A Single Cell Transcriptomic Atlas Characterizes Aging Tissues in the Mouse

Aging is characterized by a progressive loss of physiological integrity, leading to impaired function and increased vulnerability to death1. Despite rapid advances over recent years, many of the molecular and cellular processes which underlie progressive loss of healthy physiology are poorly understood2. To gain a better insight into these processes we have created a single cell transcriptomic atlas across the life span of Mus musculus which includes data from 23 tissues and organs. We discovered cell-specific changes occurring across multiple cell types and organs, as well as age related changes in the cellular composition of different organs. Using single-cell transcriptomic data we were able to assess cell type specific manifestations of different hallmarks of aging, such as senescence3, genomic instability4 and changes in the organism’s immune system2. This Tabula Muris Senis provides a wealth of new molecular information about how the most significant hallmarks of aging are reflected in a broad range of tissues and cell types.

[1]  Mauricio O. Carneiro,et al.  From FastQ Data to High‐Confidence Variant Calls: The Genome Analysis Toolkit Best Practices Pipeline , 2013, Current protocols in bioinformatics.

[2]  P. Galle,et al.  Hepatocyte-specific deletion of IL1-RI attenuates liver injury by blocking IL-1 driven autoinflammation. , 2018, Journal of hepatology.

[3]  Vincent A. Traag,et al.  From Louvain to Leiden: guaranteeing well-connected communities , 2018, Scientific Reports.

[4]  Lars Velten,et al.  Large-Scale Low-Cost NGS Library Preparation Using a Robust Tn5 Purification and Tagmentation Protocol , 2017, G3: Genes, Genomes, Genetics.

[5]  Fabian J Theis,et al.  PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells , 2019, Genome Biology.

[6]  Manuel Serrano,et al.  The Hallmarks of Aging , 2013, Cell.

[7]  Alan Ruttenberg,et al.  The Cell Ontology 2016: enhanced content, modularization, and ontology interoperability , 2016, J. Biomed. Semant..

[8]  Marcus J. Claesson,et al.  Composition, variability, and temporal stability of the intestinal microbiota of the elderly , 2010, Proceedings of the National Academy of Sciences.

[9]  Cellular senescence and cancer , 1999, The Journal of pathology.

[10]  S. Quake,et al.  A survey of human brain transcriptome diversity at the single cell level , 2015, Proceedings of the National Academy of Sciences.

[11]  J. Vijg,et al.  Genome instability and aging. , 2013, Annual review of physiology.

[12]  P. Zhang,et al.  Downregulation of ATP1A1 promotes cancer development in renal cell carcinoma , 2017, Clinical Proteomics.

[13]  Spyros Darmanis,et al.  High-affinity allergen-specific human antibodies cloned from single IgE B cell transcriptomes , 2018, Science.

[14]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[15]  James T. Webber,et al.  Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris , 2018, Nature.

[16]  M. DePristo,et al.  The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. , 2010, Genome research.

[17]  Anne E Carpenter,et al.  CellProfiler 3.0: Next-generation image processing for biology , 2018, PLoS biology.

[18]  N. Neff,et al.  Developmental Heterogeneity of Microglia and Brain Myeloid Cells Revealed by Deep Single-Cell RNA Sequencing , 2018, Neuron.

[19]  Evan Z. Macosko,et al.  Single‐Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell‐State Changes , 2019, Immunity.

[20]  Yingling Liu,et al.  Classical and alternative activation of rat hepatic sinusoidal endothelial cells by inflammatory stimuli. , 2013, Experimental and molecular pathology.

[21]  Qiang Shen,et al.  The AP-1 transcription factor regulates postnatal mammary gland development. , 2006, Developmental biology.

[22]  Jianjun Hu,et al.  The Role of IL-1 Family Members and Kupffer Cells in Liver Regeneration , 2016, BioMed research international.

[23]  Jean-Loup Guillaume,et al.  Fast unfolding of communities in large networks , 2008, 0803.0476.

[24]  Anneliese O. Speak,et al.  T cell fate and clonality inference from single cell transcriptomes , 2016, Nature Methods.

[25]  Åsa K. Björklund,et al.  Tn5 transposase and tagmentation procedures for massively scaled sequencing projects , 2014, Genome research.

[26]  K. Andersson,et al.  The link between vascular dysfunction, bladder ischemia, and aging bladder dysfunction , 2017, Therapeutic advances in urology.

[27]  Pei Li,et al.  The inflammatory cytokine TNF-α promotes the premature senescence of rat nucleus pulposus cells via the PI3K/Akt signaling pathway , 2017, Scientific Reports.

[28]  Marc Salit,et al.  Synthetic Spike-in Standards Improve Run-Specific Systematic Error Analysis for DNA and RNA Sequencing , 2012, PloS one.

[29]  H. Simon,et al.  Living and dying for inflammation: neutrophils, eosinophils, basophils. , 2013, Trends in immunology.

[30]  T. Rando Stem cells, ageing and the quest for immortality , 2006, Nature.

