Skip to main content

Interdependency Between Genetic and Epigenetic Regulatory Defects in Cancer

  • Protocol
  • First Online:
Cancer Cell Signaling

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1165))

Abstract

Epigenetic regulation is understood as heritable changes in gene expression and genome function that can occur without affecting the DNA sequence. In its in vivo context DNA is coupled to a group of small basic proteins that together with the DNA form the chromatin. The organization and regulation of the chromatin alliance with multiple nuclear functions are inconceivable without genetic information. With the advance on the understanding of the chromatin organization of the eukaryotic genome, it has been clear that not only genetics but also epigenetics influence both normal human biology and diseases. As a consequence, the basic concepts and mechanisms of cancer need to be readdressed and viewed not only locally but also at the whole genome scale or even, in the three-dimensional context of the cell nucleus space. Such a vision has a larger impact than has been previously predicted, since phenomena like aging, senescence, the entail of nutrition, stem cell biology, and cancer are orchestrated by epigenetic and genetic processes. Here I describe the relevance and central role of genetic and epigenetic defects in cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.00
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674

    Article  CAS  PubMed  Google Scholar 

  2. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW (2013) Cancer genome landscapes. Science 339:1546–1558

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Rodríguez-Paredes M, Esteller M (2011) Cancer epigenetics reaches mainstream oncology. Nat Med 17:330–339

    Article  PubMed  Google Scholar 

  4. You JS, Jones PA (2012) Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 22:9–20

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Van Speybroeck L (2002) From epigenesis to epigenetics: the case of C.H. Waddington. Ann N Y Acad Sci 981:61–81

    Article  PubMed  Google Scholar 

  6. Slack JMW (2002) Conrad Hal Waddington: the last renaissance biologist? Nat Rev Genet 3:889–895

    Article  CAS  PubMed  Google Scholar 

  7. Lim JP, Brunet A (2013) Bridging the transgenerational gap with epigenetic memory. Trends Genet 29:176–186

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Pujadas E, Feinberg AP (2012) Regulated noise in the epigenetic landscape of development and disease. Cell 148:1123–1131

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Portela A, Esteller M (2010) Epigenetic modifications and human disease. Nat Biotechnol 28:1057–1068

    Article  CAS  PubMed  Google Scholar 

  10. ENCODE Project Consortium (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74

    Article  Google Scholar 

  11. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB et al (2011) Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473:43–39

    Google Scholar 

  12. Bird A (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16:6–21

    Article  CAS  PubMed  Google Scholar 

  13. Smith ZD, Meissner A (2013) DNA methylation: roles in mammalian development. Nat Rev Genet 14:204–220

    Article  CAS  PubMed  Google Scholar 

  14. Okano M, Bell DW, Haber DA, Li E (1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for the de novo methylation and mammalian development. Cell 99:247–257

    Article  CAS  PubMed  Google Scholar 

  15. Bestor TH (2000) The DNA methyltransferases of mammals. Hum Mol Genet 9:2395–2402

    Article  CAS  PubMed  Google Scholar 

  16. Rhee I, Bachman KE, Park BH, Jair KW, Yen RW, Schuebel KE et al (2002) DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 416:552–556

    Article  CAS  PubMed  Google Scholar 

  17. Li E (2002) Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet 3:662–675

    Article  CAS  PubMed  Google Scholar 

  18. Cordaux R, Batzer MA (2009) The impact of retrotransposons on human genome evolution. Nat Rev Genet 10:691–703

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Meehan RR, Lweis JD, Bird AP (1992) Characterization of MeCP2, a vertebrate DNA binding protein with affinity to methylated DNA. Nucleic Acids Res 20:5085–5092

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Hendrich B, Bird AP (1998) Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol Cell Biol 18:6538–6547

    CAS  PubMed Central  PubMed  Google Scholar 

  21. Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N et al (1998) Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet 19:187–191

    Article  CAS  PubMed  Google Scholar 

  22. Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN et al (1998) Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393:386–389

    Article  CAS  PubMed  Google Scholar 

  23. Magdinier F, Billard LM, Wittmann G et al (2000) Regional methylation of the 5′ end CpG island of BRCA1 is associated with reduced gene expression in human somatic cells. FASEB J 14:1585–1594

    Article  CAS  PubMed  Google Scholar 

  24. Magdinier F, Wolffe AP (2001) Selective association of the methyl-CpG binding protein MBD2 with the silent p14/p16 locus in human neoplasia. Proc Natl Acad Sci USA 98:4990–4995

