Three‐dimensional chromatin re‐organization during muscle stem cell aging
暂无分享,去创建一个
Stephen C. J. Parker | C. Aguilar | P. Fraczek | Jacqueline A. Larouche | K. Sabin | Benjamin A. Yang | C. A. Aguilar
[1] Luís A. Nunes Amaral,et al. Aging is associated with a systemic length-associated transcriptome imbalance , 2022, Nature aging.
[2] Wei Zhou,et al. Decoding the spatial chromatin organization and dynamic epigenetic landscapes of macrophage cells during differentiation and immune activation , 2022, Nature Communications.
[3] Albert E. Almada,et al. Prolonged FOS activity disrupts a global myogenic transcriptional program by altering 3D chromatin architecture in primary muscle progenitor cells , 2022, Skeletal muscle.
[4] A. Brunet,et al. Ageing and rejuvenation of tissue stem cells and their niches , 2022, Nature reviews. Molecular cell biology.
[5] Wei Wang,et al. An injury-responsive Rac-to-Rho GTPase switch drives activation of muscle stem cells through rapid cytoskeletal remodeling. , 2022, Cell stem cell.
[6] L. Harries. Dysregulated RNA processing and metabolism: a new hallmark of ageing and provocation for cellular senescence , 2022, The FEBS journal.
[7] L. Mirny,et al. Dynamics of CTCF- and cohesin-mediated chromatin looping revealed by live-cell imaging , 2022, Science.
[8] Yu Xin Wang,et al. Primary cilia on muscle stem cells are critical to maintain regenerative capacity and are lost during aging , 2022, Nature Communications.
[9] Michael Q. Zhang,et al. MyoD is a 3D genome structure organizer for muscle cell identity , 2022, Nature communications.
[10] C. Mozzetta,et al. Epigenetic control of muscle stem cells: time for a new dimension. , 2022, Trends in genetics : TIG.
[11] Rafael Riudavets Puig,et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding profiles , 2021, Nucleic Acids Res..
[12] OUP accepted manuscript , 2022, Bioinformatics.
[13] P. Muñoz-Cánoves,et al. Control of satellite cell function in muscle regeneration and its disruption in ageing , 2021, Nature Reviews Molecular Cell Biology.
[14] Erika M. Deoudes,et al. Plotgardener: Cultivating precise multi-panel figures in R , 2021, bioRxiv.
[15] Xiaona Chen,et al. CRISPR/Cas9/AAV9-mediated in vivo editing identifies MYC regulation of 3D genome in skeletal muscle stem cell , 2021, Stem cell reports.
[16] David A. Selck,et al. Single-cell measurement of higher-order 3D genome organization with scSPRITE , 2021, Nature Biotechnology.
[17] J. Redondo,et al. CHD4 ensures stem cell lineage fidelity during skeletal muscle regeneration , 2021, Stem cell reports.
[18] Myungjin Kim,et al. Sestrins regulate muscle stem cell metabolic homeostasis , 2021, Stem cell reports.
[19] Jeongmoon J. Choi,et al. Murine muscle stem cell response to perturbations of the neuromuscular junction are attenuated with aging , 2021, eLife.
[20] C. Aguilar,et al. Neutrophil and natural killer cell imbalances prevent muscle stem cell–mediated regeneration following murine volumetric muscle loss , 2021, bioRxiv.
[21] Michael Q. Zhang,et al. The loss of heterochromatin is associated with multiscale three-dimensional genome reorganization and aberrant transcription during cellular senescence , 2021, Genome research.
[22] Howard Y. Chang,et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis , 2021, Nature Genetics.
[23] F. Relaix,et al. Perspectives on skeletal muscle stem cells , 2021, Nature Communications.
[24] M. Rudnicki,et al. Analysis of human satellite cell dynamics on cultured adult skeletal muscle myofibers , 2021, Skeletal Muscle.
[25] Howard Y. Chang,et al. Chromatin and gene-regulatory dynamics of the developing human cerebral cortex at single-cell resolution , 2020, Cell.
[26] Raphael Gottardo,et al. Integrated analysis of multimodal single-cell data , 2020, Cell.
[27] Fidel Ramírez,et al. pyGenomeTracks: reproducible plots for multivariate genomic datasets , 2020, Bioinform..
[28] OUP accepted manuscript , 2021, Nucleic Acids Research.
[29] Juan M. Vaquerizas,et al. FAN-C: a feature-rich framework for the analysis and visualisation of chromosome conformation capture data , 2020, Genome Biology.
