Differential DNA methylation with age displays both common and dynamic features across human tissues that are influenced by CpG landscape

Kenneth Day, Lindsay L Waite, Anna Thalacker-Mercer, Andrew West, Marcas M Bamman, James D Brooks, Richard M Myers, Devin Absher, Kenneth Day, Lindsay L Waite, Anna Thalacker-Mercer, Andrew West, Marcas M Bamman, James D Brooks, Richard M Myers, Devin Absher

Abstract

Background: DNA methylation is an epigenetic modification that changes with age in human tissues, although the mechanisms and specificity of this process are still poorly understood. We compared CpG methylation changes with age across 283 human blood, brain, kidney, and skeletal muscle samples using methylation arrays to identify tissue-specific age effects.

Results: We found age-associated CpGs (ageCGs) that are both tissue-specific and common across tissues. Tissue-specific age CGs are frequently located outside CpG islands with decreased methylation, and common ageCGs show the opposite trend. AgeCGs are significantly associated with poorly expressed genes, but those with decreasing methylation are linked with higher tissue-specific expression levels compared with increasing methylation. Therefore, tissue-specific gene expression may protect against common age-dependent methylation. Distinguished from other tissues, skeletal muscle age CGs are more associated with expression, enriched near genes related to myofiber contraction, and closer to muscle-specific CTCF binding sites. Kidney-specific ageCGs are more increasingly methylated compared to other tissues as measured by affiliation with kidney-specific expressed genes. Underlying chromatin features also mark common and tissue-specific age effects reflective of poised and active chromatin states, respectively. In contrast with decreasingly methylated ageCGs, increasingly methylated ageCGs are also generally further from CTCF binding sites and enriched within lamina associated domains.

Conclusions: Our data identified common and tissue-specific DNA methylation changes with age that are reflective of CpG landscape and suggests both common and unique alterations within human tissues. Our findings also indicate that a simple epigenetic drift model is insufficient to explain all age-related changes in DNA methylation.

Figures

Figure 1
Figure 1
Linear regression results showed an association between DNA methylation and age across four human tissues. (A) Uniform quantile-quantile (Q-Q) plots of -log P-values from linear regression tests for associations between β-score and age within blood, brain, kidney, and skeletal muscle tissue samples. (B) Scatterplots for the top five strongest associations between β-score and age across each of the four tissues. Illumina CpG ID, P-value, and R-squared results are depicted above each scatterplot.
Figure 2
Figure 2
Bar graphs displaying the percentages of ageCGs classified by CpG context and positive or negative association with age across four human tissues. (A) Percentage of total ageCGs that exhibited a positive or negative association with age in each tissue. (B,C) Percentage of positively associated (B) and negatively associated ageCGs (C) positioned within CpG islands, shores, or other. (D-F) Percentages of positively or negatively associated ageCGs each classified within CpG islands (D), CpG shores (E), or CpG other contexts (F). P-values are below bar graphs that resulted from Pearson’s Chi-squared tests between number of positive or negative associations across tissues in each respective CpG context.
Figure 3
Figure 3
Boxplots of slope magnitude grouped according to positive (+) or negative (-) ageCGs and CpG context across four tissues. Vertical red and green lines mark the smallest and largest median slope magnitude, respectively, across all four tissues. A significant three-way interaction between CpG context, tissue type, and positive or negative slope trend was found among slope magnitude values (ANOVA on Box-Cox power transformed slope values, P = 5.4 × 10-5).
