A single nuclei atlas of aging human abdominal subcutaneous white adipose tissue

Abstract White adipose tissue (WAT) is a robust energy storage and endocrine organ critical for maintaining metabolic health as we age. Our aim was to identify cell-specific transcriptional aberrations that occur in WAT with aging. We leveraged full-length snRNA-Seq to characterize the cellular landscape of human subcutaneous WAT in a prospective cohort of 10 Younger (≤ 30 years) and 10 Older individuals (≥ 65 years) balanced for sex and body mass index (BMI). We highlight that aging WAT is associated with adipocyte hypertrophy, increased proportions of resident macrophages (M2), an upregulated innate immune response and senescence profiles in specific adipocyte populations, highlighting CXCL14 as a biomarker of this process. We also identify novel markers of pre-adipocytes and track their expression levels through pre-adipocyte differentiation. We propose that aging WAT is associated with low-grade inflammation that is managed by a foundation of innate immunity to preserve the metabolic health of the WAT.

[1]  Patrik L. Ståhl,et al.  An integrated single cell and spatial transcriptomic map of human white adipose tissue , 2023, Nature Communications.

[2]  H. Ouyang,et al.  High-resolution aging niche of human adipose tissues , 2023, Signal Transduction and Targeted Therapy.

[3]  M. Chen,et al.  Metastasis suppressor 1 interacts with protein tyrosine phosphatase receptor‐δ to regulate adipogenesis , 2023, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[4]  Steven R Smith,et al.  Isolation of nuclei from frozen human subcutaneous adipose tissue for full-length single-nuclei transcriptional profiling , 2023, STAR protocols.

[5]  Janez Konc,et al.  Genome-wide screening for genetic variants in polyadenylation signal (PAS) sites in mouse selection lines for fatness and leanness , 2022, Mammalian Genome.

[6]  T. Kivisild,et al.  The role of contactin-associated protein-like 2 in neurodevelopmental disease and human cerebral cortex evolution , 2022, Frontiers in Molecular Neuroscience.

[7]  Steven R Smith,et al.  Single cell full-length transcriptome of human subcutaneous adipose tissue reveals unique and heterogeneous cell populations , 2022, iScience.

[8]  C. Lumeng,et al.  Single-nuclei Transcriptome of Human AT Reveals Metabolically Distinct Depot-Specific Adipose Progenitor Subpopulations , 2022, bioRxiv.

[9]  Aaron M. Streets,et al.  Mapping the temporal transcriptional landscape of human white and brown adipogenesis using single-nuclei RNA-seq , 2022, bioRxiv.

[10]  Zhong Sheng Sun,et al.  Putative complement control protein CSMD3 dysfunction impairs synaptogenesis and induces neurodevelopmental disorders , 2022, Brain, Behavior, and Immunity.

[11]  N. LeBrasseur,et al.  A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues , 2021, Nature Communications.

[12]  Gary D Bader,et al.  The reactome pathway knowledgebase 2022 , 2021, Nucleic Acids Res..

[13]  Bernard T. Lee,et al.  A single-cell atlas of human and mouse white adipose tissue , 2021, Nature.

[14]  C. Knibbe,et al.  Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce , 2021, Nature Medicine.

[15]  Aaron M. Streets,et al.  Single-cell multi-omic analysis of thymocyte development reveals drivers of CD4/CD8 lineage commitment , 2021, bioRxiv.

[16]  C. Lindgren,et al.  A distribution-centered approach for analyzing human adipocyte size estimates and their association with obesity-related traits and mitochondrial function , 2021, International Journal of Obesity.

[17]  H. Shan,et al.  Cooperation of ATF4 and CTCF promotes adipogenesis through transcriptional regulation , 2021, Cell Biology and Toxicology.

[18]  M. Lindsay,et al.  Divergent immunometabolic changes in adipose tissue and skeletal muscle with ageing in healthy humans , 2021, The Journal of physiology.

[19]  Santiago J. Carmona,et al.  UCell: Robust and scalable single-cell gene signature scoring , 2021, bioRxiv.

[20]  M. Pellegrini,et al.  Single cell sequencing of human white adipose tissue identifies novel cell states in health and obesity , 2021, Nature Immunology.

[21]  Lei Shen,et al.  The long noncoding RNA MALAT1 modulates adipose loss in cancer-associated cachexia by suppressing adipogenesis through PPAR-γ , 2021, Nutrition & Metabolism.

[22]  G. Rappold,et al.  SHANK2 mutations impair apoptosis, proliferation and neurite outgrowth during early neuronal differentiation in SH-SY5Y cells , 2021, Scientific Reports.

[23]  T. Quesada-López,et al.  The chemokine CXCL14 is negatively associated with obesity and concomitant type-2 diabetes in humans , 2021, International Journal of Obesity.

[24]  F. Fazi,et al.  The role of RNA-binding and ribosomal proteins as specific RNA translation regulators in cellular differentiation and carcinogenesis. , 2020, Biochimica et biophysica acta. Molecular basis of disease.

[25]  S. Mandrup,et al.  Plasticity of Epididymal Adipose Tissue in Response to Diet-Induced Obesity at Single-Nucleus Resolution. , 2020, Cell metabolism.

[26]  Anushya Muruganujan,et al.  The Gene Ontology resource: enriching a GOld mine , 2020, Nucleic Acids Res..

[27]  A. Regev,et al.  snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis , 2020, Nature.

[28]  Raphael Gottardo,et al.  Integrated analysis of multimodal single-cell data , 2020, Cell.

[29]  M. Eberl,et al.  CXCL14 Preferentially Synergizes With Homeostatic Chemokine Receptor Systems , 2020, Frontiers in Immunology.

[30]  Richard G. Lee,et al.  RIPK1 gene variants associate with obesity in humans and can be therapeutically silenced to reduce obesity in mice , 2020, Nature Metabolism.

[31]  M. D. de Angelis,et al.  Increased mitochondrial respiration of adipocytes from metabolically unhealthy obese compared to healthy obese individuals , 2020, Scientific Reports.

[32]  M. Parker,et al.  Expanding the molecular spectrum and the neurological phenotype related to CAMTA1 variants , 2020, Clinical genetics.

[33]  U. Smith,et al.  Altered PTPRD DNA methylation associates with restricted adipogenesis in healthy first-degree relatives of Type 2 diabetes subjects. , 2020, Epigenomics.

[34]  T. Quesada-López,et al.  Reduced circulating levels of chemokine CXCL14 in adolescent girls with polycystic ovary syndrome: normalization after insulin sensitization , 2020, BMJ Open Diabetes Research & Care.

[35]  H. Pan,et al.  Neural EGFL like 1 as a potential pro-chondrogenic, anti-inflammatory dual-functional disease-modifying osteoarthritis drug. , 2020, Biomaterials.

[36]  P. Malatesta,et al.  Cdh4 Down-Regulation Impairs in Vivo Infiltration and Malignancy in Patients Derived Glioblastoma Cells , 2019, International journal of molecular sciences.

[37]  I. Amit,et al.  Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner , 2019, Cell.

[38]  Yibing Liu,et al.  Transcriptome Analysis of Landrace Pig Subcutaneous Preadipocytes during Adipogenic Differentiation , 2019, Genes.

[39]  Stefano Monti,et al.  hypeR: An R Package for Geneset Enrichment Workflows , 2019, bioRxiv.

[40]  Kathleen M. Jagodnik,et al.  ChEA3: transcription factor enrichment analysis by orthogonal omics integration , 2019, Nucleic Acids Res..

[41]  Ivona Percec,et al.  Identification of a mesenchymal progenitor cell hierarchy in adipose tissue , 2019, Science.

[42]  C. Plass,et al.  Random forest-based modelling to detect biomarkers for prostate cancer progression , 2019, Clinical Epigenetics.

[43]  R. Satija,et al.  Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression , 2019, Genome Biology.

[44]  Katherine C. Hall,et al.  Zfp423 Regulates Skeletal Muscle Regeneration and Proliferation , 2019, Molecular and Cellular Biology.

[45]  Andrew J. Hill,et al.  The single cell transcriptional landscape of mammalian organogenesis , 2019, Nature.

[46]  A. Fritsche,et al.  Preadipocytes of obese humans display gender-specific bioenergetic responses to glucose and insulin , 2018, Molecular metabolism.

[47]  T. Osborne,et al.  Differential open chromatin profile and transcriptomic signature define depot-specific human subcutaneous preadipocytes: primary outcomes , 2018, Clinical Epigenetics.

[48]  T. Quesada-López,et al.  CXCL14, a Brown Adipokine that Mediates Brown-Fat-to-Macrophage Communication in Thermogenic Adaptation. , 2018, Cell metabolism.

[49]  Mengle Shao,et al.  Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice , 2018, eLife.

