Vol 24, No 5 (2017)
Original articles — Basic science and experimental cardiology
Published online: 2017-04-27

open access

Page views 5821
Article views/downloads 3544
Get Citation

Connect on Social Media

Connect on Social Media

Heart failure: Pilot transcriptomic analysis of cardiac tissue by RNA-sequencing

Concetta Schiano1, Valerio Costa, Marianna Aprile, Vincenzo Grimaldi, Ciro Maiello, Roberta Esposito, Andrea Soricelli, Vittorio Colantuoni, Francesco Donatelli, Alfredo Ciccodicola, Claudio Napoli
Pubmed: 28497843
Cardiol J 2017;24(5):539-553.

Abstract

Background: Despite left ventricular (LV) dysfunction contributing to mortality in chronic heart failure (HF), the molecular mechanisms of LV failure continues to remain poorly understood and myocardial biomarkers have yet to be identified. The aim of this pilot study was to investigate specific transcriptome changes occurring in cardiac tissues of patients with HF compared to healthy condition patients to improve diagnosis and possible treatment of affected subjects.

Methods: Unlike other studies, only dilated cardiomyopathy (DCM) (n = 2) and restrictive cardiomyopathy (RCM) (n = 2) patients who did not report family history of the disease were selected with the aim of obtaining a homogeneous population for the study. The transcriptome of all patients were studied by RNA-sequencing (RNA-Seq) and the read counts were adequately filtered and normalized using a recently developed user-friendly tool for RNA-Seq data analysis, based on a new graphical user interface (RNA-SeqGUI).

Results: By using this approach in a pairwise comparison with healthy donors, we were able to identify DCM- and RCM-specific expression signatures for protein-coding genes as well as for long noncoding RNAs (lncRNAs). Differential expression of 5 genes encoding different members of the mediator complex was disclosed in this analysis. Interestingly, a significant alteration was found for genes which had never been associated with HF until now, and 27 lncRNA/mRNA pairs that were significantly altered in HF patients.

Conclusions: The present findings revealed specific expression pattern of both protein-coding and lncRNAs in HF patients, confirming that new LV myocardial biomarkers could be reliably identified using Next-Generation Sequencing-based approaches.

Article available in PDF format

View PDF Download PDF file

References

  1. Modesto K, Sengupta PP. Myocardial mechanics in cardiomyopathies. Prog Cardiovasc Dis. 2014; 57(1): 111–124.
  2. McMurray J, Adamopoulos S, Anker SD, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. European Heart Journal. 2012; 33(14): 1787–1847.
  3. Roger VL. Epidemiology of heart failure. Circ Res. 2013; 113(6): 646–659.
  4. Ziaeian B, Fonarow G. Epidemiology and aetiology of heart failure. Nat Rev Cardiol. 2016; 13(6): 368–378.
  5. Matkovich SJ, Zhang Y, Van Booven DJ, et al. Deep mRNA sequencing for in vivo functional analysis of cardiac transcriptional regulators: application to Galphaq. Circ Res. 2010; 106(9): 1459–1467.
  6. Napoli C, Lerman LO, Sica V, et al. Microarray analysis: a novel research tool for cardiovascular scientists and physicians. Heart. 2003; 89(6): 597–604.
  7. Gora M, Kiliszek M, Burzynska B. Will global transcriptome analysis allow the detection of novel prognostic markers in coronary artery disease and heart failure? Curr Genomics. 2013; 14(6): 388–396.
  8. Nagalakshmi U, Wang Z, Waern K, et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science. 2008; 320(5881): 1344–1349.
  9. Costa V, Aprile M, Esposito R, et al. RNA-Seq and human complex diseases: recent accomplishments and future perspectives. Eur J Hum Genet. 2013; 21(2): 134–142.
  10. Herrer I, Roselló-Lletí E, Rivera M, et al. RNA-sequencing analysis reveals new alterations in cardiomyocyte cytoskeletal genes in patients with heart failure. Lab Invest. 2014; 94(6): 645–653.
  11. Hershberger RE, Siegfried JD. Update 2011: clinical and genetic issues in familial dilated cardiomyopathy. J Am Coll Cardiol. 2011; 57(16): 1641–1649.
  12. Towbin J. Inherited Cardiomyopathies. Circulation Journal. 2014; 78(10): 2347–2356.
  13. Ounzain S, Micheletti R, Beckmann T, et al. Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs. Eur Heart J. 2015; 36(6): 353–68a.
  14. di Salvo TG, Yang KC, Brittain E, et al. Right ventricular myocardial biomarkers in human heart failure. J Card Fail. 2015; 21(5): 398–411.
  15. He M, Yang Z, Abdellatif M, et al. GTPase Activating Protein (Sh3 Domain) Binding Protein 1 Regulates the Processing of MicroRNA-1 during Cardiac Hypertrophy. PLoS One. 2015; 10(12): e0145112.
  16. Chen S, Puthanveetil P, Feng B, et al. Cardiac miR-133a overexpression prevents early cardiac fibrosis in diabetes. J Cell Mol Med. 2014; 18(3): 415–421.
  17. Ganesan J, Ramanujam D, Sassi Y, et al. MiR-378 controls cardiac hypertrophy by combined repression of mitogen-activated protein kinase pathway factors. Circulation. 2013; 127(21): 2097–2106.
  18. Heymans S, Corsten MF, Verhesen W, et al. Macrophage microRNA-155 promotes cardiac hypertrophy and failure. Circulation. 2013; 128(13): 1420–1432.
  19. Ucar A, Gupta SK, Fiedler J, et al. The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy. Nat Commun. 2012; 3: 1078.
  20. da Costa Martins PA, Salic K, Gladka MM, et al. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling. Nat Cell Biol. 2010; 12(12): 1220–1227.
  21. Han P, Li W, Lin CH, et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature. 2014; 514(7520): 102–106.
  22. Kumarswamy R, Bauters C, Volkmann I, et al. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ Res. 2014; 114(10): 1569–1575.
  23. Vausort M, Wagner DR, Devaux Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res. 2014; 115(7): 668–677.
  24. Molina-Navarro MM, Roselló-Lletí E, Tarazón E, et al. Heart failure entails significant changes in human nucleocytoplasmic transport gene expression. Int J Cardiol. 2013; 168(3): 2837–2843.
  25. Rienzo M, Costa V, Scarpato M, et al. RNA-Seq for the identification of novel Mediator transcripts in endothelial progenitor cells. Gene. 2014; 547(1): 98–105.
  26. Kim D, Pertea G, Trapnell C, et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013; 14(4): R36.
  27. Russo F, Angelini C. RNASeqGUI: a GUI for analysing RNA-Seq data. Bioinformatics. 2014; 30(17): 2514–2516.
  28. Soneson C, Delorenzi M. A comparison of methods for differential expression analysis of RNA-seq data. BMC Bioinformatics. 2013; 14: 91.
  29. Kanehisa M. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000; 28(1): 27–30.
  30. Huang DaW, Sherman BT, Tan Q, et al. The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 2007; 8(9): R183.
  31. Mi H, Muruganujan A, Thomas PD. PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res. 2013; 41(Database issue): D377–D386.
  32. Karolchik D, Hinrichs AS, Furey TS, et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res. 2004; 32(Database issue): D493–D496.
  33. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010; 26(6): 841–842.
  34. Rienzo M, Schiano C, Casamassimi A, et al. Identification of valid reference housekeeping genes for gene expression analysis in tumor neovascularization studies. Clin Transl Oncol. 2013; 15(3): 211–218.
  35. Costa V, Angelini C, De Feis I, et al. Uncovering the complexity of transcriptomes with RNA-Seq. J Biomed Biotechnol. 2010; 2010: 853916.
  36. Kelwick R, Desanlis I, Wheeler GN, et al. The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol. 2015; 16: 113.
  37. Kostin S, Hein S, Arnon E, et al. The cytoskeleton and related proteins in the human failing heart. Heart Fail Rev. 2000; 5(3): 271–280.
  38. Zile MR, Green GR, Schuyler GT, et al. Cardiocyte cytoskeleton in patients with left ventricular pressure overload hypertrophy. J Am Coll Cardiol. 2001; 37(4): 1080–1084.
  39. Guo DC, Pannu H, Tran-Fadulu V, et al. Mutations in smooth muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat Genet. 2007; 39(12): 1488–1493.
  40. Chen RX, Liu F, Li Y, et al. Neuromedin S increases L-type Ca(2+) channel currents through G(i)α-protein and phospholipase C-dependent novel protein kinase C delta pathway in adult rat ventricular myocytes. Cell Physiol Biochem. 2012; 30(3): 618–630.
  41. Schiano C, Casamassimi A, Vietri MT, et al. The roles of mediator complex in cardiovascular diseases. Biochim Biophys Acta. 2014; 1839(6): 444–451.
  42. Muncke N, Jung C, Rüdiger H, et al. Missense mutations and gene interruption in PROSIT240, a novel TRAP240-like gene, in patients with congenital heart defect (transposition of the great arteries). Circulation. 2003; 108(23): 2843–2850.
  43. Grøntved L, Madsen MS, Boergesen M, et al. MED14 tethers mediator to the N-terminal domain of peroxisome proliferator-activated receptor gamma and is required for full transcriptional activity and adipogenesis. Mol Cell Biol. 2010; 30(9): 2155–2169.
  44. Wang W, Huang Lu, Huang Y, et al. Mediator MED23 links insulin signaling to the adipogenesis transcription cascade. Dev Cell. 2009; 16(5): 764–771.
  45. Kikuchi Y, Umemura H, Nishitani S, et al. Human mediator MED17 subunit plays essential roles in gene regulation by associating with the transcription and DNA repair machineries. Genes Cells. 2015; 20(3): 191–202.
  46. Femminella GD, Ferrara N, Rengo G. The emerging role of microRNAs in Alzheimer's disease. Front Physiol. 2015; 6: 40.
  47. Clarke BD, Roby JA, Slonchak A, et al. Functional non-coding RNAs derived from the flavivirus 3' untranslated region. Virus Res. 2015; 206: 53–61.
  48. 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.
  49. Juan L, Wang G, Radovich M, et al. Potential roles of microRNAs in regulating long intergenic noncoding RNAs. BMC Med Genomics. 2013; 6 Suppl 1: S7.
  50. Skroblin P, Mayr M. Circ Res. 2014; 115(7): 607–609.
  51. Philippen LE, Dirkx E, da Costa-Martins PA, et al. Non-coding RNA in control of gene regulatory programs in cardiac development and disease. J Mol Cell Cardiol. 2015; 89(Pt A): 51–58.
  52. Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell. 2008; 133(1): 38–52.
  53. Babon JJ, Lucet IS, Murphy JM, et al. The molecular regulation of Janus kinase (JAK) activation. Biochem J. 2014; 462(1): 1–13.