Figure 4
Figure 4
Jointly learned tissue-specific chromatin states across four human tissues and functional enrichments within positive and negative ageCG positions. Ten input chromatin states using Roadmap Epigenome ChIP-seq data for six histone modifications across four human tissues were used with ChromHMM software. (A) Heatmap/table shows learned emission parameters based on genome-wide combinations of histone marks. Values indicate observed frequencies of histone modifications found at genomic positions corresponding with chromatin states. (B) Transitional parameter heatmap/table shows probabilities of transitions between states (multiplied by 100). Rows show the 'from' chromatin state and columns show the 'to' chromatin state, that is, a 17% probability that chromatin state 7 transitioned into state 8. (C) Heatmap/table depicts percentage of the genome for each chromatin state (topmost row) and relative fold functional enrichments of genome category (that is, vista enhancers, lamin B1 laminB1lads, CpGs within CGIs, CGSs, CGOs, positive (+) and negative (-) ageCGs and non-ageCGs). Enrichments for chromatin states underlying ChIP-seq CTCF binding sites were determined using ENCODE data for CD14+ and CD20+ cells (merged peaks blood), kidney tissue, and myotubes (brain data not available). Overlap enrichments were determined separately for each tissue using tissue-specific segmentation files generated from the jointly learned model. Values across rows indicate relative fold enrichment, and blue color scale is based on subtraction of the minimum value in the row divided by the maximum row value for each tissue separately (vertical black lines divide table enrichments per tissue). (D) Neighborhood enrichments for RefSeq transcriptional start site annotations (TSS) within chromatin states determined using default 0-based anchor coordinates for each start site position. Fold enrichment values and color scale are according to rows. In all panels, the lower axis shows chromatin state colored to match chromatin state descriptions (1 to 10).
Figure 5
Figure 5
Validation of ageCGs by targeted capture and bisulfite sequencing of genomic regions encompassing ageCG sites. (A) Scatterplots of percentage methylation by bisulfite sequencing (Bis-seq) versus Methylation27 β-score and correlation of methylation values between the two methods across 19 kidney samples used for validation. (B) A representative Bland-Altman plot for comparison of Bis-seq and Methylation27 methylation values. Points depict the average percentage methylation between both methods plotted against the differences in methylation between the methods. Dotted lines show limits of agreement (average difference ± 1.96 standard deviation of the difference). (C, D) Comparison of methylation delta values (median percentage methylation of young minus old samples) at CpGs covered by Bis-seq (top panels) and Methylation27 arrays (middle panels). Red points represent ageCGs, and delta values shown for Bis-seq are only for the 9 youngest and 10 oldest samples. False discovery rate (q) values indicate significance level of a widespread age effect by linear mixed model results with Bis-seq data at these target regions. The DKK1 target just missed the significance threshold (q < 0.05), and the RLN1 target demonstrates an example region that contained abrupt changes in direction of delta values among neighboring CpGs. Bottom panels depict labeled gene regions (blue boxes, exons blue lines, introns), CpG islands (green boxes), and fetal kidney chromatin states (colors correspond with definitions in Figure 4). Greater details and examples of all Bis-seq targets are available in Additional files 7 and 24.

