Confronting false discoveries in single-cell differential expression

Jordan W Squair, Matthieu Gautier, Claudia Kathe, Mark A Anderson, Nicholas D James, Thomas H Hutson, Rémi Hudelle, Taha Qaiser, Kaya J E Matson, Quentin Barraud, Ariel J Levine, Gioele La Manno, Michael A Skinnider, Grégoire Courtine, Jordan W Squair, Matthieu Gautier, Claudia Kathe, Mark A Anderson, Nicholas D James, Thomas H Hutson, Rémi Hudelle, Taha Qaiser, Kaya J E Matson, Quentin Barraud, Ariel J Levine, Gioele La Manno, Michael A Skinnider, Grégoire Courtine

Abstract

Differential expression analysis in single-cell transcriptomics enables the dissection of cell-type-specific responses to perturbations such as disease, trauma, or experimental manipulations. While many statistical methods are available to identify differentially expressed genes, the principles that distinguish these methods and their performance remain unclear. Here, we show that the relative performance of these methods is contingent on their ability to account for variation between biological replicates. Methods that ignore this inevitable variation are biased and prone to false discoveries. Indeed, the most widely used methods can discover hundreds of differentially expressed genes in the absence of biological differences. To exemplify these principles, we exposed true and false discoveries of differentially expressed genes in the injured mouse spinal cord.

Conflict of interest statement

G.C. is a founder and shareholder of Onward Medical, a company with no direct relationships with the present work. The remaining authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. A systematic benchmark of differential…
Fig. 1. A systematic benchmark of differential expression in single-cell transcriptomics.
a Schematic overview of the eighteen ground-truth datasets analyzed in this study. b Statistical methods for DE analysis employed in 500 recent scRNA-seq papers. Grey bars represent DE analysis methods included in this study. “Other” includes methods used in two or fewer studies. Inset pie chart shows the total proportion of recent scRNA-seq papers that employed DE analysis methods included in this study. Source data are provided as a Source Data file. c Area under the concordance curve (AUCC) for fourteen DE methods in the eighteen ground-truth datasets shown in a. d Mean difference in the AUCC (∆ AUCC) between the fourteen DE methods shown in c. Asterisks indicate comparisons with a two-tailed t-test p-value less than 0.05. e AUCC of GO term enrichment, as evaluated using gene set enrichment analysis, in the eighteen ground-truth datasets shown in a. f Rank and statistical significance of the GO term GO:0043330 (“response to exogenous dsRNA”) in GSEA analyses of mouse bone marrow mononuclear cells stimulated with poly-I:C, a type of synthetic dsRNA, for four h, using the output of fourteen DE methods. Source data are provided as a Source Data file.
Fig. 2. Single-cell DE methods are biased…
Fig. 2. Single-cell DE methods are biased towards highly expressed genes.
a Schematic illustration of the creation of ‘pseudobulks’ from single-cell data. Top, biological replicate from which each cell was obtained. Bottom, simulated gene expression matrix. Read counts for each gene are aggregated across cells of a given type within each biological replicate. b Mean AUCCs across eighteen ground-truth datasets after dividing the transcriptome into terciles of lowly, moderately, or highly expressed genes. c Mean expression levels of the 100 top-ranked false-positive genes from each DE method. d Spearman correlation between the mean expression of 80 ERCC spike-ins expressed in at least three cells and the –log10p-value of differential expression assigned by each DE method. e Scatterplots of mean ERCC expression vs. –log10p-value for exemplary single-cell and pseudobulk DE methods. Trend lines and shaded areas show local polynomial regression and the 95% confidence interval, respectively. f Mean expression levels of the 200 top-ranked genes from each DE method in a collection of 46 scRNA-seq datasets.
Fig. 3. DE analysis of single-cell data…
Fig. 3. DE analysis of single-cell data must account for biological replicates.
