Page 94 - 2020_11-Haematologica-web
P. 94

S. Yang et al.
motes lipolysis in BM adipocytes, contributing to their remodeling into small adipocytes in the pathogenesis of AML.
Moreover, we also found that treatment with the TRPV4 agonist 4aPDD rescued BM adipocyte remodel- ing, which was correlated with increased survival in AML-bearing mice, supporting a crucial role of TRPV4 in the growth and progression of AML. Although there are unstained positive white circles in the BM of AML mice (Figure 5A), we suspect that these circles may represent an increase in blood vessels or sinuses in the BM of these animals. Leukemia cells can promote angiogenesis, which in turn contributes to the proliferation of leukemia cells.48 Of course, it cannot be excluded that a few adipocytes were not stained positively. Many studies have also reported the effect of circulating factors in obese animals on leukemia.49,50 However, in our study, both experimen- tal groups and control groups were obese mice, and their basic background was the same. This should, therefore, have allowed us to control for the impact of the circulat-
ing factors, which sould not have been responsible for observed differences in mouse survival and other vari- ables. Future studies in vivo are needed to validate the spe- cific modulatory role of GDF15 on TRPV4.
In conclusion, leukemia cells activate a transcriptional network that includes GDF15-related-PI3K/AKT activa- tion and subsequent TRPV4 downregulation, which pro- motes BM adipocyte remodeling. The morphological adaptation of BM adipocytes and the modulation of lipo- lysis may represent a novel strategy for the treatment of hematologic malignancies, especially in elderly patients, whose aging and increased adiposity of the BM microen- vironment reduce the efficacy of cytotoxic chemotherapy.
Acknowledgments
The work was supported by grants from the National Natural Science Foundation of China (grant n. 81870132), Science and Technology Commission of Shanghai Municipality (grant n.18DZ2293500) and Shanghai Sailing Program (grant n. 18YF1419100).
References
1. Asada N. Regulation of malignant hematopoiesis by bone marrow microenvi- ronment. Front Oncol. 2018;8:119.
2. Schepers K, Campbell TB, Passegue E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell. 2015;16(3):254-267.
3. Tabe Y, Yamamoto S, Saitoh K, et al. Bone marrow adipocytes facilitate fatty acid oxi- dation activating AMPK and a transcription- al network supporting survival of acute monocytic leukemia cells. Cancer Res. 2017;77(6):1453-1464.
4. Lee MKS, Al-Sharea A, Dragoljevic D, Murphy AJ. Hand of FATe: lipid metabolism in hematopoietic stem cells. Curr Opin Lipidol. 2018;29(3):240-245.
5. Lu W, Weng W, Zhu Q, et al. Small bone marrow adipocytes predict poor prognosis in acute myeloid leukemia. Haematologica. 2018;103(1):e21-e24.
6. Shafat MS, Oellerich T, Mohr S, et al. Leukemic blasts program bone marrow adipocytes to generate a protumoral microenvironment. Blood. 2017;129(10): 1320-1332.
7. Choe SS, Huh JY, Hwang IJ, Kim JI, Kim JB. Adipose tissue remodeling: its role in energy metabolism and metabolic disorders. Front Endocrinol (Lausanne). 2016;7:30.
8. Bochet L, Lehuede C, Dauvillier S, et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Cancer Res. 2013;73(18):5657-5668.
9. Herroon MK, Rajagurubandara E, Hardaway AL, et al. Bone marrow adipocytes promote tumor growth in bone
via FABP4-dependent mechanisms. Oncotarget. 2013;4 (11):2108-2123.
10. Dirat B, Bochet L, Dabek M, et al. Cancer- associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011;71(7):2455-2465.
11. Fraczak E, Olbromski M, Piotrowska A, et al. Bone marrow adipocytes in haematolog- ical malignancies. Acta Histochem. 2018;120(1):22-27.
12. Lu W, Wan Y, Li ZQ, et al. Growth differen- tiation factor 15 contributes to marrow adipocyte remodeling in response to the growth of leukemic cells. J Exp Clin Cancer Res. 2018;37(1):66.
13. Lu JM, Wang CY, Hu CL, Fang YJ, Mei YA. GDF-15 enhances intracellular Ca2+ by increasing Cav1.3 expression in rat cerebel- lar granule neurons. Biochem J. 2016;473 (13):1895-1904.
olism in macrophages blunts lipolysis during
ageing. Nature. 2017;550(7674):119-123.
23. Li JY, Gong L, Liu SZ, et al. Adipose HuR protects against diet-induced obesity and insulin resistance. Nat Commun. 2019;10(1):
2375.