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Ballestar E, Paz MF, Valle L, Wei S, Fraga MF, Espada J et al (2003) Methyl-CpG binding proteins identify novel sites of epigenetic inactivation in human cancer. EMBO J 22:6335–6345

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Baubec T, Ivánek R, Lienert F, Schübeler D (2013) Methylation-dependent and -independent genomic targeting principles of the MBD protein family. Cell 153:480–492

    Article  CAS  PubMed  Google Scholar 

  27. Antequera F (2003) Structure, function and evolution of CpG island promoters. Cell Mol Life Sci 60:1647–1658

    Article  CAS  PubMed  Google Scholar 

  28. Weber M, Hellmann I, Stadler MB, Ramos L, Pääbo S, Rebhan M et al (2007) Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 39:457–466

    Article  CAS  PubMed  Google Scholar 

  29. Lienert F, Wirbelawer C, Som I, Dean A, Mohn F, Schübeler D (2011) Identification of genetic elements that autonomously determine DNA methylation states. Nat Genet 43:1091–1097

    Article  CAS  PubMed  Google Scholar 

  30. Issa J-P (2004) CpG island methylator phenotype in cancer. Nat Rev Cancer 4:988–993

    Article  CAS  PubMed  Google Scholar 

  31. Recillas-Targa F, de la Rosa-Velázquez IA, Soto-Reyes A (2011) Insulation of tumor supresor genes by the nuclear factor CTCF. Biochem Cell Biol 89:479–488

    Article  PubMed  Google Scholar 

  32. Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P et al (2009) The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 41:178–186

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Toni-Filippini J et al (2009) Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462:315–322

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Hemberger W, Dean W, Reik W (2009) Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddingston’s canal. Nat Rev Mol Cell Biol 10:526–537

    Article  CAS  PubMed  Google Scholar 

  35. Wu H, Zhang Y (2011) Mechanisms and functions of Tet protein-mediated 5-methylcytosine oxidation. Genes Dev 25:2436–2452

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Bridno Y et al (2009) Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324:930–935

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Lund AH, van Lohuizen M (2004) Epigenetics and cancer. Genes Dev 18:315–2335

    Article  Google Scholar 

  38. Zenter GE, Henikoff S (2013) Regulation of nucleosome dynamics by histone modification. Nat Struct Mol Biol 20:259–266

    Article  Google Scholar 

  39. Fraga MF, Balletar E, Villar-Garea A, Boix-Chornet M, Espada Y, Schotta G et al (2005) Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet 37:91–400

    Article  Google Scholar 

  40. Zink D, Fischer AH, Nickerson JA (2004) Nuclear structure in cancer cells. Nat Rev Cancer 4:677–687

    Article  CAS  PubMed  Google Scholar 

  41. Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C et al (2008) Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet 40:741–750

    Article  CAS  PubMed  Google Scholar 

  42. Guil S, Esteller M (2012) Cis-acting noncoding RNAs: friends and foes. Nat Struct Mol Biol 19:1068–1075

    Article  CAS  PubMed  Google Scholar 

  43. Recillas-Targa F (2002) DNA methylation, chromatin boundaries and mechanisms of genomic imprinting. Arch Med Res 33:428–438

    Article  CAS  PubMed  Google Scholar 

  44. Henckel A, Nakabayashi K, Sanz LA, Feil R, Hata K, Arnaud P (2009) Histone methylation is mechanistically linked to DNA methylation at imprinting control regions in mammals. Hum Mol Genet 18:3375–3383

    Article  CAS  PubMed  Google Scholar 

  45. Ratnakumar K, Berstein E (2013) ATRX: the case of a peculiar chromatin remodeler. Epigenetics 8:3–9

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Esteller M (2005) DNA methylation and cancer therapy: new developments and expectations. Curr Opin Oncol 17:55–60

    Article  CAS  PubMed  Google Scholar 

  47. Esteller M, Levine R, Baylin SB, Ellenson LH, Herman JG (1998) MLH1 promoter hypermethylation is associated with the microsatellite instability phenotype in sporadic endometrial carcinomas. Oncogene 17:2413–2417

    Article  CAS  PubMed  Google Scholar 

  48. Esteller M (2005) Dormant hypermethylated tumour suppressor genes: questions and answers. J Pathol 205:172–180

    Article  CAS  PubMed  Google Scholar 

  49. DeSmet C, DeBacker O, Faraoni I, Lurquin C, Brasseur F, Boon T (1996) The activation of human gene MAGE-1in tumor cells is correlated with genome-wide demethylation. Proc Natl Acad Sci USA 93:7149–7153

