Multiplex mapping of chromatin accessibility and DNA methylation within targeted single molecules identifies epigenetic heterogeneity in neural stem cells and glioblastoma

Human tumors are comprised of heterogeneous cell populations that display diverse molecular and phenotypic features. To examine the extent to which epigenetic differences contribute to intratumoral cellular heterogeneity, we have developed a high-throughput method, termed MAPit-patch. The method uses multiplexed amplification of targeted sequences from submicrogram quantities of genomic DNA followed by next generation bisulfite sequencing. This provides highly scalable and simultaneous mapping of chromatin accessibility and DNA methylation on single molecules at high resolution. Long sequencing reads from targeted regions maintain the structural integrity of epigenetic information and provide substantial depth of coverage, detecting for the first time minority subpopulations of epigenetic configurations formerly obscured by existing genome-wide and population-ensemble methodologies. Analyzing a cohort of 71 promoters of genes with exons commonly mutated in cancer, MAPit-patch uncovered several differentially accessible and methylated promoters that are associated with altered gene expression between neural stem cell (NSC) and glioblastoma (GBM) cell populations. In addition, considering each promoter individually, substantial epigenetic heterogeneity was observed across the sequenced molecules, indicating the presence of epigenetically distinct cellular subpopulations. At the divergent MLH1/EPM2AIP1 promoter, a locus with three well-defined, nucleosome-depleted regions (NDRs), a fraction of promoter copies with inaccessible chromatin was detected and enriched upon selection of temozolomide-tolerant GBM cells. These results illustrate the biological relevance of epigenetically distinct subpopulations that in part underlie the phenotypic heterogeneity of tumor cell populations. Furthermore, these findings show that alterations in chromatin accessibility without accompanying changes in DNA methylation may constitute a novel class of epigenetic biomarker.

[1]  Peter A. Jones,et al.  OCT4 establishes and maintains nucleosome-depleted regions that provide additional layers of epigenetic regulation of its target genes , 2011, Proceedings of the National Academy of Sciences.

[2]  Carolina E. Pardo,et al.  Simultaneous Single‐Molecule Mapping of Protein‐DNA Interactions and DNA Methylation by MAPit , 2011, Current protocols in molecular biology.

[3]  Gangning Liang,et al.  Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules , 2012, Genome research.

[4]  Cizhong Jiang,et al.  Nucleosome positioning and gene regulation: advances through genomics , 2009, Nature Reviews Genetics.

[5]  G. Reifenberger,et al.  Genetic alteration and expression of the phosphoinositol‐3‐kinase/Akt pathway genes PIK3CA and PIKE in human glioblastomas , 2005, Neuropathology and applied neurobiology.

[6]  A. Verma MGMT Gene Silencing and Benefit From Temozolomide in Glioblastoma , 2006 .

[7]  A. Feinberg,et al.  Increased methylation variation in epigenetic domains across cancer types , 2011, Nature Genetics.

[8]  M. Frommer,et al.  CpG islands in vertebrate genomes. , 1987, Journal of molecular biology.

[9]  H. Mehdorn,et al.  The expression of mismatch repair proteins MLH1, MSH2 and MSH6 correlates with the Ki67 proliferation index and survival in patients with recurrent glioblastoma , 2010, Neurological research.

[10]  A. Feinberg,et al.  Stochastic epigenetic variation as a driving force of development, evolutionary adaptation, and disease , 2010, Proceedings of the National Academy of Sciences.

[11]  Carolina E. Pardo,et al.  WIF1 is a frequent target for epigenetic silencing in squamous cell carcinoma of the cervix. , 2011, Carcinogenesis.

[12]  T. Tsuruo,et al.  Promotion of glioma cell survival by acyl-CoA synthetase 5 under extracellular acidosis conditions , 2009, Oncogene.

[13]  R. Mitra,et al.  Bisulfite Patch PCR enables multiplexed sequencing of promoter methylation across cancer samples. , 2010, Genome research.

[14]  J. Visvader,et al.  Cancer stem cells in solid tumours: accumulating evidence and unresolved questions , 2008, Nature Reviews Cancer.

[15]  Peter A. Jones,et al.  A decade of exploring the cancer epigenome — biological and translational implications , 2011, Nature Reviews Cancer.

[16]  Darryl Shibata,et al.  Tumour susceptibility and spontaneous mutation in mice deficient in Mlh1, Pms1 and Pms2 DMA mismatch repair , 1998, Nature Genetics.

[17]  H. Augustin,et al.  Transcriptional profiling of human glioblastoma vessels indicates a key role of VEGF‐A and TGFβ2 in vascular abnormalization , 2012, The Journal of pathology.

[18]  J. Herman,et al.  Gene silencing in cancer in association with promoter hypermethylation. , 2003, The New England journal of medicine.

