Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells
暂无分享,去创建一个
Bertrand Z. Yeung | Kristopher L. Nazor | Eleni P. Mimitou | A. Regev | C. Lareau | R. Satija | Peter Smibert | Efthymia Papalexi | P. Thakore | K. Nazor | Y. Hao | S. Sakaguchi | V. Sankaran | J. Wing | Leif S. Ludwig | Tse-Shun Huang | Tatsuya Kibayashi | Wendy Luo | A. L. Zorzetto-Fernandes | Kelvin Y. Chen | Yusuke Takeshima | Mayu Hata
[1] Howard Y. Chang,et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis , 2021, Nature Genetics.
[2] Bertrand Z. Yeung,et al. Simultaneous single cell measurements of intranuclear proteins and gene expression , 2021, bioRxiv.
[3] Avi Srivastava,et al. Multimodal single-cell chromatin analysis with Signac , 2020, bioRxiv.
[4] Sarah E. Pierce,et al. High-throughput single-cell chromatin accessibility CRISPR screens enable unbiased identification of regulatory networks in cancer , 2020, Nature Communications.
[5] Raphael Gottardo,et al. Integrated analysis of multimodal single-cell data , 2020, Cell.
[6] Lucas T. Graybuck,et al. TEA-seq: a trimodal assay for integrated single cell measurement of transcription, epitopes, and chromatin accessibility , 2020, bioRxiv.
[7] Lucas T. Graybuck,et al. TEA-seq: a trimodal assay for integrated single cell measurement of transcription, epitopes, and chromatin accessibility , 2020, bioRxiv.
[8] Hatice S. Kaya-Okur,et al. Single-cell analysis of chromatin silencing programs in developmental and tumor progression , 2020, bioRxiv.
[9] G. Castelo-Branco,et al. Single-cell profiling of histone modifications in the mouse brain , 2020 .
[10] D. Landau,et al. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics , 2020, Nature Reviews Genetics.
[11] A. Regev,et al. Purifying Selection against Pathogenic Mitochondrial DNA in Human T Cells. , 2020, The New England journal of medicine.
[12] M. Peakman,et al. Follicular helper T cell profiles predict response to costimulation blockade in type 1 diabetes , 2020, Nature Immunology.
[13] I. Amit,et al. Coupled scRNA-Seq and Intracellular Protein Activity Reveal an Immunosuppressive Role of TREM2 in Cancer , 2020, Cell.
[14] Mark S. Anderson,et al. T cell subset-selective IL2RA enhancers shape autoimmune diabetes risk , 2020, bioRxiv.
[15] Martin J. Aryee,et al. Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling , 2020, Nature Biotechnology.
[16] Aviv Regev,et al. Chromatin Potential Identified by Shared Single-Cell Profiling of RNA and Chromatin , 2020, Cell.
[17] A. Trumpp,et al. Quantitative proteomics reveals specific metabolic features of Acute Myeloid Leukemia stem cells. , 2020, Blood.
[18] Howard Y. Chang,et al. ArchR: An integrative and scalable software package for single-cell chromatin accessibility analysis , 2020, bioRxiv.
[19] Thomas M. Norman,et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing , 2020, Nature Biotechnology.
[20] M. Akkoyunlu,et al. Disease Stage-Specific Pathogenicity of CD138 (Syndecan 1)-Expressing T Cells in Systemic Lupus Erythematosus , 2020, bioRxiv.
[21] J. Tsang,et al. Normalizing and denoising protein expression data from droplet-based single cell profiling , 2020, Nature Communications.
[22] A. Schier. Single-cell biology: beyond the sum of its parts , 2020, Nature Methods.
[23] S. Preissl,et al. Single-cell multimodal omics: the power of many , 2020, Nature Methods.
[24] Sean C. Bendall,et al. Immune monitoring using mass cytometry and related high-dimensional imaging approaches , 2019, Nature Reviews Rheumatology.
[25] Lior Pachter,et al. Highly multiplexed single-cell RNA-seq by DNA oligonucleotide tagging of cellular proteins , 2019, Nature Biotechnology.
[26] C. Lareau,et al. Longitudinal assessment of clonal mosaicism in humanhematopoiesis via mitochondrial mutation tracking. , 2019, Blood advances.
[27] Jonathan S. Packer,et al. Massively multiplex chemical transcriptomics at single-cell resolution , 2019, Science.
[28] Howard Y. Chang,et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia , 2019, Nature Biotechnology.
[29] Jean Yee Hwa Yang,et al. CiteFuse enables multi-modal analysis of CITE-seq data , 2019, bioRxiv.
[30] B. Ren,et al. An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome , 2019, Nature Structural & Molecular Biology.
[31] Jason D. Buenrostro,et al. Inference and effects of barcode multiplets in droplet-based single-cell assays , 2019, Nature Communications.
[32] Kun Zhang,et al. High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell , 2019, Nature Biotechnology.
[33] Neva C. Durand,et al. Activity-by-Contact model of enhancer-promoter regulation from thousands of CRISPR perturbations , 2019, Nature Genetics.
[34] Jason D. Buenrostro,et al. Droplet-based combinatorial indexing for massive-scale single-cell chromatin accessibility , 2019, Nature Biotechnology.
[35] Jennifer L Hu,et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices , 2019, Nature Methods.
[36] Paul J. Hoffman,et al. Comprehensive Integration of Single-Cell Data , 2018, Cell.
[37] Duhee Bang,et al. Multiplexed single-cell RNA-seq via transient barcoding for simultaneous expression profiling of various drug perturbations , 2019, Science Advances.
[38] Neville E. Sanjana,et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells , 2019, Nature Methods.
[39] Howard Y. Chang,et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion , 2019, Nature Biotechnology.
[40] Allon M Klein,et al. Scrublet: Computational Identification of Cell Doublets in Single-Cell Transcriptomic Data. , 2019, Cell systems.
[41] Hatice S. Kaya-Okur,et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells , 2019, Nature Communications.
[42] Martin J. Aryee,et al. Lineage Tracing in Humans Enabled by Mitochondrial Mutations and Single-Cell Genomics , 2019, Cell.
[43] Cole Trapnell,et al. A Genome-wide Framework for Mapping Gene Regulation via Cellular Genetic Screens , 2019, Cell.
[44] Matthew C. Canver,et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis , 2019, Nature Biotechnology.
[45] R. Satija,et al. Integrative single-cell analysis , 2019, Nature Reviews Genetics.
[46] Jacob M. Schreiber,et al. A Genome-wide Framework for Mapping Gene Regulation via Cellular Genetic Screens , 2019, Cell.
[47] Bertrand Z. Yeung,et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics , 2018, Genome Biology.
[48] Bo Li,et al. Nuclei multiplexing with barcoded antibodies for single-nucleus genomics , 2018, Nature Communications.
[49] Lior Pachter,et al. The Barcode, UMI, Set format and BUStools , 2018, bioRxiv.
[50] Fan Zhang,et al. Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.
[51] Christoph Hafemeister,et al. Comprehensive integration of single cell data , 2018, bioRxiv.
[52] Eun Sug Park,et al. Protein Barcodes Enable High-Dimensional Single-Cell CRISPR Screens , 2018, Cell.
[53] Andrew C. Adey,et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells , 2018, Science.
[54] Ansuman T. Satpathy,et al. Coupled Single-Cell CRISPR Screening and Epigenomic Profiling Reveals Causal Gene Regulatory Networks , 2018, Cell.
[55] David A. Knowles,et al. Landscape of stimulation-responsive chromatin across diverse human immune cells , 2018, Nature Genetics.
[56] Jonathan S. Weissman,et al. MULTI-seq: Scalable sample multiplexing for single-cell RNA sequencing using lipid-tagged indices , 2018, bioRxiv.
[57] Samuel L. Wolock,et al. Scrublet: computational identification of cell doublets in single-cell transcriptomic data , 2018, bioRxiv.
[58] Mauro J. Muraro,et al. Combined quantification of intracellular (phospho-)proteins and transcriptomics from fixed single cells , 2018, bioRxiv.
[59] Martin J. Aryee,et al. Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation , 2018, Cell.
[60] Howard Y. Chang,et al. Genetic determinants of co-accessible chromatin regions in activated T cells across humans , 2018, Nature Genetics.
[61] Paul Hoffman,et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.
[62] Akiko Seki,et al. Optimized RNP transfection for highly efficient CRISPR/Cas9-mediated gene knockout in primary T cells , 2018, The Journal of experimental medicine.
[63] Charlotte Soneson,et al. Bias, robustness and scalability in single-cell differential expression analysis , 2018, Nature Methods.
[64] Chun Jimmie Ye,et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation , 2017, Nature Biotechnology.
[65] Vanessa M. Peterson,et al. Multiplexed quantification of proteins and transcripts in single cells , 2017, Nature Biotechnology.
[66] Howard Y. Chang,et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation , 2017, Nature.
[67] Nicholas A. Sinnott-Armstrong,et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues , 2017, Nature Methods.
[68] William J. Greenleaf,et al. chromVAR: Inferring transcription factor-associated accessibility from single-cell epigenomic data , 2017, Nature Methods.