    Article  CAS  Google Scholar 

  50. Adorjan P, Distler J, Lipscher E, Model F, Muller J, Pelet C et al (2002) Tumour class prediction and discovery by microarray-based DNA methylation analysis. Nucleic Acids Res 30:e21

    Article  PubMed Central  PubMed  Google Scholar 

  51. Oshimo Y, Nakayama H, Ito R, Kitadai Y, Yoshida K, Chayama K et al (2003) Promoter methylation of cyclin D2 gene in gastric carcinoma. Int J Oncol 6:1663–1670

    Google Scholar 

  52. Sakai T, Toguchida J, Ohtani N, Yandell DW, Rapaport JM, Dryja TP (1991) Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene. Am J Hum Genet 48:880–888

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S et al (1994) Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci USA 91:9700–9704

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC et al (1995) 5′CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancer. Nat Med 1:686–692

    Article  CAS  PubMed  Google Scholar 

  55. De La Rosa-Velázquez IA, Rincón-Arano H, Benítez-Bribiesca L, Recillas-Targa F (2007) Epgienetic regulation of the human retinoblastoma tumor supresor gene promoter by CTCF. Cancer Res 67:2577–2585

    Article  Google Scholar 

  56. Soto-Reyes E, Recillas-Targa F (2010) Epigenetic regulation of the human p53 gene promoter by the CTCF transcription factor in transformed cell lines. Oncogene 29:2217–2227

    Article  CAS  PubMed  Google Scholar 

  57. Cunningham JM, Christensen ER, Tester DJ, Kim CY, Roche PC, Burgart LJ et al (1998) Hypermethylation of the hMLH1 promoter in colon cancer with microsatellite instability. Cancer Res 58:3455–3460

    CAS  PubMed  Google Scholar 

  58. Chen WY, Cooper TK, Zahnow CA, Overholtzer M, Zhao Z, Ladanyi M et al (2004) Epigenetic and genetic loss of Hic1 function accentuates the role of p53 in tumorigenesis. Cancer Cell 6:387–398

    Article  CAS  PubMed  Google Scholar 

  59. Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA et al (2003) Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113:703–716

    Article  CAS  PubMed  Google Scholar 

  60. Liu DX, Nath N, Chellappan SP, Greene LA (2005) Regulation of neuron survival and death by p130 and associated chromatin modifiers. Genes Dev 19:719–732

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Margueron R, Reinberg D (2011) The Polycomb complex PRC2 and its mark in life. Nature 469:343–349

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Hansen KH, Bracken AP, Pasini D, Dietrich N, Gehani SS, Monrad A et al (2008) A model for transmission of the H3K27me3 epigenetic mark. Nat Cell Biol 10:1291–1300

    Article  CAS  PubMed  Google Scholar 

  63. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG et al (2002) The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419:624–629

    Article  CAS  PubMed  Google Scholar 

  64. Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA et al (2003) EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA 100:11606–11611

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. Asangani IA, Ateeq B, Cao Q, Dodson L, Pandhi M, Kunju LP et al (2013) Characterization of the EZH2-MMSET histone methyltransferase regulatory axis in cancer. Mol Cell 49:741–750

    Google Scholar 

  66. Dávalos-Salas M, Furlan-Magaril M, González-Buendía E, Valdes-Quezada C, Ayala-Ortega E, Recillas-Targa F (2011) Gain of DNA methylation is enhanced in the absence of CTCF at the human retinoblastoma gene promoter. BMC Cancer 11:232

    Google Scholar 

  67. DiCroce L, Raker VA, Corsaro M, Fazi F, Fanelli M, Faretta M et al (2002) Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 295:1079–1082

    Article  CAS  Google Scholar 

  68. DiCroce L (2005) Chromatin modifying activity of leukaemia associated fusion proteins. Hum Mol Genet 14:R77–R84

    Article  CAS  Google Scholar 

  69. Peterson LF, Zhang DE (2004) The 8; 21 translocation in leukaemogenesis. Oncogene 23:4255–4262

    Article  CAS  PubMed  Google Scholar 

  70. Rinn JL, Chang HY (2012) Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166

    Article  CAS  PubMed  Google Scholar 

  71. Mercer TR, Mattick JS (2013) Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol 20:300–307

    Article  CAS  PubMed  Google Scholar 

  72. Ryan BM, Robles AD, Harris CC (2010) Genetic variation in microRNA networks: the implications for cancer research. Nat Rev Cancer 10:389–402

