skip to main content


Title: Spike-in normalization for single-cell RNA-seq reveals dynamic global transcriptional activity mediating anticancer drug response
Abstract The transcriptional plasticity of cancer cells promotes intercellular heterogeneity in response to anticancer drugs and facilitates the generation of subpopulation surviving cells. Characterizing single-cell transcriptional heterogeneity after drug treatments can provide mechanistic insights into drug efficacy. Here, we used single-cell RNA-seq to examine transcriptomic profiles of cancer cells treated with paclitaxel, celecoxib and the combination of the two drugs. By normalizing the expression of endogenous genes to spike-in molecules, we found that cellular mRNA abundance shows dynamic regulation after drug treatment. Using a random forest model, we identified gene signatures classifying single cells into three states: transcriptional repression, amplification and control-like. Treatment with paclitaxel or celecoxib alone generally repressed gene transcription across single cells. Interestingly, the drug combination resulted in transcriptional amplification and hyperactivation of mitochondrial oxidative phosphorylation pathway linking to enhanced cell killing efficiency. Finally, we identified a regulatory module enriched with metabolism and inflammation-related genes activated in a subpopulation of paclitaxel-treated cells, the expression of which predicted paclitaxel efficacy across cancer cell lines and in vivo patient samples. Our study highlights the dynamic global transcriptional activity driving single-cell heterogeneity during drug response and emphasizes the importance of adding spike-in molecules to study gene expression regulation using single-cell RNA-seq.  more » « less
Award ID(s):
1830961
NSF-PAR ID:
10335470
Author(s) / Creator(s):
; ; ; ; ;
Date Published:
Journal Name:
NAR Genomics and Bioinformatics
Volume:
3
Issue:
2
ISSN:
2631-9268
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Abstract

    Drug screening data from massive bulk gene expression databases can be analyzed to determine the optimal clinical application of cancer drugs. The growing amount of single-cell RNA sequencing (scRNA-seq) data also provides insights into improving therapeutic effectiveness by helping to study the heterogeneity of drug responses for cancer cell subpopulations. Developing computational approaches to predict and interpret cancer drug response in single-cell data collected from clinical samples can be very useful. We propose scDEAL, a deep transfer learning framework for cancer drug response prediction at the single-cell level by integrating large-scale bulk cell-line data. The highlight in scDEAL involves harmonizing drug-related bulk RNA-seq data with scRNA-seq data and transferring the model trained on bulk RNA-seq data to predict drug responses in scRNA-seq. Another feature of scDEAL is the integrated gradient feature interpretation to infer the signature genes of drug resistance mechanisms. We benchmark scDEAL on six scRNA-seq datasets and demonstrate its model interpretability via three case studies focusing on drug response label prediction, gene signature identification, and pseudotime analysis. We believe that scDEAL could help study cell reprogramming, drug selection, and repurposing for improving therapeutic efficacy.

