Evaluation of bisulfite kits for DNA methylation profiling in terms of DNA fragmentation and DNA recovery using digital PCR

Sam Kint, Ward De Spiegelaere, Jonas De Kesel, Linos Vandekerckhove, Wim Van Criekinge, Sam Kint, Ward De Spiegelaere, Jonas De Kesel, Linos Vandekerckhove, Wim Van Criekinge

Abstract

DNA methylation is one of the most important epigenetic modifications in the regulation of gene transcription. The current gold standard to study this modification is bisulfite sequencing. Although multiple commercial bisulfite treatment kits provide good conversion efficiencies, DNA loss and especially DNA fragmentation remain troublesome. This hampers DNA methylation profiling of long DNA sequences. Here, we explored the performance of twelve commercial bisulfite kits by an in-depth comparison of DNA fragmentation using gel electrophoresis, qPCR and digital PCR, DNA recovery by spectroscopic measurements and digital PCR and conversion efficiency by next generation sequencing. The results show a clear performance difference between the bisulfite kits, and depending on the specific goal of the study, the most appropriate kit might differ. Moreover, we demonstrated that digital PCR is a valuable method to monitor both DNA fragmentation as well as DNA recovery after bisulfite treatment.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Principle of bisulfite-mediated methylcytosine (mC)…
Fig 1. Principle of bisulfite-mediated methylcytosine (mC) mapping.
A: Deamination of cytosine (C) and mC. Sodium bisulfite deaminates C to uracil (U) (upper row) and mC to thymine (T) (lower row). The rate of mC deamination is two orders of magnitude less than that of C. B: The mapping protocol: after bisulfite treatment, mC will remain C where unmethylated C will be deaminated to U. During subsequent PCR amplification, the U deamination product templates adenine (A), which then templates T, resulting in a C to T transition at unmethylated C. By sequencing, mC can be identified as bases that remained C after bisulfite treatment [38].
Fig 2. DNA recovery of the twelve…
Fig 2. DNA recovery of the twelve bisulfite kits.
DNA recovery is shown as percentages ± SD and is calculated by the ratio of the measured output concentration of each bisulfite kit (Qubit ssDNA) to the maximal theoretical output concentration based on the input in every kit (Qubit dsDNA). The labels in the figure refer to the kit numbers in Table 1.
Fig 3. Cq values ± SD of…
Fig 3. Cq values ± SD of the smallest and the largest amplicons.
The data used are the geometric means of the average values from the five donor samples as shown in S3 Table. The data labels refer to the kit number as provided in Table 1.
Fig 4. Visual representation of the rankings…
Fig 4. Visual representation of the rankings of the dPCR experiments.
Results are given as the geometric means of the average number of intact copies per ng bisulfite treated (BT) DNA measured by dPCR ± SD of all the five donor samples as shown in S4 Table. The data labels refer to the kit number as provided in Table 1.
Fig 5. Effect of time and temperature.
Fig 5. Effect of time and temperature.
Effect of time and temperature of the conversion protocol on fragmentation between the different bisulfite conversion kits. Labels refer to the kit numbers in Table 1. The upper panels (A-B) show the effect of different conversion temperatures between the kits measured by qPCR (A) and dPCR (B). The lower panels (C-D) show the effect of different conversion times between the kits measured by qPCR (C) and dPCR (D). The values on the x-axis are normalized to show the relative amount of fragmentation: 0 is least fragmenting and 1 is most fragmenting.

References

    1. Attwood JT, Yung RL, Richardson BC. DNA methylation and the regulation of gene transcription. Cell Mol Life Sci C. 2014;59: 241–257. doi:
    1. Kass SU, Landsberger N, Wolffe a P. DNA methylation directs a time-dependent repression of transcription initiation. Curr Biol. 1997;7: 157–165. doi:
    1. Siegfried Z, Eden S, Mendelsohn M, Feng X, Tsuberi BZ, Cedar H. DNA methylation represses transcription in vivo. Nat Genet. 1999;22: 203–206. doi:
    1. Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10: 295–304. doi:
    1. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99: 247–257. doi:
    1. Pradhan S, Bacolla A, Wells RD, Roberts RJ. Recombinant human DNA (cytosine-5) methyltransferase. J Biol Chem. 1999;274: 33002–33010. doi:
    1. Shen H, Laird PW. Interplay between the cancer genome and epigenome. Cell. Elsevier; 2013;153: 38–55. doi:
    1. Bednarik DP, Mosca JD, Raj NB. Methylation as a modulator of expression of human immunodeficiency virus. J Virol. 1987;61: 1253–7.
    1. Schulze-Forster K, Götz F, Wagner H, Kröger H, Simon D. Transcription of HIV1 is inhibited by DNA methylation. Biochem Biophys Res Commun. 1990;168: 141–147. doi:
    1. Blazkova J, Trejbalova K, Gondois-Rey F, Halfon P, Philibert P, Guiguen A, et al. CpG methylation controls reactivation of HIV from latency. PLoS Pathog. 2009;5: e1000554 doi:
    1. Trejbalová K, Kovářová D, Blažková J, Machala L, Jilich D, Weber J, et al. Development of 5’ LTR DNA methylation of latent HIV-1 provirus in cell line models and in long-term-infected individuals. Clin Epigenetics. 2016;8: 19 doi:
    1. Gutekunst KA, Kashanchi F, Brady JN, Bednarik DP. Transcription of the HIV-1 LTR is regulated by the density of DNA CpG methylation. J Acquir Immune Defic Syndr. 1993;6: 541–549.
    1. Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Sci New Ser. 1975;187: 226–232.
    1. Riggs AD. X inactivation, differentiation, and DNA methylation. Cytogenet Genome Res. Karger Publishers; 1975;14: 9–25. doi:
    1. Hayatsu H. The bisulfite genomic sequencing used in the analysis of epigenetic states, a technique in the emerging environmental genotoxicology research. Mutat Res—Rev Mutat Res. 2008;659: 77–82. doi:
    1. Zhang Y, Li S-K, Yi Yang K, Liu M, Lee N, Tang X, et al. Whole genome methylation array reveals the down-regulation of IGFBP6 and SATB2 by HIV-1. Sci Rep. Nature Publishing Group; 2015;5: 10806 doi:
    1. Milavetz BI, Balakrishnan L. Viral epigenetics. Methods Mol Biol. 2015;1238: 569–596. doi:
    1. Schatz P, Dietrich D, Koenig T, Burger M, Lukas A, Fuhrmann I, et al. Development of a diagnostic microarray assay to assess the risk of recurrence of prostate cancer based on PITX2 DNA methylation. J Mol Diagn. American Society for Investigative Pathology; 2010;12: 345–53. doi:
    1. Weiss G, Cottrell S, Distler J, Schatz P, Kristiansen G, Ittmann M, et al. DNA methylation of the PITX2 gene promoter region is a strong independent prognostic marker of biochemical recurrence in patients with prostate cancer after radical prostatectomy. J Urol. 2009;181: 1678–1685. doi:
    1. Dietrich D, Hasinger O, Bañez LL, Sun L, van Leenders GJ, Wheeler TM, et al. Development and clinical validation of a real-time PCR assay for PITX2 DNA methylation to predict prostate-specific antigen recurrence in prostate cancer patients following radical prostatectomy. J Mol Diagn. 2013;15: 270–9. doi:
    1. Bañez LL, Sun L, van Leenders GJ, Wheeler TM, Bangma CH, Freedland SJ, et al. Multicenter clinical validation of PITX2 methylation as a prostate specific antigen recurrence predictor in patients with post-radical prostatectomy prostate cancer. J Urol. 2010;184: 149–156. doi:
    1. Holmes EE, Jung M, Meller S, Leisse A, Sailer V, Zech J, et al. Performance evaluation of kits for bisulfite-conversion of DNA from tissues, cell lines, FFPE tissues, aspirates, lavages, effusions, plasma, serum, and urine. PLoS One. 2014;9: e93933 doi:
    1. Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W, et al. MGMT promoter methylation in malignant gliomas: ready for personalized medicine? Nat Rev Neurol. Nature Publishing Group; 2010;6: 39–51. doi:
    1. Stewart GD, Van Neste L, Delvenne P, Delrée P, Delga A, McNeill SA, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: Results of the MATLOC study. J Urol. 2013;189: 1110–1116. doi:
    1. Kneip C, Schmidt B, Seegebarth A, Weickmann S, Fleischhacker M, Liebenberg V, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. J Thorac Oncol. 2011;6: 1632–8. doi:
    1. Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63: 317–25. doi:
    1. Grützmann R, Molnar B, Pilarsky C, Habermann JK, Schlag PM, Saeger HD, et al. Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One. 2008;3: e3759 doi:
    1. DeVos T, Tetzner R, Model F, Weiss G, Schuster M, Distler J, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem. 2009;55: 1337–1346. doi:
    1. Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg GW, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A. 1992;89: 1827–31. doi:
    1. Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci. 1996;93: 9821–9826. doi:
    1. Li Y, Tollefsbol TO. DNA methylation detection: bisulfite genomic sequencing analysis. Methods Mol Biol. NIH Public Access; 2011;791: 11–21. doi:
    1. Grunau C, Clark SJ, Rosenthal A. Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res. 2001;29: E65–5. doi:
    1. Raizis AM, Schmitt F, Jost JP. A bisulfite method of 5-methylcytosine mapping that minimizes template degradation. Anal Biochem. Academic Press; 1995;226: 161–166. doi:
    1. Tusnády GE, Simon I, Váradi A, Arányi T. BiSearch: primer-design and search tool for PCR on bisulfite-treated genomes. Nucleic Acids Res. 2005;33: e9 doi:
    1. Darst RP, Pardo CE, Ai L, Brown KD, Kladde MP. Bisulfite sequencing of DNA. Curr Protoc Mol Biol. 2010;Chapter 7: Unit–7.917. doi:
    1. Tanaka K, Okamoto A. Degradation of DNA by bisulfite treatment. Bioorg Med Chem Lett. 2007;17: 1912–1915. doi:
    1. Ehrich M, Zoll S, Sur S, van den Boom D. A new method for accurate assessment of DNA quality after bisulfite treatment. Nucleic Acids Res. Oxford University Press; 2007;35: e29 doi:
    1. Chávez L, Kauder S, Verdin E. In vivo, in vitro, and in silico analysis of methylation of the HIV-1 provirus. Methods. 2011;53: 47–53. doi:
    1. Leontiou CA, Hadjidaniel MD, Mina P, Antoniou P, Ioannides M, Patsalis PC. Bisulfite conversion of DNA: Performance comparison of different kits and methylation quantitation of epigenetic biomarkers that have the potential to be used in non-invasive prenatal testing. PLoS One. 2015;10: 1–22. doi:
    1. Izzi B, Binder AM, Michels KB. Pyrosequencing evaluation of widely available bisulfite conversion methods: considerations for application. Med Epigenetics. 2014;2: 28–36. doi:
    1. Bryzgunova O, Laktionov P, Skvortsova T, Bondar A, Morozkin E, Lebedeva A, et al. Efficacy of bisulfite modification and recovery of human genomic and circulating DNA using commercial kits. Eur J Mol Bio. 2013;1: 1–8. doi:
    1. Meissner A, Gnirke A, Bell GW, Ramsahoye B, Lander ES, Jaenisch R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. Oxford University Press; 2005;33: 5868–5877. doi:
    1. Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong CT, et al. Dynamic changes in the human methylome during differentiation. Genome Res. 2010;20: 320–331. doi:
    1. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. Nature Publishing Group; 2009;462: 315–322. doi:
    1. Jang HS, Shin WJ, Lee JE, Do JT. CpG and non-CpG methylation in epigenetic gene regulation and brain function. Genes (Basel). 2017;8: 2–20. doi:
    1. Pelizzola M, Ecker JR. The DNA methylome. FEBS Lett. 2011;585: 1994–2000. doi:
    1. Sharma G, Sowpati DT, Singh P, Khan MZ, Ganji R, Upadhyay S, et al. Genome-wide non-CpG methylation of the host genome during M. tuberculosis infection. Sci Rep. Nature Publishing Group; 2016;6: 25006 doi:
    1. Li Y, Zhu J, Tian G, Li N, Li Q, Ye M, et al. The DNA methylome of human peripheral blood mononuclear cells. PLoS Biol. 2010;8: e1000533 doi:
    1. Vynck M, Trypsteen W, Thas O, Vandekerckhove L, De Spiegelaere W. The future of digital polymerase chain reaction in virology. Mol Diagn Ther. Springer International Publishing; 2016;20: 437–447. doi:
    1. Fuso A, Ferraguti G, Scarpa S, Ferrer I, Lucarelli M. Disclosing bias in bisulfite assay: MethPrimers underestimate high DNA methylation Chiariotti L, editor. PLoS One. Public Library of Science; 2015;10: e0118318 doi:
    1. Genereux DP, Johnson WC, Burden AF, Stöger R, Laird CD. Errors in the bisulfite conversion of DNA: Modulating inappropriate- and failed-conversion frequencies. Nucleic Acids Res. 2008;36 doi:
    1. Wojdacz TK, Dobrovic A, Hansen LL. Methylation-sensitive high-resolution melting. Nat Protoc. Nature Publishing Group; 2008;3: 1903–1908. doi:
    1. Trypsteen W, Kiselinova M, Vandekerckhove L, De Spiegelaere W. Diagnostic utility of droplet digital PCR for HIV reservoir quantification. J virus Erad. 2016;2: 162–9.

Source: PubMed

3
Suscribir