scholarly journals Single Cell Sequencing Reveals Heterogeneity of Gene Expression in KMT2A Rearranged Infant ALL at Relapse Compared to Diagnosis

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2756-2756
Author(s):  
Erin Guest ◽  
Byunggil Yoo ◽  
Rumen Kostadinov ◽  
Midhat S. Farooqi ◽  
Emily Farrow ◽  
...  

Introduction Infant acute lymphoblastic leukemia (ALL) with KMT2A rearrangement (KMT2A-r) is associated with a very poor prognosis. Disease free survival from the date of diagnosis is approximately 20% to 40%, depending on age, white blood cell count, and response to induction therapy. Refractory and relapsed infant ALL is often resistant to attempts at re-induction, and second remission is difficult to both achieve and maintain. Genomic sequencing studies of infant KMT2A-r ALL clinical samples have demonstrated an average of fewer than 3 additional non-silent somatic mutations per case at diagnosis, most commonly sub-clonal variants in RAS pathway genes. We previously reported relapse-associated gains in somatic variants associated with signaling, adhesion, and B-cell development pathways (Blood 2016 128:1735). We hypothesized that relapsed infant ALL is characterized by recurrent, altered patterns of gene expression. In this analysis, we utilized single cell RNA sequencing (scRNAseq) to identify candidate genes with differential expression in diagnostic vs. relapse leukemia specimens from 3 infants with KMT2A-r ALL. Methods Cryopreserved blood or bone marrow specimens from 3 infants enrolled in the Children's Oncology Group AALL0631 trial were selected for analysis. Samples from both diagnosis (DX) and relapse (RL) time points were thawed and checked for viability (>90% of cells viable) using trypan blue staining. Samples were multiplexed and processed for single cell RNA sequencing using the Chromium Single Cell 3' Library Kit (v2) and 10x Genomics Chromium controller per manufacturer's instructions (10x Genomics, Pleasanton, CA). Single cell libraries were converted to cDNA, amplified, and sequenced on an Illumina NovaSeq instrument. Two technical replicates were performed. Samples were de-multiplexed using genotype information acquired from previous whole exome sequencing (WES) and demuxlet software. Transcript alignment and counting were performed using the Cell Ranger pipeline (10x Genomics, default settings, Version 2.2.0, GRCh37 reference). Quality control, normalization, gene expression analysis, and unsupervised clustering were performed using the Seurat R package (Version 3.0). Dimensionality reduction and visualization were performed with the UMAP algorithm. Analyses were restricted to leukemia blasts with CD19 expression by scRNAseq. Results The clinical features for each case are shown in Table 1. Cells from the 3 infant ALL samples clustered together, distinct from cells of non-infant B-ALL, T-ALL, and mixed lineage acute leukemia biospecimens in the Children's Mercy scRNAseq database, but largely did not overlap with one another. For each of the 3 infant cases, cells from DX and RL time points could be distinguished by differential patterns of gene expression (Figure 1). Individual genes with statistically significant (p<0.05) log-fold change values were examined. Figure 2 summarizes the number of genes with up-regulation of expression by scRNAseq at RL compared to DX. Only 6 genes, DYNLL1, HMGB2, HMGN2, JUN, STMN1, and TUBA1B, were significantly increased at RL across all 3 cases. We repeated this analysis, restricting to leukemia blasts with CD79A expression, and identified these same 6 genes, and 4 additional genes: H2AFZ, NUCKS1, PRDX1, and TUBB, as consistently up-regulated in RL clusters. We examined the expression of candidate genes of interest, including clinically targetable genes, to compare the distribution of expression at DX and RL (Table 2). Conclusion Genomic factors underlying the aggressive, refractory clinical phenotype of relapsed infant ALL have yet to be defined. Each of these 3 cases demonstrates unique expression patterns at relapse, readily distinguishable from both the paired diagnostic sample and the other 2 relapse samples. Thus, scRNAseq is a powerful tool to identify heterogeneity in gene expression, with the potential to discover recurrent genomic drivers within resistant disease sub-clones. Ongoing analyses include scRNAseq in additional infant ALL samples, relative quantification of transcript expression in single cells, and comparison with bulk RNAseq data. Disclosures No relevant conflicts of interest to declare.