[50]  R. Pique-Regi,et al.  Deconstructing Adipogenesis Induced by β3-Adrenergic Receptor Activation with Single-Cell Expression Profiling. , 2018, Cell metabolism.

[51]  Petra C. Schwalie,et al.  A stromal cell population that inhibits adipogenesis in mammalian fat depots , 2018, Nature.

[52]  S. Kritchevsky,et al.  Cellular Senescence Biomarker p16INK4a+ Cell Burden in Thigh Adipose is Associated With Poor Physical Function in Older Women , 2018, The journals of gerontology. Series A, Biological sciences and medical sciences.

[53]  F. Haczeyni,et al.  Causes and mechanisms of adipocyte enlargement and adipose expansion , 2018, Obesity reviews : an official journal of the International Association for the Study of Obesity.

[54]  S. Joost,et al.  Single cell transcriptomics suggest that human adipocyte progenitor cells constitute a homogeneous cell population , 2017, Stem Cell Research & Therapy.

[55]  A. Carrière,et al.  Identification of the ectoenzyme CD38 as a marker of committed preadipocytes , 2017, International Journal of Obesity.

[56]  Hannah A. Pliner,et al.  Reversed graph embedding resolves complex single-cell trajectories , 2017, Nature Methods.

[57]  A. Scialdone,et al.  Adipocyte Accumulation in the Bone Marrow during Obesity and Aging Impairs Stem Cell-Based Hematopoietic and Bone Regeneration , 2017, Cell stem cell.

[58]  B. Goodpaster,et al.  Metabolic Flexibility in Health and Disease. , 2017, Cell metabolism.

[59]  M. Jensen,et al.  How to Measure Adipose Tissue Insulin Sensitivity , 2017, The Journal of clinical endocrinology and metabolism.

[60]  K. Sakimura,et al.  PKN2 is essential for mouse embryonic development and proliferation of mouse fibroblasts , 2017, Genes to cells : devoted to molecular & cellular mechanisms.

[61]  J. Kirkland,et al.  Aging and adipose tissue: potential interventions for diabetes and regenerative medicine , 2016, Experimental Gerontology.

[62]  N. LeBrasseur,et al.  Identification of Senescent Cells in the Bone Microenvironment , 2016, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[63]  Roland Eils,et al.  Complex heatmaps reveal patterns and correlations in multidimensional genomic data , 2016, Bioinform..

[64]  Davis J. McCarthy,et al.  A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor , 2016, F1000Research.

[65]  Alexandra R. Washabaugh,et al.  Adipose tissue fibrosis, hypertrophy, and hyperplasia: Correlations with diabetes in human obesity , 2016, Obesity.

[66]  M. Gawaz,et al.  CXCL14 as an emerging immune and inflammatory modulator , 2016, Journal of Inflammation.

[67]  M. Jensen,et al.  Targeting senescent cells enhances adipogenesis and metabolic function in old age , 2015, eLife.

[68]  S. Tavazoie,et al.  PTPRN2 and PLCβ1 promote metastatic breast cancer cell migration through PI(4,5)P2‐dependent actin remodeling , 2015, The EMBO journal.

[69]  M. Jensen,et al.  JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age , 2015, Proceedings of the National Academy of Sciences.

[70]  Sean C. Bendall,et al.  Data-Driven Phenotypic Dissection of AML Reveals Progenitor-like Cells that Correlate with Prognosis , 2015, Cell.

[71]  M. Birnbaum,et al.  The Role of PDE3B Phosphorylation in the Inhibition of Lipolysis by Insulin , 2015, Molecular and Cellular Biology.

[72]  G. Shulman Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. , 2014, The New England journal of medicine.

[73]  J. Kirkland,et al.  Cellular senescence and the senescent secretory phenotype in age-related chronic diseases , 2014, Current opinion in clinical nutrition and metabolic care.

[74]  Cole Trapnell,et al.  The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells , 2014, Nature Biotechnology.

[75]  K. Nair,et al.  Adipocyte mitochondrial function is reduced in human obesity independent of fat cell size. , 2014, The Journal of clinical endocrinology and metabolism.

[76]  J. Lawrence,et al.  Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence , 2013, The Journal of cell biology.

[77]  J. Borén,et al.  Ectopic lipid storage and insulin resistance: a harmful relationship , 2013, Journal of Internal Medicine.

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

[79]  J. Campisi,et al.  Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. , 2013, The Journal of clinical investigation.

