CC BY-NC-ND 4.0 · Thromb Haemost 2020; 120(02): 229-242
DOI: 10.1055/s-0039-3401824
Coagulation and Fibrinolysis
Georg Thieme Verlag KG Stuttgart · New York

A Comprehensive Sequencing-Based Analysis of Allelic Methylation Patterns in Hemostatic Genes in Human Liver

1   Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
,
Marcela Davila Lopez
2   Bioinformatics Core Facility, University of Gothenburg, Gothenburg, Sweden
,
Sofia Klasson
1   Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
,
Lena Hansson
3   Science for Life Laboratories (SciLifeLab), Stockholm, Sweden
4   Novo Nordisk, Oxford, United Kingdom
,
Staffan Nilsson
1   Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
5   Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
,
Tara M. Stanne*
1   Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
,
Christina Jern*
1   Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
6   Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
› Author Affiliations
Funding This study was supported by the Swedish Heart and Lung Foundation (20160316), the Swedish Research Council (2018–02543), the Swedish state under the agreement between the Swedish government and the county councils (the ALF-agreement, ALFGBG-720081), the Swedish Foundation for Strategic Research (RIF14–0081), the Rune and Ulla Amlövs Foundation for Neurologic Research, the John and Brit Wennerström Foundation for Neurologic Research, the Marcus Borgströms Foundation for Neurologic Research, and the Nilsson-Ehle Endowments.
Further Information

Publication History

28 May 2019

01 November 2019

Publication Date:
30 December 2019 (online)

Abstract

Characterizing the relationship between genetic, epigenetic (e.g., deoxyribonucleic acid [DNA] methylation), and transcript variation could provide insights into mechanisms regulating hemostasis and potentially identify new drug targets. Several hemostatic factors are synthesized in the liver, yet high-resolution DNA methylation data from human liver tissue is currently lacking for these genes. Single-nucleotide polymorphisms (SNPs) can influence DNA methylation in cis which can affect gene expression. This can be analyzed through allele-specific methylation (ASM) experiments. We performed targeted genomic DNA- and bisulfite-sequencing of 35 hemostatic genes in human liver samples for SNP and DNA methylation analysis, respectively, and integrated the data for ASM determination. ASM-associated SNPs (ASM-SNPs) were tested for association to gene expression in liver using in-house generated ribonucleic acid-sequencing data. We then assessed whether ASM-SNPs associated with gene expression, plasma proteins, or other traits relevant for hemostasis using publicly available data. We identified 112 candidate ASM-SNPs. Of these, 68% were associated with expression of their respective genes in human liver or in other human tissues and 54% were associated with the respective plasma protein levels, activity, or other relevant hemostatic genome-wide association study traits such as venous thromboembolism, coronary artery disease, stroke, and warfarin dose maintenance. Our study provides the first detailed map of the DNA methylation landscape and ASM analysis of hemostatic genes in human liver tissue, and suggests that methylation regulated by genetic variants in cis may provide a mechanistic link between noncoding SNPs and variation observed in circulating hemostatic proteins, prothrombotic diseases, and drug response.

Authors' Contributions

M.O.L., T.M.S., and C.J. conceived the research design of the present study. C.J. provided funding and was responsible for sample contribution. M.O.L. and S.K. isolated gDNA and RNA and M.O.L. prepared sequencing libraries. M.O.L., L.H., and S.N. performed statistical analyses. M.O.L., T.M.S., and S.K. drafted the figures. M.O.L., C.J., and T.M.S. interpreted the data. M.O.L. and T.M.S. drafted the manuscript. L.H., M.D.L., S.K., and C.J. intellectually reviewed the manuscript. All authors contributed to the last revision process and approved the version to be published.


* Authors contributed equally.