2018 ◽  
Author(s):  
Kent A. Riemondy ◽  
Monica Ransom ◽  
Christopher Alderman ◽  
Austin E. Gillen ◽  
Rui Fu ◽  
...  

ABSTRACTSingle-cell RNA sequencing (scRNA-seq) methods generate sparse gene expression profiles for thousands of single cells in a single experiment. The information in these profiles is sufficient to classify cell types by distinct expression patterns but the high complexity of scRNA-seq libraries often prevents full characterization of transcriptomes from individual cells. To extract more focused gene expression information from scRNA-seq libraries, we developed a strategy to physically recover the DNA molecules comprising transcriptome subsets, enabling deeper interrogation of the isolated molecules by another round of DNA sequencing. We applied the method in cell-centric and gene-centric modes to isolate cDNA fragments from scRNA-seq libraries. First, we resampled the transcriptomes of rare, single megakaryocytes from a complex mixture of lymphocytes and analyzed them in a second round of DNA sequencing, yielding up to 20-fold greater sequencing depth per cell and increasing the number of genes detected per cell from a median of 1,313 to 2,002. We similarly isolated mRNAs from targeted T cells to improve the reconstruction of their VDJ-rearranged immune receptor mRNAs. Second, we isolatedCD3DmRNA fragments expressed across cells in a scRNA-seq library prepared from a clonal T cell line, increasing the number of cells with detectedCD3Dexpression from 59.7% to 100%. Transcriptome resampling is a general approach to recover targeted gene expression information from single-cell RNA sequencing libraries that enhances the utility of these costly experiments, and may be applicable to the targeted recovery of molecules from other single-cell assays.


2021 ◽  
Author(s):  
Moonyoung Kang ◽  
Yuri Choi ◽  
Hyeonjin Kim ◽  
Sang-Gyu Kim

High-throughput single-cell RNA sequencing (scRNA-seq) identifies distinct cell populations based on cell-to-cell heterogeneity in gene expression. By examining the distribution of the density of gene expression profiles, the metabolic features of each cell population can be observed. Here, we employ the scRNA-seq technique to reveal the entire biosynthetic pathway of a flower volatile. The corolla (petals) of the wild tobacco Nicotiana attenuata emits a bouquet of scents that are composed mainly of benzylacetone (BA), a rare floral volatile. Protoplasts from the N. attenuata corolla were isolated at three different time points, and the transcript levels of >16,000 genes were analyzed in 3,756 single cells. We performed unsupervised clustering analysis to determine which cell clusters were involved in BA biosynthesis. The biosynthetic pathway of BA was uncovered by analyzing gene co-expression in scRNA-seq datasets and by silencing candidate genes in the corolla. In conclusion, the high-resolution spatiotemporal atlas of gene expression provided by scRNA-seq reveals the molecular features underlying cell-type-specific metabolism in a plant.


2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Sunny Z. Wu ◽  
Daniel L. Roden ◽  
Ghamdan Al-Eryani ◽  
Nenad Bartonicek ◽  
Kate Harvey ◽  
...  