a Schematic illustration of the experiment shown in b, in which the aggregation procedure was disabled and pseudobulk DE methods were applied to individual cells. b Left, AUCC of the original fourteen DE methods, plus six pseudobulk methods applied to individual cells, in the eighteen ground-truth datasets. Right, Spearman correlation between ERCC mean expression and –log10 p-value assigned by six pseudobulk DE methods, before and after disabling the aggregation procedure. c Schematic illustration of the experiment shown in d, in which the replicate associated with each cell was shuffled to produce ‘pseudo-replicates.’ d Left, AUCC of the original fourteen DE methods, plus six pseudobulk methods applied to pseudo-replicates, in the eighteen ground-truth datasets. Right, Spearman correlation between ERCC mean expression and –log10 p-value assigned by six pseudobulk DE methods, before and after shuffling replicates to produce pseudo-replicates. e Variance of gene expression in pseudobulks formed from biological replicates and pseudo-replicates in mouse bone marrow mononuclear cells stimulated with poly-I:C. Shuffling the replicate associated with each cell produced a systematic decrease in the variance of gene expression. Right, pie chart shows the proportion of genes with increased or decreased variance in pseudo-replicates, as compared to biological replicates. f Decreases in the variance of gene expression in pseudo-replicates as compared to biological replicates across 46 scRNA-seq datasets. Points show the mean variance in biological replicates; arrowheads show the mean variance in pseudo-replicates. g Left, expression of the gene c in biological replicates (points) and pseudo-replicates (arrowheads) from unstimulated cells and cells stimulated with poly-I:C, with the range of possible pseudo-replicate expression values shown as a density. Right, mean (horizontal line) and variance (shaded area) of Txnrd3 expression in biological replicates (left) and pseudo-replicates (right). P-values were calculated by edgeR-LRT.
Fig. 4. False discoveries in single-cell DE.
Fig. 4. False discoveries in single-cell DE.
a Schematic illustration of simulation experiments. Single-cell RNA-seq datasets were simulated with varying degrees of heterogeneity between replicates. Replicates were then randomly assigned to either a ‘treatment’ or ‘control’ group, and DE analysis was performed between groups. b Number of DE genes detected in stimulation experiments with varying degrees of heterogeneity between replicates by a representative single-cell DE method, a representative pseudobulk method, and the same pseudobulk method applied to pseudo-replicates. Points and error bars show the mean and standard deviation across ten independent simulations. c Number of DE genes detected by the tests shown in b for genes divided into deciles by the magnitude of the change in variance between biological replicates and pseudo-replicates (∆-variance). d Volcano plots showing DE between T cells from random groups of unstimulated controls drawn from Kang et al. using a representative pseudobulk method, edgeR-LRT, applied to biological replicates or pseudo-replicates. Discarding information about biological replicates leads to the appearance of false discoveries. e Number of DE genes detected in comparisons of random groups of unstimulated controls from 14 scRNA-seq studies with at least six control samples. f Number of DE genes detected within spinal cord regions from control mice profiled by spatial transcriptomics using a representative pseudobulk method, edgeR-LRT (points), or a representative single-cell method, the Wilcoxon rank-sum test (arrowheads). g Mean change in variance between biological replicates and pseudo-replicates for 18 human and 20 mouse scRNA-seq datasets.
Fig. 5. True and false discoveries in…
Fig. 5. True and false discoveries in the injured mouse spinal cord.
a Synapses and projections of Vglut2ON neurons in the mouse lumbar spinal cord before and after SCI. Experiments were repeated in n = 3 mice. b Top, chronophotography of mice before and after SCI. Bottom left, principal component analysis of gait parameters for each condition (small circles). Large circles show the average per group. Bottom right, bar plot showing the average scores on principal component 1 (PC1), which quantify the locomotor performance of injured and uninjured mice. c snRNA-seq experimental design. Inset demonstrates persistent amplitude at increasing frequency of the H-reflex in response to plantar stimulation, reflecting hyperexcitability. d Uniform manifold approximation and projection (UMAP) visualization of 19,237 nuclei, revealing the major cell types of the mouse lumbar spinal cord. e UMAP visualization colored by the AUC of cell type prioritizations, as calculated by Augur. Endothelial cells are highlighted as the cell type with the highest AUC. f Pseudobulk expression levels in lumbar spinal cord endothelial cells by snRNA-seq (in counts per 10,000), left, and mean counts per Pecam1ON cell by RNA-scope, center, for Igfbp6, a gene identified as DE only by edgeR-LRT. Horizontal line and shaded area show the mean and standard deviation, respectively. Right, colocalization of Igfbp6 with the endothelial marker gene Pecam1 in RNAscope in situ hybridizations from injured and uninjured mouse spinal cords. g As in f, but for Prex2, a gene identified as DE only by the Wilcoxon rank-sum test. h Summary of in vivo screen results. Filled bars represent genes validated by RNAscope. i, Immunohistochemistry reveals Pecam1ON cells with atrophic features after SCI. Experiments were repeated in n = 3 mice. Source data are provided as a Source Data file.

References

    1. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009;10:57–63. doi: 10.1038/nrg2484.
