#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Coxsackievirus-Induced miR-21 Disrupts Cardiomyocyte Interactions via the Downregulation of Intercalated Disk Components


Coxsackievirus B3 (CVB3) is one of most common causes of heart inflammation and failure. However, the mechanism by which CVB3 induces cardiac damage has not been fully elucidated. Particularly, the involvement of microRNAs (miRNAs), a family of small RNAs controlling the progression of a wide range of diseases, in CVB3 infection is still unclear. These small RNAs are essential to understand the CVB3-caused heart muscle cell injury and have great potential to serve therapeutic purposes. Here, we systematically analyzed the miRNA changes during CVB3 infection and found that miR-21 is increased by viral infection. We further demonstrated that the CVB3-induced miR-21 triggers heart muscle cell damage by interfering with the cell-cell interactions. miR-21 suppresses the levels of components in cell-cell interactions by either promoting the degradation of those proteins or directly inhibiting the protein production. Inhibition of miR-21 can reduce the host injury caused by CVB3 infection. Our findings will shed new lights on the pathogenesis of CVB3-induced heart failure.


Vyšlo v časopise: Coxsackievirus-Induced miR-21 Disrupts Cardiomyocyte Interactions via the Downregulation of Intercalated Disk Components. PLoS Pathog 10(4): e32767. doi:10.1371/journal.ppat.1004070
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004070

Souhrn

Coxsackievirus B3 (CVB3) is one of most common causes of heart inflammation and failure. However, the mechanism by which CVB3 induces cardiac damage has not been fully elucidated. Particularly, the involvement of microRNAs (miRNAs), a family of small RNAs controlling the progression of a wide range of diseases, in CVB3 infection is still unclear. These small RNAs are essential to understand the CVB3-caused heart muscle cell injury and have great potential to serve therapeutic purposes. Here, we systematically analyzed the miRNA changes during CVB3 infection and found that miR-21 is increased by viral infection. We further demonstrated that the CVB3-induced miR-21 triggers heart muscle cell damage by interfering with the cell-cell interactions. miR-21 suppresses the levels of components in cell-cell interactions by either promoting the degradation of those proteins or directly inhibiting the protein production. Inhibition of miR-21 can reduce the host injury caused by CVB3 infection. Our findings will shed new lights on the pathogenesis of CVB3-induced heart failure.


Zdroje

1. HeL, HannonGJ (2004) MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 5: 522–531.

2. PasquinelliAE (2012) MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nat Rev Genet 13: 271–282.

3. SmallEM, FrostRJ, OlsonEN (2010) MicroRNAs add a new dimension to cardiovascular disease. Circulation 121: 1022–1032.

4. SkalskyRL, CullenBR (2010) Viruses, microRNAs, and host interactions. Annu Rev Microbiol 64: 123–141.

5. KumarswamyR, VolkmannI, ThumT (2011) Regulation and function of miRNA-21 in health and disease. RNA Biol 8: 706–713.

6. ChengY, ZhangC (2010) MicroRNA-21 in cardiovascular disease. J Cardiovasc Transl Res 3: 251–255.

7. SayedD, RaneS, LypowyJ, HeM, ChenIY, et al. (2008) MicroRNA-21 targets Sprouty2 and promotes cellular outgrowths. Mol Biol Cell 19: 3272–3282.

8. BauersachsJ (2012) miR-21: a central regulator of fibrosis not only in the broken heart. Cardiovasc Res 96: 227–229 discussion 230–223.

9. DongS, ChengY, YangJ, LiJ, LiuX, et al. (2009) MicroRNA expression signature and the role of microRNA-21 in the early phase of acute myocardial infarction. J Biol Chem 284: 29514–29525.

10. ThumT, GrossC, FiedlerJ, FischerT, KisslerS, et al. (2008) MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 456: 980–984.

11. TatsuguchiM, SeokHY, CallisTE, ThomsonJM, ChenJF, et al. (2007) Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol 42: 1137–1141.

12. ChengY, LiuX, ZhangS, LinY, YangJ, et al. (2009) MicroRNA-21 protects against the H(2)O(2)-induced injury on cardiac myocytes via its target gene PDCD4. J Mol Cell Cardiol 47: 5–14.

13. PatrickDM, MontgomeryRL, QiX, ObadS, KauppinenS, et al. (2010) Stress-dependent cardiac remodeling occurs in the absence of microRNA-21 in mice. J Clin Invest 120: 3912–3916.

14. RosatoP, AnastasiadouE, GargN, LenzeD, BoccellatoF, et al. (2012) Differential regulation of miR-21 and miR-146a by Epstein-Barr virus-encoded EBNA2. Leukemia 26: 2343–2352.

15. ChenY, ChenJ, WangH, ShiJ, WuK, et al. (2013) HCV-induced miR-21 contributes to evasion of host immune system by targeting MyD88 and IRAK1. PLoS Pathog 9: e1003248.

16. HuberSA, GaunttCJ, SakkinenP (1998) Enteroviruses and myocarditis: viral pathogenesis through replication, cytokine induction, and immunopathogenicity. Adv Virus Res 51: 35–80.

17. EckartRE, ScovilleSL, CampbellCL, ShryEA, StajduharKC, et al. (2004) Sudden death in young adults: a 25-year review of autopsies in military recruits. Ann Intern Med 141: 829–834.

18. HoBC, YuSL, ChenJJ, ChangSY, YanBS, et al. (2011) Enterovirus-induced miR-141 contributes to shutoff of host protein translation by targeting the translation initiation factor eIF4E. Cell Host Microbe 9: 58–69.

19. CorstenMF, PapageorgiouA, VerhesenW, CaraiP, LindowM, et al. (2012) MicroRNA profiling identifies microRNA-155 as an adverse mediator of cardiac injury and dysfunction during acute viral myocarditis. Circ Res 111: 415–425.

20. HemidaMG, YeX, ZhangHM, HansonPJ, LiuZ, et al. (2013) MicroRNA-203 enhances coxsackievirus B3 replication through targeting zinc finger protein-148. Cell Mol Life Sci 70: 277–291.

21. XuHF, DingYJ, ShenYW, XueAM, XuHM, et al. (2012) MicroRNA- 1 represses Cx43 expression in viral myocarditis. Mol Cell Biochem 362: 141–148.

22. LiuYL, WuW, XueY, GaoM, YanY, et al. (2013) MicroRNA-21 and -146b are involved in the pathogenesis of murine viral myocarditis by regulating TH-17 differentiation. Arch Virol 7: 593–608.

23. SheikhF, RossRS, ChenJ (2009) Cell-cell connection to cardiac disease. Trends Cardiovasc Med 19: 182–190.

24. GarrodDR, BerikaMY, BardsleyWF, HolmesD, TaberneroL (2005) Hyper-adhesion in desmosomes: its regulation in wound healing and possible relationship to cadherin crystal structure. J Cell Sci 118: 5743–5754.

25. JamoraC, FuchsE (2002) Intercellular adhesion, signalling and the cytoskeleton. Nat Cell Biol 4: E101–108.

26. NoormanM, van der HeydenMA, van VeenTA, CoxMG, HauerRN, et al. (2009) Cardiac cell-cell junctions in health and disease: Electrical versus mechanical coupling. J Mol Cell Cardiol 47: 23–31.

27. DennertR, CrijnsHJ, HeymansS (2008) Acute viral myocarditis. Eur Heart J 29: 2073–2082.

28. LewisBP, BurgeCB, BartelDP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120: 15–20.

29. DweepH, StichtC, PandeyP, GretzN (2011) miRWalk–database: prediction of possible miRNA binding sites by “walking” the genes of three genomes. J Biomed Inform 44: 839–847.

30. BaronCP, JacobsenS, PurslowPP (2004) Cleavage of desmin by cysteine proteases: Calpains and cathepsin B. Meat Sci 68: 447–456.

31. ChenF, ChangR, TrivediM, CapetanakiY, CrynsVL (2003) Caspase proteolysis of desmin produces a dominant-negative inhibitor of intermediate filaments and promotes apoptosis. J Biol Chem 278: 6848–6853.

32. CohenS, ZhaiB, GygiSP, GoldbergAL (2012) Ubiquitylation by Trim32 causes coupled loss of desmin, Z-bands, and thin filaments in muscle atrophy. J Cell Biol 198: 575–589.

33. ErnstR, MuellerB, PloeghHL, SchliekerC (2009) The otubain YOD1 is a deubiquitinating enzyme that associates with p97 to facilitate protein dislocation from the ER. Mol Cell 36: 28–38.

34. NewtonK, MatsumotoML, WertzIE, KirkpatrickDS, LillJR, et al. (2008) Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies. Cell 134: 668–678.

35. WagnerSA, BeliP, WeinertBT, ScholzC, KelstrupCD, et al. (2012) Proteomic analyses reveal divergent ubiquitylation site patterns in murine tissues. Mol Cell Proteomics 11: 1578–1585.

36. XuG, PaigeJS, JaffreySR (2010) Global analysis of lysine ubiquitination by ubiquitin remnant immunoaffinity profiling. Nat Biotechnol 28: 868–873.

37. KimW, BennettEJ, HuttlinEL, GuoA, LiJ, et al. (2011) Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol Cell 44: 325–340.

38. FujitaS, ItoT, MizutaniT, MinoguchiS, YamamichiN, et al. (2008) miR-21 Gene expression triggered by AP-1 is sustained through a double-negative feedback mechanism. J Mol Biol 378: 492–504.

39. IliopoulosD, JaegerSA, HirschHA, BulykML, StruhlK (2010) STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol Cell 39: 493–506.

40. PerdigueroE, Sousa-VictorP, Ruiz-BonillaV, JardiM, CaellesC, et al. (2011) p38/MKP-1-regulated AKT coordinates macrophage transitions and resolution of inflammation during tissue repair. J Cell Biol 195: 307–322.

41. YasukawaH, YajimaT, DuplainH, IwatateM, KidoM, et al. (2003) The suppressor of cytokine signaling-1 (SOCS1) is a novel therapeutic target for enterovirus-induced cardiac injury. J Clin Invest 111: 469–478.

42. JensenKJ, GarmaroudiFS, ZhangJ, LinJ, BoroomandS, et al. (2013) An ERK-p38 subnetwork coordinates host cell apoptosis and necrosis during coxsackievirus B3 infection. Cell Host Microbe 13: 67–76.

43. BanerjeeI, FuselerJW, PriceRL, BorgTK, BaudinoTA (2007) Determination of cell types and numbers during cardiac development in the neonatal and adult rat and mouse. Am J Physiol Heart Circ Physiol 293: H1883–1891.

44. ShiY, ChenC, LisewskiU, WrackmeyerU, RadkeM, et al. (2009) Cardiac deletion of the Coxsackievirus-adenovirus receptor abolishes Coxsackievirus B3 infection and prevents myocarditis in vivo. J Am Coll Cardiol 53: 1219–1226.

45. RoyS, KhannaS, HussainSR, BiswasS, AzadA, et al. (2009) MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue. Cardiovasc Res 82: 21–29.

46. AbstonED, CoronadoMJ, BucekA, OnyimbaJA, BrandtJE, et al. (2013) TLR3 deficiency induces chronic inflammatory cardiomyopathy in resistant mice following coxsackievirus B3 infection: role for IL-4. Am J Physiol Regul Integr Comp Physiol 304: R267–277.

47. GuiJ, YueY, ChenR, XuW, XiongS (2012) A20 (TNFAIP3) alleviates CVB3-induced myocarditis via inhibiting NF-kappaB signaling. PLoS One 7: e46515.

48. Smigielska-CzepielK, van den BergA, JellemaP, Slezak-ProchazkaI, MaatH, et al. (2013) Dual role of miR-21 in CD4+ T-cells: activation-induced miR-21 supports survival of memory T-cells and regulates CCR7 expression in naive T-cells. PLoS One 8: e76217.

49. JuY, WangT, LiY, XinW, WangS, et al. (2007) Coxsackievirus B3 affects endothelial tight junctions: possible relationship to ZO-1 and F-actin, as well as p38 MAPK activity. Cell Biol Int 31: 1207–1213.

50. KartenbeckJ, FrankeWW, MoserJG, StoffelsU (1983) Specific attachment of desmin filaments to desmosomal plaques in cardiac myocytes. EMBO J 2: 735–742.

51. McLendonPM, RobbinsJ (2011) Desmin-related cardiomyopathy: an unfolding story. Am J Physiol Heart Circ Physiol 301: H1220–1228.

52. BaloghJ, MerisckayM, LiZ, PaulinD, ArnerA (2002) Hearts from mice lacking desmin have a myopathy with impaired active force generation and unaltered wall compliance. Cardiovasc Res 53: 439–450.

53. LiZ, Colucci-GuyonE, Pincon-RaymondM, MericskayM, PourninS, et al. (1996) Cardiovascular lesions and skeletal myopathy in mice lacking desmin. Dev Biol 175: 362–366.

54. MilnerDJ, WeitzerG, TranD, BradleyA, CapetanakiY (1996) Disruption of muscle architecture and myocardial degeneration in mice lacking desmin. J Cell Biol 134: 1255–1270.

55. GoncharovT, NiessenK, de AlmagroMC, Izrael-TomasevicA, FedorovaAV, et al. (2013) OTUB1 modulates c-IAP1 stability to regulate signalling pathways. EMBO J 32: 1103–1114.

56. SunXX, ChallagundlaKB, DaiMS (2012) Positive regulation of p53 stability and activity by the deubiquitinating enzyme Otubain 1. EMBO J 31: 576–592.

57. XuP, DuongDM, SeyfriedNT, ChengD, XieY, et al. (2009) Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell 137: 133–145.

58. PickartCM, FushmanD (2004) Polyubiquitin chains: polymeric protein signals. Curr Opin Chem Biol 8: 610–616.

59. MarchantD, SiX, LuoH, McManusB, YangD (2008) The impact of CVB3 infection on host cell biology. Curr Top Microbiol Immunol 323: 177–198.

60. Zemljic-HarpfAE, MillerJC, HendersonSA, WrightAT, MansoAM, et al. (2007) Cardiac-myocyte-specific excision of the vinculin gene disrupts cellular junctions, causing sudden death or dilated cardiomyopathy. Mol Cell Biol 27: 7522–7537.

61. RalfkiaerU, HagedornPH, BangsgaardN, LovendorfMB, AhlerCB, et al. (2011) Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL). Blood 118: 5891–5900.

62. MarchantD, DouY, LuoH, GarmaroudiFS, McDonoughJE, et al. (2009) Bosentan enhances viral load via endothelin-1 receptor type-A-mediated p38 mitogen-activated protein kinase activation while improving cardiac function during coxsackievirus-induced myocarditis. Circ Res 104: 813–821.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 4
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#