References

    1. Scharf AN, Imhof A. Every methyl counts-epigenetic calculus. FEBS Lett. 2011;14:2001–2007. doi: 10.1016/j.febslet.2010.11.029.
    1. Jin B, Li Y, Robertson KD. DNA methylation: superior or subordinate in the epigenetic hierarchy? Genes Cancer. 2011;14:607–617. doi: 10.1177/1947601910393957.
    1. Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol. 1987;14:261–282. doi: 10.1016/0022-2836(87)90689-9.
    1. Kafri T, Ariel M, Brandeis M, Shemer R, Urven L, McCarrey J, Cedar H, Razin A. Developmental pattern of gene-specific DNA methylation in the mouse embryo and germ line. Genes Dev. 1992;14:705–714. doi: 10.1101/gad.6.5.705.
    1. Smith ZD, Chan MM, Mikkelsen TS, Gu H, Gnirke A, Regev A, Meissner A. A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature. 2012;14:339–344. doi: 10.1038/nature10960.
    1. Trowbridge JJ, Orkin SH. DNA methylation in adult stem cells: New insights into self-renewal. Epigenetics. 2010;14:189–193. doi: 10.4161/epi.5.3.11374.
    1. Bocker MT, Hellwig I, Breiling A, Eckstein V, Ho AD, Lyko F. Genome-wide promoter DNA methylation dynamics of human hematopoietic progenitor cells during differentiation and aging. Blood. 2011;14:e182–e189. doi: 10.1182/blood-2011-01-331926.
    1. Berdasco M, Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Dev Cell. 2010;14:698–711. doi: 10.1016/j.devcel.2010.10.005.
    1. Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci USA. 2006;14:1412–1417. doi: 10.1073/pnas.0510310103.
    1. Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, Schubeler D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet. 2007;14:457–466. doi: 10.1038/ng1990.
    1. Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, Herb B, Ladd-Acosta C, Rho J, Loewer S, Miller J, Schlaeger T, Daley GQ, Feinberg AP. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet. 2009;14:1350–1353. doi: 10.1038/ng.471.
    1. Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, Ji H, Potash JB, Sabunciyan S, Feinberg AP. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;14:178–186. doi: 10.1038/ng.298.
    1. Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, Savage DA, Mueller-Holzner E, Marth C, Kocjan G, Gayther SA, Jones A, Beck S, Wagner W, Laird PW, Jacobs IJ, Widschwendter M. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010;14:440–446. doi: 10.1101/gr.103606.109.
    1. Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet. 2006;14:21–33. doi: 10.1038/nrg1748.
    1. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;14:484–492. doi: 10.1038/nrg3230.
    1. Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong CT, Low HM, Kin Sung KW, Rigoutsos I, Loring J, Wei CL. Dynamic changes in the human methylome during differentiation. Genome Res. 2010;14:320–331. doi: 10.1101/gr.101907.109.
    1. Wareham KA, Lyon MF, Glenister PH, Williams ED. Age related reactivation of an X-linked gene. Nature. 1987;14:725–727. doi: 10.1038/327725a0.
    1. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;14:10604–10609. doi: 10.1073/pnas.0500398102.
    1. Raj A, van Oudenaarden A. Nature, nurture, or chance: stochastic gene expression and its consequences. Cell. 2008;14:216–226. doi: 10.1016/j.cell.2008.09.050.
    1. Martin GM. Epigenetic gambling and epigenetic drift as an antagonistic pleiotropic mechanism of aging. Aging Cell. 2009;14:761–764. doi: 10.1111/j.1474-9726.2009.00515.x.
    1. Pujadas E, Feinberg AP. Regulated noise in the epigenetic landscape of development and disease. Cell. 2012;14:1123–1131. doi: 10.1016/j.cell.2012.02.045.
    1. Guo JU, Ma DK, Mo H, Ball MP, Jang MH, Bonaguidi MA, Balazer JA, Eaves HL, Xie B, Ford E, Zhang K, Ming GL, Gao Y, Song H. Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat Neurosci. 2011;14:1345–1351. doi: 10.1038/nn.2900.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;14:118–127. doi: 10.1093/biostatistics/kxj037.
    1. Rakyan VK, Down TA, Maslau S, Andrew T, Yang TP, Beyan H, Whittaker P, McCann OT, Finer S, Valdes AM, Leslie RD, Deloukas P, Spector TD. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 2010;14:434–439. doi: 10.1101/gr.103101.109.
    1. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, Deconde R, Chen M, Rajapakse I, Friend S, Ideker T, Zhang K. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013;14:359–367. doi: 10.1016/j.molcel.2012.10.016.
    1. Numata S, Ye T, Hyde TM, Guitart-Navarro X, Tao R, Wininger M, Colantuoni C, Weinberger DR, Kleinman JE, Lipska BK. DNA methylation signatures in development and aging of the human prefrontal cortex. Am J Hum Genet. 2012;14:260–272. doi: 10.1016/j.ajhg.2011.12.020.
    1. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;14:44–57.
    1. da Huang W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;14:1–13. doi: 10.1093/nar/gkn923.
    1. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold BJ, Pachter L. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;14:511–515. doi: 10.1038/nbt.1621.
    1. Ernst J, Kellis M. ChromHMM: automating chromatin-state discovery and characterization. Nat Methods. 2012;14:215–216. doi: 10.1038/nmeth.1906.
    1. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;14:315–326. doi: 10.1016/j.cell.2006.02.041.
    1. Hawkins RD, Hon GC, Lee LK, Ngo Q, Lister R, Pelizzola M, Edsall LE, Kuan S, Luu Y, Klugman S, Antosiewicz-Bourget J, Ye Z, Espinoza C, Agarwahl S, Shen L, Ruotti V, Wang W, Stewart R, Thomson JA, Ecker JR, Ren B. Distinct epigenomic landscapes of pluripotent and lineage-committed human cells. Cell Stem Cell. 2010;14:479–491. doi: 10.1016/j.stem.2010.03.018.
    1. Zentner GE, Tesar PJ, Scacheri PC. Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 2011;14:1273–1283. doi: 10.1101/gr.122382.111.
    1. Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, Hawkins RD, Barrera LO, Van Calcar S, Qu C, Ching KA, Wang W, Weng Z, Green RD, Crawford GE, Ren B. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet. 2007;14:311–318. doi: 10.1038/ng1966.
    1. Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, Hanna J, Lodato MA, Frampton GM, Sharp PA, Boyer LA, Young RA, Jaenisch R. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci USA. 2010;14:21931–21936. doi: 10.1073/pnas.1016071107.
    1. Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Talhout W, Eussen BH, de Klein A, Wessels L, de Laat W, van Steensel B. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature. 2008;14:948–951. doi: 10.1038/nature06947.
    1. Christensen BC, Houseman EA, Marsit CJ, Zheng S, Wrensch MR, Wiemels JL, Nelson HH, Karagas MR, Padbury JF, Bueno R, Sugarbaker DJ, Yeh RF, Wiencke JK, Kelsey KT. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet. 2009;14:e1000602. doi: 10.1371/journal.pgen.1000602.
    1. Hernandez DG, Nalls MA, Gibbs JR, Arepalli S, van der Brug M, Chong S, Moore M, Longo DL, Cookson MR, Traynor BJ, Singleton AB. Distinct DNA methylation changes highly correlated with chronological age in the human brain. Hum Mol Genet. 2011;14:1164–1172. doi: 10.1093/hmg/ddq561.
    1. Koch CM, Suschek CV, Lin Q, Bork S, Goergens M, Joussen S, Pallua N, Ho AD, Zenke M, Wagner W. Specific age-associated DNA methylation changes in human dermal fibroblasts. PLoS One. 2011;14:e16679. doi: 10.1371/journal.pone.0016679.
    1. Bork S, Pfister S, Witt H, Horn P, Korn B, Ho AD, Wagner W. DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells. Aging Cell. 2010;14:54–63. doi: 10.1111/j.1474-9726.2009.00535.x.
    1. Schellenberg A, Lin Q, Schuler H, Koch CM, Joussen S, Denecke B, Walenda G, Pallua N, Suschek CV, Zenke M, Wagner W. Replicative senescence of mesenchymal stem cells causes DNA-methylation changes which correlate with repressive histone marks. Aging (Albany NY) 2011;14:873–888.
    1. Maegawa S, Hinkal G, Kim HS, Shen L, Zhang L, Zhang J, Zhang N, Liang S, Donehower LA, Issa JP. Widespread and tissue specific age-related DNA methylation changes in mice. Genome Res. 2010;14:332–340. doi: 10.1101/gr.096826.109.
    1. Thompson RF, Atzmon G, Gheorghe C, Liang HQ, Lowes C, Greally JM, Barzilai N. Tissue-specific dysregulation of DNA methylation in aging. Aging Cell. 2010;14:506–518. doi: 10.1111/j.1474-9726.2010.00577.x.
    1. Heyn H, Li N, Ferreira HJ, Moran S, Pisano DG, Gomez A, Diez J, Sanchez-Mut JV, Setien F, Carmona FJ, Puca AA, Sayols S, Pujana MA, Serra-Musach J, Iglesias-Platas I, Formiga F, Fernandez AF, Fraga MF, Heath SC, Valencia A, Gut IG, Wang J, Esteller M. Distinct DNA methylomes of newborns and centenarians. Proc Natl Acad Sci USA. 2012;14:10522–10527. doi: 10.1073/pnas.1120658109.
    1. Song J, Teplova M, Ishibe-Murakami S, Patel DJ. Structure-based mechanistic insights into DNMT1-mediated maintenance DNA methylation. Science. 2012;14:709–712. doi: 10.1126/science.1214453.
    1. Chu MW, Siegmund KD, Eckstam CL, Kim JY, Yang AS, Kanel GC, Tavare S, Shibata D. Lack of increases in methylation at three CpG-rich genomic loci in non-mitotic adult tissues during aging. BMC Med Genet. 2007;14:50.
    1. Sen GL, Reuter JA, Webster DE, Zhu L, Khavari PA. DNMT1 maintains progenitor function in self-renewing somatic tissue. Nature. 2010;14:563–567. doi: 10.1038/nature08683.
    1. Levi BP, Morrison SJ. Stem cells use distinct self-renewal programs at different ages. Cold Spring Harb Symp Quant Biol. 2008;14:539–553. doi: 10.1101/sqb.2008.73.049.
    1. Bröske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R, Nerlov C, Leutz A, Andrade-Navarro MA, Jacobsen SE, Rosenbauer F. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet. 2009;14:1207–1215. doi: 10.1038/ng.463.
    1. Rossi DJ, Bryder D, Zahn JM, Ahlenius H, Sonu R, Wagers AJ, Weissman IL. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci U S A. 2005;14:9194–9199. doi: 10.1073/pnas.0503280102.
    1. Serrano AL, Munoz-Canoves P. Regulation and dysregulation of fibrosis in skeletal muscle. Exp Cell Res. 2010;14:3050–3058. doi: 10.1016/j.yexcr.2010.05.035.
    1. Torres VE, Leof EB. Fibrosis, regeneration, and aging: playing chess with evolution. J Am Soc Nephrol. 2011;14:1393–1396. doi: 10.1681/ASN.2011060603.
    1. Peake J, Della Gatta P, Cameron-Smith D. Aging and its effects on inflammation in skeletal muscle at rest and following exercise-induced muscle injury. Am J Physiol Regul Integr Comp Physiol. 2010;14:R1485–R1495. doi: 10.1152/ajpregu.00467.2009.
    1. Rodwell GE, Sonu R, Zahn JM, Lund J, Wilhelmy J, Wang L, Xiao W, Mindrinos M, Crane E, Segal E, Myers BD, Brooks JD, Davis RW, Higgins J, Owen AB, Kim SK. A transcriptional profile of aging in the human kidney. PLoS Biol. 2004;14:e427. doi: 10.1371/journal.pbio.0020427.
    1. Zahn JM, Sonu R, Vogel H, Crane E, Mazan-Mamczarz K, Rabkin R, Davis RW, Becker KG, Owen AB, Kim SK. Transcriptional profiling of aging in human muscle reveals a common aging signature. PLoS Genet. 2006;14:e115. doi: 10.1371/journal.pgen.0020115.
    1. Jin B, Ernst J, Tiedemann RL, Xu H, Sureshchandra S, Kellis M, Dalton S, Liu C, Choi JH, Robertson KD. Linking DNA methyltransferases to epigenetic marks and nucleosome structure genome-wide in human tumor cells. Cell Rep. 2012;14:1411–1424. doi: 10.1016/j.celrep.2012.10.017.
    1. Herold M, Bartkuhn M, Renkawitz R. CTCF: insights into insulator function during development. Development. 2012;14:1045–1057. doi: 10.1242/dev.065268.
    1. Wang H, Maurano MT, Qu H, Varley KE, Gertz J, Pauli F, Lee K, Canfield T, Weaver M, Sandstrom R, Thurman RE, Kaul R, Myers RM, Stamatoyannopoulos JA. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 2012;14:1680–1688. doi: 10.1101/gr.136101.111.
    1. Haithcock E, Dayani Y, Neufeld E, Zahand AJ, Feinstein N, Mattout A, Gruenbaum Y, Liu J. Age-related changes of nuclear architecture in Caenorhabditis elegans. Proc Natl Acad Sci USA. 2005;14:16690–16695. doi: 10.1073/pnas.0506955102.
    1. Guarda A, Bolognese F, Bonapace IM, Badaracco G. Interaction between the inner nuclear membrane lamin B receptor and the heterochromatic methyl binding protein, MeCP2. Exp Cell Res. 2009;14:1895–1903. doi: 10.1016/j.yexcr.2009.01.019.
    1. Sun Z, Chai HS, Wu Y, White WM, Donkena KV, Klein CJ, Garovic VD, Therneau TM, Kocher JP. Batch effect correction for genome-wide methylation data with Illumina Infinium platform. BMC Med Genomics. 2011;14:84. doi: 10.1186/1755-8794-4-84.
    1. Inline barcodes design and demuxing software for Illumina next generation sequencing. .
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;14:R25. doi: 10.1186/gb-2009-10-3-r25.
    1. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, Philippakis AA, del Angel G, Rivas MA, Hanna M, McKenna A, Fennell TJ, Kernytsky AM, Sivachenko AY, Cibulskis K, Gabriel SB, Altshuler D, Daly MJ. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;14:491–498. doi: 10.1038/ng.806.
    1. RNA-Seq of human individual tissues and mixture of 16 tissues: Illumina Body Map. .
    1. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;14:1105–1111. doi: 10.1093/bioinformatics/btp120.
    1. NIH Roadmap Epigenomics Project Data Listings.
    1. ChromHMM. Chromatin state discovery and characterization.
    1. Encyclopedia of DNA Elements.

Source: PubMed

3
Subscribe