#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Long non-coding RNAs and latent HIV – A search for novel targets for latency reversal


Autoři: Wim Trypsteen aff001;  Cory H. White aff002;  Amey Mukim aff003;  Celsa A. Spina aff003;  Ward De Spiegelaere aff005;  Steve Lefever aff006;  Vicente Planelles aff007;  Alberto Bosque aff008;  Christopher H. Woelk aff002;  Linos Vandekerckhove aff001;  Nadejda Beliakova-Bethell aff003
Působiště autorů: HIV Cure Research Center, Department of Internal Medicine, Ghent University and Ghent University Hospital, Ghent, Belgium aff001;  Faculty of Medicine, University of Southampton, Southampton, Hants, United Kingdom aff002;  San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA, United States of America aff003;  Department of Pathology, University of California San Diego, La Jolla, CA, United States of America aff004;  Department of Morphology, Faculty of Veterinary Sciences, Ghent University, Ghent, Belgium aff005;  Center for Medical Genetics, Ghent University, Ghent, Belgium aff006;  Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States of America aff007;  Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States of America aff008;  Department of Medicine, University of California San Diego, La Jolla, CA, United States of America aff009
Vyšlo v časopise: PLoS ONE 14(11)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0224879

Souhrn

The latent cellular reservoir of HIV is recognized as the major barrier to cure from HIV infection. Long non-coding RNAs (lncRNAs) are more tissue and cell type-specific than protein coding genes, and may represent targets of choice for HIV latency reversal. Using two in vitro primary T-cell models, we identified lncRNAs dysregulated in latency. PVT1 and RP11-347C18.3 were up-regulated in common between the two models, and RP11-539L10.2 was down-regulated. The major component of the latent HIV reservoir, memory CD4+ T-cells, had higher expression of these lncRNAs, compared to naïve T-cells. Guilt-by-association analysis demonstrated that lncRNAs dysregulated in latency were associated with several cellular pathways implicated in HIV latency establishment and maintenance: proteasome, spliceosome, p53 signaling, and mammalian target of rapamycin (MTOR). PVT1, RP11-347C18.3, and RP11-539L10.2 were down-regulated by latency reversing agents, suberoylanilide hydroxamic acid and Romidepsin, suggesting that modulation of lncRNAs is a possible secondary mechanism of action of these compounds. These results will facilitate prioritization of lncRNAs for evaluation as targets for HIV latency reversal. Importantly, our study provides insights into regulatory function of lncRNA during latent HIV infection.

Klíčová slova:

Gene expression – HIV infections – Long non-coding RNAs – Proteasomes – T cells – Viral persistence and latency – Spliceosomes


Zdroje

1. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda DJ, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science. 2009;323(5919):1304–7. doi: 10.1126/science.1165706 19265012

2. Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DI, et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell. 2013;155(3):540–51. doi: 10.1016/j.cell.2013.09.020 24243014

3. Castro-Gonzalez S, Colomer-Lluch M, Serra-Moreno R. Barriers for HIV cure: the latent reservoir. AIDS Res Hum Retroviruses. 2018;34(9):739–59. doi: 10.1089/AID.2018.0118 30056745

4. Rasmussen TA, Lewin SR. Shocking HIV out of hiding: where are we with clinical trials of latency reversing agents? Curr Opin HIV AIDS. 2016;11(4):394–401. doi: 10.1097/COH.0000000000000279 26974532

5. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 2012;22(9):1775–89. doi: 10.1101/gr.132159.111 22955988

6. Zhang Q, Chen C-Y, Yedavalli VS, Jeang K-T. NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio. 2013;4(1):e00596–12. doi: 10.1128/mBio.00596-12 23362321

7. Liu H, Hu P-W, Couturier J, Lewis DE, Rice AP. HIV-1 replication in CD4+ T cells exploits the down-regulation of antiviral NEAT1 long non-coding RNAs following T cell activation. Virology. 2018;522:193–8. https://doi.org/10.1016/j.virol.2018.07.020 30036787

8. Li J, Chen C, Ma X, Geng G, Liu B, Zhang Y, et al. Long noncoding RNA NRON contributes to HIV-1 latency by specifically inducing tat protein degradation. Nat Commun. 2016;7:11730. doi: 10.1038/ncomms11730 27291871

9. Imam H, Shahr Bano A, Patel P, Holla P, Jameel S. The lncRNA NRON modulates HIV-1 replication in a NFAT-dependent manner and is differentially regulated by early and late viral proteins. Sci Rep. 2015;5:8639. doi: 10.1038/srep08639 25728138

10. Cron RQ, Bartz SR, Clausell A, Bort SJ, Klebanoff SJ, Lewis DB. NFAT1 enhances HIV-1 gene expression in primary human CD4 T cells. Clin Immunol. 2000;94(3):179–91. doi: 10.1006/clim.1999.4831 10692237

11. Huan C, Li Z, Ning S, Wang H, Yu X-F, Zhang W. Long noncoding RNA uc002yug.2 activates HIV-1 latency through regulation of mRNA levels of various RUNX1 isoforms and increased Tat expression. J Virol. 2018;92(9). doi: 10.1128/jvi.01844-17 29491162

12. Ma L, Sun L, Jin X, Qu D, Sun W-W, Wang J-H, et al. Long noncoding RNA MALAT1 releases epigenetic silencing of HIV-1 replication by displacing the polycomb repressive complex 2 from binding to the LTR promoter. Nucleic Acids Res. 2019;47(6):3013–3027. doi: 10.1093/nar/gkz117 30788509

13. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629–41. doi: 10.1016/j.cell.2009.02.006 19239885

14. Trypsteen W, Mohammadi P, Van Hecke C, Mestdagh P, Lefever S, Saeys Y, et al. Differential expression of lncRNAs during the HIV replication cycle: an underestimated layer in the HIV-host interplay. Sci Rep. 2016;6:36111. doi: 10.1038/srep36111 27782208

15. Peng X, Sova P, Green RR, Thomas MJ, Korth MJ, Proll S, et al. Deep sequencing of HIV-infected cells: insights into nascent transcription and host-directed therapy. J Virol. 2014;88(16):8768–82. doi: 10.1128/JVI.00768-14 24850744

16. Martins LJ, Bonczkowski P, Spivak AM, De Spiegelaere W, Novis CL, DePaula-Silva AB, et al. Modeling HIV-1 latency in primary T cells using a replication-competent virus. AIDS Res Hum Retroviruses. 2016;32(2):187–93. doi: 10.1089/aid.2015.0106 26171776

17. Bosque A, Planelles V. Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood. 2009;113(1):58–65. doi: 10.1182/blood-2008-07-168393 18849485

18. Spina CA, Anderson J, Archin NM, Bosque A, Chan J, Famiglietti M, et al. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog. 2013;9(12). doi: 10.1371/journal.ppat.1003834 24385908

19. Beliakova-Bethell N, Mukim A, White CH, Deshmukh S, Abewe H, Richman DD, et al. Histone deacetylase inhibitors induce complex host responses that contribute to differential potencies of these compounds in HIV reactivation. J Biol Chem. 2019;294(14):5576–89. doi: 10.1074/jbc.RA118.005185 30745362

20. Bosque A, Famiglietti M, Weyrich AS, Goulston C, Planelles V. Homeostatic proliferation fails to efficiently reactivate HIV-1 latently infected central memory CD4+ T cells. PLoS Pathog. 2011;7(10):e1002288. doi: 10.1371/journal.ppat.1002288 21998586

21. White CH, Moesker B, Beliakova-Bethell N, Martins LJ, Spina CA, Margolis DM, et al. Transcriptomic analysis implicates the p53 signaling pathway in the establishment of HIV-1 latency in central memory CD4 T cells in an In vitro model. PLoS Pathog. 2016;12(11):e1006026. doi: 10.1371/journal.ppat.1006026 27898737

22. Schoggins JW, Rice CM. Interferon-stimulated genes and their antiviral effector functions. Curr Opin Virol. 2011;1(6):519–25. doi: 10.1016/j.coviro.2011.10.008 22328912

23. Monroe KM, Yang Z, Johnson JR, Geng X, Doitsh G, Krogan NJ, et al. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science. 2014;343(6169):428–32. Epub 12/19. doi: 10.1126/science.1243640 24356113

24. Soriano-Sarabia N, Bateson RE, Dahl NP, Crooks AM, Kuruc JD, Margolis DM, et al. Quantitation of replication-competent HIV-1 in populations of resting CD4+ T cells. J Virol. 2014;88(24):14070–7. doi: 10.1128/JVI.01900-14 25253353

25. Chun T-W, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387(6629):183–8. doi: 10.1038/387183a0 9144289

26. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9(6):727–8. doi: 10.1038/nm880 12754504

27. Spina CA, Prince HE, Richman DD. Preferential replication of HIV-1 in the CD45RO memory cell subset of primary CD4 lymphocytes in vitro. J Clin Invest. 1997;99(7):1774–85. doi: 10.1172/JCI119342 9120023

28. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15(8):893–900. doi: 10.1038/nm.1972 19543283

29. Kaczmarek Michaels K, Natarajan M, Euler Z, Alter G, Viglianti G, Henderson AJ. Blimp-1, an intrinsic factor that represses HIV-1 proviral transcription in memory CD4+ T cells. J Immunol. 2015;194(7):3267–74. doi: 10.4049/jimmunol.1402581 25710909

30. Miller LK, Kobayashi Y, Chen C-C, Russnak TA, Ron Y, Dougherty JP. Proteasome inhibitors act as bifunctional antagonists of human immunodeficiency virus type 1 latency and replication. Retrovirology. 2013;10:120. doi: 10.1186/1742-4690-10-120 24156270

31. Lin J, Zhang X, Lu W, Xu X, Pan X, Liang T, et al. PR-957, a selective immunoproteasome inhibitor, reactivates latent HIV-1 through p-TEFb activation mediated by HSF-1. Biochem Pharmacol. 2018. doi: 10.1016/j.bcp.2018.08.042 30170098

32. Besnard E, Hakre S, Kampmann M, Lim HW, Hosmane NN, Martin A, et al. The mTOR complex controls HIV latency. Cell Host Microbe. 2016;20(6):785–97. doi: 10.1016/j.chom.2016.11.001 27978436

33. Lassen KG, Ramyar KX, Bailey JR, Zhou Y, Siliciano RF. Nuclear retention of multiply spliced HIV-1 RNA in resting CD4+ T cells. PLoS Pathog. 2006;2:e68. doi: 10.1371/journal.ppat.0020068 16839202

34. Yukl SA, Kaiser P, Kim P, Telwatte S, Joshi SK, Vu M, et al. HIV latency in isolated patient CD4+ T cells may be due to blocks in HIV transcriptional elongation, completion, and splicing. Sci Transl Med. 2018;10(430). doi: 10.1126/scitranslmed.aap9927 29491188

35. Mahiet C, Swanson CM. Control of HIV-1 gene expression by SR proteins. Biochem Soc Trans. 2016;44(5):1417–25. doi: 10.1042/BST20160113 27911724

36. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Nat Acad Sci U S A. 2009;106(28):11667–72. doi: 10.1073/pnas.0904715106 19571010

37. Zhang R, Ni F, Fu B, Wu Y, Sun R, Tian Z, et al. A long noncoding RNA positively regulates CD56 in human natural killer cells. Oncotarget. 2016;7(45):72546–58. doi: 10.18632/oncotarget.12466 27713137

38. Zhai N, Xia Y, Yin R, Liu J, Gao F. A negative regulation loop of long noncoding RNA HOTAIR and p53 in non-small-cell lung cancer. OncoTargets Ther. 2016;9:5713–20. doi: 10.2147/OTT.S110219 27695348

39. Xu M-d, Wang Y, Weng W, Wei P, Qi P, Zhang Q, et al. A positive feedback loop of lncRNA PVT1 and FOXM1 facilitates gastric cancer growth and invasion. Clin Cancer Res. 2017;23(8):2071–80. doi: 10.1158/1078-0432.CCR-16-0742 27756785

40. Erkelenz S, Hillebrand F, Widera M, Theiss S, Fayyaz A, Degrandi D, et al. Balanced splicing at the Tat-specific HIV-1 3'ss A3 is critical for HIV-1 replication. Retrovirology. 2015;12:29. doi: 10.1186/s12977-015-0154-8 25889056

41. Tranell A, Fenyö EM, Schwartz S. Serine- and arginine-rich proteins 55 and 75 (SRp55 and SRp75) induce production of HIV-1 vpr mRNA by inhibiting the 5'-splice site of exon 3. J Biol Chem. 2010;285(41):31537–47. doi: 10.1074/jbc.M109.077453 20685659

42. Tranell A, Tingsborg S, Fenyö EM, Schwartz S. Inhibition of splicing by serine-arginine rich protein 55 (SRp55) causes the appearance of partially spliced HIV-1 mRNAs in the cytoplasm. Virus Res. 2011;157(1):82–91. https://doi.org/10.1016/j.virusres.2011.02.010 21345357

43. Shkreta L, Blanchette M, Toutant J, Wilhelm E, Bell B, Story BA, et al. Modulation of the splicing regulatory function of SRSF10 by a novel compound that impairs HIV-1 replication. Nucleic Acids Res. 2017;45(7):4051–67. doi: 10.1093/nar/gkw1223 27928057

44. Guo J, Hao C, Wang C, Li L. Long noncoding RNA PVT1 modulates hepatocellular carcinoma cell proliferation and apoptosis by recruiting EZH2. Cancer Cell Int. 2018;18:98. doi: 10.1186/s12935-018-0582-3 30008615

45. Tripathy MK, McManamy MEM, Burch BD, Archin NM, Margolis DM. H3K27 demethylation at the proviral promoter sensitizes latent HIV to the effects of vorinostat in ex-vivo cultures of resting CD4+ T cells. J Virol. 2015;89:8392–405. doi: 10.1128/JVI.00572-15 26041287

46. Zhao Y, Zhao J, Guo X, She J, Liu Y. Long non-coding RNA PVT1, a molecular sponge for miR-149, contributes aberrant metabolic dysfunction and inflammation in IL-1β-simulated osteoarthritic chondrocytes. Biosci Rep. 2018. doi: 10.1042/bsr20180576 30126849

47. Fu C, Li D, Zhang X, Liu N, Chi G, Jin X. LncRNA PVT1 facilitates tumorigenesis and progression of glioma via regulation of miR-128-3p/GREM1 axis and BMP signaling pathway. Neurotherapeutics. 2018. doi: 10.1007/s13311-018-0649-9 30120709

48. He F, Song Z, Chen H, Chen Z, Yang P, Li W, et al. Long noncoding RNA PVT1-214 promotes proliferation and invasion of colorectal cancer by stabilizing Lin28 and interacting with miR-128. Oncogene. 2018. doi: 10.1038/s41388-018-0432-8 30076414

49. Huang F, Chen W, Peng J, Li Y, Zhuang Y, Zhu Z, et al. LncRNA PVT1 triggers Cyto-protective autophagy and promotes pancreatic ductal adenocarcinoma development via the miR-20a-5p/ULK1 Axis. Mol Cancer. 2018;17:98. doi: 10.1186/s12943-018-0845-6 30001707

50. Yang Q, Yu Y, Sun Z, Pan Y. Long non-coding RNA PVT1 promotes cell proliferation and invasion through regulating miR-133a in ovarian cancer. Biomed Pharmacother. 2018;106:61–7. https://doi.org/10.1016/j.biopha.2018.06.112 29957467

51. Chang Z, Cui J, Song Y. Long noncoding RNA PVT1 promotes EMT via mediating microRNA-186 targeting of Twist1 in prostate cancer. Gene. 2018;654:36–42. https://doi.org/10.1016/j.gene.2018.02.036 29452232

52. Henderson LJ, Sharma A, Monaco MCG, Major EO, Al-Harthi L. Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription through its intact core and cysteine-rich domains inhibits Wnt/β-catenin signaling in astrocytes: relevance to HIV neuropathogenesis. J Neurosci. 2012;32(46):16306–13. doi: 10.1523/JNEUROSCI.3145-12.2012 23152614

53. Farivar S, Shaabanpour Aghamaleki F. Effects of major epigenetic factors on systemic lupus erythematosus. Iran Biomed J. 2018;22(5):294–302. doi: 10.29252/ibj.22.5.294 29803202

54. Rai R, Chauhan SK, Singh VV, Rai M, Rai G. RNA-seq analysis reveals unique transcriptome signatures in systemic lupus erythematosus patients with distinct autoantibody specificities. PLoS ONE. 2016;11(11):e0166312–e. doi: 10.1371/journal.pone.0166312 27835693

55. Beliakova-Bethell N, Zhang J, Singhania A, Lee V, Terry V, Richman DD, et al. Suberoylanilide hydroxamic acid induces limited changes in the transcriptome of primary CD4+ T cells. AIDS. 2013;27:29–37. doi: 10.1097/QAD.0b013e32835b3e26 23221426

56. Wozniak MB, Villuendas R, Bischoff JR, Aparicio CB, Martínez Leal JF, de La Cueva P, et al. Vorinostat interferes with the signaling transduction pathway of T-cell receptor and synergizes with phosphoinositide-3 kinase inhibitors in cutaneous T-cell lymphoma. Haematologica. 2010;95(4):613–21. doi: 10.3324/haematol.2009.013870 20133897

57. LaBonte M, Wilson P, Fazzone W, Groshen S, Lenz H-J, Ladner R. DNA microarray profiling of genes differentially regulated by the histone deacetylase inhibitors vorinostat and LBH589 in colon cancer cell lines. BMC Med Genomics. 2009;2:67. doi: 10.1186/1755-8794-2-67 19948057

58. White CH, Johnston HE, Moesker B, Manousopoulou A, Margolis DM, Richman DD, et al. Mixed effects of suberoylanilide hydroxamic acid (SAHA) on the host transcriptome and proteome and their implications for HIV reactivation from latency. Antiviral Res. 2015;123:78–85. doi: 10.1016/j.antiviral.2015.09.002 26343910

59. Macedo AB, Resop RS, Martins LJ, Szaniawski MA, Sorensen ES, Spivak AM, et al. Influence of biological sex, age, and HIV status in an in vitro primary cell model of HIV latency using a CXCR4 tropic virus. AIDS Res Hum Retroviruses. 2018;34(9):769–77. doi: 10.1089/AID.2018.0098 29926732

60. Noyce RS, Taylor K, Ciechonska M, Collins SE, Duncan R, Mossman KL. Membrane perturbation elicits an IRF3-dependent, interferon-independent antiviral response. J Virol. 2011;85(20):10926–31. doi: 10.1128/JVI.00862-11 21813605

61. Noyce RS, Collins SE, Mossman KL. Identification of a novel pathway essential for the immediate-early, interferon-independent antiviral response to enveloped virions. J Virol. 2006;80(1):226–35. doi: 10.1128/JVI.80.1.226-235.2006 16352547

62. Rossio JL, Esser MT, Suryanarayana K, Schneider DK, Bess JW Jr., Vasquez GM, et al. Inactivation of human immunodeficiency virus type 1 infectivity with preservation of conformational and functional integrity of virion surface proteins. J Virol. 1998;72:7992–8001. 9733838

63. Park J, Lim CH, Ham S, Kim SS, Choi B-S, Roh T-Y. Genome-wide analysis of histone modifications in latently HIV-1 infected T cells. AIDS. 2014;28(12):1719–28. doi: 10.1097/QAD.0000000000000309 24762674

64. Abdel-Mohsen M, Wang C, Strain MC, Lada SM, Deng X, Cockerham LR, et al. Select host restriction factors are associated with HIV persistence during antiretroviral therapy. AIDS. 2015;29(4):411–20. doi: 10.1097/QAD.0000000000000572 25602681

65. Chen H, Li C, Huang J, Cung T, Seiss K, Beamon J, et al. CD4(+) T cells from elite controllers resist HIV-1 infection by selective upregulation of p21. J Clin Invest. 2011;121(4):1549–60. doi: 10.1172/JCI44539 21403397

66. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25(9):1105–11. doi: 10.1093/bioinformatics/btp120 19289445

67. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9. doi: 10.1093/bioinformatics/btu638 25260700

68. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25–R. doi: 10.1186/gb-2009-10-3-r25 19261174

69. Robinson MD, McCarthy DJ, Smyth GK. EdgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. doi: 10.1093/bioinformatics/btp616 19910308

70. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat Biotechnol. 2014;32(9):896–902. doi: 10.1038/nbt.2931 25150836

71. R: A language and environment for statistical computing. Vienna, Austria: R Foundation of Statistical Computing; R Core Team, 2017.

72. Lawrence M, Huber W, Pagès H, Aboyoun P, Carlson M, Gentleman R, et al. Software for computing and annotating genomic ranges. PLoS Comput Biol. 2013;9(8):e1003118. doi: 10.1371/journal.pcbi.1003118 23950696

73. Beliakova-Bethell N, Massanella M, White C, Lada SM, Du P, Vaida F, et al. The effect of cell subset isolation method on gene expression in leukocytes. Cytometry Part A. 2014;85A(1):94–104. doi: 10.1002/cyto.a.22352 24115734

74. Lefever S, Anckaert J, Volders P-J, Luypaert M, Vandesompele J, Mestdagh P. decodeRNA—predicting non-coding RNA functions using guilt-by-association. Database (Oxford). 2017;2017:bax042. doi: 10.1093/database/bax042 29220434

75. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50. doi: 10.1073/pnas.0506580102 16199517

76. Nishimura D. BioCarta. Biotech Softw Internet Rep. 2001;2(3):117–20. doi: 10.1089/152791601750294344

77. Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000;28(1):27–30. doi: 10.1093/nar/28.1.27 10592173

78. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. doi: 10.1101/gr.1239303 14597658

79. Therneau T, Hart S, Kocher J-P. Calculating sample size estimates for RNA Seq studies. R package version 1.6.0.

80. Pinheiro J, Bates D, DebRoy S, Sarkar D, Team RC. Nlme: linear and nonlinear mixed effects models. R package version 3.1–127. 2016.


Článek vyšel v časopise

PLOS One


2019 Číslo 11
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#