contexts. A benchmark comparison on 16 human brain datasets of 10x Visium spatial transcriptomics shows that RESEPT outperforms other state-of-the-art methods in different normalization settings. On an in-house data of Alzheimer's Disease (AD) brain, RESEPT contributed significantly to identifying layers 2 and 3 of human postmortem middle temporal gyrus (MTG) and understanding the mechanism of the cellular and regional vulnerability of MTG in early AD. RESEPT was also applied to glioblastoma tumor in our study, which contributes to identifying infiltrating tumor and provides a potential clinical usage on prognosis.

scDesign2: a transparent simulator that generates high-fidelity single-cell gene expression count data with gene correlations captured
COSI: Special Session 05
  • Dongyuan Song, Interdepartmental Program of Bioinformatics, UCLA, United States
  • Jingyi Jessica Li, Department of Statistics, UCLA, United States
  • Tianyi Sun, Department of Statistics, UCLA, United States
  • Wei Vivian Li, Department of Biostatistics and Epidemiology, Rutgers School of Public Health, United States

Short Abstract: A pressing challenge in single-cell transcriptomics is to benchmark experimental protocols and computational methods. A solution is to use computational simulators, but existing simulators cannot simultaneously achieve three goals: preserving genes, capturing gene correlations, and generating any number of cells with varying sequencing depths. To fill this gap, we propose scDesign2, a transparent simulator that achieves all three goals and generates high-fidelity synthetic data for multiple single-cell gene expression count-based technologies. In particular, scDesign2 is advantageous in its transparent use of probabilistic models and its ability to capture gene correlations via copulas.

scJoint: transfer learning for data integration of atlas-scale single-cell RNA-seq and ATAC-seq
COSI: Special Session 05
  • Yingxin Lin, The University of Sydney, Australia
  • Tung-Yu Wu, Stanford University, United States
  • Sheng Wan, National Chiao Tung University, Taiwan
  • Jean Yang, The University of Sydney, Australia
  • Wing Wong, Stanford University, United States
  • Rachel Wang, The University of Sydney, Australia

Short Abstract: Single-cell multi-omics data continues to grow at an unprecedented pace, and effectively integrating different modalities holds the promise for better characterization of cell identities. Although a number of methods have demonstrated promising results in integrating multiple modalities from the same tissue, the complexity and scale of data compositions typically present in cell atlases still pose a significant challenge for existing methods. Here we present scJoint, a transfer learning method to integrate atlas-scale, heterogeneous collections of scRNA-seq and scATAC-seq data. scJoint leverages information from annotated scRNA-seq data in a semi-supervised framework and uses a neural network to simultaneously train labeled and unlabeled data, enabling label transfer and joint visualization in an integrative framework. Using multiple atlas data and a biologically varying multi-modal data, we demonstrate scJoint is computationally efficient and consistently achieves significantly higher cell type label accuracy than existing methods while providing meaningful joint visualizations. This suggests scJoint is effective in overcoming the heterogeneity in different modalities towards a more comprehensive understanding of cellular phenotypes.

SIEVE: Joint Inference of Tumor Phylogeny and Variant Calling from Single-cell DNA Sequencing Data
COSI: Special Session 05
  • Senbai Kang, University of Warsaw, Poland
  • Nico Borgsmueller, ETH Zurich, Switzerland
  • Monica Valecha, University of Vigo, Spain
  • Jack Kuipers, ETH Zurich, D-BSSE, Computational Biology Group, Switzerland
  • Niko Beerenwinkel, ETH Zurich, Switzerland
  • David Posada, University of Vigo, Spain
  • Ewa Szczurek, University of Warsaw, Poland

Short Abstract: Understanding intra-tumor heterogeneity is the cornerstone of developing effective cancer treatment and precision medicine. The development of single-cell DNA sequencing technology remarkably increases the resolution of DNA profiles to single-cell level. This facilitates the inference of phylogenetic trees with individual tumor cells as leaves, providing an evolutionary model of the mechanism behind intra-tumor heterogeneity. However, most of the methods proposed for tree reconstruction from single-cell data are based on infinite-sites assumption, which is often violated in reality due to evolutionary events like loss of heterozygosity.

Here, we develop a novel computational model, called Sieve, to jointly infer tumor phylogeny and call variants under finite-sites assumption from single-cell data. We propose a novel rate matrix, with states representing genotypes corresponding to heterozygous and homozygous mutations. To properly integrate the noisy single-cell sequencing data, we develop a Dirichlet-Multinomial based probabilistic model of the sequencing coverage and nucleotide read counts. The model accounts for allelic dropouts. To acquire accurate branch lengths, acquisition bias correction is applied. We prove that Sieve outperforms existing approaches on simulated data, especially regarding branch lengths and calling homozygous mutations. Sieve is then applied to publicly available real datasets. Sieve is implemented as a package of Beast 2.