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  73. Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B et al (2008) Genomic loss of microRNA.101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science 322:1695–1699

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  74. Friedman JM, Liang G, Liu CC, Wolff EM, Tsai YC, Ye W et al (2009) The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2. Cancer Res 69:2623–2629

    Article  CAS  PubMed  Google Scholar 

  75. Fabbii M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E et al (2007) MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA 104:15805–15810

    Article  Google Scholar 

  76. Baer C, Claus R, Plass C (2012) Genome-wide epigenetic regulation of microRNAs in cancer. Cancer Res 73:473–477

    Article  Google Scholar 

  77. Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato S, Setien F et al (2007) Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res 67:1424–1429

    Article  CAS  PubMed  Google Scholar 

  78. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H et al (2012) The GENCODE v7 catalog of human long noncoding RNAs; analysis of their gene structure, evolution and expression. Genome Res 22:1775–1789

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  79. Escamilla-Del-Arenal M, da Rocha ST, Heard E (2011) Evolutionary diversity and developmental regulation of X-chromosome inactivation. Hum Genet 130:307–327

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  80. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA et al (2007) Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129:1311–1323

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  81. Gutpa RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ et al (2010) Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464:1071–1076

    Article  Google Scholar 

  82. Yap KL, Lis S, Muñoz-Cabello AM, Raguz S, Zeng L, Mujtaba S et al (2010) Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 4 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell 38:662–674

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  83. Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D et al (2010) A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142:409–419

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  84. Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y et al (2011) A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472:120–124

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  85. Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L et al (2013) Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell 25:69–80

    Article  CAS  PubMed  Google Scholar 

  86. Cremer T, Cremer C (2001) Chromosomes territories, nuclear architecture and gene regulation in mammalian cells. Nat Rev Genet 2:292–301

    Article  CAS  PubMed  Google Scholar 

  87. Fraser P, Bickmore W (2007) Nuclear organization of the genome and the potential for gene regulation. Nature 447:413–417

    Article  CAS  PubMed  Google Scholar 

  88. Bulger M, Groudine M (1999) Looping versus linking: toward a model for long-distance gene activation. Genes Dev 13:2465–2477

    Article  CAS  PubMed  Google Scholar 

  89. Reddy KL, Feinberg AP (2012) Higher order chromatin organization in cancer. Semin Cancer Biol 23:109–115

    Article  PubMed Central  PubMed  Google Scholar 

  90. Coolen MW, Stirzaker C, Song JZ, Statham AL, Kassir Z, Moreno CS et al (2010) Consolidation of the cancer genome into domains of repressive chromatin by long-range epigenetic silencing (LRES) reduces transcriptional plasticity. Nat Cell Biol 12:235–246

    CAS  PubMed Central  PubMed  Google Scholar 

  91. Bert SA, Robinson MD, Strbenac D, Statham AL, Song JZ, Hulf T et al (2013) Regional activation of the cancer genome by long-range epigenetic remodeling. Cancer Cell 2:9–22

    Article  Google Scholar 

  92. Peric-Hupkes D, Meuleman W, Pagie L, Bruggeman SW, Solovei I, Brugman W et al (2010) Molecular maps of the reorganization of genome-nuclear lamina interaction during differentiation. Mol Cell 38:603–613

    Article  CAS  PubMed  Google Scholar 

  93. Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Talhout W et al (2008) Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature 453:948–951

    Article  CAS  PubMed  Google Scholar 

  94. Azad N, Zahnow CA, Rudin CM, Baylin SB (2013) The future of epigenetic therapy in solid tumors-lessons from the past. Nat Rev Clin Oncol 10:256–266

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by the Dirección General de Asuntos del Personal Académico-Universidad Nacional Autónoma de México (IN209403 and IN203811), Consejo Nacional de Ciencia y Tecnología, México (CONACyT; 42653-Q and 128464).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Félix Recillas-Targa .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this protocol

Cite this protocol

Recillas-Targa, F. (2014). Interdependency Between Genetic and Epigenetic Regulatory Defects in Cancer. In: Robles-Flores, M. (eds) Cancer Cell Signaling. Methods in Molecular Biology, vol 1165. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-0856-1_4

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-0856-1_4

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-0855-4

  • Online ISBN: 978-1-4939-0856-1

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics