Page 54 - 2020_01-Haematologica-web
P. 54

Y-H. Ho and S. Méndez-Ferrer et al.
shows higher affinity for noradrenaline over adrenaline, the opposite is true for β2-AR.117 Another possibility might be that BM concentrations of both neurotransmitters dif- fer in aging, leading to imbalanced stimulation of the two receptors. The observation that old HSC home in the BM away from endosteal regions suggests that HSC-driven niche remodeling mainly occurs in non-endosteal domains. For instance, skewed myelopoiesis leading to increased numbers of neutrophils and defective phagocy- tosis of marrow macrophages might modulate the microenvironment favoring myeloid bias during aging.81 Increased myeloid cells might provide an additional source of catecholamines in a feed-forward loop promot- ing megakaryocyte differentiation by activating β2-AR. We propose that accumulation of old HSC results in microenvironmental remodeling by reinforcing adrenergic activity, expansion of non-endosteal niches and enhanced myeloid/megakaryocyte differentiation. As a secondary outcome, alteration of megakaryocyte niches (increased megakaryocyte numbers, proplatelet formation and appo- sition to sinusoids) might release HSC from their state of quiescence, further promoting HSC proliferation with age.
Niche alterations might predispose to hematologic neoplasms
The risk of developing myeloid malignancies increases significantly in individuals harboring clonal hematopoiesis-related somatic mutations. In fact some of these mutations are oncogenic drivers of myeloid malig- nancies.118 However, the factors limiting clonal expansion or, instead, allowing the mutant clones to become domi- nant and, in some cases, cause disease, remain unclear. Interestingly, similar niche alterations are shared between aging and myeloid disorders associated with age, which is
an important selection pressure to expand aberrant HSC clones. For instance, a damaged neuro-MSC circuit pro- motes the development of a cytokine storm created by the mutant HSC, aggravating the progression of myeloprolif- erative neoplasms.75 In myelodysplastic syndromes, abnormal production of cytokines from the microenviron- ment, dysfunction of MSC and osteolineage cells, and vas- cular remodeling have been associated with disease initia- tion and progression.119,120 Targeting the abnormal microen- vironment could be a promising complementary thera- peutic approach to treat hematologic cancers in the future, especially when early diagnosis becomes available.
Conclusive remarks
Aging of the hematopoietic system might result from both HSC-intrinsic and microenvironmental alterations with changes in the location, function and regulation of HSC and their progeny. Future studies will determine the relative contribution of the aged microenviroment to altered hematopoiesis and increased incidence of age- related disorders originating in the BM.
Acknowledgments
Y-HH received fellowships from Alborada Scholarship (University of Cambridge), Trinity-Henry Barlow Scholarship (University of Cambridge) and R.O.C. Government Scholarship to Study Abroad (GSSA). This work was supported by core sup- port grants from the Wellcome Trust and the MRC to the Cambridge Stem Cell Institute, National Health Service Blood and Transplant (United Kingdom), European Union’s Horizon 2020 research (ERC-2014-CoG-64765) and a Programme Foundation Award from Cancer Research UK to SM-F The authors regret that some relevant literature could not be discussed because of space constrictions.
References
1. Carrelha J, Meng Y, Kettyle LM, et al. Hierarchically related lineage-restricted fates of multipotent haematopoietic stem cells. Nature. 2018;554(7690):106-111.
2. Weksberg DC, Chambers SM, Boles NC, Goodell MA. CD150- side population cells represent a functionally distinct population of long-term hematopoietic stem cells. Blood. 2008;111(4):2444-2451.
3. Kiel MJ, Yilmaz OH, Morrison SJ. CD150- cells are transiently reconstituting multipo- tent progenitors with little or no stem cell activity. Blood. 2008;111(8):4413-4414; author reply 4414-4415.
4. Kent DG, Copley MR, Benz C, et al. Prospective isolation and molecular charac- terization of hematopoietic stem cells with durable self-renewal potential. Blood. 2009;113(25):6342-6350.
5. Montecino-Rodriguez E, Kong Y, Casero D, et al. Lymphoid-biased hematopoietic stem cells are maintained with age and efficiently generate lymphoid progeny. Stem Cell Reports. 2019;12(3):584-596.
6. Liang Y, Van Zant G, Szilvassy SJ. Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood. 2005;106(4):1479-1487.
7. Mohrin M, Shin J, Liu Y, et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoi- etic stem cell aging. Science. 2015;347(6228): 1374-1377.
8. Rossi DJ, Bryder D, Zahn JM, et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci U S A. 2005;102(26):9194-9199.
9. Sudo K, Ema H, Morita Y, Nakauchi H. Age- associated characteristics of murine hematopoietic stem cells. J Exp Med. 2000;192(9):1273-1280.
10. Dykstra B, Olthof S, Schreuder J, Ritsema M, de Haan G. Clonal analysis reveals mul- tiple functional defects of aged murine hematopoietic stem cells. J Exp Med. 2011;208(13):2691-2703.
11. Chambers SM, Shaw CA, Gatza C, et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregula- tion. PLoS Biol. 2007;5(8):e201.
12. Flach J, Bakker ST, Mohrin M, et al. Replication stress is a potent driver of func- tional decline in ageing haematopoietic stem cells. Nature. 2014;512(7513):198-202.
13. Rossi DJ, Bryder D, Seita J, et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature. 2007;447(7145):725-729.
14. Florian MC, Nattamai KJ, Dorr K, et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature. 2013;503(7476):392-396.
15. Ito K, Suda T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat Rev Mol Cell Biol. 2014;15(4):243-256.
16. Chandel NS, Jasper H, Ho TT, Passegue E. Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing. Nat Cell Biol. 2016;18(8):823-832.
17. Ho TT, Warr MR, Adelman ER, et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature. 2017;543(7644):205-210.
18. Vilchez D, Simic MS, Dillin A. Proteostasis and aging of stem cells. Trends Cell Biol. 2014;24(3):161-170.
19. Ergen AV, Boles NC, Goodell MA. Rantes/Ccl5 influences hematopoietic stem cell subtypes and causes myeloid skewing. Blood. 2012;119(11):2500-2509.
20. Taichman RS, Emerson SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimu- lating factor. J Exp Med. 1994;179(5):1677- 1682.
21. Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and con- trol of the niche size. Nature. 2003;425
44
haematologica | 2020; 105(1)


































































































   52   53   54   55   56