Page 59 - 2019_05-HaematologicaMondo-web
P. 59

Single cell lncRNAs in hematopoiesis
caution. Nevertheless, our results were in agreement with reported microarray data from 183 MDS patients, which related abnormal lncRNAs with gene expression, cancer, and malignancy.52 Also, differentially expressed lncRNAs in monosomy 7 cells were involved in similar pathways as their mRNA counterparts in our previous study.31
Our results are not a complete profile of lncRNAs due to several limitations, especially the use of only polyA- enriched RNAs,8 and the limited cell numbers from a few individuals due to the high cost of scRNA-seq. Additionally, annotation of novel lncRNAs is context dependent. We adopted commonly used pipelines,12-14,16-18 but annotation might vary using different algorithms. Nevertheless, our work creates a model for future profil- ing of the repertoire of lncRNAs in other cell types. Lineage signatures of lncRNAs are comparison-based, and thus may vary when such comparisons are made among different subsets. Others have categorized HSCs versus cells of specific lineages and among differentiated cells or distinct subsets.12-18 In contrast, we defined lncRNA signa- tures by making comparisons among subsets within a rel- atively homogeneous HSPC population, which may com- promise our power to detect differences. Furthermore, pseudotime ordering reconstructs the hematopoietic hier- archy based on bioinformatic analysis of transcriptome similarity, and it has demonstrated high agreement with purified cell compartments;44 however, dynamic gene expression in hematopoiesis might be preferably assessed in purified cell populations obtained after physical sorting based on membrane proteins, including after induction of
differentiation or other in vitro perturbations. Given the high cell-type specificity of lncRNAs, signature lncRNAs may be superior to mRNAs in discriminating and differen- tiating cell subsets or new cell types that cannot be easily distinguished based on cell surface markers. We did not compare the efficacy of lncRNAs and mRNAs in defining cell types due to a lack of detailed surface marker informa- tion for single cells. Future studies with larger cell num- bers, complete surface marker characterization, and whole transcriptome expression data should be of great interest in defining new cells/subtypes.
Rapid evolution and low species conservation are fea- tures of lncRNAs,10,11 making a human catalog a prerequi- site to successful, clinically relevant lncRNA studies. Based on next-generation sequencing and single cell tech- nology, we provide a global database that should be foun- dational for future studies of lncRNA biology in human HSPCs.
Acknowledgments
The authors acknowledge the support of the Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA). We thank patients and healthy volunteers who donated bone marrow. Sequencing and technical support were provided by the DNA Sequencing and Genomics Core of NHLBI. FACS sorting was performed by Keyvan Keyvanfar and the Flow Cytometry Core of NHLBI. This research was supported by an Intramural Research Program of the National Heart, Lung, and Blood Institute.
References
1. Alvarez-Dominguez JR, Lodish HF. Emerging mechanisms of long noncoding RNA function during normal and malignant hematopoiesis. Blood. 2017;130(18):1965- 1975.
2. Satpathy AT, Chang HY. Long noncoding RNA in hematopoiesis and immunity. Immunity. 2015;42(5):792-804.
3. Engreitz JM, Ollikainen N, Guttman M. Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression. Nat Rev Mol Cell Biol. 2016;17(12):756-770.
4. Derrien T, Johnson R, Bussotti G, et al. The GENCODE v7 catalog of human long non- coding RNAs: analysis of their gene struc- ture, evolution, and expression. Genome Res. 2012;22(9):1775-1789.
5. Cabili MN, Dunagin MC, McClanahan PD, et al. Localization and abundance analysis of human lncRNAs at single-cell and single- molecule resolution. Genome Biol. 2015; 16:20.
6. Djebali S, Davis CA, Merkel A, et al. Landscape of transcription in human cells. Nature. 2012;489(7414):101-108.
7. Ulitsky I, Bartel DP. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154 (1):26-46.
8. Cabili MN, Trapnell C, Goff L, et al. Integrative annotation of human large inter- genic noncoding RNAs reveals global prop- erties and specific subclasses. Genes Dev. 2011;25(18):1915-1927.
9. Washietl S, Kellis M, Garber M.
Evolutionary dynamics and tissue specificity of human long noncoding RNAs in six mammals. Genome Res. 2014;24(4):616- 628.
10. Pang KC, Frith MC, Mattick JS. Rapid evolu- tion of noncoding RNAs: lack of conserva- tion does not mean lack of function. Trends Genet. 2006;22(1):1-5.
11. Wang J, Zhang J, Zheng H, et al. Mouse tran- scriptome: neutral evolution of 'non-coding' complementary DNAs. Nature. 2004;431 (7010):1 p following 757; discussion follow- ing 757.
12. Luo M, Jeong M, Sun D, et al. Long non-cod- ing RNAs control hematopoietic stem cell function. Cell Stem Cell. 2015;16(4):426- 438.
13. Alvarez-Dominguez JR, Hu W, Yuan B, et al. Global discovery of erythroid long noncod- ing RNAs reveals novel regulators of red cell maturation. Blood. 2014;123(4):570-581.
14. Paralkar VR, Mishra T, Luan J, et al. Lineage and species-specific long noncoding RNAs during erythro-megakaryocytic develop- ment. Blood. 2014;123(12):1927-1937.
Nat Immunol. 2015;16(3):318-325.
18. Brazão TF, Johnson JS, Müller J, et al. Long noncoding RNAs in B-cell development and
activation. Blood. 2016;128(7):e10-19.
19. Collier SP, Collins PL, Williams CL, et al. Cutting edge: influence of Tmevpg1, a long intergenic noncoding RNA, on the expres- sion of Ifng by Th1 cells. J Immunol.
2012;189(5):2084-2088.
20. Vigneau S, Rohrlich PS, Brahic M, et al.
Tmevpg1, a candidate gene for the control of Theiler's virus persistence, could be impli- cated in the regulation of gamma interferon. J Virol. 2003;77(10):5632-5638.
21. Iyer MK, Niknafs YS, Malik R, et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 2015;47 (3):199-208.
22. Lei L, Xia S, Liu D, Li X, et al. Genome-wide characterization of lncRNAs in acute myeloid leukemia. Brief Bioinform. 2018. 19(4):627-635.
23. Heward JA, Lindsay MA. Long non-coding RNAs in the regulation of the immune response. Trends Immunol. 2014;35(9):408-
15. Schwarzer A, Emmrich S, Schmidt F, et al. 419.
The non-coding RNA landscape of human hematopoiesis and leukemia. Nat Commun. 2017;8(1):218.
16. Hu G, Tang Q, Sharma S, et al. Expression and regulation of intergenic long noncoding RNAs during T cell development and differ- entiation. Nat Immunol. 2013;14(11):1190- 1198.
17. Ranzani V, Rossetti G, Panzeri I, et al. The long intergenic noncoding RNA landscape of human lymphocytes highlights the regula- tion of T cell differentiation by linc-MAF-4.
24. Ulitsky I, Bartel DP. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154 (1):26-46.
25. Cabili MN, Trapnell C, Goff L, et al. Integrative annotation of human large inter- genic noncoding RNAs reveals global prop- erties and specific subclasses. Genes Dev. 2011;25(18):1915-1927.
26. Wang J, Roy B. Single-cell RNA-seq reveals lincRNA expression differences in Hela-S3 cells. Biotechnol Lett. 2017;39(3):359-366.
27. Kim DH, Marinov GK, Pepke S, et al. Single-
haematologica | 2019; 104(5)
905


































































































   57   58   59   60   61