Dnmt3a and Dnmt3b-Decommissioned Fetal Enhancers are Linked to Kidney Disease.

BACKGROUND Cytosine methylation is an epigenetic mark that dictates cell fate and response to stimuli. The timing and establishment of methylation logic during kidney development remains unknown. DNA methyltransferase 3a and 3b are the enzymes capable of establishing de novo methylation. METHODS We generated mice with genetic deletion of Dnmt3a and Dnmt3b in nephron progenitor cells (Six2 Cre Dnmt3a/3b) and kidney tubule cells (Ksp Cre Dnmt3a/3b). We characterized Ksp Cre Dnmt3a/3b mice at baseline and after injury. Unbiased omics profiling, such as whole genome bisulfite sequencing, reduced representation bisulfite sequencing and RNA sequencing were performed on whole-kidney samples and isolated renal tubule cells. RESULTS Ksp Cre Dnmt3a/3b mice showed no obvious morphologic and functional alterations at baseline. Knockout animals exhibited increased resistance to cisplatin-induced kidney injury, but not to folic acid-induced fibrosis. Whole-genome bisulfite sequencing indicated that Dnmt3a and Dnmt3b play an important role in methylation of gene regulatory regions that act as fetal-specific enhancers in the developing kidney but are decommissioned in the mature kidney. Loss of Dnmt3a and Dnmt3b resulted in failure to silence developmental genes. We also found that fetal-enhancer regions methylated by Dnmt3a and Dnmt3b were enriched for kidney disease genetic risk loci. Methylation patterns of kidneys from patients with CKD showed defects similar to those in mice with Dnmt3a and Dnmt3b deletion. CONCLUSIONS Our results indicate a potential locus-specific convergence of genetic, epigenetic, and developmental elements in kidney disease development.

[1]  Jacob M. Keaton,et al.  Mapping eGFR loci to the renal transcriptome and phenome in the VA Million Veteran Program , 2019, Nature Communications.

[2]  Lin Sun,et al.  Aberrant DNA methylation of mTOR pathway genes promotes inflammatory activation of immune cells in diabetic kidney disease. , 2019, Kidney international.

[3]  A. Verma,et al.  Functional methylome analysis of human diabetic kidney disease. , 2019, JCI insight.

[4]  Hongzhe Li,et al.  Kidney cytosine methylation changes improve renal function decline estimation in patients with diabetic kidney disease , 2019, Nature Communications.

[5]  V. M. V. Program A catalog of genetic loci associated with kidney function from analyses of a million individuals , 2019 .

[6]  Mitsuo Kato,et al.  Epigenetics and epigenomics in diabetic kidney disease and metabolic memory , 2019, Nature Reviews Nephrology.

[7]  K. Suszták,et al.  DNMT1 in Six2 Progenitor Cells Is Essential for Transposable Element Silencing and Kidney Development. , 2019, Journal of the American Society of Nephrology : JASN.

[8]  James M. Eales,et al.  Trans-ethnic kidney function association study reveals putative causal genes and effects on kidney-specific disease aetiologies , 2019, Nature Communications.

[9]  Victor G. Puelles,et al.  DNA Methyltransferase 1 Controls Nephron Progenitor Cell Renewal and Differentiation. , 2018, Journal of the American Society of Nephrology : JASN.

[10]  Helen E. Parkinson,et al.  The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019 , 2018, Nucleic Acids Res..

[11]  Mark Gerstein,et al.  GENCODE reference annotation for the human and mouse genomes , 2018, Nucleic Acids Res..

[12]  Christopher D. Brown,et al.  Renal compartment-specific genetic variation analyses identify new pathways in chronic kidney disease , 2018, Nature Medicine.

[13]  William S. DeWitt,et al.  A Single-Cell Atlas of In Vivo Mammalian Chromatin Accessibility , 2018, Cell.

[14]  P. Park,et al.  EED, a member of the polycomb group, is required for nephron differentiation and the maintenance of nephron progenitor cells , 2018, Development.

[15]  Tomi Pastinen,et al.  Pioneer factor Pax7 deploys a stable enhancer repertoire for specification of cell fate , 2018, Nature Genetics.

[16]  A. El-Osta,et al.  Epigenetics in diabetic nephropathy, immunity and metabolism , 2017, Diabetologia.

[17]  Manolis Kellis,et al.  Chromatin-state discovery and genome annotation with ChromHMM , 2017, Nature Protocols.

[18]  Meng-Pin Weng,et al.  modPhEA: model organism Phenotype Enrichment Analysis of eukaryotic gene sets , 2017, Bioinform..

[19]  Christopher D. Brown,et al.  Genetic-Variation-Driven Gene-Expression Changes Highlight Genes with Important Functions for Kidney Disease. , 2017, American journal of human genetics.

[20]  H. Pavenstädt,et al.  Developmental signalling pathways in renal fibrosis: the roles of Notch, Wnt and Hedgehog , 2016, Nature Reviews Nephrology.

[21]  A. McMahon,et al.  Differential regulation of mouse and human nephron progenitors by the Six family of transcriptional regulators , 2016, Development.

[22]  Hongbo Liu,et al.  Systematic identification and annotation of human methylation marks based on bisulfite sequencing methylomes reveals distinct roles of cell type-specific hypomethylation in the regulation of cell identity genes , 2015, Nucleic acids research.

[23]  M. Martí-Renom,et al.  Chromatin and RNA Maps Reveal Regulatory Long Noncoding RNAs in Mouse , 2015, Molecular and Cellular Biology.

[24]  M. Daly,et al.  Genetic and Epigenetic Fine-Mapping of Causal Autoimmune Disease Variants , 2014, Nature.

[25]  Y. Ko,et al.  Epigenetics: a new way to look at kidney diseases. , 2014, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[26]  D. Zheng,et al.  DNA Methylation is Developmentally Regulated for Genes Essential for Cardiogenesis , 2014, Journal of the American Heart Association.

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

[28]  Katalin Susztak,et al.  Understanding the epigenetic syntax for the genetic alphabet in the kidney. , 2014, Journal of the American Society of Nephrology : JASN.

[29]  G. Hon,et al.  Adult tissue methylomes harbor epigenetic memory at embryonic enhancers , 2013, Nature Genetics.

[30]  S. Pfaff,et al.  Transposable elements as genetic regulatory substrates in early development. , 2013, Trends in cell biology.

[31]  Francine E. Garrett-Bakelman,et al.  An optimized algorithm for detecting and annotating regional differential methylation , 2013, BMC Bioinformatics.

[32]  A. Kohlmann,et al.  The molecular profile of adult T‐cell acute lymphoblastic leukemia: Mutations in RUNX1 and DNMT3A are associated with poor prognosis in T‐ALL , 2013, Genes, chromosomes & cancer.

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

[34]  Francine E. Garrett-Bakelman,et al.  methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles , 2012, Genome Biology.

[35]  J. Berg,et al.  Dnmt3a is essential for hematopoietic stem cell differentiation , 2011, Nature Genetics.

[36]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[37]  Felix Krueger,et al.  Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications , 2011, Bioinform..

[38]  Yong-mei Zhu,et al.  Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia , 2011, Nature Genetics.

[39]  Li Ding,et al.  Recurrent DNMT3A Mutations in Patients with Myelodysplastic Syndromes , 2011, Leukemia.

[40]  Joshua F. McMichael,et al.  DNMT3A mutations in acute myeloid leukemia. , 2010, The New England journal of medicine.

[41]  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.

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

[43]  W. Birchmeier,et al.  Interplay of cadherin-mediated cell adhesion and canonical Wnt signaling. , 2010, Cold Spring Harbor perspectives in biology.

[44]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[45]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[46]  A. McMahon,et al.  Six2 defines and regulates a multipotent self-renewing nephron progenitor population throughout mammalian kidney development. , 2008, Cell stem cell.

[47]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[48]  Michael B. Stadler,et al.  Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome , 2007, Nature Genetics.

[49]  S. Fitzgerald,et al.  Combined prenatal and postnatal protein restriction influences adult kidney structure, function, and arterial pressure. , 2007, American journal of physiology. Regulatory, integrative and comparative physiology.

[50]  G. Dressler,et al.  Six2 is required for suppression of nephrogenesis and progenitor renewal in the developing kidney , 2006, The EMBO journal.

[51]  C. Osmond,et al.  Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth , 2004, Diabetologia.

[52]  M. Busslinger,et al.  Nephric lineage specification by Pax2 and Pax8. , 2002, Genes & development.

[53]  J. V. Moran,et al.  Initial sequencing and analysis of the human genome. , 2001, Nature.

[54]  W. Hörl,et al.  Recent insights into the molecular genetics of the homocysteine metabolism. , 2001, Kidney international. Supplement.

[55]  D. Haber,et al.  DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development , 1999, Cell.