Overcoming reprogramming resistance of Fanconi anemia cells

LUW Müller, MD Milsom, CE Harris… - Blood, The Journal …, 2012 - ashpublications.org
LUW Müller, MD Milsom, CE Harris, R Vyas, KM Brumme, K Parmar, LA Moreau…
Blood, The Journal of the American Society of Hematology, 2012ashpublications.org
Fanconi anemia (FA) is a recessive syndrome characterized by progressive fatal BM failure
and chromosomal instability. FA cells have inactivating mutations in a signaling pathway that
is critical for maintaining genomic integrity and protecting cells from the DNA damage
caused by cross-linking agents. Transgenic expression of the implicated genes corrects the
phenotype of hematopoietic cells, but previous attempts at gene therapy have failed largely
because of inadequate numbers of hematopoietic stem cells available for gene correction …
Abstract
Fanconi anemia (FA) is a recessive syndrome characterized by progressive fatal BM failure and chromosomal instability. FA cells have inactivating mutations in a signaling pathway that is critical for maintaining genomic integrity and protecting cells from the DNA damage caused by cross-linking agents. Transgenic expression of the implicated genes corrects the phenotype of hematopoietic cells, but previous attempts at gene therapy have failed largely because of inadequate numbers of hematopoietic stem cells available for gene correction. Induced pluripotent stem cells (iPSCs) constitute an alternate source of autologous cells that are amenable to ex vivo expansion, genetic correction, and molecular characterization. In the present study, we demonstrate that reprogramming leads to activation of the FA pathway, increased DNA double-strand breaks, and senescence. We also demonstrate that defects in the FA DNA-repair pathway decrease the reprogramming efficiency of murine and human primary cells. FA pathway complementation reduces senescence and restores the reprogramming efficiency of somatic FA cells to normal levels. Disease-specific iPSCs derived in this fashion maintain a normal karyotype and are capable of hematopoietic differentiation. These data define the role of the FA pathway in reprogramming and provide a strategy for future translational applications of patient-specific FA iPSCs.
ashpublications.org