[30] Wenqing Cai,et al. Enhancer dependence of cell-type–specific gene expression increases with developmental age , 2020, Proceedings of the National Academy of Sciences.
[31] Jeongmoon J. Choi,et al. Dissecting Murine Muscle Stem Cell Aging through Regeneration Using Integrative Genomic Analysis , 2020, Cell reports.
[32] Irving L. Weissman,et al. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse , 2020, Nature.
[33] Jackson Jones,et al. KLF3 Mediates Epidermal Differentiation through the Epigenomic Writer CBP , 2020, iScience.
[34] S. Benitah,et al. Circadian Regulation of Adult Stem Cell Homeostasis and Aging. , 2020, Cell stem cell.
[35] Rolf Backofen,et al. Galaxy HiCExplorer 3: a web server for reproducible Hi-C, capture Hi-C and single-cell Hi-C data analysis, quality control and visualization , 2020, Nucleic Acids Res..
[36] M. Looso,et al. Beyond accessibility: ATAC-seq footprinting unravels kinetics of transcription factor binding during zygotic genome activation , 2019, bioRxiv.
[37] V. Weaver,et al. Wnt4 from the Niche Controls the Mechano-Properties and Quiescent State of Muscle Stem Cells. , 2019, Cell stem cell.
[38] A. Uezumi,et al. Sustained expression of HeyL is critical for the proliferation of muscle stem cells in overloaded muscle , 2019, eLife.
[39] J. Herrero,et al. Enhancer accessibility and CTCF occupancy underlie asymmetric TAD architecture and cell type specific genome topology , 2019, Nature Communications.
[40] Anshul Kundaje,et al. The ENCODE Blacklist: Identification of Problematic Regions of the Genome , 2019, Scientific Reports.
[41] Paul J. Hoffman,et al. Comprehensive Integration of Single-Cell Data , 2018, Cell.
[42] Jing Wang,et al. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs , 2019, Nucleic Acids Res..
[43] K. Tan,et al. Spatial Genome Re-organization between Fetal and Adult Hematopoietic Stem Cells , 2019, bioRxiv.
[44] Andrew C. Adey,et al. The accessible chromatin landscape of the murine hippocampus at single-cell resolution , 2019, Genome research.
[45] Param Priya Singh,et al. The Genetics of Aging: A Vertebrate Perspective , 2019, Cell.
[46] Yu Xin Wang,et al. EGFR-Aurka Signaling Rescues Polarity and Regeneration Defects in Dystrophin-Deficient Muscle Stem Cells by Increasing Asymmetric Divisions. , 2019, Cell stem cell.
[47] Andrew J. Hill,et al. The single cell transcriptional landscape of mammalian organogenesis , 2019, Nature.
[48] M. Gessler,et al. Cell-autonomous and redundant roles of Hey1 and HeyL in muscle stem cells: HeyL requires Hes1 to bind diverse DNA sites , 2019, Development.
[49] Shawn C. Little,et al. Oncogenic Notch promotes long-range regulatory interactions within hyperconnected 3D cliques , 2019, bioRxiv.
[50] S. Dell’Orso,et al. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions , 2019, Development.
[51] Lai Guan Ng,et al. Dimensionality reduction for visualizing single-cell data using UMAP , 2018, Nature Biotechnology.
[52] R. Nagatomi,et al. The Ubiquitin-Proteasome System Is Indispensable for the Maintenance of Muscle Stem Cells , 2018, Stem cell reports.
[53] Fan Zhang,et al. Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.
[54] Thomas Pengo,et al. Muscle Satellite Cell Cross-Talk with a Vascular Niche Maintains Quiescence via VEGF and Notch Signaling. , 2018, Cell stem cell.
[55] Andrew C. Adey,et al. Cicero Predicts cis-Regulatory DNA Interactions from Single-Cell Chromatin Accessibility Data. , 2018, Molecular cell.
[56] Kevin R. Moon,et al. Recovering Gene Interactions from Single-Cell Data Using Data Diffusion , 2018, Cell.
[57] Zev J. Gartner,et al. DoubletFinder: Doublet detection in single-cell RNA sequencing data using artificial nearest neighbors , 2018, bioRxiv.
[58] Neva C. Durand,et al. The Energetics and Physiological Impact of Cohesin Extrusion , 2018, Cell.
[59] Tom H. Cheung,et al. High-dimensional single-cell cartography reveals novel skeletal muscle resident cell populations , 2018, bioRxiv.
[60] Kyoung-Jae Won,et al. Rev-erbα dynamically modulates chromatin looping to control circadian gene transcription , 2018, Science.
[61] F. Naef,et al. Clock-dependent chromatin topology modulates circadian transcription and behavior , 2018, Genes & development.
[62] Shin Fujimaki,et al. Notch1 and Notch2 Coordinately Regulate Stem Cell Function in the Quiescent and Activated States of Muscle Satellite Cells , 2018, Stem cells.
[63] J. Ellenberg,et al. Topologically associating domains and chromatin loops depend on cohesin and are regulated by CTCF, WAPL, and PDS5 proteins , 2017, The EMBO journal.
[64] C. Fan,et al. A CREB-MPP7-AMOT Regulatory Axis Controls Muscle Stem Cell Expansion and Self-Renewal Competence. , 2017, Cell reports.
[65] A. Tanay,et al. Multiscale 3D Genome Rewiring during Mouse Neural Development , 2017, Cell.
[66] Zhaoyang Feng,et al. A conserved KLF-autophagy pathway modulates nematode lifespan and mammalian age-associated vascular dysfunction , 2017, Nature Communications.
[67] Bing Ren,et al. The Three-Dimensional Organization of Mammalian Genomes. , 2017, Annual review of cell and developmental biology.
[68] Erez Lieberman Aiden,et al. Cohesin Loss Eliminates All Loop Domains , 2017, Cell.
[69] Nuno A. Fonseca,et al. Two independent modes of chromatin organization revealed by cohesin removal , 2017, Nature.
[70] J. I. Izpisúa Belmonte,et al. Regulation of Stem Cell Aging by Metabolism and Epigenetics. , 2017, Cell metabolism.
[71] Erez Lieberman Aiden,et al. Myc Regulates Chromatin Decompaction and Nuclear Architecture during B Cell Activation. , 2017, Molecular cell.
[72] Vikki M. Weake,et al. Transcriptional Signatures of Aging. , 2017, Journal of molecular biology.
[73] B. Chabot,et al. The emerging role of alternative splicing in senescence and aging , 2017, Aging cell.
[74] L. Mirny,et al. Targeted Degradation of CTCF Decouples Local Insulation of Chromosome Domains from Genomic Compartmentalization , 2017, Cell.
[75] B. Olwin,et al. Regulation of skeletal muscle stem cells by fibroblast growth factors , 2017, Developmental dynamics : an official publication of the American Association of Anatomists.
[76] Kin Chung Lam,et al. High-resolution TADs reveal DNA sequences underlying genome organization in flies , 2017, bioRxiv.
[77] K. Patel,et al. R-spondin1 Controls Muscle Cell Fusion through Dual Regulation of Antagonistic Wnt Signaling Pathways , 2017, Cell reports.
[78] Ilya M. Flyamer,et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition , 2017, Nature.
[79] D. Cacchiarelli,et al. Transcriptional and Chromatin Dynamics of Muscle Regeneration after Severe Trauma , 2016, Stem cell reports.
[80] Ichiro Manabe,et al. Klf5 regulates muscle differentiation by directly targeting muscle-specific genes in cooperation with MyoD in mice , 2016, eLife.
[81] James T. Robinson,et al. Juicebox Provides a Visualization System for Hi-C Contact Maps with Unlimited Zoom. , 2016, Cell systems.
[82] Neva C. Durand,et al. Juicer Provides a One-Click System for Analyzing Loop-Resolution Hi-C Experiments. , 2016, Cell systems.
[83] Peter H. L. Krijger,et al. Cell-of-Origin-Specific 3D Genome Structure Acquired during Somatic Cell Reprogramming , 2016, Cell stem cell.
[84] Yizhen Wang,et al. KLF7 Regulates Satellite Cell Quiescence in Response to Extracellular Signaling , 2016, Stem cells.
[85] Fidel Ramírez,et al. deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..
[86] Nicola Neretti,et al. Reorganization of chromosome architecture in replicative cellular senescence , 2016, Science Advances.
[87] Ashok Kumar,et al. TRAF6 regulates satellite stem cell self-renewal and function during regenerative myogenesis. , 2016, The Journal of clinical investigation.
[88] T. Rando,et al. Stem cells and healthy aging , 2015, Science.
[89] B. Lenhard,et al. GenomicInteractions: An R/Bioconductor package for manipulating and investigating chromatin interaction data , 2015, BMC Genomics.
[90] H. Blau,et al. The central role of muscle stem cells in regenerative failure with aging , 2015, Nature Medicine.
[91] Qing-Yu He,et al. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization , 2015, Bioinform..