Abstract Background High throughput single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for exploring cellular heterogeneity among complex human cancers. scRNA-Seq studies using fresh human surgical tissue are logistically difficult, preclude histopathological triage of samples, and limit the ability to perform batch processing. This hindrance can often introduce technical biases when integrating patient datasets and increase experimental costs. Although tissue preservation methods have been previously explored to address such issues, it is yet to be examined on complex human tissues, such as solid cancers and on high throughput scRNA-Seq platforms. Methods Using the Chromium 10X platform, we sequenced a total of ~ 120,000 cells from fresh and cryopreserved replicates across three primary breast cancers, two primary prostate cancers and a cutaneous melanoma. We performed detailed analyses between cells from each condition to assess the effects of cryopreservation on cellular heterogeneity, cell quality, clustering and the identification of gene ontologies. In addition, we performed single-cell immunophenotyping using CITE-Seq on a single breast cancer sample cryopreserved as solid tissue fragments. Results Tumour heterogeneity identified from fresh tissues was largely conserved in cryopreserved replicates. We show that sequencing of single cells prepared from cryopreserved tissue fragments or from cryopreserved cell suspensions is comparable to sequenced cells prepared from fresh tissue, with cryopreserved cell suspensions displaying higher correlations with fresh tissue in gene expression. We showed that cryopreservation had minimal impacts on the results of downstream analyses such as biological pathway enrichment. For some tumours, cryopreservation modestly increased cell stress signatures compared to freshly analysed tissue. Further, we demonstrate the advantage of cryopreserving whole-cells for detecting cell-surface proteins using CITE-Seq, which is impossible using other preservation methods such as single nuclei-sequencing. Conclusions We show that the viable cryopreservation of human cancers provides high-quality single-cells for multi-omics analysis. Our study guides new experimental designs for tissue biobanking for future clinical single-cell RNA sequencing studies.


iScience ◽  
2021 ◽  
Vol 24 (4) ◽  
pp. 102357
Author(s):  
Brenda Morsey ◽  
Meng Niu ◽  
Shetty Ravi Dyavar ◽  
Courtney V. Fletcher ◽  
Benjamin G. Lamberty ◽  
...  

PLoS ONE ◽  
2018 ◽  
Vol 13 (10) ◽  
pp. e0205883 ◽  
Author(s):  
Joseph C. Mays ◽  
Michael C. Kelly ◽  
Steven L. Coon ◽  
Lynne Holtzclaw ◽  
Martin F. Rath ◽  
...  

2019 ◽  
Author(s):  
Imad Abugessaisa ◽  
Shuhei Noguchi ◽  
Melissa Cardon ◽  
Akira Hasegawa ◽  
Kazuhide Watanabe ◽  
...  

AbstractAnalysis and interpretation of single-cell RNA-sequencing (scRNA-seq) experiments are compromised by the presence of poor quality cells. For meaningful analyses, such poor quality cells should be excluded to avoid biases and large variation. However, no clear guidelines exist. We introduce SkewC, a novel quality-assessment method to identify poor quality single-cells in scRNA-seq experiments. The method is based on the assessment of gene coverage for each single cell and its skewness as a quality measure. To validate the method, we investigated the impact of poor quality cells on downstream analyses and compared biological differences between typical and poor quality cells. Moreover, we measured the ratio of intergenic expression, suggesting genomic contamination, and foreign organism contamination of single-cell samples. SkewC is tested in 37,993 single-cells generated by 15 scRNA-seq protocols. We envision SkewC as an indispensable QC method to be incorporated into scRNA-seq experiment to preclude the possibility of scRNA-seq data misinterpretation.


2016 ◽  
Author(s):  
Hannah R. Dueck ◽  
Rizi Ai ◽  
Adrian Camarena ◽  
Bo Ding ◽  
Reymundo Dominguez ◽  
...  

AbstractRecently, measurement of RNA at single cell resolution has yielded surprising insights. Methods for single-cell RNA sequencing (scRNA-seq) have received considerable attention, but the broad reliability of single cell methods and the factors governing their performance are still poorly known. Here, we conducted a large-scale control experiment to assess the transfer function of three scRNA-seq methods and factors modulating the function. All three methods detected greater than 70% of the expected number of genes and had a 50% probability of detecting genes with abundance greater than 2 to 4 molecules. Despite the small number of molecules, sequencing depth significantly affected gene detection. While biases in detection and quantification were qualitatively similar across methods, the degree of bias differed, consistent with differences in molecular protocol. Measurement reliability increased with expression level for all methods and we conservatively estimate the measurement transfer functions to be linear above ~5-10 molecules. Based on these extensive control studies, we propose that RNA-seq of single cells has come of age, yielding quantitative biological information.


Sign in / Sign up

Export Citation Format

Share Document