Page 125 - 2019_05-HaematologicaMondo-web
P. 125

POLG inhibition promotes AML differentiation
primary AML model may reflect protection from the BM niche, or that the tested sample was resistant to alovudine. Further studies will be needed to clarify the basis for this sensitivity and resistance.
In clinical trials with patients with AIDS and HIV, alovu- dine reduced viral load but its development for HIV did not progress beyond phase II and small scale phase III tri- als.17,30,36 In these trials, alovudine was associated with dose-dependent reversible leukopenia and anemia.19 Although myelosuppression was not an acceptable toxici- ty for HIV, it may be a more manageable side effect for an anti-leukemic therapy. Moreover, our data suggest that alovudine can preferentially target AML cells over normal hematopoeitic cells. Thus, given the known toxicology and pharmacology of alovudine, along with the prior experience of its clinical use, it could potentially be rapidly repositioned for the treatment of AML.
Using a combination of pharmacological and genetic approaches, we discovered that inhibition of POLG induces the monocytic differentiation of AML cells. Thus, we identified new mechanisms by which mito- chondrial pathways control differentiation in AML and highlight alovudine as a novel potential therapeutic agent for AML. Most therapies currently in use or under investigation for AML are cytotoxic and induce cell death. A less explored therapeutic strategy is to promote the differentiation of AML cells into more mature proge- ny. Upon differentiation, the leukemic cells will cease to proliferate or die. Targeting the block in differentiation is the standard therapy for acute promyelocytic leukemia (APL)-AML.39 There is growing evidence that targeting mitochondrial pathways can impact cell fate and differ- entiation of AML beyond the subset of APL.22-24 For example, the cytosolic isocitrate dehydrogenase 1 (IDH1) and its mitochondrial homolog IDH2 encode enzymes that convert isocitrate to a-ketoglutarate (aKG), a co-fac- tor for TET2 that demethylates DNA. Mutations in IDH1 and IDH2 modify the affinity between the encoded
enzymes and their substrates, resulting in conversion of aKG to R-2-hydroxyglutarate (R-2-HG) which inhibits both TET2 and other aKG-dependent enzymes.40 The result is an increase in DNA methylation, altered gene expression, and a block in differentiation. Inhibiting mutant IDH1/2 restores normal IDH function, decreases DNA methylation, and promotes the differentiation of AML cells in vitro and in vivo.21,41
In this study, we report a new mechanism by which mitochondrial pathways control differentiation. We demonstrated that inhibition of POLG and/or reductions in levels of mitochondrial DNA that were not sufficient to impair oxidative phosphorylation led to increased mono- cytoid differentiation. Previous studies have reported that mitochondrial stress such as inhibition of the mitochondr- ial protease ClpP42,43 or oxidative stress44 can result in translocation of mitochondrial proteins to the nucleus and alter gene expression. Potentially similar mechanisms could occur upon inhibition of POLG and lead to translo- cation of proteins from the mitochondria to the nucleus, impacting gene expression to promote differentiation.
Thus, we have identified a novel mechanism by which mitochondria regulate AML fate and differentiation inde- pendently of oxidative phosphorylation. Moreover, we highlight POLG inhibitors such as alovudine as potential therapeutic agents for AML.
Acknowledgments
We thank Jill Flewelling (Princess Margaret Cancer Center) for administrative assistance.
Funding
This work was supported by Medivir AB, the Leukemia and Lymphoma Society, the Canadian Institutes of Health Research, the Princess Margaret Cancer Centre Foundation, and the Ministry of Long Term Health and Planning in the Province of Ontario. ADS holds the Barbara Baker Chair in Leukemia and Related Diseases
References
1. Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136-1152.
2. Lang BF, Gray MW, Burger G. Mitochondrial genome evolution and the origin of eukaryotes. Annu Rev Genet. 1999;33(1):351-397.
Leveraging increased cytoplasmic nucleo- side kinase activity to target mtDNA and oxidative phosphorylation in AML. Blood. 2017;129(19):2657-2666.
7. Gaur R, Grasso D, Datta PP, et al. A single mammalian mitochondrial translation initi- ation factor functionally replaces two bac- terial factors. Mol Cell. 2008;29(2):180-190.
8. Christian BE, Spremulli LL. Mechanism of protein biosynthesis in mammalian mito- chondria. Biochim Biophys Acta. 2012;1819: 1035-1054.
9. Birky CW. Eukaryotic Origins. Science. 1994;266(5183):309-310.
13. Lane AN, Fan TW-M. Regulation of mam- malian nucleotide metabolism and biosyn- thesis. Nucleic Acids Res. 2015;43(4):2466- 2485.
14. Mathews CK. Deoxyribonucleotide metabolism, mutagenesis and cancer. Nat Rev Cancer. 2015;15(9):528-539.
15. Kakuda TN. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther. 2000;22(6):685-708.
16. Gandhi VV, Samuels DC. A review com- paring deoxyribonucleoside triphosphate (dNTP) concentrations in the mitochondri- al and cytoplasmic compartments of nor- mal and transformed cells. Nucleosides Nucleotides Nucleic Acids. 2011;30(5):317-
3. Lagadinou ED, Sach A, Callahan K, et al. BCL-2 inhibition targets oxidative phos- phorylation and selectively eradicates qui- escent human leukemia stem cells. Cell Stem Cell. 2013;12(3):329-341.
4. Skrtić M, Sriskanthadevan S, Jhas B, et al. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell. 2011; 20(5):674-688.
10. Chan SSL, Copeland WC. DNA poly-
merase gamma and mitochondrial disease: Understanding the consequence of POLG
mutations. Biochim Biophys Acta - 339.
5. SriskanthadevanS,JeyarajuDV,ChungTE, et al. AML cells have low spare reserve capacity in their respiratory chain that ren- ders them susceptible to oxidative meta- bolic stress. Blood. 2015;125(13):2120- 2130.
6. Liyanage SU, Hurren R, Voisin V, et al.
Bioenerg. 2009;1787(5):312-319.
11. Bogenhagen DF. Mitochondrial DNA nucleoid structure. Biochim Biophys Acta.
2012;1819(9-10):914-920.
12. Carling PJ, Cree LM, Chinnery PF. The
implications of mitochondrial DNA copy number regulation during embryogenesis. Mitochondrion. 2011;11(5):686-692.
17. Katlama C, Ghosn J, Tubiana R, et al. MIV- 310 reduces HIV viral load in patients fail- ing multiple antiretroviral therapy: results from a 4-week phase II study. AIDS. 2004;18(9):1299-1304.
18. Rusconi S. Alovudine Medivir. Curr Opin Investig Drugs. 2003;4(2):219-223.
19. Flexner C, van der Horst C, Jacobson MA,
haematologica | 2019; 104(5)
971


































































































   123   124   125   126   127