scholarly journals MapCell: Learning a Comparative Cell Type Distance Metric With Siamese Neural Nets With Applications Toward Cell-Type Identification Across Experimental Datasets

Author(s):  
Winston Koh ◽  
Shawn Hoon

Large collections of annotated single-cell RNA sequencing (scRNA-seq) experiments are being generated across different organs, conditions and organisms on different platforms. The diversity, volume and complexity of this aggregated data requires new analysis techniques to extract actionable knowledge. Fundamental to most analysis are key abilities such as: identification of similar cells across different experiments and transferring annotations from an annotated dataset to an unannotated one. There have been many strategies explored in achieving these goals, and they focuses primarily on aligning and re-clustering datasets of interest. In this work, we are interested in exploring the applicability of deep metric learning methods as a form of distance function to capture similarity between cells and facilitate the transfer of cell type annotation for similar cells across different experiments. Toward this aim, we developed MapCell, a few-shot training approach using Siamese Neural Networks (SNNs) to learn a generalizable distance metric that can differentiate between single cell types. Requiring only a small training set, we demonstrated that SNN derived distance metric can perform accurate transfer of annotation across different scRNA-seq platforms, batches, species and also aid in flagging novel cell types.

2019 ◽  
Author(s):  
Winston Koh ◽  
Shawn Hoon

ABSTRACTLarge collections of annotated single-cell RNA sequencing (scRNA-seq) experiments are being generated across different organs, conditions and organisms on different platforms. Transferring annotations from this growing database of single cell expression data to a new unannotated experimental dataset can accelerate insights into the underlying biology. There have been many approaches towards aligning and unifying such heterogeneous datasets. In our work, we recognized the need for a robust data driven distance metric to map annotation across datasets. Towards this aim, we applied a one-shot training approach, Siamese Neural Networks (SNN), to learn a distance metric that can differentiate between known annotated single cell types. Requiring only a small training set, we demonstrated that the SNN can perform predictions across different scRNA-seq platforms, identify novel cell types and transfer annotations across samples.


2019 ◽  
Author(s):  
Matthew N. Bernstein ◽  
Zhongjie Ma ◽  
Michael Gleicher ◽  
Colin N. Dewey

SummaryCell type annotation is a fundamental task in the analysis of single-cell RNA-sequencing data. In this work, we present CellO, a machine learning-based tool for annotating human RNA-seq data with the Cell Ontology. CellO enables accurate and standardized cell type classification by considering the rich hierarchical structure of known cell types, a source of prior knowledge that is not utilized by existing methods. Furthemore, CellO comes pre-trained on a novel, comprehensive dataset of human, healthy, untreated primary samples in the Sequence Read Archive, which to the best of our knowledge, is the most diverse curated collection of primary cell data to date. CellO’s comprehensive training set enables it to run out-of-the-box on diverse cell types and achieves superior or competitive performance when compared to existing state-of-the-art methods. Lastly, CellO’s linear models are easily interpreted, thereby enabling exploration of cell type-specific expression signatures across the ontology. To this end, we also present the CellO Viewer: a web application for exploring CellO’s models across the ontology.HighlightWe present CellO, a tool for hierarchically classifying cell type from single-cell RNA-seq data against the graph-structured Cell OntologyCellO is pre-trained on a comprehensive dataset comprising nearly all bulk RNA-seq primary cell samples in the Sequence Read ArchiveCellO achieves superior or comparable performance with existing methods while featuring a more comprehensive pre-packaged training setCellO is built with easily interpretable models which we expose through a novel web application, the CellO Viewer, for exploring cell type-specific signatures across the Cell OntologyGraphical Abstract


2020 ◽  
Author(s):  
Feng Tian ◽  
Fan Zhou ◽  
Xiang Li ◽  
Wenping Ma ◽  
Honggui Wu ◽  
...  

SummaryBy circumventing cellular heterogeneity, single cell omics have now been widely utilized for cell typing in human tissues, culminating with the undertaking of human cell atlas aimed at characterizing all human cell types. However, more important are the probing of gene regulatory networks, underlying chromatin architecture and critical transcription factors for each cell type. Here we report the Genomic Architecture of Cells in Tissues (GeACT), a comprehensive genomic data base that collectively address the above needs with the goal of understanding the functional genome in action. GeACT was made possible by our novel single-cell RNA-seq (MALBAC-DT) and ATAC-seq (METATAC) methods of high detectability and precision. We exemplified GeACT by first studying representative organs in human mid-gestation fetus. In particular, correlated gene modules (CGMs) are observed and found to be cell-type-dependent. We linked gene expression profiles to the underlying chromatin states, and found the key transcription factors for representative CGMs.HighlightsGenomic Architecture of Cells in Tissues (GeACT) data for human mid-gestation fetusDetermining correlated gene modules (CGMs) in different cell types by MALBAC-DTMeasuring chromatin open regions in single cells with high detectability by METATACIntegrating transcriptomics and chromatin accessibility to reveal key TFs for a CGM


eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Alexander J Tarashansky ◽  
Jacob M Musser ◽  
Margarita Khariton ◽  
Pengyang Li ◽  
Detlev Arendt ◽  
...  

