Page 127 - 2020_08-Haematologica-web
P. 127

Myostatin propeptide expands primitive CML cells
stimulation with TGF-b1, we found SMAD2/3 phospho- rylation as well as reduced growth and survival of pri- mary CD34+ CML cells. This suggests that MSTNpp- induced SMAD2/3 activation is an unlikely cause of the growth-promoting effects. Instead, the increased STAT5 phosphorylation is a probable mechanism by which MSTNpp expands primitive CML cells. Although STAT5 phosphorylation was high in the unstimulated control cells (consistent with STAT5 being a well-known media- tor and downstream target of the BCR-ABL1 fusion in CML)2,42 MSTNpp stimulation further activated STAT5. However, cross-talk between STAT5, TGF-b family sig- naling members and other signaling pathways might also contribute to the growth-promoting effects of MSTNpp in primitive CML cells.
In conclusion, we here identify several novel positive regulators of primitive CML cells using a high-content cytokine screen. We show that the myostatin antagonist MSTNpp binds to the surface of CML cells, induces acti- vation of STAT5 and SMAD2/3, and has a previously unrecognized growth-promoting effect on primitive CML cells and corresponding normal cells. Further studies are needed to investigate whether interfering with MSTNpp
would translate into new therapeutic opportunities in CML.
Acknowledgments
We wish to thank Anna Hammarberg and the Multipark Cellomics Platform, Lund University, Lund, for help with the screening. We also thank Drs. Henrik Hjort-Hansen and Kourosh Lofti and the Nordic CML Study Group for providing clinical samples and patient characteristics.
Funding
This work was supported by the Swedish Cancer Society, the Swedish Children’s Cancer Foundation, the Medical Faculty of Lund University, the Swedish Research Council, the ISREC Foundation by a joint grant to Swiss Cancer Center, Lusanne, CREATE Health Cancer Center, from the Biltema foundation, the Medical Faculty of Lund University and the Knut and Alice Wallenberg Foundation. Grant support was also received from a Terry Fox Foundation New Frontiers Program Project (#1074), a Stem Cell Network of Centres of Excellence grant (#F17/DT2), and grants from the Canadian Cancer Society Research Institute (#704257 and #705047) and the Leukemia & Lymphoma Society of Canada (#417871).
References
1. Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer. 2005;5(3):172- 183.
2. Holyoake TL, Vetrie D. The chronic myeloid leukemia stem cell: stemming the tide of persistence. Blood. 2017;129(12): 1595-1606.
3. Schepers K, Campbell TB, Passegue E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell. 2015;16(3):254-267.
4. Nievergall E, Reynolds J, Kok CH, et al. TGF-alpha and IL-6 plasma levels selectively identify CML patients who fail to achieve an early molecular response or progress in the first year of therapy. Leukemia. 2016;30(6):1263-1272.
5. Anand M, Chodda SK, Parikh PM, Nadkarni JS. Abnormal levels of proinflammatory cytokines in serum and monocyte cultures from patients with chronic myeloid leukemia in different stages, and their role in prognosis. Hematol Oncol. 1998;16(4):143- 154.
6. Wetzler M, Kurzrock R, Estrov Z, et al. Altered levels of interleukin-1 beta and inter- leukin-1 receptor antagonist in chronic myelogenous leukemia: clinical and prog- nostic correlates. Blood. 1994;84(9):3142- 3147.
7. Pollyea DA, Jordan CT. Therapeutic target- ing of acute myeloid leukemia stem cells. Blood. 2017;129(12):1627-1635.
8. Agerstam H, Hansen N, von Palffy S, et al. IL1RAP antibodies block IL-1-induced expansion of candidate CML stem cells and mediate cell killing in xenograft models. Blood. 2016;128(23):2683-2693.
9. Zhang B, Chu S, Agarwal P, et al. Inhibition of interleukin-1 signaling enhances elimination of tyrosine kinase inhibitor-treated CML stem cells. Blood. 2016;128(23):2671-2682.
Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in xenograft mod- els of acute myeloid leukemia. Proc Natl Acad Sci U S A. 2015;112(34):10786-10791.