Simultaneous dimensionality reduction and integration for single-cell ATAC-seq data using deep learning
COSI: Special Session 05
  • Uwe Ohler, Max Delbrueck Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Germany
  • Wolfgang Kopp, Max Delbrueck Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Germany
  • Altuna Akalin, Max Delbrueck Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Germany

Short Abstract: Advances in single-cell technologies enable the routine interrogation of chromatin accessibility
for tens of thousands of single cells, elucidating gene regulatory processes at an unprecedented resolution. Meanwhile, size, sparsity and high dimensionality of the resulting data continue to pose challenges for its computational analysis, and specifically the integration of data from different sources.
We have developed a dedicated computational approach, a variational auto-encoder using a noise model specifically designed for single-cell ATAC-seq data, which facilitates simultaneous dimensionality reduction and batch correction via an adversarial learning strategy. We showcase its benefits for detailed cell type characterization on individual real and simulated data sets as well as for integrating multiple complex datasets.

Single cell RNA-Seq data analysis on mice renal fibrosis
COSI: Special Session 05
  • Hyojin Kim, Heidelberg University and Heidelberg University Hospital; RWTH Aachen University, Germany
  • Christoph Kuppe, Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Germany
  • Javier Perales-Patón, Heidelberg University and Heidelberg University Hospital; RWTH Aachen University, Germany
  • Julio Saez-Rodriguez, Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Germany
  • Rafael Kramann, Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Germany

Short Abstract: Renal fibrosis is the hallmark of chronic kidney disease and incompletely understood. The kidney is a complex organ with a large heterogeneity of cell type such as glomerular cells, tubule-epithelial cells, mesenchymal cells, neuronal cells, endothelial cells and immune cells. The vascular niche is of key importance in kidney fibrosis since it harbours fibrosis-driving cells and capillary loss is thought to be one of the main drivers of disease progression. To understand mechanisms of fibrosis and capillary loss, we performed single cell RNA-sequencing and fate tracing of the major cell types of the vascular / perivascular niche including Gli1, Ng2, Myh11, Pdgfrb and Cd31+ cells in transgenic mice subjected to Unilateral Ureteral Obstruction (UUO) versus sham. Ten different 10x single cell datasets generated from genetically tagged sorted cells. Datasets were integrated for studying cell type-wise functional activity such as pathway activity, intercellular communication and cell differentiation. Several meaningful biological stories were identified supported by prior studies, including Pdgfa-Pdgfrb interactions with JAK-STAT pathway. As well, about 170 genes across data sets were found as driver candidate genes for the cell differentiation in renal fibrosis. Literature studies proved that 40 genes of them are driver genes or fibrosis-related genes.

Sparcle: assigning transcripts to cells in multiplexed images
COSI: Special Session 05
  • Sandhya Prabhakaran, Moffitt Cancer Center, United States
  • Tal Nawy, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, United States
  • Dana Pe'Er, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, United States

Short Abstract: Imaging-based spatial transcriptomics has the power to reveal patterns of single-cell gene expression by detecting mRNA transcripts as individually resolved spots in multiplexed images. However, molecular quantification has been severely limited by the computational challenges of segmenting poorly outlined, overlapping cells, and of overcoming technical noise; the majority of transcripts are routinely discarded because they fall outside the segmentation boundaries. This lost information leads to less accurate gene count matrices and weakens downstream analyses, such as cell type or gene program identification.
Here, we present Sparcle, a probabilistic model that reassigns transcripts to cells based on gene covariation patterns and incorporates spatial features such as distance to nucleus. We demonstrate its utility on multiplexed error-robust fluorescence in situ hybridization (MERFISH), single-molecule FISH (smFISH) data, and spatially-resolved transcript amplicon readout mapping (STARmap).
Sparcle improves transcript assignment, providing more realistic per-cell quantification of each gene, better delineation of cell boundaries, and improved cluster assignments. Critically, our approach does not require an accurate segmentation and is agnostic to technological platform.