[92] Howard Y. Chang,et al. Single-cell chromatin accessibility reveals principles of regulatory variation , 2015, Nature.
[93] C. Colnot,et al. Role of Muscle Stem Cells During Skeletal Regeneration , 2015, Stem cells.
[94] P. Blackshear,et al. Post-transcriptional regulation of satellite cell quiescence by TTP-mediated mRNA decay , 2015, eLife.
[95] Jing Liang,et al. Chromatin architecture reorganization during stem cell differentiation , 2015, Nature.
[96] Philip A. Ewels,et al. Global Reorganization of the Nuclear Landscape in Senescent Cells , 2015, Cell reports.
[97] Matthew E. Ritchie,et al. limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.
[98] Neva C. Durand,et al. A 3D Map of the Human Genome at Kilobase Resolution Reveals Principles of Chromatin Looping , 2014, Cell.
[99] Jill M Dowen,et al. Control of Cell Identity Genes Occurs in Insulated Neighborhoods in Mammalian Chromosomes , 2014, Cell.
[100] Björn Usadel,et al. Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..
[101] Charity W. Law,et al. voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.
[102] Howard Y. Chang,et al. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position , 2013, Nature Methods.
[103] Carlos A Aguilar,et al. Micro- and nanoscale devices for the investigation of epigenetics and chromatin dynamics. , 2013, Nature nanotechnology.
[104] Robert Gentleman,et al. Software for Computing and Annotating Genomic Ranges , 2013, PLoS Comput. Biol..
[105] Tom H. Cheung,et al. Chromatin Modifications as Determinants of Muscle Stem Cell Quiescence and Chronological Aging , 2013, Cell reports.
[106] Manuel Serrano,et al. The Hallmarks of Aging , 2013, Cell.
[107] M. Matunis,et al. SUMO: a multifaceted modifier of chromatin structure and function. , 2013, Developmental cell.
[108] Thomas R. Gingeras,et al. STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..
[109] M. Rudnicki,et al. Wnt signaling in myogenesis. , 2012, Trends in cell biology.
[110] B. Maher. ENCODE: The human encyclopaedia , 2012, Nature.
[111] Jesse R. Dixon,et al. Topological Domains in Mammalian Genomes Identified by Analysis of Chromatin Interactions , 2012, Nature.
[112] Steven L Salzberg,et al. Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.
[113] Andrew E. Jaffe,et al. Bioinformatics Applications Note Gene Expression the Sva Package for Removing Batch Effects and Other Unwanted Variation in High-throughput Experiments , 2022 .
[114] Yu Xin Wang,et al. Satellite cells, the engines of muscle repair , 2011, Nature Reviews Molecular Cell Biology.
[115] A. Uezumi,et al. Hesr1 and Hesr3 are essential to generate undifferentiated quiescent satellite cells and to maintain satellite cell numbers , 2011, Development.
[116] M. Montminy,et al. CREB Is Activated by Muscle Injury and Promotes Muscle Regeneration , 2011, PloS one.
[117] Kenneth S Boockvar,et al. Recent trends in chronic disease, impairment and disability among older adults in the United States , 2011, BMC geriatrics.
[118] Colin N. Dewey,et al. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.
[119] Marcel Martin. Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .
[120] E. Nishida,et al. ERK5 regulates muscle cell fusion through Klf transcription factors. , 2011, Developmental cell.
[121] J. Crook,et al. G-protein Coupled Receptors in Stem Cell Self-Renewal and Differentiation , 2010, Stem Cell Reviews and Reports.
[122] C. Glass,et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.
[123] I. Amit,et al. Comprehensive mapping of long range interactions reveals folding principles of the human genome , 2011 .
[124] Gonçalo R. Abecasis,et al. The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..
[125] M. Rudnicki,et al. Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. , 2009, Cell stem cell.
[126] Clifford A. Meyer,et al. Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.
[127] Jeanne Shen,et al. A temporal switch from notch to Wnt signaling in muscle stem cells is necessary for normal adult myogenesis. , 2008, Cell stem cell.
[128] Dustin E. Schones,et al. High-Resolution Profiling of Histone Methylations in the Human Genome , 2007, Cell.
[129] Pablo Tamayo,et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.
[130] J. Black,et al. Sp1 and krüppel‐like factor family of transcription factors in cell growth regulation and cancer , 2001, Journal of cellular physiology.
[131] Marc Montminy,et al. Transcriptional regulation by the phosphorylation-dependent factor CREB , 2001, Nature Reviews Molecular Cell Biology.