Int J Stem Cells.  2019 Nov;12(3):457-462. 10.15283/ijsc19074.

Inactivation of the NHEJ Activity of DNA-PKcs Prevents Fanconi Anemia Pre-Leukemic HSC Expansion

Affiliations
  • 1Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA. Qishen.pang@cchmc.org
  • 2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.

Abstract

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure and high risk of cancer particularly leukemia. Here we show that inactivation of the non-homologous end-joining (NHEJ) activity of DNA-PKcs prevented DNA damage-induced expansion of FA pre-leukemic hematopoietic stem cells (HSCs). Furthermore, we performed serial BM transplantation to demonstrate that the DNA damage-induced expanded FA HSC compartment contained pre-leukemic stem cells that required the NHEJ activity of DNA-PKcs to induce leukemia in the secondary recipients. These results suggest that NHEJ may collaborate with FA deficiency to promote DNA damage-induced expansion of pre-leukemic HSCs.

Keyword

DNA damage; Fanconi anemia; Leukemic stem cells; Non-homologous end-joining

MeSH Terms

Bone Marrow
DNA
DNA Damage
Fanconi Anemia*
Hematopoietic Stem Cells
Leukemia
Stem Cells
DNA

Figure

  • Fig. 1 Inactivation of the NHEJ activity of DNA-PKcs prevents the expansion of pre-leukemic Fanca−/− HSCs. (A) Inactivation of the NHEJ activity of DNA-PKcs prevents MMC-induced expansion of the Fanca−/− HSCs. 2,000 LSK cells from E14.5 embryos with the indicated genotype, along with 200,000 c-Kit-depleted protector cells, were transplanted into lethally irradiated BoyJ (CD45.1+) recipients. The recipients were i.p. injected with low dose of MMC (0.3 mg/kg) weekly for 6 weeks. Two days after the final injection, donor-derived (CD45.2+) BM LSK cells were gated for analysis of the SLAM (LSKCD150+CD48−) cell population. (B) MMC induces a significant increase in apoptosis in DNA-PKcs3A/3A, Fanca−/− and DNA-PKcs3A/3AFanca−/− SLAM cells. Donor-derived (CD45.2+) BM LSK cells were gated for SLAM population and analyzed for apoptosis by Annexin V and 7AAD. (C) MMC causes increased cell cycling in Fanca−/− HSCs. Donor-derived (CD45.2+) BM LSK cells were gated for the SLAM population and analyzed for the cell cycle using Hochest 33342/Ki67 staining. (D) Survival of transplant recipients. 2,000 LSK cells from E14.5 embryos with the indicated genotype, along with 200,000 c-Kit-depleted protector cells, were transplanted into lethally irradiated BoyJ recipients (n=10 per group). MMC (0.3 mg/kg) was then administrated to the recipients weekly for 6 weeks. Survival of the recipients plotted by the Kaplan-Meier curve method and analyzed by the log-rank test. (E~G) Anemia but no leukemia developed in primary recipient mice. The moribund mice described in (D) were subjected to analysis for peripheral blood count (E), and splenomegaly (F) and infiltration of mature myeloid cells (E) in the spleens. *p<0.05; **p<0.01.

  • Fig. 2 The NHEJ activity of DNA-PKcs is required for the pre-leukemic Fanca−/− HSCs to induce leukemia in the secondary recipients. (A~C) Inactivation of the NHEJ activity of DNA-PKcs restores normal levels of BM cellularity and phenotypic HSCs in the BM of the secondary recipients. 2,000 LSK cells from E14.5 embryos with the indicated genotype, along with 200,000 c-Kit-depleted protector cells, were transplanted into lethally irradiated BoyJ (CD45.1+) recipients. The recipients were i.p. injected with low dose of MMC (0.3 mg/kg) weekly for 6 weeks. Two days after the final injection, the mice were sacrificed and analyzed for BM cellularity (A), total donor (CD45.2) engraftment (B), and donor-derived SLAM (LSKCD150+CD48−) cells (C). (D) Fanca−/− secondary recipients develop lethal leukemia. 2 million BM cells from the primary recipient mice described in (A) were transplanted into sublethally irradiated secondary CD45.1+ recipient mice. Survival of the recipients was monitored and plotted by the Kaplan-Meier curve method. n=8~10 mice for each group. (E, F) Increased myeloid infiltration in the spleen of the leukemic mice. Splenocytes from the recipient mice described in (D) were subjected to flow cytometry analysis for mature myeloid cells (E) and immature myeloid blasts (F). (G) Photomicrographs of Wright–Giemsa stain show increased blast cells in the peripheral blood smear from secondary recipient mice transplanted with Fanca−/− donor cells. *p<0.05; **p<0.01.