Comparing single-cell transcriptomic atlases from diverse organisms can elucidate the origins of cellular diversity and assist the annotation of new cell atlases. Yet, comparison between distant relatives is hindered by complex gene histories and diversifications in expression programs. Previously, we introduced the self-assembling manifold (SAM) algorithm to robustly reconstruct manifolds from single-cell data (Tarashansky et al., 2019). Here, we build on SAM to map cell atlas manifolds across species. This new method, SAMap, identifies homologous cell types with shared expression programs across distant species within phyla, even in complex examples where homologous tissues emerge from distinct germ layers. SAMap also finds many genes with more similar expression to their paralogs than their orthologs, suggesting paralog substitution may be more common in evolution than previously appreciated. Lastly, comparing species across animal phyla, spanning mouse to sponge, reveals ancient contractile and stem cell families, which may have arisen early in animal evolution.


2020 ◽  
Author(s):  
Mohit Goyal ◽  
Guillermo Serrano ◽  
Ilan Shomorony ◽  
Mikel Hernaez ◽  
Idoia Ochoa

AbstractSingle-cell RNA-seq is a powerful tool in the study of the cellular composition of different tissues and organisms. A key step in the analysis pipeline is the annotation of cell-types based on the expression of specific marker genes. Since manual annotation is labor-intensive and does not scale to large datasets, several methods for automated cell-type annotation have been proposed based on supervised learning. However, these methods generally require feature extraction and batch alignment prior to classification, and their performance may become unreliable in the presence of cell-types with very similar transcriptomic profiles, such as differentiating cells. We propose JIND, a framework for automated cell-type identification based on neural networks that directly learns a low-dimensional representation (latent code) in which cell-types can be reliably determined. To account for batch effects, JIND performs a novel asymmetric alignment in which the transcriptomic profile of unseen cells is mapped onto the previously learned latent space, hence avoiding the need of retraining the model whenever a new dataset becomes available. JIND also learns cell-type-specific confidence thresholds to identify and reject cells that cannot be reliably classified. We show on datasets with and without batch effects that JIND classifies cells more accurately than previously proposed methods while rejecting only a small proportion of cells. Moreover, JIND batch alignment is parallelizable, being more than five or six times faster than Seurat integration. Availability: https://github.com/mohit1997/JIND.


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.


PLoS ONE ◽  
2021 ◽  
Vol 16 (7) ◽  
pp. e0254194
Author(s):  
Hong-Tae Park ◽  
Woo Bin Park ◽  
Suji Kim ◽  
Jong-Sung Lim ◽  
Gyoungju Nah ◽  
...  

Mycobacterium avium subsp. paratuberculosis (MAP) is a causative agent of Johne’s disease, which is a chronic and debilitating disease in ruminants. MAP is also considered to be a possible cause of Crohn’s disease in humans. However, few studies have focused on the interactions between MAP and human macrophages to elucidate the pathogenesis of Crohn’s disease. We sought to determine the initial responses of human THP-1 cells against MAP infection using single-cell RNA-seq analysis. Clustering analysis showed that THP-1 cells were divided into seven different clusters in response to phorbol-12-myristate-13-acetate (PMA) treatment. The characteristics of each cluster were investigated by identifying cluster-specific marker genes. From the results, we found that classically differentiated cells express CD14, CD36, and TLR2, and that this cell type showed the most active responses against MAP infection. The responses included the expression of proinflammatory cytokines and chemokines such as CCL4, CCL3, IL1B, IL8, and CCL20. In addition, the Mreg cell type, a novel cell type differentiated from THP-1 cells, was discovered. Thus, it is suggested that different cell types arise even when the same cell line is treated under the same conditions. Overall, analyzing gene expression patterns via scRNA-seq classification allows a more detailed observation of the response to infection by each cell type.


2018 ◽  
Author(s):  
Douglas Abrams ◽  
Parveen Kumar ◽  
R. Krishna Murthy Karuturi ◽  
Joshy George

AbstractBackgroundThe advent of single cell RNA sequencing (scRNA-seq) enabled researchers to study transcriptomic activity within individual cells and identify inherent cell types in the sample. Although numerous computational tools have been developed to analyze single cell transcriptomes, there are no published studies and analytical packages available to guide experimental design and to devise suitable analysis procedure for cell type identification.ResultsWe have developed an empirical methodology to address this important gap in single cell experimental design and analysis into an easy-to-use tool called SCEED (Single Cell Empirical Experimental Design and analysis). With SCEED, user can choose a variety of combinations of tools for analysis, conduct performance analysis of analytical procedures and choose the best procedure, and estimate sample size (number of cells to be profiled) required for a given analytical procedure at varying levels of cell type rarity and other experimental parameters. Using SCEED, we examined 3 single cell algorithms using 48 simulated single cell datasets that were generated for varying number of cell types and their proportions, number of genes expressed per cell, number of marker genes and their fold change, and number of single cells successfully profiled in the experiment.ConclusionsBased on our study, we found that when marker genes are expressed at fold change of 4 or more than the rest of the genes, either Seurat or Simlr algorithm can be used to analyze single cell dataset for any number of single cells isolated (minimum 1000 single cells were tested). However, when marker genes are expected to be only up to fC 2 upregulated, choice of the single cell algorithm is dependent on the number of single cells isolated and proportion of rare cell type to be identified. In conclusion, our work allows the assessment of various single cell methods and also aids in examining the single cell experimental design.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 45-46
Author(s):  
Christian Pohlkamp ◽  
Kapil Jhalani ◽  
Niroshan Nadarajah ◽  
Inseok Heo ◽  
William Wetton ◽  
...  

Background: Cytomorphology is the gold standard for quick assessment of peripheral blood and bone marrow samples in hematological neoplasms. It is a broadly-accepted method for orchestrating more specific diagnostics including immunophenotyping or genetics. Inter-/intra-observer-reproducibility of single cell classification is only 75 to 90%. Only a limited number of cells (100 - 500 cells/smear) is read in a time-consuming procedure. Machine learning (ML) is more reliable where human skills are limited, i.e. in handling large amounts of data or images. We here tested ML to differentiate peripheral blood leukocytes in a high throughput hematology laboratory. Aim: To establish an ML-based cell classifier capable of identifying healthy and pathologic cells in digitalized peripheral blood smear scans at an accuracy competitive with or outperforming human expert level. Methods: We selected >2,600 smears out of our unique archive of > 250,000 peripheral blood smears from hematological neoplasms. Depending on quality, we scanned up to 1,000 single cell images per smear. For image acquisition, a Metafer Scanning System (Zeiss Axio Imager.Z2 microscope, automatic slide feeder and automatic oiling device) from MetaSystems (Altlussheim, GER) was used. Areas of interest were defined by pre-scan in 10x magnification followed by high resolution scan in 40x to generate cell images for analysis. Average capture times for 300/500 cells were 3:43/4:37 min We set up a supervised ML-learning model using colour images (144x144 pixels) as input, outputting predicted probabilities of 21 predefined classes. We used ImageNet-pretrained Xception as our base model. We trained, evaluated and deployed the model using Amazon SageMaker on a subset of 82,974 images randomly selected from 514,183 cells captured and labelled for this study. 20 different cell types and one garbage class were classified. We included cell type categories referring to the critical importance of detecting rare leukemia subtypes (e.g. APL). Numbers of images from respective 21 classes ranged from 1,830 to 14,909 (median: 2,945). Minority classes were up-sampledto handle imbalances. Each picture was labelled by highly skilled technicians (median years practicing in this laboratory: 5) and two independent hematologists (median years at microscope: 20). Results: On a separate test set of 8,297 cells, our classifier was able to predict any of the five cell types occurring in the peripheral blood of healthy individuals (PMN, lymphocytes, monocytes, eosinophils, basophils) at very high median accuracy (97.0%) Median prediction accuracy of 15 rare or pathological cell types was 91.3%. For six critical pathological cell forms (myeloblasts, atypical/bilobulated promyelocytes in APL/APLv, hairy cells, lymphoma cells,plasma cells), median accuracy was 93.4% (sensitivity 93.8%). We saw a very high "T98 accuracy" for these cell types (98.5%) which is the accuracy of cell type predictions with prediction probability >0.98 (achieved in 2231/2417 cases), implicating that critical cells predicted with probability <0.98 should be flagged for human expert validation with priority. For all 21 classes median accuracy was 91.7%. Accuracy was lower for cells representing consecutive steps of maturation, e.g. promyelo-/myelo-/metamyelocytes, reproducing inconsistencies from the human-built phenotypic classification system (s.Fig.). Conclusions: We demonstrate an automated workflow using automatic microscopic cell capturing and ML-driven cell differentiation in samples of hematologic patients. Reproducibility, accuracy, sensitivity and specificity are above 90%, for many cell types above 98%. By flagging suspicious cells for humanvalidation, this tool can support even experienced hematology professionals, especially in detecting rare cell types. Given an appropriate scanning speed, it clearly outperforms human investigators in terms of examination time and number of differentiated cells. An ML-based intelligence can make its skills accessible to hematology laboratories on site or after upload of scanned cell images, independent of time/location. A cloud-based infrastructure is available. A prospective head to head challenge between ML-based classifier and human experts comparing sensitivity and accuracy for detection of all cell classes in peripheral blood will be tested to proof suitability for routine use (NCT 4466059). Figure Disclosures Heo: AWS: Current Employment. Wetton:AWS: Current Employment. Drescher:MetaSystems: Current Employment. Hänselmann:MetaSystems: Current Employment. Lörch:MetaSystems: Current equity holder in private company.


Sign in / Sign up

Export Citation Format

Share Document