[31]  O. Devuyst,et al.  Uromodulin is expressed in the distal convoluted tubule, where it is critical for regulation of the sodium chloride cotransporter NCC. , 2018, Kidney international.

[32]  J. Campisi Aging, cellular senescence, and cancer. , 2013, Annual review of physiology.

[33]  Kajia Cao,et al.  Lamin B1 depletion in senescent cells triggers large-scale changes in gene expression and the chromatin landscape , 2013, Genes & development.

[34]  O. Dreesen,et al.  Biomarkers of Cellular Senescence and Skin Aging , 2018, Front. Genet..

[35]  C. Weyand,et al.  Understanding immunosenescence to improve responses to vaccines , 2013, Nature Immunology.

[36]  M. Demaria,et al.  Hallmarks of Cellular Senescence. , 2018, Trends in cell biology.

[37]  A. van Oudenaarden,et al.  Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations , 2017, Nature Methods.

[38]  O. Devuyst,et al.  Uromodulin: from physiology to rare and complex kidney disorders , 2017, Nature Reviews Nephrology.

[39]  E. Verdin,et al.  Sirtuins: critical regulators at the crossroads between cancer and aging , 2007, Oncogene.

[40]  Principal Investigators,et al.  Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris , 2018 .

[41]  James T. Webber,et al.  The murine transcriptome reveals global aging nodes with organ-specific phase and amplitude , 2019, bioRxiv.

[42]  T. Finkel,et al.  The metabolic regulation of aging , 2015, Nature Medicine.

[43]  E. Srivatsan,et al.  Cellular senescence and tumor suppressor gene p16 , 2012, International journal of cancer.

[44]  Kerstin B. Meyer,et al.  BBKNN: fast batch alignment of single cell transcriptomes , 2019, Bioinform..

[45]  Quy H. Nguyen,et al.  Experimental Considerations for Single-Cell RNA Sequencing Approaches , 2018, Front. Cell Dev. Biol..

[46]  Kyung‐Jin Min,et al.  Sirtuin signaling in cellular senescence and aging , 2019, BMB reports.

[47]  Anne-Marie Mes-Masson,et al.  Regulation of E2Fs and senescence by PML nuclear bodies. , 2011, Genes & development.

[48]  Irving L. Weissman,et al.  Hoxb5 marks long-term haematopoietic stem cells revealing a homogenous perivascular niche , 2016, Nature.

[49]  Mark S. Schmidt,et al.  Aging-related inflammation driven by cellular senescence enhances NAD consumption via activation of CD38+ pro-inflammatory macrophages , 2019, bioRxiv.

[50]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[51]  A. Nakashima,et al.  Identification of cellular senescence-specific genes by comparative transcriptomics , 2016, Scientific Reports.

[52]  Nomeda Girnius,et al.  The cJUN NH2-terminal kinase (JNK) pathway contributes to mouse mammary gland remodeling during involution , 2018, Cell Death & Differentiation.

[53]  Phillip G. Popovich,et al.  Novel Markers to Delineate Murine M1 and M2 Macrophages , 2015, PloS one.

[54]  Fabian J. Theis,et al.  PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells , 2017, Genome Biology.

[55]  A. Suskind The Aging Overactive Bladder: a Review of Aging-Related Changes from the Brain to the Bladder , 2017, Current Bladder Dysfunction Reports.

[56]  E. Lane,et al.  Lamin B1 fluctuations have differential effects on cellular proliferation and senescence , 2013, The Journal of cell biology.

[57]  D. Grundy,et al.  Age-related changes in afferent pathways and urothelial function in the male mouse bladder , 2014, The Journal of physiology.

[58]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[59]  A. Carnero,et al.  Markers of cellular senescence. , 2013, Methods in molecular biology.

[60]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[61]  Åsa K. Björklund,et al.  Smart-seq2 for sensitive full-length transcriptome profiling in single cells , 2013, Nature Methods.

[62]  T. Dudnakova,et al.  Methods Molecular Biology , 2016 .

[63]  J. Nikolich-Žugich The twilight of immunity: emerging concepts in aging of the immune system , 2017, Nature Immunology.

[64]  G. Christ,et al.  Age-related alterations in regeneration of the urinary bladder after subtotal cystectomy. , 2013, The American journal of pathology.

[65]  T. Newman,et al.  Thymic involution and rising disease incidence with age , 2018, Proceedings of the National Academy of Sciences.

[66]  C. Weyand,et al.  Successful and Maladaptive T Cell Aging. , 2017, Immunity.

[67]  P. Linsley,et al.  MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data , 2015, Genome Biology.

[68]  Y. Isaka Epidermal growth factor as a prognostic biomarker in chronic kidney diseases. , 2016, Annals of translational medicine.

[69]  David A. Knowles,et al.  Batch effects and the effective design of single-cell gene expression studies , 2016, bioRxiv.

[70]  David A. Knowles,et al.  Batch effects and the effective design of single-cell gene expression studies , 2016, Scientific Reports.