PL EN


Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników
2018 | 23 |

Tytuł artykułu

MicroRNA-495 inhibits the high glucoseinduced inflammation, differentiation and extracellular matrix accumulation of cardiac fibroblasts through downregulation of NOD1

Autorzy

Warianty tytułu

Języki publikacji

EN

Abstrakty

EN
Background: MicroRNAs (miRNAs) have physiological and pathophysiological functions that are involved in the regulation of cardiac fibrosis. This study aimed to investigate the effects of miR-495 on high glucose-induced cardiac fibrosis in human cardiac fibroblasts (CFs) and to establish the mechanism underlying these effects. Methods: Human CFs were transfected with an miR-495 inhibitor or mimic and incubated with high glucose. The levels of NOD1 and miR-495 were then determined via quantitative RT-PCR. Pro-inflammatory cytokine levels, cell differentiation and extracellular matrix accumulation were respectively detected using ELISA, quantitative RT-PCR and western blot assays. The luciferase reporter assay, quantitative RT-PCR and western blot were used to explore whether NOD1 was a target of miR-495. The effects of miR-495 on the NF-κB and TGF-β1/Smad signaling pathways were also detected via western blot. Results: Our results show that high glucose can significantly increase the expression of NOD1 in a time-dependent manner. Upregulation of miR-495 significantly alleviated the high glucose-induced increases in cell differentiation and collagen accumulation of CFs. Moreover, the bioinformatics analysis predicted that NOD1 was a potential target gene for miR-495. The luciferase reporter assay showed that miR-495 can directly target NOD1. The introduction of miR-495 could significantly inhibit the high glucose-activated NF-κB and TGF-β1/Smad signaling pathways. Conclusion: Upregulation of miR-495 ameliorates the high glucose-induced inflammatory, cell differentiation and extracellular matrix accumulation of human CFs by modulating both the NF-κB and TGF-β1/Smad signaling pathways through downregulation of NOD1 expression. These results provide further evidence for the protective effect of miR-495 overexpression in cases of high glucose-induced cardiac fibrosis.

Słowa kluczowe

Wydawca

-

Rocznik

Tom

23

Opis fizyczny

p.1-13,fig.,ref.

Twórcy

autor
  • Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai 201707, People’s Republic of China
autor
  • Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai 201707, People’s Republic of China
autor
  • Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai 201707, People’s Republic of China
autor
  • Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai 201707, People’s Republic of China

