Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Genomics ; 24(1): 722, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38030970

ABSTRACT

Cell type-specific differential gene expression analyses based on single-cell transcriptome datasets are sensitive to the presence of cell-free mRNA in the droplets containing single cells. This so-called ambient RNA contamination may differ between samples obtained from patients and healthy controls. Current ambient RNA correction methods were not developed specifically for single-cell differential gene expression (sc-DGE) analyses and might therefore not sufficiently correct for ambient RNA-derived signals. Here, we show that ambient RNA levels are highly sample-specific. We found that without ambient RNA correction, sc-DGE analyses erroneously identify transcripts originating from ambient RNA as cell type-specific disease-associated genes. We therefore developed a computationally lean and intuitive correction method, Fast Correction for Ambient RNA (FastCAR), optimized for sc-DGE analysis of scRNA-Seq datasets generated by droplet-based methods including the 10XGenomics Chromium platform. FastCAR uses the profile of transcripts observed in libraries that likely represent empty droplets to determine the level of ambient RNA in each individual sample, and then corrects for these ambient RNA gene expression values. FastCAR can be applied as part of the data pre-processing and QC in sc-DGE workflows comparing scRNA-Seq data in a health versus disease experimental design. We compared FastCAR with two methods previously developed to remove ambient RNA, SoupX and CellBender. All three methods identified additional genes in sc-DGE analyses that were not identified in the absence of ambient RNA correction. However, we show that FastCAR performs better at correcting gene expression values attributed to ambient RNA, resulting in a lower frequency of false-positive observations. Moreover, the use of FastCAR in a sc-DGE workflow increases the cell-type specificity of sc-DGE analyses across disease conditions.


Subject(s)
Gene Expression Profiling , RNA , Humans , RNA/metabolism , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Transcriptome , Research Design , Single-Cell Analysis/methods
2.
Cell Syst ; 14(7): 620-628.e3, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37473732

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) massively profiles transcriptomes of individual cells encapsulated in barcoded droplets in parallel. However, in real-world scRNA-seq data, many barcoded droplets do not contain cells, but instead, they capture a fraction of ambient RNAs released from damaged or lysed cells. A typical first step to analyze scRNA-seq data is to filter out cell-free droplets and isolate cell-containing droplets, but distinguishing them is often challenging; incorrect filtering may mislead the downstream analysis substantially. We propose SiftCell, a suite of software tools to identify and visualize cell-containing and cell-free droplets in manifold space via randomization (SiftCell-Shuffle) to classify between the two types of droplets (SiftCell-Boost) and to quantify the contribution of ambient RNAs for each droplet (SiftCell-Mix). By applying our method to datasets obtained by various single-cell platforms, we show that SiftCell provides a streamlined way to perform upstream quality control of scRNA-seq, which is more comprehensive and accurate than existing methods.


Subject(s)
Single-Cell Analysis , Software , Sequence Analysis, RNA/methods , Base Sequence , Single-Cell Analysis/methods , RNA-Seq , RNA/genetics
3.
Genome Biol ; 24(1): 140, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37337297

ABSTRACT

BACKGROUND: In droplet-based single-cell and single-nucleus RNA-seq experiments, not all reads associated with one cell barcode originate from the encapsulated cell. Such background noise is attributed to spillage from cell-free ambient RNA or barcode swapping events. RESULTS: Here, we characterize this background noise exemplified by three scRNA-seq and two snRNA-seq replicates of mouse kidneys. For each experiment, cells from two mouse subspecies are pooled, allowing to identify cross-genotype contaminating molecules and thus profile background noise. Background noise is highly variable across replicates and cells, making up on average 3-35% of the total counts (UMIs) per cell and we find that noise levels are directly proportional to the specificity and detectability of marker genes. In search of the source of background noise, we find multiple lines of evidence that the majority of background molecules originates from ambient RNA. Finally, we use our genotype-based estimates to evaluate the performance of three methods (CellBender, DecontX, SoupX) that are designed to quantify and remove background noise. We find that CellBender provides the most precise estimates of background noise levels and also yields the highest improvement for marker gene detection. By contrast, clustering and classification of cells are fairly robust towards background noise and only small improvements can be achieved by background removal that may come at the cost of distortions in fine structure. CONCLUSIONS: Our findings help to better understand the extent, sources and impact of background noise in single-cell experiments and provide guidance on how to deal with it.


Subject(s)
RNA , Single-Cell Analysis , Animals , Mice , Sequence Analysis, RNA/methods , RNA-Seq/methods , RNA/genetics , Genotype , Single-Cell Analysis/methods , Gene Expression Profiling/methods , Cluster Analysis
4.
Neuron ; 110(24): 4043-4056.e5, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36240767

ABSTRACT

Ambient RNA contamination in single-cell and single-nuclei RNA sequencing (snRNA-seq) is a significant problem, but its consequences are poorly understood. Here, we show that ambient RNAs in brain snRNA-seq datasets have a nuclear or non-nuclear origin with distinct gene set signatures. Both ambient RNA signatures are predominantly neuronal, and we find that some previously annotated neuronal cell types are distinguished by ambient RNA contamination. We detect pervasive neuronal ambient RNA contamination in all glial cell types unless glia and neurons are physically separated prior to sequencing. We demonstrate that this contamination can be removed in silico and show that previous single-nuclei RNA-seq-based annotations of immature oligodendrocytes are glial nuclei contaminated with ambient RNAs. After ambient RNA removal, we detect rare, committed oligodendrocyte progenitor cells not annotated in most previous adult human brain datasets. Together, these results provide an in-depth analysis of ambient RNA contamination in brain single-nuclei datasets.


Subject(s)
Cell Nucleus , RNA , Adult , Humans , RNA/metabolism , Cell Nucleus/metabolism , Neurons/metabolism , Sequence Analysis, RNA/methods , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Solitary Nucleus , Single-Cell Analysis/methods
SELECTION OF CITATIONS
SEARCH DETAIL