[19]  A. Vescovi,et al.  Evidence for label-retaining tumour-initiating cells in human glioblastoma. , 2011, Brain : a journal of neurology.

[20]  A. Monks,et al.  Characterization of MLH1 and MSH2 DNA mismatch repair proteins in cell lines of the NCI anticancer drug screen , 2000, Cancer Chemotherapy and Pharmacology.

[21]  Carolina E. Pardo,et al.  DNA methyltransferase probing of chromatin structure within populations and on single molecules. , 2009, Methods in molecular biology.

[22]  H. Yoshioka,et al.  Ubiquitous Expression of the α1(XIX) Collagen Gene (Col19a1) during Mouse Embryogenesis Becomes Restricted to a Few Tissues in the Adult Organism* , 1997, The Journal of Biological Chemistry.

[23]  Carolina E. Pardo,et al.  Epigenetic diversity of Kaposi’s sarcoma–associated herpesvirus , 2013, Nucleic acids research.

[24]  Carolina E. Pardo,et al.  DNA methyltransferase accessibility protocol for individual templates by deep sequencing. , 2012, Methods in enzymology.

[25]  S. Rafii,et al.  Alternative promoters regulate transcription of the gene that encodes stem cell surface protein AC133. , 2004, Blood.

[26]  François Fuks,et al.  DNA methylation and histone modifications: teaming up to silence genes. , 2005, Current opinion in genetics & development.

[27]  M. Kool,et al.  Mismatch repair deficiency: a temozolomide resistance factor in medulloblastoma cell lines that is uncommon in primary medulloblastoma tumours , 2012, British Journal of Cancer.

[28]  J. V. Van Etten,et al.  Cloning, characterization and expression of the gene coding for a cytosine-5-DNA methyltransferase recognizing GpC. , 1998, Nucleic acids research.

[29]  Ian M. Carr,et al.  MethylViewer: computational analysis and editing for bisulfite sequencing and methyltransferase accessibility protocol for individual templates (MAPit) projects , 2010, Nucleic Acids Res..

[30]  Peter A. Jones,et al.  Gene Reactivation by 5-Aza-2′-Deoxycytidine–Induced Demethylation Requires SRCAP–Mediated H2A.Z Insertion to Establish Nucleosome Depleted Regions , 2012, PLoS genetics.

[31]  Peter A. Jones,et al.  Hypomethylation of a LINE-1 Promoter Activates an Alternate Transcript of the MET Oncogene in Bladders with Cancer , 2010, PLoS genetics.

[32]  S. Markowitz,et al.  Mismatch repair mutations override alkyltransferase in conferring resistance to temozolomide but not to 1,3-bis(2-chloroethyl)nitrosourea. , 1996, Cancer research.

[33]  M. Kladde,et al.  Gal4p-Mediated Chromatin Remodeling Depends on Binding Site Position in Nucleosomes but Does Not Require DNA Replication , 1998, Molecular and Cellular Biology.

[34]  Brent A Reynolds,et al.  Isolation, expansion, and differentiation of adult Mammalian neural stem and progenitor cells using the neurosphere assay. , 2009, Methods in molecular biology.

[35]  D. Ward,et al.  Mutation in the DNA mismatch repair gene homologue hMLH 1 is associated with hereditary non-polyposis colon cancer , 1994, Nature.

[36]  Scott A. Hoose,et al.  Single-molecule and population probing of chromatin structure using DNA methyltransferases. , 2007, Methods.

[37]  M. Kladde,et al.  Direct study of DNA‐protein interactions in repressed and active chromatin in living cells. , 1996, The EMBO journal.

[38]  M. Esteller Cancer epigenomics: DNA methylomes and histone-modification maps , 2007, Nature Reviews Genetics.

[39]  M. S. Kallos,et al.  Bioreactor expansion of human neural precursor cells in serum‐free media retains neurogenic potential , 2009, Biotechnology and bioengineering.

[40]  Martin J. van den Bent,et al.  Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. , 2005, The New England journal of medicine.

[41]  Erik Sundström,et al.  Cellular composition of long‐term human spinal cord‐ and forebrain‐derived neurosphere cultures , 2006, Journal of neuroscience research.

[42]  Austin G Smith,et al.  CD133 (Prominin) Negative Human Neural Stem Cells Are Clonogenic and Tripotent , 2009, PloS one.

[43]  Daiya Takai,et al.  Comprehensive analysis of CpG islands in human chromosomes 21 and 22 , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[44]  S. Schreiber,et al.  Signaling Network Model of Chromatin , 2002, Cell.

[45]  Ben S. Wittner,et al.  A Chromatin-Mediated Reversible Drug-Tolerant State in Cancer Cell Subpopulations , 2010, Cell.

[46]  G. Reifenberger,et al.  Promoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomas , 2011, International journal of cancer.