Bibliografia

  • 1. Zatalia SR, Sanusi H. The role of antioxidants in the pathophysiology, complications, and management of diabetes mellitus. Acta Med Indones. 2013;45:141–7.
  • 2. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 2014;71:549–74.
  • 3. Val-Blasco A, Prieto P, Gonzalez-Ramos S, Benito G, Vallejo-Cremades MT, Pacheco I, González-Peramato P, Agra N, Terrón V, Delgado C, Martín-Sanz P, Boscá L, Fernández-Velasco M. NOD1 activation in cardiac fibroblasts induces myocardial fibrosis in a murine model of type 2 diabetes. Biochem J. 2017;474:399–410.
  • 4. García R, Merino D, Gómez JM, Nistal JF, Hurlé MA, Cortajarena AL, Villar AV. Extracellular heat shock protein 90 binding to TGFβ receptor I participates in TGFβ-mediated collagen production in myocardial fibroblasts. Cell Signal. 2016;28:1563–79.
  • 5. Karri VV, Kuppusamy G, Talluri SV, Mannemala SS, Kollipara R, Wadhwani AD, Mulukutla S, Raju KR, Malayandi R. Curcumin loaded chitosan nanoparticles impregnated into collagen-alginate scaffolds for diabetic wound healing. Int J Biol Macromol. 2016;9:1519–29.
  • 6. Schertzer JD, Tamrakar AK, Magalhães JG, Pereira S, Bilan PJ, Fullerton MD, Liu Z, Steinberg GR, Giacca A, Philpott DJ, Klip A. NOD1 activators link innate immunity to insulin resistance. Diabetes. 2011;60:2206–15.
  • 7. Prieto P, Vallejo-Cremades MT, Benito G, González-Peramato P, Francés D, Agra N, Terrón V, Gónzalez-Ramos S, Delgado C, Ruiz-Gayo M, Pacheco I, Velasco-Martín JP, Regadera J, Martín-Sanz P, López-Collazo E, Boscá L, Fernández-Velasco M. NOD1 receptor is up-regulated in diabetic human and murine myocardium. Clin Sci (Lond). 2014;127:665–77.
  • 8. Chi W, Dao D, Lau TC, Henriksbo BD, Cavallari JF, Foley KP, Schertzer JD. Bacterial peptidoglycan stimulates adipocyte lipolysis via NOD1. PLoS One. 2014;9:e97675.
  • 9. Prajapati B, Jena PK, Rajput P, Purandhar K, Seshadri S. Understanding and modulating the toll like receptors (TLRs) and NOD like receptors (NLRs) cross talk in type 2 diabetes. Curr Diabetes Rev. 2014;10:190–200.
  • 10. Zhou YJ, Liu C, Li CL, Song YL, Tang YS, Zhou H, Li A, Li Y, Weng Y, Zheng FP. Increased NOD1, but not NOD2, activity in subcutaneous adipose tissue from patients with metabolic syndrome. Obesity (Silver Spring). 2015;23:1394–400.
  • 11. Fernández-Velasco M, Prieto P, Terrón V, Benito G, Flores JM, Delgado C, Zaragoza C, Lavin B, Gómez-Parrizas M, López-Collazo E, Martín-Sanz P, Boscá L. NOD1 activation induces cardiac dysfunction and modulates cardiac fibrosis and cardiomyocyte apoptosis. PLoS One. 2012;7:e45260.
  • 12. Mennigen JA, Zhang D. Micro trout: a comprehensive, genome-wide miRNA target prediction framework for rainbow trout, Oncorhynchus mykiss. Comp Biochem Physiol Part D Genomics Proteomics. 2016;20:19–26.
  • 13. Wang Y, Ouyang M, Wang Q, Jian Z. MicroRNA-142-3p inhibits hypoxia/reoxygenation-induced apoptosis and fibrosis of cardiomyocytes by targeting high mobility group box 1. Int J Mol Med. 2016;38:1377–86.
  • 14. Cheng R, Dang R, Zhou Y, Ding M, Hua H. MicroRNA-98 inhibits TGF-β1-induced differentiation and collagen production of cardiac fibroblasts by targeting TGFBR1. Hum Cell. 2017;30:192–200.
  • 15. Rawal S, Munasinghe PE, Nagesh PT, Lew JKS, Jones GT, Williams MJA, Davis P, Bunton D, Galvin IF, Manning P, Lamberts RR, Katare R. Down-regulation of miR-15a/b accelerates fibrotic remodeling in the type 2 diabetic human and mouse heart. Clin Sci (Lond). 2017;131:847–63.
  • 16. Geng H, Guan J. MiR-18a-5p inhibits endothelial-mesenchymal transition and cardiac fibrosis through the Notch2 pathway. Biochem Biophys Res Commun. 2017;491:329–36.
  • 17. Zhong F, Chen H, Han L, Jin Y, Wang W. Curcumin attenuates lipopolysaccharide-induced renal inflammation. Biol Pharm Bull. 2011;34:226–32.
  • 18. Lee KY, Ito K, Hayashi R, Jazrawi EP, Barnes PJ, Adcock IM. NF-kappaB and activator protein 1 response elements and the role of histone modifications in IL-1beta-induced TGF-beta1 gene transcription. J Immunol. 2006;176:603–15.
  • 19. Schmierer B, Hill CS. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007;8:970–82.
  • 20. Burstein B, Nattel S. Atrial fibrosis: mechanisms and clinical relevance in atrial fibrillation. J Am Coll Cardiol. 2008;51:802–9.
  • 21. Heydari B, Abdullah S, Pottala JV, Shah R, Abbasi S, Mandry D, Francis SA, Lumish H, Ghoshhajra BB, Hoffmann U, Appelbaum E, Feng JH, Blankstein R, Steigner M, McConnell JP, Harris W, Antman EM, Jerosch-Herold M, Kwong RY. Effect of Omega-3 acid ethyl esters on left ventricular remodeling after acute myocardial infarction: the OMEGA-REMODEL randomized clinical trial. Circulation. 2016;134:378–91.
  • 22. Dong H, Dong S, Zhang L, Gao X, Lv G, Chen W, Shao S. MicroRNA-214 exerts a cardio-protective effect by inhibition of fibrosis. Anat Rec (Hoboken). 2016;299:1348–57.
  • 23. Wang J, Liew OW, Richards AM, Chen YT. Overview of MicroRNAs in cardiac hypertrophy, fibrosis, and apoptosis. Int J Mol Sci. 2016;17:E749.
  • 24. Zhang D, Cui Y, Li B, Luo X, Li B, Tang Y. miR-155 regulates high glucose-induced cardiac fibrosis via the TGF-β signaling pathway. Mol BioSyst. 2016;13:215–24.
  • 25. Yuan J, Chen H, Ge D, Xu Y, Xu H, Yang Y, Gu M, Zhou Y, Zhu J, Ge T, Chen Q, Gao Y, Wang Y, Li X, Zhao Y. Mir-21 promotes cardiac fibrosis after myocardial infarction via targeting Smad7. Cell Physiol Biochem. 2017;42:2207–19.
  • 26. Zhang W, Chang H, Zhang H, Zhang L. MiR-30e attenuates isoproterenol-induced cardiac fibrosis through suppressing Snai1/TGF-β signaling. J Cardiovasc Pharmacol. 2017;70:362–8.
  • 27. Cucoranu I, Clempus R, Dikalova A, Phelan PJ, Ariyan S, Dikalov S, Sorescu D. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ Res. 2005;97:900–7.
  • 28. Chen X, Liu G, Zhang W, Zhang J, Yan Y, Dong W, Liang E, Zhang Y, Zhang M. Inhibition of MEF2A prevents hyperglycemia-induced extracellular matrix accumulation by blocking Akt and TGF-β1/Smad activation in cardiac fibroblasts. Int J Biochem Cell Biol. 2015;69:52–61.
  • 29. Li Y, Ma J, Zhu H, Singh M, Hill D, Greer PA, Arnold JM, Abel ED, Peng T. Targeted inhibition of calpain reduces myocardial hypertrophy and fibrosis in mouse models of type 1 diabetes. Diabetes. 2011;60:2985–94.
  • 30. Li CJ, Lv L, Li H, Yu DM. Cardiac fibrosis and dysfunction in experimental diabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc Diabetol. 2012;11:73.
  • 31. Pelouch V, Dixon IM, Golfman L, Beamish RE, Dhalla NS. Role of extracellular matrix proteins in heart function. Mol Cell Biochem. 1993;129:101–20.
  • 32. Kanneganti TD, Lamkanfi M, Núñez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007;27:549–59.
  • 33. Fritz JH, Ferrero RL, Philpott DJ, Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nat Immunol. 2006;7:1250–7.
  • 34. Hysi P, Kabesch M, Moffatt MF, Schedel M, Carr D, Zhang Y, Boardman B, von Mutius E, Weiland SK, Leupold W, Fritzsch C, Klopp N, Musk AW, James A, Nunez G, Inohara N, Cookson WO. NOD1 variation, immunoglobulin E and asthma. Hum Mol Genet. 2005;14:935–41.
  • 35. Weidinger S, Klopp N, Rummler L, Wagenpfeil S, Novak N, Baurecht HJ, Groer W, Darsow U, Heinrich J, Gauger A, Schafer T, Jakob T, Behrendt H, Wichmann HE, Ring J, Illig T. Association of NOD1 polymorphisms with atopic eczema and related phenotypes. J Allergy Clin Immunol. 2005;116:177–84.
  • 36. da Silva CJ, Miranda Y, Austin-Brown N, Hsu J, Mathison J, Xiang R, Zhou H, Li Q, Han J, Ulevitch RJ. Nod1-dependent control of tumor growth. Proc Natl Acad Sci U S A. 2006;103:1840–5.
  • 37. Girardin SE, Tournebize R, Mavris M, Page AL, Li X, Stark GR, Bertin J, DiStefano PS, Yaniv M, Sansonetti PJ, Philpott DJ. CARD4/Nod1 mediates NF-kappaB and JNK activation by invasive Shigella flexneri. EMBO Rep. 2001;2:736–42.
  • 38. Takeshita K, Hayashi M, Iino S, Kondo T, Inden Y, Iwase M, Kojima T, Hirai M, Ito M, Loskutoff DJ, Saito H, Murohara T, Yamamoto K. Increased expression of plasminogen activator inhibitor-1 in cardiomyocytes contributes to cardiac fibrosis after myocardial infarction. Am J Pathol. 2004;164:449–56.

Typ dokumentu

Bibliografia

Identyfikatory

Identyfikator YADDA

bwmeta1.element.agro-d455b63e-a60c-45e0-bd21-57cc28751a72
JavaScript jest wyłączony w Twojej przeglądarce internetowej. Włącz go, a następnie odśwież stronę, aby móc w pełni z niej korzystać.