Gene body DNA hydroxymethylation restricts the magnitude of transcriptional changes during aging

DNA hydroxymethylation (5hmC) is the most abundant oxidative derivative of DNA methylation (5mC) and is typically enriched at enhancers and gene bodies of transcriptionally active and tissue-specific genes. Although aberrant genomic 5hmC has been implicated in many age-related diseases, the functional role of the modification in aging remains largely unknown. Here, we report that 5hmC is stably enriched in multiple aged organs. Using the liver and cerebellum as model organs, we show that 5hmC accumulates in gene bodies associated with tissue-specific function and thereby restricts the magnitude of gene expression changes during aging. Mechanistically, we found that 5hmC decreases binding affinity of splicing factors compared to unmodified cytosine and 5mC, and is correlated with age-related alternative splicing events, suggesting RNA splicing as a potential mediator of 5hmC’s transcriptionally restrictive function. Furthermore, we show that various age-related contexts, such as prolonged quiescence and senescence, are partially responsible for driving the accumulation of 5hmC with age. We provide evidence that this age-related function is conserved in mouse and human tissues, and further show that the modification is altered by regimens known to modulate lifespan. Our findings reveal that 5hmC is a regulator of tissue-specific function and may play a role in regulating longevity.

[1]  H. Herzog,et al.  Sex-specific changes in metabolism during the transition from chow to high-fat diet feeding are abolished in response to dieting in C57BL/6J mice , 2022, International Journal of Obesity.

[2]  Margarita V. Meer,et al.  Tick tock, tick tock: Mouse culture and tissue aging captured by an epigenetic clock , 2022, Aging cell.

[3]  M. Brenowitz,et al.  High throughput and low bias DNA methylation and hydroxymethylation analysis by direct injection mass spectrometry. , 2021, Analytica chimica acta.

[4]  P. Gadue,et al.  Loss of TBX3 enhances pancreatic progenitor generation from human pluripotent stem cells , 2021, Stem cell reports.

[5]  Manolis Maragkakis,et al.  TERA-Seq: true end-to-end sequencing of native RNA molecules for transcriptome characterization , 2021, Nucleic acids research.

[6]  Payel Sen,et al.  Epigenetic dysregulation in cardiovascular aging and disease , 2021, The journal of cardiovascular aging.

[7]  Tong Chen,et al.  EVenn: Easy to create repeatable and editable Venn diagrams and Venn networks online. , 2021, Journal of genetics and genomics = Yi chuan xue bao.

[8]  A. Feuchtinger,et al.  Liver-fibrosis-activated transcriptional networks govern hepatocyte reprogramming and intra-hepatic communication. , 2021, Cell metabolism.

[9]  D. Calvisi,et al.  TBX3 functions as a tumor suppressor downstream of activated CTNNB1 mutants during hepatocarcinogenesis. , 2021, Journal of hepatology.

[10]  C. Yi,et al.  Tissue-specific 5-hydroxymethylcytosine landscape of the human genome , 2020, Nature Communications.

[11]  M. Brenowitz,et al.  DNA methylation and hydroxymethylation analysis using a high throughput and low bias direct injection mass spectrometry platform. , 2021, MethodsX.

[12]  S. Ferrari,et al.  Author contributions , 2021 .

[13]  A. Bergamaschi,et al.  A human tissue map of 5-hydroxymethylcytosines exhibits tissue specificity through gene and enhancer modulation , 2020, Nature Communications.

[14]  Margarita V. Meer,et al.  Reprogramming to recover youthful epigenetic information and restore vision , 2020, Nature.

[15]  J. Lebeck,et al.  Sex-Specific Effect of High-Fat Diet on Glycerol Metabolism in Murine Adipose Tissue and Liver , 2020, Frontiers in Endocrinology.

[16]  S. Sidoli,et al.  Guide for protein fold change and p-value calculation for non-experts in proteomics. , 2020, Molecular omics.

[17]  R. de Cabo,et al.  Disulfiram Treatment Normalizes Body Weight in Obese Mice. , 2020, Cell metabolism.

[18]  L. Freeman,et al.  Sex differences in response to a high fat, high sucrose diet in both the gut microbiome and hypothalamic astrocytes and microglia , 2020, Nutritional neuroscience.

[19]  A. Salminen,et al.  ER stress activates immunosuppressive network: implications for aging and Alzheimer’s disease , 2020, Journal of Molecular Medicine.

[20]  Kian Peng Koh,et al.  Coordination of germ layer lineage choice by TET1 during primed pluripotency , 2020, Genes & development.

[21]  Clifford A. Meyer,et al.  Lisa: inferring transcriptional regulators through integrative modeling of public chromatin accessibility and ChIP-seq data , 2020, Genome Biology.

[22]  A. Leung,et al.  Structure-Mediated RNA Decay by UPF1 and G3BP1. , 2020, Molecular cell.

[23]  Angela N. Brooks,et al.  Full-length transcript characterization of SF3B1 mutation in chronic lymphocytic leukemia reveals downregulation of retained introns , 2018, Nature Communications.

[24]  Chen-Yu Liao,et al.  Alpha-ketoglutarate, an endogenous metabolite, extends lifespan and compresses morbidity in aging mice , 2019, bioRxiv.

[25]  A. Kundaje,et al.  The ENCODE Blacklist: Identification of Problematic Regions of the Genome , 2019, Scientific Reports.

[26]  Xiaolan Fan,et al.  Alpha-ketoglutarate extends Drosophila lifespan by inhibiting mTOR and activating AMPK , 2019, Aging.

[27]  C. Ling,et al.  Epigenetics in Human Obesity and Type 2 Diabetes , 2019, Cell metabolism.

[28]  Angela N. Brooks,et al.  Nanopore native RNA sequencing of a human poly(A) transcriptome , 2018, bioRxiv.

[29]  P. Sen,et al.  The senescent cell epigenome , 2018, Aging.

[30]  Trey Ideker,et al.  DNA Methylation Clocks in Aging: Categories, Causes, and Consequences. , 2018, Molecular cell.

[31]  Ratnesh K Singh,et al.  Immunohistochemical Detection of 5-Methylcytosine and 5-Hydroxymethylcytosine in Developing and Postmitotic Mouse Retina , 2018, Journal of visualized experiments : JoVE.

[32]  G. Pfeifer,et al.  Gene body profiles of 5-hydroxymethylcytosine: potential origin, function and use as a cancer biomarker , 2018, Epigenomics.

[33]  Gordon K Smyth,et al.  The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads , 2018, bioRxiv.

[34]  Steve Horvath,et al.  DNA methylation-based biomarkers and the epigenetic clock theory of ageing , 2018, Nature Reviews Genetics.

[35]  Heng Li,et al.  Minimap2: pairwise alignment for nucleotide sequences , 2017, Bioinform..

[36]  E. Masliah,et al.  ER-associated degradation regulates Alzheimer’s amyloid pathology and memory function by modulating γ-secretase activity , 2017, Nature Communications.

[37]  J. Menezes,et al.  The role of alternative splicing coupled to nonsense-mediated mRNA decay in human disease. , 2017, The international journal of biochemistry & cell biology.

[38]  R. Zukin,et al.  The emerging field of epigenetics in neurodegeneration and neuroprotection , 2017, Nature Reviews Neuroscience.

[39]  Fabian J Theis,et al.  Impulse model-based differential expression analysis of time course sequencing data , 2017, bioRxiv.

[40]  R. Young,et al.  Transcriptional Addiction in Cancer , 2017, Cell.

[41]  Maureen A. Sartor,et al.  annotatr: Genomic regions in context , 2016, bioRxiv.

[42]  I. Lin,et al.  Correlated 5-Hydroxymethylcytosine (5hmC) and Gene Expression Profiles Underpin Gene and Organ-Specific Epigenetic Regulation in Adult Mouse Brain and Liver , 2017, PloS one.

[43]  J. Ule,et al.  Major Shifts in Glial Regional Identity Are a Transcriptional Hallmark of Human Brain Aging , 2017, Cell reports.

[44]  S. Berger,et al.  Epigenetic Mechanisms of Longevity and Aging , 2016, Cell.

[45]  Siddharth S. Dey,et al.  Single-cell 5hmC sequencing reveals chromosome-wide cell-to-cell variability and enables lineage reconstruction , 2016, Nature Biotechnology.

[46]  M. Robinson,et al.  DRIMSeq: a Dirichlet-multinomial framework for multivariate count outcomes in genomics , 2016, F1000Research.

[47]  A. Brunet,et al.  The Aging Epigenome. , 2016, Molecular cell.

[48]  K. Helin,et al.  Role of TET enzymes in DNA methylation, development, and cancer , 2016, Genes & development.

[49]  Cheng Luo,et al.  Structural insight into substrate preference for TET-mediated oxidation , 2015, Nature.

[50]  William A. Pastor,et al.  Simultaneous deletion of the methylcytosine oxidases Tet1 and Tet3 increases transcriptome variability in early embryogenesis , 2015, Proceedings of the National Academy of Sciences.

[51]  M. Futschik,et al.  Novel Human Embryonic Stem Cell Regulators Identified by Conserved and Distinct CpG Island Methylation State , 2015, PloS one.

[52]  B. Kennedy,et al.  H3K36 methylation promotes longevity by enhancing transcriptional fidelity , 2015, Genes & development.

[53]  Haiyuan Yu,et al.  Trimethylation of Lys36 on H3 restricts gene expression change during aging and impacts life span , 2015, Genes & development.

[54]  Edwin Cuppen,et al.  Sambamba: fast processing of NGS alignment formats , 2015, Bioinform..

[55]  V. Bordignon,et al.  Endoplasmic Reticulum Stress, Genome Damage, and Cancer , 2015, Front. Oncol..

[56]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[57]  Lan Lin,et al.  rMATS: Robust and flexible detection of differential alternative splicing from replicate RNA-Seq data , 2014, Proceedings of the National Academy of Sciences.

[58]  Stephanie A. Tammen,et al.  Aging Alters Hepatic DNA Hydroxymethylation, as Measured by Liquid Chromatography/Mass Spectrometry , 2014, Journal of cancer prevention.

[59]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[60]  J. Cadet,et al.  Hydroxyl-radical-induced oxidation of 5-methylcytosine in isolated and cellular DNA , 2014, Nucleic acids research.

[61]  Hilary A. Godwin,et al.  The metabolite α-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR , 2014, Nature.

[62]  Fidel Ramírez,et al.  deepTools: a flexible platform for exploring deep-sequencing data , 2014, Nucleic Acids Res..

[63]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

[64]  Miguel Beato,et al.  bwtool: a tool for bigWig files , 2014, Bioinform..

[65]  Xiaoying Ye,et al.  Roles for Tbx3 in regulation of two-cell state and telomere elongation in mouse ES cells , 2013, Scientific Reports.

[66]  J. Thomson,et al.  Edinburgh Research Explorer 5-hydroxymethylcytosine profiling as an indicator of cellular state , 2013 .

[67]  K. Shirahige,et al.  PPARγ-induced PARylation promotes local DNA demethylation by production of 5-hydroxymethylcytosine , 2013, Nature Communications.

[68]  Zechen Chong,et al.  Ascorbic acid enhances Tet-mediated 5-methylcytosine oxidation and promotes DNA demethylation in mammals. , 2013, Journal of the American Chemical Society.

[69]  Mohammad M. Karimi,et al.  Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells , 2013, Nature.

[70]  Ellen T. Gelfand,et al.  The Genotype-Tissue Expression (GTEx) project , 2013, Nature Genetics.

[71]  A. H. Smits,et al.  Dynamic Readers for 5-(Hydroxy)Methylcytosine and Its Oxidized Derivatives , 2013, Cell.

[72]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[73]  N. Heintz,et al.  MeCP2 binds to 5hmc enriched within active genes and accessible chromatin in the nervous system , 2012, Cell.

[74]  Kevin W Eliceiri,et al.  NIH Image to ImageJ: 25 years of image analysis , 2012, Nature Methods.

[75]  Colm E. Nestor,et al.  Tissue type is a major modifier of the 5-hydroxymethylcytosine content of human genes. , 2012, Genome research.

[76]  R. Stark,et al.  DiffBind : Differential binding analysis of ChIP-Seq peak data , 2012 .

[77]  Yi Zhang,et al.  Replication-Dependent Loss of 5-Hydroxymethylcytosine in Mouse Preimplantation Embryos , 2011, Science.

[78]  Weiwei Li,et al.  Distribution of 5-Hydroxymethylcytosine in Different Human Tissues , 2011, Journal of nucleic acids.

[79]  D. Selkoe Alzheimer's disease. , 2011, Cold Spring Harbor perspectives in biology.

[80]  M. Biel,et al.  Tissue Distribution of 5-Hydroxymethylcytosine and Search for Active Demethylation Intermediates , 2010, PloS one.

[81]  Yi Zhang,et al.  Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification , 2010, Nature.

[82]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[83]  R. Meehan,et al.  Enzymatic approaches and bisulfite sequencing cannot distinguish between 5-methylcytosine and 5-hydroxymethylcytosine in DNA. , 2010, BioTechniques.

[84]  Peter A. Jones,et al.  Epigenetics in cancer. , 2010, Carcinogenesis.

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

[86]  David R. Liu,et al.  Conversion of 5-Methylcytosine to 5- Hydroxymethylcytosine in Mammalian DNA by the MLL Partner TET1 , 2009 .

[87]  N. Heintz,et al.  The Nuclear DNA Base 5-Hydroxymethylcytosine Is Present in Purkinje Neurons and the Brain , 2009, Science.

[88]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[89]  Xiang-Dong Fu,et al.  The splicing factor SC35 has an active role in transcriptional elongation , 2008, Nature Structural &Molecular Biology.

[90]  H. Willenbring,et al.  A reproducible and well-tolerated method for 2/3 partial hepatectomy in mice , 2008, Nature Protocols.

[91]  P. Rabinovitch,et al.  Transcriptional response to aging and caloric restriction in heart and adipose tissue , 2007, Aging cell.

[92]  Tyson A. Clark,et al.  Ultraconserved elements are associated with homeostatic control of splicing regulators by alternative splicing and nonsense-mediated decay. , 2007, Genes & development.

[93]  Angela N. Brooks,et al.  The coupling of alternative splicing and nonsense-mediated mRNA decay. , 2007, Advances in experimental medicine and biology.

[94]  Y. Benjamini,et al.  Adaptive linear step-up procedures that control the false discovery rate , 2006 .

[95]  A. Kornblihtt,et al.  Multiple links between transcription and splicing. , 2004, RNA.

[96]  S. Brenner,et al.  Evidence for the widespread coupling of alternative splicing and nonsense-mediated mRNA decay in humans , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[97]  B. Chatterjee,et al.  Impacts of transcriptional regulation on aging and senescence , 2002, Ageing Research Reviews.

[98]  Qiang Zhou,et al.  Stimulatory effect of splicing factors on transcriptional elongation , 2001, Nature.

[99]  J. Tazi,et al.  RasGAP-Associated Endoribonuclease G3BP: Selective RNA Degradation and Phosphorylation-Dependent Localization , 2001, Molecular and Cellular Biology.

[100]  D B Allison,et al.  Influences of aging and caloric restriction on the transcriptional profile of skeletal muscle from rhesus monkeys , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[101]  A. Bird,et al.  MeCP2 Is a Transcriptional Repressor with Abundant Binding Sites in Genomic Chromatin , 1997, Cell.