DNA repair mechanisms and cellular reprogramming - criteria for successful generation of human induced pluripotent stem cells and in vitro disease modelling

  • Miodrag Stojkovic Human Genetics Department, Faculty of Medical Sciences, University of Kragujevac, Serbia SPEBO MEDICAL, Leskovac, Serbia
  • Lyle Armstrong Institute of Genetic Medicine, Newcastle University, Newcastle NE1 3BZ, UK
  • Majlinda Lako Institute of Genetic Medicine, Newcastle University, Newcastle NE1 3BZ, UK

Abstract


All stem cells, whether they are pluripotent or multipotent tissue specific cells, need to be able to eliminate genomic mutations if they threaten the ability of the cell to complete its function in vivo.  For adult stem cells whose purpose is the lifelong repair and regeneration of a specific organ system, this is of great importance.  It takes only a little imagination to envisage the potential harm that could accrue in the haematopoietic system if only a small fraction of the haematopoietic stem cells (HSCs), resident in the endosteal niche of the bone marrow (BM) were allowed to accumulate mutations that altered their ability to generate lymphoid or myeloid lineages.  The daily requirement for new blood cells is of the order of one billion, so damage of this type would soon become apparent and the patient’s health would decline.  Important though this is, the impact of mutations occurring in adult life is minor compared to the possible disruptions that could arise if the genome of the early embryo was damaged to the point that embryonic development was restricted.  The inner cell mass (ICM) of blastocyst stage embryos gives rise to every tissue found in the adult but consists of a very small number of cells (this can be as few as 12 cells).  If a mutation is allowed to persist in any of the ICM cells, it will be transferred to the tissues and organs that arise from that cell and could have a profound effect on organ function. The accumulation of mutations in the ICM must be prevented at all costs.

References

Hyslop LA, Stojkovic M and Lako M. (2005) Human embryonic stem cell biology and clinical implications. Expert reviews in Molecular Medicine 7:-21

Takahashi K, Okita K, Nakagawa M, Yamanaka S. (2007) Induction of pluripotent stem cells from fibroblast cultures. Nat Protoc. 2(12):3081-9.

Yamanaka S.(2008) Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif. 41 Suppl 1:51-6

Ohnuki M, Takahashi K, Yamanaka S. (2009) Generation and characterization of human induced pluripotent stem cells. Curr Protoc Stem Cell Biol. Chapter 4:Unit 4A.2

Saretzki G, Armstrong L, Leake A, Lako M, von Zglinicki T.(2004) Stress defense in murine embryonic stem cells is superior to that of various differentiated murine cells. Stem Cells.22(6):962-71

Saretzki G, Walter T, Atkinson S, Passos JF, Bareth B, Keith WN, Stewart R, Hoare S, Stojkovic M, Armstrong L, von Zglinicki T, Lako M. (2008) Downregulation of multiple stress defense mechanisms during differentiation of human embryonic stem cells. Stem Cells. 26(2):455-64

Armstrong L, Tilgner K, Saretzki G, Atkinson SP, Stojkovic M, Moreno R, Przyborski S, Lako M. (2010) Human induced pluripotent stem cell lines show stress defense mechanisms and mitochondrial regulation similar to those of human embryonic stem cells. Stem Cells 28(4):661-73.

Kenyon J, Gerson SL. The role of DNA damage repair in aging of adult stem cells, Nucleic Acids Res 2007; 35: 7557–7565.

Nouspikel T, Hanawalt PC (2000) Terminally differentiated human neurons repair transcribed genes but display attenuated global DNA repair and modulation of repair gene expression, Mol. Cell. Biol. 20: 1562–1570.

Momcilovic O, Knobloch L, Fornsaglio J, Varum S, Easley C, Schatten G (2010) DNA damage responses in human induced pluripotent stem cells and embryonic stem cells, PLoS ONE 5 :: e13410.

Mathur D, Danford TW, Boyer LA, Young RA, Gifford DK, Jaenisch R. (2008) Analysis of the mouse embryonic stem cell regulatory networks obtained by ChIP-chip and ChIP-PET. Genome Biol. 9(8):R126

Fong YW, Inouye C, Yamaguchi T, Cattoglio C, Grubisic I, Tjian R. (2011) A DNA repair complex functions as an Oct4/Sox2 coactivator in embryonic stem cells. Cell. 147(1):120-31

Zhan Q, Carrier F, Fornace AJ Jr. (1993) Induction of cellular p53 activity by DNA-damaging agents and growth arrest. Mol Cell Biol. 13(7):4242-50.

