Page 70 - Haematologica March 2020
P. 70

C. Zhang et al. 342(6159):705-706.
10. Sankaran VG, Orkin SH. Genome-wide association studies of hematologic pheno- types: a window into human hematopoiesis. Curr Opin Genet Dev. 2013;23(3):339-344.
11. De Gobbi M, Viprakasit V, Hughes JR, et al. A regulatory SNP causes a human genetic disease by creating a new transcriptional promoter. Science. 2006;312(5777):1215- 1217.
12. McVicker G, van de Geijn B, Degner JF, et al. Identification of genetic variants that affect histone modifications in human cells. Science. 2013;342(6159):747-749.
13. Kilpinen H, Waszak SM, Gschwind AR, et al. Coordinated effects of sequence varia- tion on DNA binding, chromatin structure, and transcription. Science. 2013; 342(6159):744-747.
14. Kasowski M, Kyriazopoulou- Panagiotopoulou S, Grubert F, et al. Extensive variation in chromatin states across humans. Science. 2013; 342(6159):750-752.
15. Heinz S, Romanoski CE, Benner C, et al. Effect of natural genetic variation on enhancer selection and function. Nature. 2013;503(7477):487-492.
16. Van Zant G, Liang Y. Natural genetic diver- sity as a means to uncover stem cell regula- tory pathways. Ann N Y Acad Sci. 2009; 1176:170-177.
17. Liang Y, Jansen M, Aronow B, et al. The quantitative trait gene latexin influences the size of the hematopoietic stem cell pop- ulation in mice. Nat Genet. 2007;39(2):178- 188.
18. de Haan G. Latexin is a newly discovered regulator of hematopoietic stem cells. Nat Genet. 2007;39(2):141-142.
19. Scadden DT. Nice Neighborhood: Emerging Concepts of the Stem Cell Niche. Cell. 2014;157(1):41-50.
20. Muthusamy V, Premi S, Soper C, et al. The hematopoietic stem cell regulatory gene latexin has tumor-suppressive properties in malignant melanoma. J Invest Dermatol. 2013;133(7):1827-1833.
21. Abd Elmageed ZY, Moroz K, Kandil E. Clinical significance of CD146 and latexin during different stages of thyroid cancer. Mol Cell Biochem. 2013;381(1-2):95-103.
22. Mitsunaga K, Kikuchi J, Wada T, el al. Latexin regulates the abundance of multi- ple cellular proteins in hematopoietic stem cells. J Cell Physiol. 2012;227(3):1138-1147.
23. Liu Y, Howard D, Rector K, et al. Latexin is
down-regulated in hematopoietic malig- nancies and restoration of expression inhibits lymphoma growth. PLoS One. 2012;7(9):e44979.
24. Li Y, Basang Z, Ding H, et al. Latexin expression is downregulated in human gas- tric carcinomas and exhibits tumor sup- pressor potential. BMC Cancer. 2011; 11:121.
25. Jin C, Zang C, Wei G, et al. H3.3/H2A.Z double variant-containing nucleosomes mark 'nucleosome-free regions' of active promoters and other regulatory regions. Nat Genet. 2009;41(8):941-945.
26. Creyghton MP, Markoulaki S, Levine SS, et al. H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment. Cell. 2008; 135(4):649-661.
27. Andang M, Hjerling-Leffler J, Moliner A, et al. Histone H2AX-dependent GABA(A) receptor regulation of stem cell prolifera- tion. Nature. 2008;451(7177):460-464.
28. Lee MG, Villa R, Trojer P, et al. Demethylation of H3K27 regulates poly- comb recruitment and H2A ubiquitination. Science. 2007;318(5849):447-450.
29. Gevry N, Chan HM, Laflamme L, et al. p21 transcription is regulated by differential localization of histone H2A.Z. Genes Dev. 2007;21(15):1869-1881.
30. Pusterla T, de Marchis F, Palumbo R, et al. High mobility group B2 is secreted by myeloid cells and has mitogenic and chemoattractant activities similar to high mobility group B1. Autoimmunity. 2009; 42(4):308-310.
31. Pfannkuche K, Summer H, Li O, et al. The high mobility group protein HMGA2: a co- regulator of chromatin structure and pluripotency in stem cells? Stem Cell Rev. 2009;5(3):224-230.
32. Laurent B, Randrianarison-Huetz V, Marechal V, et al. High-mobility group pro- tein HMGB2 regulates human erythroid differentiation through trans-activation of GFI1B transcription. Blood. 2010; 115(3):687-695.
33. Nishino J, Kim I, Chada K, et al. Hmga2 promotes neural stem cell self-renewal in young but not old mice by reducing p16Ink4a and p19Arf Expression. Cell. 2008;135(2):227-239.
34. Yanai H, Ban T, Wang Z, et al. HMGB pro- teins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature. 2009;462(7269):99-103.
35. Ye ZJ, Kluger Y, Lian Z, et al. Two types of
precursor cells in a multipotential hematopoietic cell line. Proc Natl Acad Sci USA. 2005;102(51):18461-18466.
36. Lemieux ME, Cheng Z, Zhou Q, et al. Inactivation of a single copy of Crebbp selectively alters pre-mRNA processing in mouse hematopoietic stem cells. PloS One. 2011;6(8):e24153.
37. Kutlesa S, Zayas J, Valle A, et al. T-cell dif- ferentiation of multipotent hematopoietic cell line EML in the OP9-DL1 coculture sys- tem. Exp Hematol. 2009;37(8):909-923.
38. Liu Y, Zhang C, Li Z, et al. Latexin Inactivation Enhances Survival and Long- Term Engraftment of Hematopoietic Stem Cells and Expands the Entire Hematopoietic System in Mice. Stem Cell Rep. 2017;8(4):991-1004.
39. You Y, Wen R, Pathak R, et al. Latexin sen- sitizes leukemogenic cells to gamma-irradi- ation-induced cell-cycle arrest and cell death through Rps3 pathway. Cell Death Dis. 2014;5:e1493.
40. Consortium GT, Laboratory DA. Coordinating Center -Analysis Working G, Statistical Methods groups-Analysis Working G, Enhancing Gg, Fund NIHC, et al. Genetic effects on gene expression across human tissues. Nature. 2017; 550(7675):204-213.
41. Abraham AB, Bronstein R, Reddy AS, et al. Aberrant neural stem cell proliferation and increased adult neurogenesis in mice lack- ing chromatin protein HMGB2. PLoS One. 2013;8(12):e84838.
42. Taniguchi N, Carames B, Ronfani L, et al. Aging-related loss of the chromatin protein HMGB2 in articular cartilage is linked to reduced cellularity and osteoarthritis. Proc Natl Acad Sci U S A. 2009;106(4):1181- 1186.
43. Zhou X, Li M, Huang H, et al. HMGB2 reg- ulates satellite-cell-mediated skeletal mus- cle regeneration through IGF2BP2. J Cell Sci. 2016;129(22):4305-4316.
44. Aird KM, Iwasaki O, Kossenkov AV, et al. HMGB2 orchestrates the chromatin land- scape of senescence-associated secretory phenotype gene loci. J Cell Biol. 2016; 215(3):325-334.
45. Nacarelli T, Liu P, Zhang R. Epigenetic Basis of Cellular Senescence and Its Implications in Aging. Genes (Basel). 2017;24;8(12).
46. Kloth M, Goering W, Ribarska T, et al. The SNP rs6441224 influences transcriptional activity and prognostically relevant hyper- methylation of RARRES1 in prostate can- cer. Int J Cancer. 2012;131(6):E897-904.
584
haematologica | 2020; 105(3)


































































































   68   69   70   71   72