Spatially resolved transcriptomic profiles of mouse early organogenesis
COSI: Special Session 05
  • Abhishek Sampath Kumar, Max Planck Institute for Molecular Genetics, Germany
  • Luyi Tian, Broad Institute of MIT & Harvard, United States
  • Robert Stickels, Broad Institute of MIT & Harvard, United States
  • Adriano Bolondi, Max Planck Institute for Molecular Genetics, Germany
  • Evan Macosko, Broad Institute of MIT & Harvard, United States
  • Alexander Meissner, Max Planck Institute for Molecular Genetics, Germany
  • Fei Chen, Broad Institute of MIT & Harvard, United States

Short Abstract: The fast development of single-cell transcriptome sequencing has advanced our knowledge of embryonic development. However, the spatial context is lost during cell dissociation, which is important for understanding tissue formation and cell-cell interaction. Here, we applied Slide-seqV2 on multiple mouse embryo sections in different stages to build a spatial map of mouse early organogenesis with near-cellular resolution. We performed joint clustering of spatial location and gene expression patterns at different scales. By comparing with the single-cell transcriptome atlas we showed our approach not just accurately characterized cell types, but also resolved transcriptionally similar cell states with spatial patterns. We found known and novel genes with transcriptional biases along the neural crest and neural tube axis. And we further investigated the role of Tbx6 in specification of paraxial mesoderm, revealed the impact of Tbx6 knock out on neural tube patterning. In sum, we spatially mapped the emerging cell states in different scales from whole embryo to specific regions in mouse organogenesis.

Squidpy: a scalable framework for spatial single cell analysis
COSI: Special Session 05
  • Mohammad Lotfollahi, Helmholtz Center Munich, Germany
  • Ignacio Ibarra, Helmholtz Center Munich, Germany
  • Fabian Theis, Helmholtz Center Munich, Germany
  • Louis Kuemmerle, Helmholtz Center Munich, Germany
  • Giovanni Palla, Helmholtz Center Munich, Germany
  • Hannah Spitzer, Helmholtz Center Munich, Germany
  • Michal Klein, Helmholtz Center Munich, Germany
  • Anna Schaar, Helmholtz Center Munich, Germany
  • Sergei Rybakov, Helmholtz Center Munich, Germany
  • Olle Holmberg, Helmholtz Center Munich, Germany
  • Isaac Virshup, University of Melbourne, Australia
  • Sabrina Richter, Helmholtz Center Munich, Germany

Short Abstract: Spatial omics data are advancing the study of tissue organization and cellular communication at an unprecedented scale. For this purpose we developed “Spatial Quantification of Molecular Data in Python” (Squidpy), a python-based framework for the analysis of spatially-resolved omics data. Squidpy aims to bring the diversity of spatial data in a common data representation and provide a common set of analysis and interactive visualization tools. Such infrastructure is useful in a variety of analysis settings, for different data types, and it explicitly leverages the additional information that spatial data provides: the spatial coordinates and, when available, the tissue image. Squidpy is built on top of Scanpy and Anndata, and it relies on several scientific computing libraries in Python, such as Scikit-image and Napari. Its modularity makes it suitable to be interfaced with a variety of additional tools in the python data science and machine learning ecosystem, as well as several single-cell data analysis packages. It allows to quickly explore spatial datasets and lays the foundations for both spatial omics data analysis as well as novel methods development.

Subspace clustering to identify cell-type stoichiometric constraints
COSI: Special Session 05
  • Angela Huang, University of Pennsylvania, United States
  • Junhyong Kim, University of Pennsylvania, United States

Short Abstract: With the advent of single-cell RNA-sequencing, researchers now have the ability to define cell types from large amounts of transcriptome information. Over the years, various clustering algorithms have been designed. Currently, most clustering algorithms measure cell-to-cell similarities using distance metrics based on the assumption that each cluster is comprised of “nearby” neighbors. In effect, clusters are a collection of similar cells in the embedded metric. Here, we propose that biological cluster should be comprised of sets of cells that satisfy a set of stochiometric constraints, whose intersections define a cell type. We propose to model each cell population with a single affine subspace, where all cells of the same type share a common set of linear constraints. We present an algorithm that leverages this subspace structure and learns a cell-to-cell affinity matrix based on notions of subspace similarity. We simulate scRNA-seq data according to the subspace model and benchmark our algorithm against pre-existing methods. We further test our algorithm on an in-house C. elegans dataset and show recovery of information on both cell type and developmental time. Lastly, we demonstrate how the subspace model allows us to compactly recover the major genes involved in an organism’s development.



International Society for Computational Biology
525-K East Market Street, RM 330
Leesburg, VA, USA 20176

ISCB On the Web

Twitter Facebook Linkedin
Flickr Youtube