    1. Stark R, Grzelak M, Hadfield J. RNA sequencing: the teenage years. Nat. Rev. Genet. 2019;20:631–656. doi: 10.1038/s41576-019-0150-2.
    1. Srinivasan K, et al. Untangling the brain’s neuroinflammatory and neurodegenerative transcriptional responses. Nat. Commun. 2016;7:11295. doi: 10.1038/ncomms11295.
    1. Chen X, Teichmann SA, Meyer KB. From tissues to cell types and back: single-cell gene expression analysis of tissue architecture. Annu. Rev. Biomed. Data Sci. 2018;1:29–51. doi: 10.1146/annurev-biodatasci-080917-013452.
    1. Kang HM, et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 2018;36:89–94. doi: 10.1038/nbt.4042.
    1. Mathys H, et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature. 2019;570:332–337. doi: 10.1038/s41586-019-1195-2.
    1. Finak G, et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 2015;16:278. doi: 10.1186/s13059-015-0844-5.
    1. Kharchenko PV, Silberstein L, Scadden DT. Bayesian approach to single-cell differential expression analysis. Nat. Methods. 2014;11:740–742. doi: 10.1038/nmeth.2967.
    1. Zimmerman, K. D., Espeland, M. A. & Langefeld, C. D. Pseudoreplication bias in single-cell studies; a practical solution. BioRxiv (2020) 10.1101/2020.01.15.906248.
    1. Crowell HL, et al. muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data. Nat. Commun. 2020;11:6077. doi: 10.1038/s41467-020-19894-4.
    1. Mehta T, Tanik M, Allison DB. Towards sound epistemological foundations of statistical methods for high-dimensional biology. Nat. Genet. 2004;36:943–947. doi: 10.1038/ng1422.
    1. Hagai T, et al. Gene expression variability across cells and species shapes innate immunity. Nature. 2018;563:197–202. doi: 10.1038/s41586-018-0657-2.
    1. Cano-Gamez E, et al. Single-cell transcriptomics identifies an effectorness gradient shaping the response of CD4+ T cells to cytokines. Nat. Commun. 2020;11:1801. doi: 10.1038/s41467-020-15543-y.
    1. Angelidis I, et al. An atlas of the aging lung mapped by single cell transcriptomics and deep tissue proteomics. Nat. Commun. 2019;10:963. doi: 10.1038/s41467-019-08831-9.
    1. Reyfman PA, et al. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2019;199:1517–1536. doi: 10.1164/rccm.201712-2410OC.
    1. Irizarry RA, et al. Multiple-laboratory comparison of microarray platforms. Nat. Methods. 2005;2:345–350. doi: 10.1038/nmeth756.
    1. Soneson C, Robinson MD. Bias, robustness and scalability in single-cell differential expression analysis. Nat. Methods. 2018;15:255–261. doi: 10.1038/nmeth.4612.
    1. Lun ATL, Marioni JC. Overcoming confounding plate effects in differential expression analyses of single-cell RNA-seq data. Biostatistics. 2017;18:451–464. doi: 10.1093/biostatistics/kxw055.
    1. Rapaport F, et al. Comprehensive evaluation of differential gene expression analysis methods for RNA-seq data. Genome Biol. 2013;14:R95. doi: 10.1186/gb-2013-14-9-r95.
    1. Tarazona S, García-Alcalde F, Dopazo J, Ferrer A, Conesa A. Differential expression in RNA-seq: a matter of depth. Genome Res. 2011;21:2213–2223. doi: 10.1101/gr.124321.111.
    1. Jiang L, et al. Synthetic spike-in standards for RNA-seq experiments. Genome Res. 2011;21:1543–1551. doi: 10.1101/gr.121095.111.
    1. Tung P-Y, et al. Batch effects and the effective design of single-cell gene expression studies. Sci. Rep. 2017;7:39921. doi: 10.1038/srep39921.
    1. Ståhl PL, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78–82. doi: 10.1126/science.aaf2403.
    1. Maniatis S, et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science. 2019;364:89–93. doi: 10.1126/science.aav9776.
    1. van den Brand R, et al. Restoring voluntary control of locomotion after paralyzing spinal cord injury. Science. 2012;336:1182–1185. doi: 10.1126/science.1217416.
    1. Beauparlant J, et al. Undirected compensatory plasticity contributes to neuronal dysfunction after severe spinal cord injury. Brain. 2013;136:3347–3361. doi: 10.1093/brain/awt204.