Marión RM, Strati K, Li H, Murga M, Blanco R, Ortega S, Fernandez-Capetillo O, Serrano M, Blasco MA.(2009) A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 460(7259):1149-53

Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA.(2010) Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science. 329(5987):78-82

Caldecott KW (2008) Single-strand break repair and genetic disease. Nat Rev Genet. 9(8):619-31

Niedernhofer LJ, Lalai AS, Hoeijmakers JH.(2005) Fanconi anemia (cross)linked to DNA repair. Cell.123(7):1191-1198.

Kim Y, Lach FP, Desetty R, Hanenberg H, Auerbach AD, Smogorzewska A.(2011) Mutations of the SLX4 gene in Fanconi anemia. Nat Genet. 43(2):142-146.

Auerbach AD, Wolman SR.(1976) Susceptibility of Fanconi's anaemia fibroblasts to chromosome damage by carcinogens. Nature. 261(5560):494-6.

Raya A, Rodríguez-Pizà I, Guenechea G, Vassena R, Navarro S, Barrero MJ, Consiglio A, Castellà M, Río P, Sleep E, González F, Tiscornia G, Garreta E, Aasen T, Veiga A, Verma IM, Surrallés J, Bueren J, Izpisúa Belmonte JC.(2009) Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature. 460(7251):53-9.

Müller LU, Milsom MD, Harris CE, Vyas R, Brumme KM, Parmar K, Moreau LA, Schambach A, Park IH, London WB, Strait K, Schlaeger T, Devine AL, Grassman E, D'Andrea A, Daley GQ, Williams DA. (2012) Overcoming reprogramming resistance of Fanconi anemia cells. Blood. 119(23):5449-57

Turinetto V, Orlando L, Sanchez-Ripoll Y, Kumpfmueller B, Storm MP, Porcedda P, Minieri V, Saviozzi S, Accomasso L, Cibrario Rocchietti E, Moorwood K, Circosta P, Cignetti A, Welham MJ, Giachino C. (2012) High basal γH2AX levels sustain self-renewal of mouse embryonic and induced pluripotent stem cells. Stem Cells. 30(7):1414-23.

Yung SK, Tilgner K, Ledran MH, Habibollah S, Neganova I, Singhapol C, Saretzki G, g Stojkovic M, Armstrong L, Przyborski S, Lako M (2012) Human pluripotent stem cell models of Fanconi Anaemia deficiency reveal animportant role for Fanconi Anaemia proteins in cellular reprogramming and survival of haematopoietic progenitors. Stem Cells (in press).

Buck D, Malivert L, de Chasseval R, et al (2006). Cernunnos, a novel nonhomologous end-joining factor, is mutated in human immunodeficiency with microcephaly. Cell. 124(2):287-99.

Tilgner K, Neganova I, Singhapol C, Saretzki G, Evans J, Gorbunova V, Gennery A, Przyborski S, Stojkovic M, Armstrong L, Jeggo P and Lako M. (2012) A human induced pluripotent stem cell model of Cernunnos deficiency reveals an important role for XLF in the survival of most primitive haematopoietic Progenitors. (under review with Stem Cells)

Seluanov A, Mao Z, Gorbunova V. (2002) Analysis of DNA double-strand break (DSB) repair in mammalian cells. J. Vis. Exp. 43:pii:2002.

O'Driscoll M, Cerosaletti KM, Girard PM, Dai Y, Stumm M, Kysela B, et al.(2001) DNA ligase IV mutations identified in patients exhibiting developmental delay and immunodeficiency. Mol Cell 8(6): 1175-1185

Riballo E, Critchlow SE, Teo SH, Doherty AJ, Priestley A, Broughton B, et al (1999). Identification of a defect in DNA ligase IV in a radiosensitive leukaemia patient. Curr Biol. 9(13): 699-702

Tilgner K, Neganova I, Yung S, Singhapol C, Saretzki G, Evans J, Gorbunova V, Gennery A, Przyborski S, Stojkovic M, Armstrong L, Jeggo P and Lako M (2012) A human iPSC model of Ligase IV deficiency reveals an important role for NHEJ-mediated-DSB repair in the survival and genomic stability of induced pluripotent stem cells and emerging haematopoietic progenitors. Cell Death and Differentiation (under final review).

Published
2013/02/24
Section
Review Paper