Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 149 No. 0102 (2019)

The hypoxic kidney: pathogenesis and noncoding RNA-based therapeutic strategies

  • George Haddad
  • Malte Kölling
  • Johan M. Lorenzen
DOI
https://doi.org/10.4414/smw.2019.14703
Cite this as:
Swiss Med Wkly. 2019;149:w14703
Published
13.01.2019

Summary

Acute kidney injury (AKI) is a disease entity of major importance, affecting approximately 6% of all patients on the intensive care unit. The mortality rate exceeds 60%. AKI is related to several underlying conditions, including sepsis, nephrotoxicity or major surgery. Ischaemia reperfusion injury or hypoxic conditions may lead to severe injury of the kidney and is associated with a steep decline in survival rates of patients. At present, AKI is diagnosed on the basis of creatinine levels and urine output. Novel markers and knowledge of their pathophysiological role is of major importance for targeted therapeutic interventions. Noncoding RNAs (ncRNAs) have recently been introduced and are the subject of intensive research initiatives. They are arbitrarily separated into small ncRNAs (≤200 nucleotides) and long ncRNAs (lncRNAs, ≥200 nucleotides). Whereas small ncRNAs such as microRNAs have been extensively studied over the past several years, investigations into the role of linear lncRNAs and circular RNAs (circRNAs) are largely lacking. The present review article therefore aims to elucidate in detail the role of microRNAs, lncRNAs and circRNAs in animal models as well as patients with ischaemic AKI and to describe their use as biomarkers as well as their potential use as therapeutics.

References

  1. Barrantes F, Tian J, Vazquez R, Amoateng-Adjepong Y, Manthous CA. Acute kidney injury criteria predict outcomes of critically ill patients. Crit Care Med. 2008;36(5):1397–403. doi:.https://doi.org/10.1097/CCM.0b013e318168fbe0
  2. Mehta RL, Kellum JA, Shah SV, Molitoris BA, Ronco C, Warnock DG, et al.; Acute Kidney Injury Network. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care. 2007;11(2):R31. doi:.https://doi.org/10.1186/cc5713
  3. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, et al., Beginning and Ending Supportive Therapy for the Kidney (BEST Kidney) Investigators. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA. 2005;294(7):813–8. doi:.https://doi.org/10.1001/jama.294.7.813
  4. Kelly KJ. Acute renal failure: much more than a kidney disease. Semin Nephrol. 2006;26(2):105–13. doi:.https://doi.org/10.1016/j.semnephrol.2005.09.003
  5. Bon D, Chatauret N, Giraud S, Thuillier R, Favreau F, Hauet T. New strategies to optimize kidney recovery and preservation in transplantation. Nat Rev Nephrol. 2012;8(6):339–47. doi:.https://doi.org/10.1038/nrneph.2012.83
  6. Weight SC, Bell PR, Nicholson ML. Renal ischaemia--reperfusion injury. Br J Surg. 1996;83(2):162–70. doi:.https://doi.org/10.1002/bjs.1800830206
  7. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. doi:. https://doi.org/10.1038/nature11247
  8. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012;489(7414):101–8. doi:.https://doi.org/10.1038/nature11233
  9. Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 2011;121(11):4210–21. doi:.https://doi.org/10.1172/JCI45161
  10. Lorenzen JM, Batkai S, Thum T. Regulation of cardiac and renal ischemia-reperfusion injury by microRNAs. Free Radic Biol Med. 2013;64:78–84. doi:.https://doi.org/10.1016/j.freeradbiomed.2013.06.044
  11. Lorenzen JM, Haller H, Thum T. MicroRNAs as mediators and therapeutic targets in chronic kidney disease. Nat Rev Nephrol. 2011;7(5):286–94. doi:.https://doi.org/10.1038/nrneph.2011.26
  12. Ambros V. A hierarchy of regulatory genes controls a larva-to-adult developmental switch in C. elegans. Cell. 1989;57(1):49–57. doi:.https://doi.org/10.1016/0092-8674(89)90171-2
  13. Chalfie M, Horvitz HR, Sulston JE. Mutations that lead to reiterations in the cell lineages of C. elegans. Cell. 1981;24(1):59–69. doi:.https://doi.org/10.1016/0092-8674(81)90501-8
  14. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP. Vertebrate microRNA genes. Science. 2003;299(5612):1540. doi:.https://doi.org/10.1126/science.1080372
  15. Lorenzen JM, Thum T. Circulating and urinary microRNAs in kidney disease. Clin J Am Soc Nephrol. 2012;7(9):1528–33. doi:.https://doi.org/10.2215/CJN.01170212
  16. Wei Q, Bhatt K, He HZ, Mi QS, Haase VH, Dong Z. Targeted deletion of Dicer from proximal tubules protects against renal ischemia-reperfusion injury. J Am Soc Nephrol. 2010;21(5):756–61. doi:.https://doi.org/10.1681/ASN.2009070718
  17. Zhang L, Xu Y, Xue S, Wang X, Dai H, Qian J, et al. Implications of dynamic changes in miR-192 expression in ischemic acute kidney injury. Int Urol Nephrol. 2017;49(3):541–50. doi:.https://doi.org/10.1007/s11255-016-1485-7
  18. Lorenzen JM, Kaucsar T, Schauerte C, Schmitt R, Rong S, Hübner A, et al. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J Am Soc Nephrol. 2014;25(12):2717–29. doi:.https://doi.org/10.1681/ASN.2013121329
  19. Godwin JG, Ge X, Stephan K, Jurisch A, Tullius SG, Iacomini J. Identification of a microRNA signature of renal ischemia reperfusion injury. Proc Natl Acad Sci USA. 2010;107(32):14339–44. doi:.https://doi.org/10.1073/pnas.0912701107
  20. Shapiro MD, Bagley J, Latz J, Godwin JG, Ge X, Tullius SG, et al. MicroRNA expression data reveals a signature of kidney damage following ischemia reperfusion injury. PLoS One. 2011;6(8):e23011. doi:.https://doi.org/10.1371/journal.pone.0023011
  21. Aguado-Fraile E, Ramos E, Sáenz-Morales D, Conde E, Blanco-Sánchez I, Stamatakis K, et al. miR-127 protects proximal tubule cells against ischemia/reperfusion: identification of kinesin family member 3B as miR-127 target. PLoS One. 2012;7(9):e44305. doi:.https://doi.org/10.1371/journal.pone.0044305
  22. Cantaluppi V, Gatti S, Medica D, Figliolini F, Bruno S, Deregibus MC, et al. Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney Int. 2012;82(4):412–27. doi:.https://doi.org/10.1038/ki.2012.105
  23. Xu X, Kriegel AJ, Liu Y, Usa K, Mladinov D, Liu H, et al. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int. 2012;82(11):1167–75. doi:.https://doi.org/10.1038/ki.2012.241
  24. Wilflingseder J, Sunzenauer J, Toronyi E, Heinzel A, Kainz A, Mayer B, et al. Molecular pathogenesis of post-transplant acute kidney injury: assessment of whole-genome mRNA and miRNA profiles. PLoS One. 2014;9(8):e104164. doi:.https://doi.org/10.1371/journal.pone.0104164
  25. Wilflingseder J, Jelencsics K, Bergmeister H, Sunzenauer J, Regele H, Eskandary F, et al. miR-182-5p Inhibition Ameliorates Ischemic Acute Kidney Injury. Am J Pathol. 2017;187(1):70–9. doi:.https://doi.org/10.1016/j.ajpath.2016.09.011
  26. Bijkerk R, van Solingen C, de Boer HC, van der Pol P, Khairoun M, de Bruin RG, et al. Hematopoietic microRNA-126 protects against renal ischemia/reperfusion injury by promoting vascular integrity. J Am Soc Nephrol. 2014;25(8):1710–22. doi:.https://doi.org/10.1681/ASN.2013060640
  27. Bellinger MA, Bean JS, Rader MA, Heinz-Taheny KM, Nunes JS, Haas JV, et al. Concordant changes of plasma and kidney microRNA in the early stages of acute kidney injury: time course in a mouse model of bilateral renal ischemia-reperfusion. PLoS One. 2014;9(4):e93297. doi:.https://doi.org/10.1371/journal.pone.0093297
  28. Wang JF, Zha YF, Li HW, Wang F, Bian Q, Lai XL, et al. Screening plasma miRNAs as biomarkers for renal ischemia-reperfusion injury in rats. Med Sci Monit. 2014;20:283–9. doi:.https://doi.org/10.12659/MSM.889937
  29. Yu L, Gu T, Shi E, Wang Y, Fang Q, Wang C. Dysregulation of renal microRNA expression after deep hypothermic circulatory arrest in rats. Eur J Cardiothorac Surg. 2016;49(6):1725–31. doi:.https://doi.org/10.1093/ejcts/ezv460
  30. Saikumar J, Hoffmann D, Kim TM, Gonzalez VR, Zhang Q, Goering PL, et al. Expression, circulation, and excretion profile of microRNA-21, -155, and -18a following acute kidney injury. Toxicol Sci. 2012;129(2):256–67. doi:.https://doi.org/10.1093/toxsci/kfs210
  31. Kito N, Endo K, Ikesue M, Weng H, Iwai N. miRNA Profiles of Tubular Cells: Diagnosis of Kidney Injury. BioMed Res Int. 2015;2015:465479. doi:.https://doi.org/10.1155/2015/465479
  32. Lorenzen JM, Kielstein JT, Hafer C, Gupta SK, Kümpers P, Faulhaber-Walter R, et al. Circulating miR-210 predicts survival in critically ill patients with acute kidney injury. Clin J Am Soc Nephrol. 2011;6(7):1540–6. doi:.https://doi.org/10.2215/CJN.00430111
  33. Lorenzen JM, Volkmann I, Fiedler J, Schmidt M, Scheffner I, Haller H, et al. Urinary miR-210 as a mediator of acute T-cell mediated rejection in renal allograft recipients. Am J Transplant. 2011;11(10):2221–7. doi:.https://doi.org/10.1111/j.1600-6143.2011.03679.x
  34. Lorenzen JM, Thum T. Long noncoding RNAs in kidney and cardiovascular diseases. Nat Rev Nephrol. 2016;12(6):360–73. doi:.https://doi.org/10.1038/nrneph.2016.51
  35. Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43(6):904–14. doi:.https://doi.org/10.1016/j.molcel.2011.08.018
  36. Lin J, Zhang X, Xue C, Zhang H, Shashaty MG, Gosai SJ, et al. The long noncoding RNA landscape in hypoxic and inflammatory renal epithelial injury. Am J Physiol Renal Physiol. 2015;309(11):F901–13. doi:.https://doi.org/10.1152/ajprenal.00290.2015
  37. Yu TM, Palanisamy K, Sun KT, Day YJ, Shu KH, Wang IK, et al. RANTES mediates kidney ischemia reperfusion injury through a possible role of HIF-1α and LncRNA PRINS. Sci Rep. 2016;6(1):18424. doi:.https://doi.org/10.1038/srep18424
  38. Arvaniti E, Moulos P, Vakrakou A, Chatziantoniou C, Chadjichristos C, Kavvadas P, et al. Whole-transcriptome analysis of UUO mouse model of renal fibrosis reveals new molecular players in kidney diseases. Sci Rep. 2016;6(1):26235. doi:.https://doi.org/10.1038/srep26235
  39. Zhou Q, Huang XR, Yu J, Yu X, Lan HY. Long Noncoding RNA Arid2-IR Is a Novel Therapeutic Target for Renal Inflammation. Mol Ther. 2015;23(6):1034–43. doi:.https://doi.org/10.1038/mt.2015.31
  40. Kölling M, Genschel C, Kaucsar T, Hübner A, Rong S, Schmitt R, et al. Hypoxia-induced long non-coding RNA Malat1 is dispensable for renal ischemia/reperfusion-injury. Sci Rep. 2018;8(1):3438. doi:.https://doi.org/10.1038/s41598-018-21720-3
  41. Qiu J, Chen Y, Huang G, Zhang Z, Chen L, Na N. Transforming growth factor-β activated long non-coding RNA ATB plays an important role in acute rejection of renal allografts and may impacts the postoperative pharmaceutical immunosuppression therapy. Nephrology (Carlton). 2017;22(10):796–803. doi:.https://doi.org/10.1111/nep.12851
  42. Lorenzen JM, Schauerte C, Kielstein JT, Hübner A, Martino F, Fiedler J, et al. Circulating long noncoding RNATapSaki is a predictor of mortality in critically ill patients with acute kidney injury. Clin Chem. 2015;61(1):191–201. doi:.https://doi.org/10.1373/clinchem.2014.230359
  43. Lorenzen JM, Schauerte C, Kölling M, Hübner A, Knapp M, Haller H, et al. Long Noncoding RNAs in Urine Are Detectable and May Enable Early Detection of Acute T Cell-Mediated Rejection of Renal Allografts. Clin Chem. 2015;61(12):1505–14. doi:.https://doi.org/10.1373/clinchem.2015.243600
  44. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nat Biotechnol. 2014;32(5):453–61. doi:.https://doi.org/10.1038/nbt.2890
  45. Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, et al. circRNA biogenesis competes with pre-mRNA splicing. Mol Cell. 2014;56(1):55–66. doi:.https://doi.org/10.1016/j.molcel.2014.08.019
  46. Starke S, Jost I, Rossbach O, Schneider T, Schreiner S, Hung LH, et al. Exon circularization requires canonical splice signals. Cell Rep. 2015;10(1):103–11. doi:.https://doi.org/10.1016/j.celrep.2014.12.002
  47. Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8. doi:.https://doi.org/10.1038/nature11928
  48. Li XF, Lytton J. A circularized sodium-calcium exchanger exon 2 transcript. J Biol Chem. 1999;274(12):8153–60. doi:.https://doi.org/10.1074/jbc.274.12.8153
  49. Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, et al. The RNA binding protein quaking regulates formation of circRNAs. Cell. 2015;160(6):1125–34. doi:.https://doi.org/10.1016/j.cell.2015.02.014
  50. Khan MA, Reckman YJ, Aufiero S, van den Hoogenhof MM, van der Made I, Beqqali A, et al. RBM20 Regulates Circular RNA Production From the Titin Gene. Circ Res. 2016;119(9):996–1003. doi:.https://doi.org/10.1161/CIRCRESAHA.116.309568
  51. Ivanov A, Memczak S, Wyler E, Torti F, Porath HT, Orejuela MR, et al. Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals. Cell Rep. 2015;10(2):170–7. doi:.https://doi.org/10.1016/j.celrep.2014.12.019
  52. Salzman J, Gawad C, Wang PL, Lacayo N, Brown PO. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS One. 2012;7(2):e30733. doi:.https://doi.org/10.1371/journal.pone.0030733
  53. Alhasan AA, Izuogu OG, Al-Balool HH, Steyn JS, Evans A, Colzani M, et al. Circular RNA enrichment in platelets is a signature of transcriptome degradation. Blood. 2016;127(9):e1–11. doi:.https://doi.org/10.1182/blood-2015-06-649434
  54. Preußer C, Hung LH, Schneider T, Schreiner S, Hardt M, Moebus A, et al. Selective release of circRNAs in platelet-derived extracellular vesicles. J Extracell Vesicles. 2018;7(1):1424473. doi:.https://doi.org/10.1080/20013078.2018.1424473
  55. Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, et al. Circular RNA is enriched and stable in exosomes: a promising biomarker for cancer diagnosis. Cell Res. 2015;25(8):981–4. doi:.https://doi.org/10.1038/cr.2015.82
  56. Memczak S, Papavasileiou P, Peters O, Rajewsky N. Identification and Characterization of Circular RNAs As a New Class of Putative Biomarkers in Human Blood. PLoS One. 2015;10(10):e0141214. doi:.https://doi.org/10.1371/journal.pone.0141214
  57. Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z, Sharpless NE. Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet. 2010;6(12):e1001233. doi:.https://doi.org/10.1371/journal.pgen.1001233
  58. Lukiw WJ. Circular RNA (circRNA) in Alzheimer’s disease (AD). Front Genet. 2013;4:307. doi:.https://doi.org/10.3389/fgene.2013.00307
  59. Li P, Chen S, Chen H, Mo X, Li T, Shao Y, et al. Using circular RNA as a novel type of biomarker in the screening of gastric cancer. Clin Chim Acta. 2015;444:132–6. doi:.https://doi.org/10.1016/j.cca.2015.02.018
  60. Chao CW, Chan DC, Kuo A, Leder P. The mouse formin (Fmn) gene: abundant circular RNA transcripts and gene-targeted deletion analysis. Mol Med. 1998;4(9):614–28. doi:.https://doi.org/10.1007/BF03401761
  61. Xu T, Wu J, Han P, Zhao Z, Song X. Circular RNA expression profiles and features in human tissues: a study using RNA-seq data. BMC Genomics. 2017;18(S6, Suppl 6):680. doi:.https://doi.org/10.1186/s12864-017-4029-3
  62. Zhou J, Chen H, Fan Y. Systematic analysis of the expression profile of non-coding RNAs involved in ischemia/reperfusion-induced acute kidney injury in mice using RNA sequencing. Oncotarget. 2017;8(59):100196–215. doi:.https://doi.org/10.18632/oncotarget.22130
  63. Kölling M, Seeger H, Haddad G, Kistler A, Nowak A, Faulhaber-Walter R, et al. The circular RNA ciRs-126 predicts survival in critically ill patients with acute kidney injury. Kidney Int Rep. 2018;3(5):1144–52. doi:.https://doi.org/10.1016/j.ekir.2018.05.012
  64. Kölling M, Kaucsar T, Schauerte C, Hübner A, Dettling A, Park JK, et al. Therapeutic miR-21 silencing ameliorates diabetic kidney disease in mice. Mol Ther. 2017;25(1):165–80. doi:.https://doi.org/10.1016/j.ymthe.2016.08.001
  65. Schauerte C, Hübner A, Rong S, Wang S, Shushakova N, Mengel M, et al. Antagonism of profibrotic microRNA-21 improves outcome of murine chronic renal allograft dysfunction. Kidney Int. 2017;92(3):646–56. doi:.https://doi.org/10.1016/j.kint.2017.02.012
  66. Lorenzen JM, Kaucsar T, Schauerte C, Schmitt R, Rong S, Hübner A, et al. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J Am Soc Nephrol. 2014;25(12):2717–29. doi:.https://doi.org/10.1681/ASN.2013121329