Supplementary Material

 
  • References

  • 1 Kerkel K, Spadola A, Yuan E. , et al. Genomic surveys by methylation-sensitive SNP analysis identify sequence-dependent allele-specific DNA methylation. Nat Genet 2008; 40 (07) 904-908
  • 2 Zhang Y, Rohde C, Reinhardt R, Voelcker-Rehage C, Jeltsch A. Non-imprinted allele-specific DNA methylation on human autosomes. Genome Biol 2009; 10 (12) R138
  • 3 Schalkwyk LC, Meaburn EL, Smith R. , et al. Allelic skewing of DNA methylation is widespread across the genome. Am J Hum Genet 2010; 86 (02) 196-212
  • 4 Paliwal A, Temkin AM, Kerkel K. , et al. Comparative anatomy of chromosomal domains with imprinted and non-imprinted allele-specific DNA methylation. PLoS Genet 2013; 9 (08) e1003622
  • 5 Goll MG, Bestor TH. Eukaryotic cytosine methyltransferases. Annu Rev Biochem 2005; 74: 481-514
  • 6 Robertson KD. DNA methylation and human disease. Nat Rev Genet 2005; 6 (08) 597-610
  • 7 Lister R, Pelizzola M, Dowen RH. , et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009; 462 (7271): 315-322
  • 8 Brenet F, Moh M, Funk P. , et al. DNA methylation of the first exon is tightly linked to transcriptional silencing. PLoS One 2011; 6 (01) e14524
  • 9 Aran D, Toperoff G, Rosenberg M, Hellman A. Replication timing-related and gene body-specific methylation of active human genes. Hum Mol Genet 2011; 20 (04) 670-680
  • 10 Jjingo D, Conley AB, Yi SV, Lunyak VV, Jordan IK. On the presence and role of human gene-body DNA methylation. Oncotarget 2012; 3 (04) 462-474
  • 11 Wagner JR, Busche S, Ge B, Kwan T, Pastinen T, Blanchette M. The relationship between DNA methylation, genetic and expression inter-individual variation in untransformed human fibroblasts. Genome Biol 2014; 15 (02) R37
  • 12 Yang X, Han H, De Carvalho DD, Lay FD, Jones PA, Liang G. Gene body methylation can alter gene expression and is a therapeutic target in cancer. Cancer Cell 2014; 26 (04) 577-590
  • 13 Kurdyukov S, Bullock M. DNA methylation analysis: choosing the right method. Biology (Basel) 2016; 5 (01) 3
  • 14 Kint S, De Spiegelaere W, De Kesel J, Vandekerckhove L, Van Criekinge W. Evaluation of bisulfite kits for DNA methylation profiling in terms of DNA fragmentation and DNA recovery using digital PCR. PLoS One 2018; 13 (06) e0199091
  • 15 Grundberg E, Meduri E, Sandling JK. , et al; Multiple Tissue Human Expression Resource Consortium. Global analysis of DNA methylation variation in adipose tissue from twins reveals links to disease-associated variants in distal regulatory elements. Am J Hum Genet 2013; 93 (05) 876-890
  • 16 Kaplow IM, MacIsaac JL, Mah SM, McEwen LM, Kobor MS, Fraser HB. A pooling-based approach to mapping genetic variants associated with DNA methylation. Genome Res 2015; 25 (06) 907-917
  • 17 Kang EY, Martin LJ, Mangul S. , et al. Discovering single nucleotide polymorphisms regulating human gene expression using allele specific expression from RNA-seq data. Genetics 2016; 204 (03) 1057-1064
  • 18 Olsson Lindvall M, Hansson L, Klasson S, Davila Lopez M, Jern C, Stanne TM. Hemostatic genes exhibit a high degree of allele-specific regulation in liver. Thromb Haemost 2019; 119 (07) 1072-1083
  • 19 Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9 (04) 357-359
  • 20 Staley JR, Blackshaw J, Kamat MA. , et al. PhenoScanner: a database of human genotype-phenotype associations. Bioinformatics 2016; 32 (20) 3207-3209
  • 21 GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 2015; 348 (6235): 648-660
  • 22 Battle A, Brown CD, Engelhardt BE, Montgomery SB. ; GTEx Consortium; Laboratory, Data Analysis &Coordinating Center (LDACC)—Analysis Working Group; Statistical Methods groups—Analysis Working Group; Enhancing GTEx (eGTEx) groups; NIH Common Fund; NIH/NCI; NIH/NHGRI; NIH/NIMH; NIH/NIDA; Biospecimen Collection Source Site—NDRI; Biospecimen Collection Source Site—RPCI; Biospecimen Core Resource—VARI; Brain Bank Repository—University of Miami Brain Endowment Bank; Leidos Biomedical—Project Management; ELSI Study; Genome Browser Data Integration &Visualization—EBI; Genome Browser Data Integration &Visualization—UCSC Genomics Institute, University of California Santa Cruz; Lead analysts; Laboratory, Data Analysis &Coordinating Center (LDACC); NIH program management; Biospecimen collection; Pathology; eQTL manuscript working group. Genetic effects on gene expression across human tissues. Nature 2017; 550 (7675): 204-213
  • 23 Eicher JD, Landowski C, Stackhouse B. , et al. GRASP v2.0: an update on the Genome-Wide Repository of Associations between SNPs and phenotypes. Nucleic Acids Res 2015; 43 (Database issue): D799-D804
  • 24 Franzén O, Ermel R, Cohain A. , et al. Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases. Science 2016; 353 (6301): 827-830
  • 25 Stanne TM, Olsson M, Lorentzen E. , et al. A genome-wide study of common and rare genetic variants associated with circulating thrombin activatable fibrinolysis inhibitor. Thromb Haemost 2018; 118 (02) 298-308
  • 26 Olsson M, Stanne TM, Pedersen A. , et al. Genome-wide analysis of genetic determinants of circulating factor VII-activating protease (FSAP) activity. J Thromb Haemost 2018; 16 (10) 2024-2034
  • 27 Gaunt TR, Shihab HA, Hemani G. , et al. Systematic identification of genetic influences on methylation across the human life course. Genome Biol 2016; 17: 61
  • 28 Ward LD, Kellis M. HaploReg v4: systematic mining of putative causal variants, cell types, regulators and target genes for human complex traits and disease. Nucleic Acids Res 2016; 44 (D1): D877-D881
  • 29 Kundaje A, Meuleman W, Ernst J. , et al; Roadmap Epigenomics Consortium. Integrative analysis of 111 reference human epigenomes. Nature 2015; 518 (7539): 317-330
  • 30 Teichert M, Eijgelsheim M, Uitterlinden AG. , et al. Dependency of phenprocoumon dosage on polymorphisms in the VKORC1, CYP2C9, and CYP4F2 genes. Pharmacogenet Genomics 2011; 21 (01) 26-34
  • 31 Peden JFHJ, Saleheen D, Chambers JC. , et al; Coronary Artery Disease (C4D) Genetics Consortium. A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat Genet 2011; 43 (04) 339-344
  • 32 Ikram MA, Seshadri S, Bis JC. , et al. Genome-wide association studies of stroke. N Engl J Med 2009; 360 (17) 1718-1728
  • 33 Do C, Shearer A, Suzuki M. , et al. Genetic-epigenetic interactions in cis: a major focus in the post-GWAS era. Genome Biol 2017; 18 (01) 120
  • 34 Kulis M, Heath S, Bibikova M. , et al. Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat Genet 2012; 44 (11) 1236-1242
  • 35 Varley KE, Gertz J, Bowling KM. , et al. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res 2013; 23 (03) 555-567
  • 36 Hannon E, Spiers H, Viana J. , et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat Neurosci 2016; 19 (01) 48-54
  • 37 Gutierrez-Arcelus M, Lappalainen T, Montgomery SB. , et al. Passive and active DNA methylation and the interplay with genetic variation in gene regulation. eLife 2013; 2: e00523
  • 38 Gutierrez-Arcelus M, Ongen H, Lappalainen T. , et al. Tissue-specific effects of genetic and epigenetic variation on gene regulation and splicing. PLoS Genet 2015; 11 (01) e1004958
  • 39 Fu J, Wolfs MG, Deelen P. , et al. Unraveling the regulatory mechanisms underlying tissue-dependent genetic variation of gene expression. PLoS Genet 2012; 8 (01) e1002431
  • 40 Bettscheider M, Kuczynska A, Almeida O, Spengler D. Optimized analysis of DNA methylation and gene expression from small, anatomically-defined areas of the brain. J Vis Exp 2012; (65) e3938
  • 41 Gamazon ER, Badner JA, Cheng L. , et al. Enrichment of cis-regulatory gene expression SNPs and methylation quantitative trait loci among bipolar disorder susceptibility variants. Mol Psychiatry 2013; 18 (03) 340-346
  • 42 Do C, Lang CF, Lin J. , et al. Mechanisms and disease associations of haplotype-dependent allele-specific DNA methylation. Am J Hum Genet 2016; 98 (05) 934-955
  • 43 Volkov P, Olsson AH, Gillberg L. , et al. A genome-wide mQTL analysis in human adipose tissue identifies genetic variants associated with DNA methylation, gene expression and metabolic traits. PLoS One 2016; 11 (06) e0157776
  • 44 Pirmohamed M, Burnside G, Eriksson N. , et al; EU-PACT Group. A randomized trial of genotype-guided dosing of warfarin. N Engl J Med 2013; 369 (24) 2294-2303
  • 45 Kimmel SE, French B, Kasner SE. , et al; COAG Investigators. A pharmacogenetic versus a clinical algorithm for warfarin dosing. N Engl J Med 2013; 369 (24) 2283-2293
  • 46 Cullell N, Carrera C, Muiño E, Torres N, Krupinski J, Fernandez-Cadenas I. Pharmacogenetic studies with oral anticoagulants. Genome-wide association studies in vitamin K antagonist and direct oral anticoagulants. Oncotarget 2018; 9 (49) 29238-29258
  • 47 Luo Z, Liu R, Sun B. , et al. Identification of gene modules associated with warfarin dosage by a genome-wide DNA methylation study. Pharmazie 2018; 73 (05) 288-293
  • 48 Gallego-Fabrega C, Carrera C, Reny JL. , et al. PPM1A methylation is associated with vascular recurrence in aspirin-treated patients. Stroke 2016; 47 (07) 1926-1929
  • 49 Cotto KC, Wagner AH, Feng YY. , et al. DGIdb 3.0: a redesign and expansion of the drug-gene interaction database. Nucleic Acids Res 2018; 46 (D1): D1068-D1073
  • 50 Yang Y, Scott SA. DNA methylation profiling using long-read single molecule real-time bisulfite sequencing (SMRT-BS). Methods Mol Biol 2017; 1654: 125-134