     
    more » « less
  2. Abstract Background Pancreatic cancer is a complex disease with a desmoplastic stroma, extreme hypoxia, and inherent resistance to therapy. Understanding the signaling and adaptive response of such an aggressive cancer is key to making advances in therapeutic efficacy. Redox factor-1 (Ref-1), a redox signaling protein, regulates the conversion of several transcription factors (TFs), including HIF-1α, STAT3 and NFκB from an oxidized to reduced state leading to enhancement of their DNA binding. In our previously published work, knockdown of Ref-1 under normoxia resulted in altered gene expression patterns on pathways including EIF2, protein kinase A, and mTOR. In this study, single cell RNA sequencing (scRNA-seq) and proteomics were used to explore the effects of Ref-1 on metabolic pathways under hypoxia. Methods scRNA-seq comparing pancreatic cancer cells expressing less than 20% of the Ref-1 protein was analyzed using left truncated mixture Gaussian model and validated using proteomics and qRT-PCR. The identified Ref-1’s role in mitochondrial function was confirmed using mitochondrial function assays, qRT-PCR, western blotting and NADP assay. Further, the effect of Ref-1 redox function inhibition against pancreatic cancer metabolism was assayed using 3D co-culture in vitro and xenograft studies in vivo. Results Distinct transcriptional variation in central metabolism, cell cycle, apoptosis, immune response, and genes downstream of a series of signaling pathways and transcriptional regulatory factors were identified in Ref-1 knockdown vs Scrambled control from the scRNA-seq data. Mitochondrial DEG subsets downregulated with Ref-1 knockdown were significantly reduced following Ref-1 redox inhibition and more dramatically in combination with Devimistat in vitro. Mitochondrial function assays demonstrated that Ref-1 knockdown and Ref-1 redox signaling inhibition decreased utilization of TCA cycle substrates and slowed the growth of pancreatic cancer co-culture spheroids. In Ref-1 knockdown cells, a higher flux rate of NADP + consuming reactions was observed suggesting the less availability of NADP + and a higher level of oxidative stress in these cells. In vivo xenograft studies demonstrated that tumor reduction was potent with Ref-1 redox inhibitor similar to Devimistat. Conclusion Ref-1 redox signaling inhibition conclusively alters cancer cell metabolism by causing TCA cycle dysfunction while also reducing the pancreatic tumor growth in vitro as well as in vivo. 
    more » « less
  3. null (Ed.)
    Abstract The ability to predict the efficacy of cancer treatments is a longstanding goal of precision medicine that requires improved understanding of molecular interactions with drugs and the discovery of biomarkers of drug response. Identifying genes whose expression influences drug sensitivity can help address both of these needs, elucidating the molecular pathways involved in drug efficacy and providing potential ways to predict new patients’ response to available therapies. In this study, we integrated cancer type, drug treatment, and survival data with RNA-seq gene expression data from The Cancer Genome Atlas to identify genes and gene sets whose expression levels in patient tumor biopsies are associated with drug-specific patient survival using a log-rank test comparing survival of patients with low vs. high expression for each gene. This analysis was successful in identifying thousands of such gene–drug relationships across 20 drugs in 14 cancers, several of which have been previously implicated in the respective drug’s efficacy. We then clustered significant genes based on their expression patterns across patients and defined gene sets that are more robust predictors of patient outcome, many of which were significantly enriched for target genes of one or more transcription factors, indicating several upstream regulatory mechanisms that may be involved in drug efficacy. We identified a large number of genes and gene sets that were potentially useful as transcript-level biomarkers for predicting drug-specific patient survival outcome. Our gene sets were robust predictors of drug-specific survival and our results included both novel and previously reported findings, suggesting that the drug-specific survival marker genes reported herein warrant further investigation for insights into drug mechanisms and for validation as biomarkers to aid cancer therapy decisions. 
    more » « less
  4. Abstract Chinese hamster ovary (CHO) cell lines are widely used to manufacture biopharmaceuticals. However, CHO cells are not an optimal expression host due to the intrinsic plasticity of the CHO genome. Genome plasticity can lead to chromosomal rearrangements, transgene exclusion, and phenotypic drift. A poorly understood genomic element of CHO cell line instability is extrachromosomal circular DNA (eccDNA) in gene expression and regulation. EccDNA can facilitate ultra-high gene expression and are found within many eukaryotes including humans, yeast, and plants. EccDNA confers genetic heterogeneity, providing selective advantages to individual cells in response to dynamic environments. In CHO cell cultures, maintaining genetic homogeneity is critical to ensuring consistent productivity and product quality. Understanding eccDNA structure, function, and microevolutionary dynamics under various culture conditions could reveal potential engineering targets for cell line optimization. In this study, eccDNA sequences were investigated at the beginning and end of two-week fed-batch cultures in an ambr ® 250 bioreactor under control and lactate-stressed conditions. This work characterized structure and function of eccDNA in a CHO-K1 clone. Gene annotation identified 1551 unique eccDNA genes including cancer driver genes and genes involved in protein production. Furthermore, RNA-seq data is integrated to identify transcriptionally active eccDNA genes. 
    more » « less
  5. null (Ed.)
    Single cell RNA-sequencing (scRNA-seq) technology enables comprehensive transcriptomic profiling of thousands of cells with distinct phenotypic and physiological states in a complex tissue. Substantial efforts have been made to characterize single cells of distinct identities from scRNA-seq data, including various cell clustering techniques. While existing approaches can handle single cells in terms of different cell (sub)types at a high resolution, identification of the functional variability within the same cell type remains unsolved. In addition, there is a lack of robust method to handle the inter-subject variation that often brings severe confounding effects for the functional clustering of single cells. In this study, we developed a novel data denoising and cell clustering approach, namely CIBS, to provide biologically explainable functional classification for scRNA-seq data. CIBS is based on a systems biology model of transcriptional regulation that assumes a multi-modality distribution of the cells’ activation status, and it utilizes a Boolean matrix factorization approach on the discretized expression status to robustly derive functional modules. CIBS is empowered by a novel fast Boolean Matrix Factorization method, namely PFAST, to increase the computational feasibility on large scale scRNA-seq data. Application of CIBS on two scRNA-seq datasets collected from cancer tumor micro-environment successfully identified subgroups of cancer cells with distinct expression patterns of epithelial-mesenchymal transition and extracellular matrix marker genes, which was not revealed by the existing cell clustering analysis tools. The identified cell groups were significantly associated with the clinically confirmed lymph-node invasion and metastasis events across different patients. Index Terms—Cell clustering analysis, Data denoising, Boolean matrix factorization, Cancer microenvirionment, Metastasis. 
    more » « less