[80]  M. McNurlan,et al.  Peripheral fat loss and decline in adipogenesis in older humans. , 2013, Metabolism: clinical and experimental.

[81]  S. Kawamoto,et al.  Rbfox3-regulated alternative splicing of Numb promotes neuronal differentiation during development , 2013, The Journal of cell biology.

[82]  V. Korolchuk,et al.  Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response , 2012, Aging cell.

[83]  O. Lesur,et al.  Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans. , 2012, Seminars in immunology.

[84]  G. Fisher,et al.  Receptor type protein tyrosine phosphatases (RPTPs) – roles in signal transduction and human disease , 2012, Journal of Cell Communication and Signaling.

[85]  Yuanyuan Wu,et al.  A modified protocol to maximize differentiation of human preadipocytes and improve metabolic phenotypes , 2012, Obesity.

[86]  O. MacDougald,et al.  Adipose tissue stem cells meet preadipocyte commitment: going back to the future[S] , 2012, Journal of Lipid Research.

[87]  Angel Pan,et al.  A new function of Nell-1 protein in repressing adipogenic differentiation. , 2011, Biochemical and biophysical research communications.

[88]  F. Westermann,et al.  CAMTA1, a 1p36 tumor suppressor candidate, inhibits growth and activates differentiation programs in neuroblastoma cells. , 2011, Cancer research.

[89]  M. Jensen,et al.  Fat tissue, aging, and cellular senescence , 2010, Aging cell.

[90]  B. Spiegelman,et al.  Transcriptional Control of Preadipocyte Determination by Zfp423 , 2010, Nature.

[91]  Robert Ross,et al.  Age-related changes in total and regional fat distribution , 2009, Ageing Research Reviews.

[92]  H. Saito,et al.  Age-associated increase in cytokine production during systemic inflammation: adipose tissue as a major source of IL-6. , 2009, The journals of gerontology. Series A, Biological sciences and medical sciences.

[93]  Judith Campisi,et al.  Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor , 2008, PLoS biology.

[94]  R. Hammer,et al.  White Fat Progenitor Cells Reside in the Adipose Vasculature , 2008, Science.

[95]  Yutaka Takahashi,et al.  CXCL14 enhances insulin-dependent glucose uptake in adipocytes and is related to high-fat diet-induced obesity. , 2007, Biochemical and biophysical research communications.

[96]  A. Merrill,et al.  Aging Up-Regulates Expression of Inflammatory Mediators in Mouse Adipose Tissue1 , 2007, The Journal of Immunology.

[97]  J. Campisi,et al.  Cellular senescence: when bad things happen to good cells , 2007, Nature Reviews Molecular Cell Biology.

[98]  Carlos Cordon-Cardo,et al.  Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas , 2007, Nature.

[99]  M. Desai,et al.  Obesity is associated with macrophage accumulation in adipose tissue. , 2003, The Journal of clinical investigation.

[100]  J. Leszyk,et al.  Mitochondrial Biogenesis and Remodeling during Adipogenesis and in Response to the Insulin Sensitizer Rosiglitazone , 2003, Molecular and Cellular Biology.

[101]  C. Smas,et al.  Function of pref-1 as an inhibitor of adipocyte differentiation , 2000, International Journal of Obesity.

[102]  A. P. Soler,et al.  Interaction of alpha-actinin with the cadherin/catenin cell-cell adhesion complex via alpha-catenin , 1995, The Journal of cell biology.

[103]  E. Wang,et al.  Senescent human fibroblasts resist programmed cell death, and failure to suppress bcl2 is involved. , 1995, Cancer research.

[104]  L. Groop,et al.  Glucose and free fatty acid metabolism in non-insulin-dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. , 1989, The Journal of clinical investigation.

[105]  K. Iyama,et al.  Electron microscopical studies on the genesis of white adipocytes: Differentiation of immature pericytes into adipocytes in transplanted preadipose tissue , 1979, Virchows Archiv. B, Cell pathology including molecular pathology.

[106]  J. Mesirov,et al.  The Molecular Signatures Database (MSigDB) hallmark gene set collection. , 2015, Cell systems.

[107]  Brent L Fogel,et al.  Orchestration of neurodevelopmental programs by RBFOX1: implications for autism spectrum disorder. , 2013, International review of neurobiology.

[108]  S. Farmer Regulation of PPARgamma activity during adipogenesis. , 2005, International journal of obesity.

[109]  Hiroyuki Ogata,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 1999, Nucleic Acids Res..