[69] H. Swerdlow,et al. Large-scale simultaneous measurement of epitopes and transcriptomes in single cells , 2017, Nature Methods.
[70] M. Hurles,et al. Somatic mutations reveal asymmetric cellular dynamics in the early human embryo , 2017, Nature.
[71] Haiyan Tan,et al. Integrative Proteomics and Phosphoproteomics Profiling Reveals Dynamic Signaling Networks and Bioenergetics Pathways Underlying T Cell Activation , 2017, Immunity.
[72] L. Steinmetz,et al. Human haematopoietic stem cell lineage commitment is a continuous process , 2017, Nature Cell Biology.
[73] I. Amit,et al. Dissecting Immune Circuits by Linking CRISPR-Pooled Screens with Single-Cell RNA-Seq , 2016, Cell.
[74] Thomas M. Norman,et al. Perturb-Seq: Dissecting Molecular Circuits with Scalable Single-Cell RNA Profiling of Pooled Genetic Screens , 2016, Cell.
[75] Grace X. Y. Zheng,et al. Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.
[76] Howard Y. Chang,et al. Lineage-specific and single cell chromatin accessibility charts human hematopoiesis and leukemia evolution , 2016, Nature Genetics.
[77] Neville E Sanjana,et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening , 2016, Nature Protocols.
[78] Lior Pachter,et al. Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.
[79] R. Elkon,et al. Functional genetic screens for enhancer elements in the human genome using CRISPR-Cas9 , 2016, Nature Biotechnology.
[80] Jennifer A. Doudna,et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins , 2015, Proceedings of the National Academy of Sciences.
[81] Evan Z. Macosko,et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.
[82] G. Kaplan,et al. Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns. , 2013, The Journal of infectious diseases.
[83] M. Hatano,et al. CXCR4 Expression on Activated B Cells Is Downregulated by CD63 and IL-21 , 2011, The Journal of Immunology.
[84] S. Green,et al. The known and the unknown , 2010, Journal of stem cells & regenerative medicine.
[85] Helen Bevan,et al. The power of many , 2010, Optician.
[86] Clifford A. Meyer,et al. Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.
[87] H. Schneider,et al. CTLA-4 trafficking and surface expression. , 2008, Trends in immunology.
[88] Alexander Hoffmann,et al. Generation and Activation of Multiple Dimeric Transcription Factors within the NF-κB Signaling System , 2008, Molecular and Cellular Biology.
[89] M. Bokarewa,et al. Phenotypic and functional characterization of human CD25+ B cells , 2006, Immunology.
[90] F. Macian,et al. NFAT proteins: key regulators of T-cell development and function , 2005, Nature Reviews Immunology.
[91] F. Sánchez‐Madrid,et al. CD69 is an immunoregulatory molecule induced following activation. , 2005, Trends in immunology.
[92] Dan R. Littman,et al. The Role of CXCR4 in Maintaining Peripheral B Cell Compartments and Humoral Immunity , 2004, The Journal of experimental medicine.
[93] E. Wherry,et al. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells , 2003, Nature Immunology.
[94] Lin Chen,et al. Transcriptional regulation by calcium, calcineurin, and NFAT. , 2003, Genes & development.
[95] J. Allison,et al. ICOS co-stimulatory receptor is essential for T-cell activation and function , 2001, Nature.
[96] G. Freeman,et al. Mouse Inducible Costimulatory Molecule (ICOS) Expression Is Enhanced by CD28 Costimulation and Regulates Differentiation of CD4+ T Cells1 , 2000, The Journal of Immunology.
[97] Johannes Gerdes,et al. The Ki‐67 protein: From the known and the unknown , 2000, Journal of cellular physiology.
[98] B. Alarcón,et al. Triggering the TCR complex causes the downregulation of nonengaged receptors by a signal transduction-dependent mechanism. , 2000, Immunity.
[99] G. Schuler,et al. Generation of large numbers of fully mature and stable dendritic cells from leukapheresis products for clinical application. , 1999, Journal of immunological methods.
[100] J. Bluestone,et al. Regulation of CTLA-4 expression during T cell activation. , 1996, Journal of immunology.
[101] S. Tonegawa,et al. Surface expression of alternative forms of the TCR/CD3 complex. , 1991, Proceedings of the National Academy of Sciences of the United States of America.
[102] E. Lander,et al. Genomic mapping by fingerprinting random clones: a mathematical analysis. , 1988, Genomics.
[103] F. Harrisson. Beyond the sum of its parts: practice, education and community , 2008 .
[104] A. Balsari,et al. Flow cytometric analysis of activation markers on stimulated T cells and their correlation with cell proliferation. , 1997, Cytometry.
[105] H. Piaggio. Mathematical Analysis , 1955, Nature.