scholarly journals Sensitivity to sequencing depth in single-cell cancer genomics

2017 ◽  
Author(s):  
João M. Alves ◽  
David Posada

AbstractBackgroundQuerying cancer genomes at single-cell resolution is expected to provide a powerful framework to understand in detail the dynamics of cancer evolution. However, given the high costs currently associated with single-cell sequencing, together with the inevitable technical noise arising from single-cell genome amplification, cost-effective strategies that maximize the quality of single-cell data are critically needed. Taking advantage of five published single-cell whole-genome and whole-exome cancer datasets, we studied the impact of sequencing depth and sampling effort towards single-cell variant detection, including structural and driver mutations, genotyping accuracy, clonal inference and phylogenetic reconstruction, using recent tools specifically designed for single-cell data.ResultsAltogether, our results suggest that, for relatively large sample sizes (25 or more cells), sequencing single tumor cells at depths >5x does not drastically improve somatic variant discovery, the characterization of clonal genotypes or the estimation of phylogenies from single tumor cells.ConclusionsWe demonstrate that sequencing many individual tumor cells at a modest depth represents an effective alternative to explore the mutational landscape and clonal evolutionary patterns of cancer genomes, without the excessively high costs associated with high-coverage genome sequencing.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1566-1566
Author(s):  
Cathelijne Fokkema ◽  
Madelon M.E. de Jong ◽  
Sabrin Tahri ◽  
Zoltan Kellermayer ◽  
Chelsea den Hollander ◽  
...  

Abstract Introduction The introduction of new treatment regimens has significantly increased the progression free survival (PFS) of newly diagnosed multiple myeloma (MM) patients. However, even with these novel treatments, for some the disease remains refractory, highlighting the need to identify the pathobiology of high-risk MM. In MM patients, high levels of circulating tumor cells (CTCs) is associated with an inferior prognosis independent of high-risk cytogenetics (Chakraborty et al., 2016), suggesting that CTC numbers are a relevant reflection of tumor cell biology. We hypothesized that high levels of CTCs in MM patients are either the result of a transcriptionally distinct tumor clone with enhanced migration capacities, or driven by transcriptional differences present in the bone marrow (BM) tumor cells. To test these hypotheses, we 1) compared MM cells from paired blood and BM samples, and 2) compared BM tumor cells of patients with high and low CTC levels, using single cell RNA-sequencing. Results We isolated plasma cell (PCs) from viably frozen mononuclear cells of paired peripheral blood (PB) and BM aspirates from five newly diagnosed MM patients (0.5%-8% CTCs) to determine the presence of a distinct CTC subclone. We generated single cell transcriptomes from 44,779 CTCs and 35,697 BM PCs. In the total 9 clusters common to BM PCs and CTCs were identified upon single cell data integration, but no cluster specific for either source was detected. Only 25 genes were significantly differential expressed between CTCs and BM PCs. The absence of transcriptional clusters unique to either CTCs or BM PCs, and the transcriptional similarity between these two anatomical sites makes it highly unlikely that CTC levels are driven by the presence of a transcriptionally-primed migratory clone. We next set out to identify possible transcriptional differences in BM PCs from eight patients with high (2-22%) versus thirteen patients with low (0.004%-0.08%) percentages of CTCs. Recurrent high-risk mutations were present in both groups. Single cell transcriptomes were generated from 74,830 BM PCs. Single cell data integration across all patients led to the identification of 8 distinct PC clusters, one of which was characterized by enhanced proliferation as defined by STMN1 and MKI67 transcription. Interestingly, this proliferative cluster was increased in patients with a high percentage of CTCs. Furthermore, cell cycle analyses based on canonical G2M and S phase markers revealed that actively cycling PCs were more frequent in the BM of patients with a high percentage of CTCs (64% versus 30%, p<0.001), irrespective of the transcriptional cluster of origin. We hypothesized that plasma cell-extrinsic cues from the bone marrow micro-environment might be driving tumor proliferation. In order to substantiate this, we isolated BM immune cells from the same 21 patients and generated a library of 301,045 single immune cell transcriptomes. This library contained all major immune cell subsets, including CD4 + and CD8 + T cells, NK cells, B cells and monocytes. Comparative analyses of these cell populations in patients with either high or low levels of CTC are ongoing. Conclusion Through single cell transcriptomic analyses, we demonstrate that CTCs and BM PCs are transcriptionally similar. Importantly, we identify increased BM PC proliferation as a significant difference between patients with high and low levels of CTCs, implicating an increased tumor proliferation as one of the potential mechanisms driving CTC levels and MM disease pathobiology. The relation of the BM immune micro-environment to this altered proliferative state is currently under investigation. Disclosures van der Velden: Janssen: Other: Service Level Agreement; BD Biosciences: Other: Service Level Agreement; Navigate: Other: Service Level Agreement; Agilent: Research Funding; EuroFlow: Other: Service Level Agreement, Patents & Royalties: for network, not personally. Sonneveld: SkylineDx: Honoraria, Research Funding; Karyopharm: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Celgene/BMS: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding. Broyl: Sanofi: Honoraria; Janssen Pharmaceuticals: Honoraria; Celgene: Honoraria; Bristol-Meyer Squibb: Honoraria; Amgen: Honoraria.


2017 ◽  
Vol 42 ◽  
pp. 22-32 ◽  
Author(s):  
Daphne Tsoucas ◽  
Guo-Cheng Yuan

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
James Ding ◽  
Samantha L. Smith ◽  
Gisela Orozco ◽  
Anne Barton ◽  
Steve Eyre ◽  
...  

AbstractCD4+ T-cells represent a heterogeneous collection of specialised sub-types and are a key cell type in the pathogenesis of many diseases due to their role in the adaptive immune system. By investigating CD4+ T-cells at the single cell level, using RNA sequencing (scRNA-seq), there is the potential to identify specific cell states driving disease or treatment response. However, the impact of sequencing depth and cell numbers, two important factors in scRNA-seq, has not been determined for a complex cell population such as CD4+ T-cells. We therefore generated a high depth, high cell number dataset to determine the effect of reduced sequencing depth and cell number on the ability to accurately identify CD4+ T-cell subtypes. Furthermore, we investigated T-cell signatures under resting and stimulated conditions to assess cluster specific effects of stimulation. We found that firstly, cell number has a much more profound effect than sequencing depth on the ability to classify cells; secondly, this effect is greater when cells are unstimulated and finally, resting and stimulated samples can be combined to leverage additional power whilst still allowing differences between samples to be observed. While based on one individual, these results could inform future scRNA-seq studies to ensure the most efficient experimental design.


2018 ◽  
Author(s):  
Subarna Palit ◽  
Fabian J. Theis ◽  
Christina E. Zielinski

AbstractRecent advances in cytometry have radically altered the fate of single-cell proteomics by allowing a more accurate understanding of complex biological systems. Mass cytometry (CyTOF) provides simultaneous single-cell measurements that are crucial to understand cellular heterogeneity and identify novel cellular subsets. High-dimensional CyTOF data were traditionally analyzed by gating on bivariate dot plots, which are not only laborious given the quadratic increase of complexity with dimension but are also biased through manual gating. This review aims to discuss the impact of new analysis techniques for in-depths insights into the dynamics of immune regulation obtained from static snapshot data and to provide tools to immunologists to address the high dimensionality of their single-cell data.


Nature ◽  
2021 ◽  
Author(s):  
Sohrab Salehi ◽  
Farhia Kabeer ◽  
Nicholas Ceglia ◽  
Mirela Andronescu ◽  
Marc J. Williams ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3488-3488
Author(s):  
Hope L Mumme ◽  
Swati S Bhasin ◽  
Beena E Thomas ◽  
Bhakti Dwivedi ◽  
Deborah DeRyckere ◽  
...  

Abstract Introduction: The emergence and optimization of single cell profiling as a powerful tool to characterize the tumor microenvironment has revealed the heterogeneity of pediatric cancers, particularly different leukemia types/subtypes. Ease of access and analysis of the data from studies on different leukemia types is critical for improving diagnosis as well as therapy.Currently, there are no single cell data based resources available for pediatric leukemias. We have developed a comprehensive resource, Pediatric Single Cell Cancer Atlas (PedScAtlas), with the goal of developing a pan-leukemia genomics signature as well as highlighting the heterogeneity of different types of leukemia. This resource facilitates exploration and visualization of expression signatures in different leukemias without requiring extensive analysis and bioinformatics support. Methods: The PedScAtlas was built based on single cell data from various leukemias and normal bone marrow (BM) cells that have been pre-processed and analyzed using a uniform approach to generate normalized expression data (M. Bhasin et al. Blood 2020 (ASH), S. S. Bhasin et al. Blood 2020 (ASH), Panigraphy et al. JCI 2019, Stroopinsky et al. Haematologica 2021, Thomas et al. Blood 2020 (ASH)). The current version of PedScAtlas contains data from 30 local leukemia samples (Bhasin, et al. Blood 2020 (ASH), Thomas et al. Blood 2020 (ASH)) and those available on public resources such as GEO (Bailur et al. JCI Insight 2020). The PedScAtlas dataset and the Immune cell dataset each underwent quality control, integration, normalization, and dimensionality reduction using the Uniform Manifold Approximation and Projection (UMAP) method. Unsupervised UMAP analysis identified cellular clusters with similar transcriptome profiles that were annotated based on expression of cell-specific markers. Differential expression comparing different types of leukemia including acute myeloid leukemia (AML), B-cell acute lymphoblastic leukemia (B-ALL), T-cell acute lymphoblastic leukemia (T-ALL), and mixed phenotype acute leukemia (MPAL) samples was performed. To further ascertain genes specifically expressed in malignant blasts, the atlas also contains data from normal BM samples (Bailur et al. JCI Insight 2020) and healthy immune cells from the census of Immune Cells by the Human Cell Atlas Project (https://data.humancellatlas.org/). The web resource source code is written in R programming language and the interactive webserver has been implemented using the R Shiny package (Fig 1). The tool has been extensively tested on multiple operating systems (Linux, Mac, Windows) and web-browsers (Chrome, FireFox, and Safari). The tool is currently hosted on a 64bit CentOS 6 backend server running the Shiny Server program designed to host R Shiny applications. Results: The PedScAtlas contains data that facilitate exploration of gene expression profiles across leukemia types/subtypes and tumor microenvironment (TME) cell types. The atlas includes data from 33,930 AML, 25,744 T-ALL, 13,404 MPAL, and 6,252 B-ALL blast cells. It also contains single cell profiles of healthy BM samples from publicly available studies. The user can select data sets from the 5 major types of leukemia and normal BM in any combination of their choice to explore the expression profile of a gene of interest. The data can be visualized as UMAP (Fig. 1), or violin plots with annotations based on cluster ID, cell type, disease type, sample ID, and future continuous remission or relapse outcome. The UMAP with gene expression analyses allows the user to visualize the distribution of cell expressing a given gene on the UMAP plot. The Biomarker tool shows expression of different leukemia biomarker gene sets in the entire leukemia dataset. The Immune Cell section contains BM data from the Human Cell Atlas Project; the purpose of this section is to validate leukemia biomarkers by checking that the gene does not have significant expression in the healthy immune microenvironment. Conclusions: The PedScAtlas resource provides a unique and straightforward tool for biomarker identification, analysis of leukemia subtype heterogeneity, and transcriptome profile of the immune cell microenvironment. The resource is available online at https://bhasinlab.bmi.emory.edu/PediatricSC/. Figure 1 Figure 1. Disclosures DeRyckere: Meryx: Other: Equity ownership. Graham: Meryx: Membership on an entity's Board of Directors or advisory committees, Other: Equity ownership.


2021 ◽  
Author(s):  
Marc J Williams ◽  
Tyler Funnell ◽  
Ciara O'Flanagan ◽  
Andrew McPherson ◽  
Sohrab Salehi ◽  
...  

Cancer genomes exhibit extensive chromosomal copy number changes and structural variation, yet how allele specific alterations drive cancer genome evolution remains unclear. Here, through application of a new computational approach we report allele specific copy number alterations in 11,097 single cell whole genomes from genetically engineered mammary epithelial cells and 21,852 cells from high grade serous ovarian and triple negative breast cancers. Resolving single cell copy number profiles to individual alleles uncovered genomic background distributions of gains, losses and loss of heterozygosity, yielding evidence of positive selection of specific chromosomal alterations. In addition specific genomic loci in maternal and paternal alleles were commonly found to be altered in parallel with convergent phenotypic transcriptional effects. Finally we show that haplotype specific alterations trace the cyclical etiology of high level amplifications and reveal clonal haplotype decomposition of complex structures. Together, our results illuminate how allele and haplotype specific alterations, here determined across thousands of single cell cancer genomes, impact the etiology and evolution of structural variations in human tumours.


2015 ◽  
Vol 25 (10) ◽  
pp. 1499-1507 ◽  
Author(s):  
Nicholas E. Navin

Sign in / Sign up

Export Citation Format

Share Document