    1. Skinnider MA, et al. Cell type prioritization in single-cell data. Nat. Biotechnol. 2021;39:30–34. doi: 10.1038/s41587-020-0605-1.
    1. Squair JW, Skinnider MA, Gautier M, Foster LJ, Courtine G. Prioritization of cell types responsive to biological perturbations in single-cell data with Augur. Nat. Protoc. 2021;16:3836–3873. doi: 10.1038/s41596-021-00561-x.
    1. Robinson MD, McCarthy DJ, Smyth G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. Wang F, et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagn. 2012;14:22–29. doi: 10.1016/j.jmoldx.2011.08.002.
    1. Samanta D, Semenza GL. Maintenance of redox homeostasis by hypoxia-inducible factors. Redox Biol. 2017;13:331–335. doi: 10.1016/j.redox.2017.05.022.
    1. Zhang C, et al. IGF binding protein-6 expression in vascular endothelial cells is induced by hypoxia and plays a negative role in tumor angiogenesis. Int. J. Cancer. 2012;130:2003–2012. doi: 10.1002/ijc.26201.
    1. Li Y, et al. Pericytes impair capillary blood flow and motor function after chronic spinal cord injury. Nat. Med. 2017;23:733–741. doi: 10.1038/nm.4331.
    1. Zimmerman KD, Espeland MA, Langefeld CD. A practical solution to pseudoreplication bias in single-cell studies. Nat. Commun. 2021;12:738. doi: 10.1038/s41467-021-21038-1.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888–1902.e21. doi: 10.1016/j.cell.2019.05.031.
    1. Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 2018;19:15. doi: 10.1186/s13059-017-1382-0.
    1. Svensson, V., da Veiga Beltrame, E. & Pachter, L. A curated database reveals trends in single-cell transcriptomics. Database (Oxford)2020, (2020).
    1. Zhang JM, Kamath GM, Tse DN. Valid post-clustering differential analysis for single-cell RNA-Seq. Cell Syst. 2019;9:383–392.e6. doi: 10.1016/j.cels.2019.07.012.
    1. Ntranos V, Yi L, Melsted P, Pachter L. A discriminative learning approach to differential expression analysis for single-cell RNA-seq. Nat. Methods. 2019;16:163–166. doi: 10.1038/s41592-018-0303-9.
    1. McDavid A, et al. Data exploration, quality control and testing in single-cell qPCR-based gene expression experiments. Bioinformatics. 2013;29:461–467. doi: 10.1093/bioinformatics/bts714.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 2012;40:4288–4297. doi: 10.1093/nar/gks042.
    1. Lun ATL, Chen Y, Smyth GK. It’s DE-licious: a recipe for differential expression analyses of RNA-seq experiments using quasi-likelihood methods in edgeR. Methods Mol. Biol. 2016;1418:391–416. doi: 10.1007/978-1-4939-3578-9_19.
    1. Law CW, Chen Y, Shi W, Smyth G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29. doi: 10.1186/gb-2014-15-2-r29.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Sergushichev, A. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. BioRxiv (2016) 10.1101/060012.
    1. Zappia L, Phipson B, Oshlack A. Splatter: simulation of single-cell RNA sequencing data. Genome Biol. 2017;18:174. doi: 10.1186/s13059-017-1305-0.
    1. Wu YE, Pan L, Zuo Y, Li X, Hong W. Detecting activated cell populations using single-cell RNA-Seq. Neuron. 2017;96:313–329.e6. doi: 10.1016/j.neuron.2017.09.026.
    1. Hrvatin S, et al. Single-cell analysis of experience-dependent transcriptomic states in the mouse visual cortex. Nat. Neurosci. 2018;21:120–129. doi: 10.1038/s41593-017-0029-5.
    1. Sathyamurthy A, et al. Massively parallel single nucleus transcriptional profiling defines spinal cord neurons and their activity during behavior. Cell Rep. 2018;22:2216–2225. doi: 10.1016/j.celrep.2018.02.003.
    1. Grubman A, et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat. Neurosci. 2019;22:2087–2097. doi: 10.1038/s41593-019-0539-4.
    1. Rossi MA, et al. Obesity remodels activity and transcriptional state of a lateral hypothalamic brake on feeding. Science. 2019;364:1271–1274. doi: 10.1126/science.aax1184.
    1. Smillie CS, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative Colitis. Cell. 2019;178:714–730.e22. doi: 10.1016/j.cell.2019.06.029.
    1. Tran NM, et al. Single-cell profiles of retinal ganglion cells differing in resilience to injury reveal neuroprotective genes. Neuron. 2019;104:1039–1055.e12. doi: 10.1016/j.neuron.2019.11.006.
    1. Goldfarbmuren KC, et al. Dissecting the cellular specificity of smoking effects and reconstructing lineages in the human airway epithelium. Nat. Commun. 2020;11:2485. doi: 10.1038/s41467-020-16239-z.
    1. Nagy C, et al. Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons. Nat. Neurosci. 2020;23:771–781. doi: 10.1038/s41593-020-0621-y.
    1. Huang B, et al. Mucosal profiling of pediatric-onset Colitis and IBD reveals common pathogenics and therapeutic pathways. Cell. 2019;179:1160–1176.e24. doi: 10.1016/j.cell.2019.10.027.
    1. Wilk AJ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020;26:1070–1076. doi: 10.1038/s41591-020-0944-y.
    1. Asboth L, et al. Cortico-reticulo-spinal circuit reorganization enables functional recovery after severe spinal cord contusion. Nat. Neurosci. 2018;21:576–588. doi: 10.1038/s41593-018-0093-5.
    1. Wenger N, et al. Spatiotemporal neuromodulation therapies engaging muscle synergies improve motor control after spinal cord injury. Nat. Med. 2016;22:138–145. doi: 10.1038/nm.4025.
    1. Anderson MA, et al. Required growth facilitators propel axon regeneration across complete spinal cord injury. Nature. 2018;561:396–400. doi: 10.1038/s41586-018-0467-6.
    1. Scheff SW, Rabchevsky AG, Fugaccia I, Main JA, Lumpp JE. Experimental modeling of spinal cord injury: characterization of a force-defined injury device. J. Neurotrauma. 2003;20:179–193. doi: 10.1089/08977150360547099.
    1. Squair, J. W. et al. Integrated systems analysis reveals conserved gene networks underlying response to spinal cord injury. elife7, (2018).
    1. Courtine G, et al. Transformation of nonfunctional spinal circuits into functional states after the loss of brain input. Nat. Neurosci. 2009;12:1333–1342. doi: 10.1038/nn.2401.
    1. Takeoka A, Vollenweider I, Courtine G, Arber S. Muscle spindle feedback directs locomotor recovery and circuit reorganization after spinal cord injury. Cell. 2014;159:1626–1639. doi: 10.1016/j.cell.2014.11.019.
    1. Dominici N, et al. Versatile robotic interface to evaluate, enable and train locomotion and balance after neuromotor disorders. Nat. Med. 2012;18:1142–1147. doi: 10.1038/nm.2845.
    1. La Manno, G. et al. RNA velocity of single cells. Nature560, 494–498 (2018).
    1. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:296. doi: 10.1186/s13059-019-1874-1.
    1. Zeisel A, et al. Molecular architecture of the mouse nervous system. Cell. 2018;174:999–1014.e22. doi: 10.1016/j.cell.2018.06.021.
    1. Grimm D, et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 2008;82:5887–5911. doi: 10.1128/JVI.00254-08.
    1. Anderson MA, et al. Astrocyte scar formation aids central nervous system axon regeneration. Nature. 2016;532:195–200. doi: 10.1038/nature17623.
    1. Tomer R, Ye L, Hsueh B, Deisseroth K. Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nat. Protoc. 2014;9:1682–1697. doi: 10.1038/nprot.2014.123.
    1. Voigt FF, et al. The mesoSPIM initiative: open-source light-sheet microscopes for imaging cleared tissue. Nat. Methods. 2019;16:1105–1108. doi: 10.1038/s41592-019-0554-0.
    1. Arneson D, et al. Single cell molecular alterations reveal target cells and pathways of concussive brain injury. Nat. Commun. 2018;9:3894. doi: 10.1038/s41467-018-06222-0.
    1. Avey D, et al. Single-cell RNA-Seq uncovers a robust transcriptional response to morphine by Glia. Cell Rep. 2018;24:3619–3629.e4. doi: 10.1016/j.celrep.2018.08.080.
    1. Aztekin C, et al. Identification of a regeneration-organizing cell in the Xenopus tail. Science. 2019;364:653–658. doi: 10.1126/science.aav9996.
    1. Bhattacherjee A, et al. Cell type-specific transcriptional programs in mouse prefrontal cortex during adolescence and addiction. Nat. Commun. 2019;10:4169. doi: 10.1038/s41467-019-12054-3.
    1. Brenner E, et al. Single cell transcriptome profiling of the human alcohol-dependent brain. Hum. Mol. Genet. 2020;29:1144–1153. doi: 10.1093/hmg/ddaa038.
    1. Cheng C-W, et al. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell. 2019;178:1115–1131.e15. doi: 10.1016/j.cell.2019.07.048.
    1. Co, M., Hickey, S. L., Kulkarni, A., Harper, M. & Konopka, G. Cortical foxp2 supports behavioral flexibility and developmental dopamine D1 receptor expression. Cereb. Cortex30, 1855–1870 (2020).
    1. Davie K, et al. A single-cell transcriptome Atlas aging Drosophilla brain. Cell. 2018;174:982–998.e20. doi: 10.1016/j.cell.2018.05.057.
    1. Denisenko E, et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 2020;21:130. doi: 10.1186/s13059-020-02048-6.
    1. Der E, et al. Tubular cell and keratinocyte single-cell transcriptomics applied to lupus nephritis reveal type I IFN and fibrosis relevant pathways. Nat. Immunol. 2019;20:915–927. doi: 10.1038/s41590-019-0386-1.
    1. Gunner G, et al. Sensory lesioning induces microglial synapse elimination via ADAM10 and fractalkine signaling. Nat. Neurosci. 2019;22:1075–1088. doi: 10.1038/s41593-019-0419-y.
    1. Haber AL, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551:333–339. doi: 10.1038/nature24489.
    1. Hashimoto K, et al. Single-cell transcriptomics reveals expansion of cytotoxic CD4 T cells in supercentenarians. Proc. Natl Acad. Sci. USA. 2019;116:24242–24251. doi: 10.1073/pnas.1907883116.
    1. Hu P, et al. Dissecting cell-type composition and activity-dependent transcriptional state in mammalian brains by massively parallel single-nucleus RNA-seq. Mol. Cell. 2017;68:1006–1015.e7. doi: 10.1016/j.molcel.2017.11.017.
    1. Jaitin DA, et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell. 2019;178:686–698.e14. doi: 10.1016/j.cell.2019.05.054.
    1. Jäkel S, et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature. 2019;566:543–547. doi: 10.1038/s41586-019-0903-2.
    1. Kim D-W, et al. Multimodal analysis of cell types in a hypothalamic node controlling social behavior. Cell. 2019;179:713–728.e17. doi: 10.1016/j.cell.2019.09.020.
    1. Kotliarov Y, et al. Broad immune activation underlies shared set point signatures for vaccine responsiveness in healthy individuals and disease activity in patients with lupus. Nat. Med. 2020;26:618–629. doi: 10.1038/s41591-020-0769-8.
    1. Madissoon E, et al. scRNA-seq assessment of the human lung, spleen, and esophagus tissue stability after cold preservation. Genome Biol. 2019;21:1. doi: 10.1186/s13059-019-1906-x.
    1. Nault R, Fader KA, Bhattacharya S, Zacharewski TR. Single-nuclei RNA sequencing assessment of the hepatic effects of 2,3,7,8-Tetrachlorodibenzo-p-dioxin. Cell. Mol. Gastroenterol. Hepatol. 2021;11:147–159. doi: 10.1016/j.jcmgh.2020.07.012.
    1. Ordovas-Montanes J, et al. Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature. 2018;560:649–654. doi: 10.1038/s41586-018-0449-8.
    1. Reyes M, et al. An immune-cell signature of bacterial sepsis. Nat. Med. 2020;26:333–340. doi: 10.1038/s41591-020-0752-4.
    1. Schafflick D, et al. Integrated single cell analysis of blood and cerebrospinal fluid leukocytes in multiple sclerosis. Nat. Commun. 2020;11:247. doi: 10.1038/s41467-019-14118-w.
    1. Schirmer L, et al. Neuronal vulnerability and multilineage diversity in multiple sclerosis. Nature. 2019;573:75–82. doi: 10.1038/s41586-019-1404-z.
    1. Wagner DE, et al. Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo. Science. 2018;360:981–987. doi: 10.1126/science.aar4362.
    1. Wang S, et al. Single-cell transcriptomic atlas primate ovarian aging. Cell. 2020;180:585–600.e19. doi: 10.1016/j.cell.2020.01.009.
    1. Wirka RC, et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat. Med. 2019;25:1280–1289. doi: 10.1038/s41591-019-0512-5.
    1. Ximerakis M, et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat. Neurosci. 2019;22:1696–1708. doi: 10.1038/s41593-019-0491-3.

Source: PubMed

3
Subscribe