24. Wang F, Ren XF, Chen Z, et al. The N-termi-
nal His-tag affects the triglyceride lipase activity of hormone-sensitive lipase in testis. J Cell Biochem. 2019;120(8):13706-13716.
25. Eguchi J, Wang X, Yu ST, et al. Transcriptional control of adipose lipid han- dling by IRF4. Cell Metab. 2011,13(3):249-
14. Zemel M, Thompson W, Milstead A, Morris
K, Campbell P. Calcium and dairy accelera-
tion of weight and fat loss during energy
restriction in obese adults. Obes Res. 259.
2004;12(4):582-590.
15. Shi H, Dirienzo D, Zemel MB. Effects of
dietary calcium on adipocyte lipid metabo- lism and body weight regulation in energy restricted aP2-agouti transgenic mice. FASEB J. 2001;15(2):291-293.
16. Hashimoto R, Katoh Y, Miyamoto Y, et al. High extracellular Ca2+ enhances the adipocyte accumulation of bone marrow stromal cells through a decrease in cAMP. Cell Calcium. 2017;67:74-80.
17. Xue B, Greenberg AG, Kraemer FB, Zemel MB. Mechanism of intracellular calcium ([Ca2+]i) inhibition of lipolysis in human adipocytes. FASEB J. 2001;15(13):2527-2529.
18. Kim JH,Mynatt RL, Moore JW, Woychik RP, Moustaid N, Zemel MB. The effects of calci- um channel blockade on agouti-induced obesity. FASEB J. 1996;10(14):1646-1652.
19. He YH, Zhang HQ, Teng JH, Huang LN, Li Y, Sun CH. Involvement of calcium-sensing receptor in inhibition of lipolysis through intracellular cAMP and calcium pathways in human adipocytes. Biochem Biophys Res Commun. 2011;404(1):393-399.
20. Alexander R, Kerby A, Aubdool AA, et al. 4alpha-phorbol 12,13-didecanoate activates cultured mouse dorsal root ganglia neurons independently of TRPV4. Br J Pharmacol. 2013;168(3):761-772.
26. Liu DD, Lu JM, Zhao QR, Hu CL, Mei YA. Growth differentiation factor-15 promotes glutamate release in medial prefrontal cortex of mice through upregulation of T-type cal- cium channels. Sci Rep. 2016;6:28653.
27. Xu MY, Pang QQ, Xu SQ, et al. Hypoxia- inducible factor-1a activates transforming growth factor-β1/Smad signaling and increases collagen deposition in dermal fibroblasts. Oncotarget. 2017;9(3):3188- 3197.
28. Bansal V, De D, An J, et al. Chemical induced conversion of mouse fibroblasts and human adipose-derived stem cells into skeletal muscle-like cells. Biomaterials. 2019;193:30-46.
29. Herberman RB, Holden HT, Ting CC, Lavrin DL, Kirchner H. Cell-mediated immunity to leukemia virus- and tumor-associated anti- gens in mice. Cancer Res. 1976;36:615-621.
30. Toft-Bertelsen TL, Krizaj D, MacAulay N. When size matters: transient receptor poten- tial vanilloid 4 channel as a volume-sensor rather than an osmo-sensor. J Physiol. 2017;595(11):3287-3302.
31. Rübenhagen R, Rönsch H, Jung H, Krämer R, Morbach S. Osmosensor and osmoregu- lator properties of the betaine carrier BetP from Corynebacterium glutamicum in pro- teoliposomes. J Biol Chem. 2000;275(2):735-
21. Sergeev IN, Li S, Ho CT, Rawson NE, 741.
Dushenkov S. Polymethoxyflavones acti- vate Ca2+-dependent apoptotic targets in adipocytes. J Agric Food Chem. 2009;57(13): 5771-5776.
22. Camell CD, Sander J, Spadaro O, et al. Inflammasome-driven catecholamine catab-
32. Heide T, Stuart MC, Poolman B. On the osmotic signal and osmosensing mechanism of an ABC transport system for glycine betaine. EMBO J. 2001;20(24):7022-7032.
33. Ramsey IS, Delling M, Clapham DE. An introduction to TRP channels. Annu Rev
2582
haematologica | 2020; 105(11)


































































































   92   93   94   95   96