IL1RAP expression as a measure of leukemic stem cell burden at diagnosis of chronic myeloid leukemia predicts therapy out- come. Leukemia. 2016;30(1):253-257.
20. Koschmieder S, Gottgens B, Zhang P, et al. Inducible chronic phase of myeloid leukemia with expansion of hematopoietic stem cells in a transgenic model of BCR-ABL leukemogenesis. Blood. 2005;105(1):324-
10. Agerstam H, Karlsson C, Hansen N, et al.
12. Jiang X, Fujisaki T, Nicolini F, et al. Autonomous multi-lineage differentiation in vitro of primitive CD34+ cells from patients with chronic myeloid leukemia. Leukemia. 2000;14(6):1112-1121.
13. Jiang X, Lopez A, Holyoake T, Eaves A, Eaves C. Autocrine production and action of IL-3 and granulocyte colony-stimulating fac- tor in chronic myeloid leukemia. Proc Natl Acad Sci U S A. 1999;96(22):12804-12809.
14. Emanuel PD, Bates LJ, Castleberry RP, Gualtieri RJ, Zuckerman KS. Selective hypersensitivity to granulocyte-macrophage colony-stimulating factor by juvenile chron- ic myeloid leukemia hematopoietic progen- itors. Blood. 1991;77(5):925-929.
15. Welner Robert S, Amabile G, Bararia D, et al. Treatment of Chronic Myelogenous Leukemia by Blocking Cytokine Alterations Found in Normal Stem and Progenitor Cells. Cancer Cell. 2015;27(5):671-681.
16. Reynaud D, Pietras E, Barry-Holson K, et al. IL-6 controls leukemic multipotent progeni- tor cell fate and contributes to chronic myel- ogenous leukemia development. Cancer Cell. 2011;20(5):661-673.
17. Schurch C, Riether C, Amrein MA, Ochsenbein AF. Cytotoxic T cells induce proliferation of chronic myeloid leukemia stem cells by secreting interferon-gamma. J Exp Med. 2013;210(3):605-621.
18. Jaras M, Johnels P, Hansen N, et al. Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein. Proc Natl Acad Sci U S A. 2010;107(37):16280-16285.
19. Landberg N, Hansen N, Askmyr M, et al.
21. Kishi K. A new leukemia cell line with Philadelphia chromosome characterized as basophil precursors. Leuk Res. 1985;9(3): 381-390.
22. Han HQ, Zhou X, Mitch WE, Goldberg AL. Myostatin/activin pathway antagonism: molecular basis and therapeutic potential. Int J Biochem Cell Biol. 2013;45(10):2333- 2347.
23. von Laer D, Corovic A, Vogt B, et al. Loss of CD38 antigen on CD34+CD38+ cells during short-term culture. Leukemia. 2000;14(5): 947-948.
24. Boitano AE, Wang J, Romeo R, et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329(5997):1345- 1348.
25. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387(6628):83-90.
26. Schuelke M, Wagner KR, Stolz LE, et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med. 2004;350(26):2682-2688.
27. Furihata T, Kinugawa S, Fukushima A, et al. Serum myostatin levels are independently associated with skeletal muscle wasting in patients with heart failure. Int J Cardiol. 2016;220:483-487.
28. Han DS, Huang CH, Chen SY, Yang WS. Serum reference value of two potential dop- ing candidates-myostatin and insulin-like growth factor-I in the healthy young male. J Int Soc Sports Nutr. 2017;14:2.
29. Blank U, Karlsson S. The role of Smad sig-
11. Pietras EM, Mirantes-Barbeito C, Fong S, et
al. Chronic interleukin-1 exposure drives haematopoietic stem cells towards preco-
cious myeloid differentiation at the expense
of self-renewal. Nat Cell Biol. 2016;18(6):607-618. 334.
haematologica | 2020; 105(8)
2103


































































































   125   126   127   128   129