Reference

References

1. Bagby GC Jr. Genetic basis of Fanconi anemia. Curr Opin Hematol. 2003; 10:68–76. DOI: 10.1097/00062752-200301000-00011. PMID: 12483114.
Article
2. Green AM, Kupfer GM. Fanconi anemia. Hematol Oncol Clin North Am. 2009; 23:193–214. DOI: 10.1016/j.hoc.2009.01.008. PMID: 19327579. PMCID: PMC5912671.
Article
3. Bogliolo M, Surrallés J. Fanconi anemia: a model disease for studies on human genetics and advanced therapeutics. Curr Opin Genet Dev. 2015; 33:32–40. DOI: 10.1016/j.gde.2015.07.002. PMID: 26254775.
Article
4. Kottemann MC, Smogorzewska A. Fanconi anaemia and the repair of Watson and Crick DNA crosslinks. Nature. 2013; 493:356–363. DOI: 10.1038/nature11863. PMID: 23325218. PMCID: PMC3700363.
Article
5. Dong H, Nebert DW, Bruford EA, Thompson DC, Joenje H, Vasiliou V. Update of the human and mouse Fanconi anemia genes. Hum Genomics. 2015; 9:32. DOI: 10.1186/s40246-015-0054-y. PMID: 26596371. PMCID: PMC4657327.
Article
6. Mamrak NE, Shimamura A, Howlett NG. Recent discoveries in the molecular pathogenesis of the inherited bone marrow failure syndrome Fanconi anemia. Blood Rev. 2017; 31:93–99. DOI: 10.1016/j.blre.2016.10.002. PMCID: PMC5391297.
Article
7. Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, Giampietro PF, Hanenberg H, Auerbach AD. A 20-year perspective on the International Fanconi Anemia Registry (IFAR). Blood. 2003; 101:1249–1256. DOI: 10.1182/blood-2002-07-2170.
Article
8. Vélez-Ruelas MA, Martínez-Jaramillo G, Arana-Trejo RM, Mayani H. Hematopoietic changes during progression from Fanconi anemia into acute myeloid leukemia: case report and brief review of the literature. Hematology. 2006; 11:331–334. DOI: 10.1080/10245330500397703.
Article
9. Mehta P, Locatelli F, Stary J, Smith FO. Bone marrow transplantation for inherited bone marrow failure syndromes. Pediatr Clin North Am. 2010; 57:147–170. DOI: 10.1016/j.pcl.2010.01.002. PMID: 20307716.
Article
10. Schneider M, Chandler K, Tischkowitz M, Meyer S. Fanconi anaemia: genetics, molecular biology, and cancer – implications for clinical management in children and adults. Clin Genet. 2015; 88:13–24. DOI: 10.1111/cge.12517.
Article
11. Adamo A, Collis SJ, Adelman CA, Silva N, Horejsi Z, Ward JD, Martinez-Perez E, Boulton SJ, La Volpe A. Preventing nonhomologous end joining suppresses DNA repair defects of Fanconi anemia. Mol Cell. 2010; 39:25–35. DOI: 10.1016/j.molcel.2010.06.026. PMID: 20598602.
Article
12. Bunting SF, Nussenzweig A. Dangerous liaisons: Fanconi anemia and toxic nonhomologous end joining in DNA crosslink repair. Mol Cell. 2010; 39:164–166. DOI: 10.1016/j.molcel.2010.07.016. PMID: 20670885. PMCID: PMC6591721.
Article
13. Pace P, Mosedale G, Hodskinson MR, Rosado IV, Sivasubramaniam M, Patel KJ. Ku70 corrupts DNA repair in the absence of the Fanconi anemia pathway. Science. 2010; 329:219–223. DOI: 10.1126/science.1192277. PMID: 20538911.
Article
14. Bunting SF, Callén E, Wong N, Chen HT, Polato F, Gunn A, Bothmer A, Feldhahn N, Fernandez-Capetillo O, Cao L, Xu X, Deng CX, Finkel T, Nussenzweig M, Stark JM, Nussenzweig A. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010; 141:243–254. DOI: 10.1016/j.cell.2010.03.012. PMID: 20362325. PMCID: PMC2857570.
Article
15. Zhang S, Yajima H, Huynh H, Zheng J, Callen E, Chen HT, Wong N, Bunting S, Lin YF, Li M, Lee KJ, Story M, Gapud E, Sleckman BP, Nussenzweig A, Zhang CC, Chen DJ, Chen BP. Congenital bone marrow failure in DNA-PKcs mutant mice associated with deficiencies in DNA repair. J Cell Biol. 2011; 193:295–305. DOI: 10.1083/jcb.201009074. PMID: 21482716. PMCID: PMC3080267.
Article
16. Carreau M, Gan OI, Liu L, Doedens M, McKerlie C, Dick JE, Buchwald M. Bone marrow failure in the Fanconi anemia group C mouse model after DNA damage. Blood. 1998; 91:2737–2744. DOI: 10.1182/blood.V91.8.2737.2737_2737_2744. PMID: 9531583.
Article
17. Du W, Amarachintha S, Wilson A, Pang Q. The immune receptor Trem1 cooperates with diminished DNA damage response to induce preleukemic stem cell expansion. Leukemia. 2017; 31:423–433. DOI: 10.1038/leu.2016.242.
Article
18. Du W, Amarachintha S, Wilson AF, Pang Q. Hyper-active non-homologous end joining selects for synthetic lethality resistant and pathological Fanconi anemia hematopoietic stem and progenitor cells. Sci Rep. 2016; 6:22167. DOI: 10.1038/srep22167. PMID: 26916217. PMCID: PMC4768158.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr