Osmotin Protects H9c2 Cells from Simulated Ischemia-Reperfusion Injury through AdipoR1/PI3K/AKT Signaling Pathway

Jianhua Liu, Hua Sui, Jianlin Zhao, Yan Wang, Jianhua Liu, Hua Sui, Jianlin Zhao, Yan Wang

Abstract

Objective: This study aimed to investigate the effect of osmotin on myocardial ischemia/reperfusion (I/R), as well as the underlying mechanisms. Methods:In vitro I/R injury model was established on rat cardiac myoblast H9c2 cells by oxygen and glucose deprivation followed by reperfusion (OGD/R). Cells were administrated with osmotin, and transfected with small interfering RNAs (siRNAs) which specifically target adiponectin receptor 1 or 2 (AdipoR1/2). Besides, the cells were incubated with or without LY294002 as inhibitor of phosphatidylinositol 3-kinase (PI3K) under OGD/R condition. Cell viability, apoptosis, expressions of apoptosis-related proteins and inflammatory factors were analyzed. Results: The results showed that osmotin significantly increased H9c2 cells viability compared with the cells treated with vehicle (P < 0.05), and decreased H9c2 cells apoptosis by regulating expressions of apoptosis-related proteins. Moreover, we observed that osmotin statistically reduced the release of proinflammatory factors and increased the release of anti-inflammatory factors in H9c2 cells (P < 0.05). However, these effects were markedly reversed by AdipoR1 silence but not AdipoR2. Furthermore, osmotin dramatically upregulated the phosphorylation levels of PI3K, AKT, ERK, and downregulated the phosphorylation level of NF-κB (P < 0.05). While administration of LY294002 reduced cell viability, increased cell apoptosis, and aggravated inflammatory response (P < 0.05). Conclusion: Our results suggested that the protective effect of osmotin on the simulated OGD/R injured H9c2 cells might be associated with AdipoR1/PI3K/AKT signaling pathway.

Keywords: AKT signaling pathway; AdipoR1/PI3K signaling pathway; acute myocardial infarction; ischemia-reperfusion injury; osmotin.

Figures

Figure 1
Figure 1
The time axis of OGD/R exposure and osmotin administration with or without LY294002 treatment. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; siRNA, small interfering RNA; semi-qRT-PCR, Semi-quantitative real-time reverse transcriptase polymerase chain reaction; LDH, lactate dehydrogenase; ROS, reactive oxidative stress; MTT, 3-(4, 5-dimethylthiazol-yl)-2, 5-diphenyl-2-H-tetrazolium bromide.
Figure 2
Figure 2
Effects of osmotin combine with AdipoRs silence on H9c2 cell viability. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or transfection with specifically siRNAs for AdipoR1 or AdipoR2. (A) Effects of osmotin on the cell viability (n = 5); (B–D). Transfection efficiency of siRNAs for AdipoR1 and AdipoR2 were measured by semi-qRT-PCR (n = 5), and western blot assay. GAPDH acted as internal control; (E) Effects of osmotin and/or combine with AdipoR1 or AdipoR2 siRNAs transfection on cell viability (n = 5). (F) Relative LDH release of H9c2 cells after corresponding administrations (n = 5). *P < 0.05 compare to corresponding control; ns, no significant compared to the osmotin group. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; NC, negative control; siRNA, small interfering RNA; semi-qRT-PCR, semi-quantitative Real-time reverse transcriptase polymerase chain reaction; LDH, lactate dehydrogenase.
Figure 3
Figure 3
Effects of osmotin and silencing of AdipoRs on apoptosis of H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or transfection with specifically siRNAs for AdipoR1 or AdipoR2. (A) Effects of osmotin and silencing of AdipoRs on the percentages of apoptotic cells (n = 5); (B) The expressions of apoptosis-related proteins were measured by Wearer blot. (C–E) Relative protein expression levels of apoptotic related proteins (n = 5). (F) Effects of osmotin and silencing of AdipoRs on the number of ROS positive cells (n = 5). *P < 0.05 compare to corresponding control; ns, no significant compared to the osmotin group. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; Bcl, B-cell lymphoma; siRNA, small interfering RNA; ROS, reactive oxidative stress; ns, non-significant.
Figure 4
Figure 4
Effects of osmotin and silencing of AdipoRs on expressions of proinflammatory factors in H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or transfection with specifically siRNAs for AdipoR1 or AdipoR2. (A) Expressions of proinflammatory factors including IL-1β, IL-6, IL-8, and TNF-αmeasured by western blot. (B–E), Relative protein expression levels of proinflammatory factors IL-1β, IL-6, IL-8, and TNF-α(n = 5). *P < 0.05 compare to corresponding control; ns, no significant compared to the osmotin group. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; IL, interleukin; TNF, tumor necrosis factor; NF-κB, nuclear factor-kappa B; siRNA, small interfering RNA; ns, non-significant.
Figure 5
Figure 5
Effects of osmotin and silencing of AdipoRs on inflammatory factors in H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or transfection with specifically siRNAs for AdipoR1 or AdipoR2. (A–D). The relative contents of proinflammatory factors IL-1β, IL-6, IL-8, and TNF-α were measured by ELISA assay (n = 5). (E) Protein expressions of anti-inflammatory factors IL-4, IL-10 and IL-13 were measured by western blot. *P < 0.05 compare to corresponding control; ns, no significant compared to the osmotin group. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; IL, interleukin; TNF, tumor necrosis factor; siRNA, small interfering RNA; ns, non-significant.
Figure 6
Figure 6
Effects of osmotin and silencing of AdipoRs on the expressions of PI3K, AKT, NF-κB, and ERK in H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or transfection with specifically siRNAs for AdipoR1 or AdipoR2. (A) Expressions of p/t-PI3K and p/t-AKT in H9c2 cells measured by western blot after corresponding administrations. (B) The ratio of p/t PI3K (n = 5). (C) The ratio of p/t AKT (n = 5). (D) Expressions of p/t-NF-κB and p/t-ERK1/2 in H9c2 cells measured by western blot after corresponding administrations. (E) The ratio of p/t NF-κB. (F) The ratio of p/t ERK1/2 *P < 0.05 compare to corresponding control; ns, no significant compared to the osmotin group. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; PI3K, phosphatidylinositol 3-kinase; NF-κB, nuclear factor kappa B; siRNA, small interfering RNA; ns, non-significant.
Figure 7
Figure 7
Effects of osmotin and administration of LY294002 on H9c2 cell viability and apoptosis. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or LY294002 as inhibitor of PI3K. (A) Effects of osmotin and/or LY294002 administration on cell viability (n = 5); (B) Relative LDH release of administrated H9c2 cells (n = 5). (C) Effects of osmotin and administration of LY294002 on cell apoptosis (n = 5). (D) Expressions of apoptotic related factors measured by western bolt. GAPDH acted as internal control. (E–G) Relative expression levels of apoptotic related factors (n = 5). (H) Effects of osmotin and/or LY294002 administration on the number of ROS positive cells (n = 5). *P < 0.05 compare to corresponding control. OGD/R, oxygen and glucose deprivation/reperfusion; LY294002, inhibitor of phosphatidylinositol 3-kinase (PI3K); LDH, lactate dehydrogenase; ROS, reactive oxidative stress; LY294002, inhibitor of phosphatidylinositol 3-kinase (PI3K).
Figure 8
Figure 8
Effects of osmotin and administration of LY294002 on the proinflammatory factors expressions in H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or LY294002 as inhibitor of PI3K. (A) Expressions of IL-1β, IL-6, IL-8, and TNF-α in H9c2 cells after administration were measured by western blot. (B–E) Relative expression levels of IL-1β, IL-6, IL-8, and TNF-α(n = 5). *P < 0.05 compare to corresponding control. OGD/R, oxygen and glucose deprivation/reperfusion; LY294002, inhibitor of phosphatidylinositol 3-kinase (PI3K); IL, interleukin; TNF, tumor necrosis factor.
Figure 9
Figure 9
Effects of osmotin and administration of LY294002 on the inflammatory factors contents in H9c2 cells. H9c2 cells were pre-treated with OGD/R, and then been administrated with osmotin and/or LY294002 as inhibitor of PI3K. (A–D) Results of ELISA assay about the contents of proinflammatory factors IL-1β, IL-6, IL-8, and TNF-α(n = 5). (E) Protein expressions of anti-inflammatory factors IL-4, IL-10 and IL-13 were detected by western blot *P < 0.05 compare to corresponding control. OGD/R, oxygen and glucose deprivation/reperfusion; AdipoR, adiponectin receptor; LY294002, inhibitor of phosphatidylinositol 3-kinase (PI3K); IL, interleukin; TNF, tumor necrosis factor.

References

    1. Ali T., Yoon G. H., Shah S. A., Lee H. Y., Kim M. O. (2015). Osmotin attenuates amyloid beta-induced memory impairment, tau phosphorylation and neurodegeneration in the mouse hippocampus. Sci. Rep. 5:11708. 10.1038/srep11708
    1. Anil K. S., Hima K. P., Shravan K. G., Mohanalatha C., Kavi Kishor P. B. (2015). Osmotin: a plant sentinel and a possible agonist of mammalian adiponectin. Front. Plant Sci. 6:163 10.3389/fpls.2015.00163
    1. Arslan F., Lai R. C., Smeets M. B., Akeroyd L., Choo A., Aguor E. N. E., et al. . (2013). Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 10, 301–312. 10.1016/j.scr.2013.01.002
    1. Badshah H., Ali T., Kim M. O. (2016). Osmotin attenuates LPS-induced neuroinflammation and memory impairments via the TLR4/NFκB signaling pathway. Sci. Rep. 6:24493. 10.1038/srep24493
    1. Baumgarten G. (2001). In vivo expression of proinflammatory mediators in the adult heart after endotoxin administration: the role of toll-like receptor-4. J. Inf. Dis. 183, 1617–1624. 10.1086/320712
    1. Cantley L. C. (2002). The phosphoinositide 3-kinase pathway. Science 296, 1655–1657. 10.1126/science.296.5573.1655
    1. Chan K. H., Lam K. S., Cheng O. Y., Kwan J. S., Ho P. W., Cheng K. K., et al. . (2012). Adiponectin is protective against oxidative stress induced cytotoxicity in amyloid-beta neurotoxicity. PLoS ONE 7:e52354. 10.1371/journal.pone.0052354
    1. Ding G., Qin Q., He N., Francis-David S. C., Hou J., Liu J., et al. . (2007). Adiponectin and its receptors are expressed in adult ventricular cardiomyocytes and upregulated by activation of peroxisome proliferator-activated receptor γ. J. Mol. Cell. Cardiol. 43, 73–84. 10.1016/j.yjmcc.2007.04.014
    1. Fang F., Li D., Pan H., Chen D., Qi L., Zhang R., et al. . (2011). Luteolin inhibits apoptosis and improves cardiomyocyte contractile function through the PI3K/Akt pathway in simulated ischemia/reperfusion. Pharmacology 88, 149–158. 10.1159/000330068
    1. Frangogiannis N. G., Smith C. W., Entman M. L. (2002). The inflammatory response in myocardial infarction. Cardiovasc. Res. 53, 31–47. 10.1016/S0008-6363(01)00434-5
    1. Frantz S., Kobzik L., Kim Y. D., Fukazawa R., Medzhitov R., Lee R. T., et al. . (1999). Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J. Clin. Invest. 104, 271–280. 10.1172/JCI6709
    1. Fujio Y., Nguyen T. D., Kitsis R., Walsh K. (2000). Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. Circulation 101, 660–667. 10.1161/01.CIR.101.6.660
    1. Gonon A. T., Widegren U., Bulhak A., Salehzadeh F., Persson J., Sjöquist P. O., et al. . (2008). Adiponectin protects against myocardial ischaemia-reperfusion injury via AMP-activated protein kinase, Akt, and nitric oxide. Cardiovasc. Res. 78, 116–122. 10.1093/cvr/cvn017
    1. Ha T., Fang H., Liu X., Ma J., Mcmullen J. R., Shioi T., et al. . (2008). Lipopolysaccharide-induced myocardial protection against ischaemia/reperfusion injury is mediated through a PI3K/Akt-dependent mechanism. Cardiovasc. Res. 78, 546–553. 10.1093/cvr/cvn037
    1. Heusch G. (2013). Cardioprotection: chances and challenges of its translation to the clinic. Lancet 381, 166–175. 10.1016/S0140-6736(12)60916-7
    1. Inada T., Kubo K., Shingu K. (2011). Possible link between cyclooxygenase-inhibiting and antitumor properties of propofol. J. Anesth. 25, 569–575. 10.1007/s00540-011-1163-y
    1. Ishii H., Kamikawa S., Hirohata S., Mizutani A., Abe K., Seno M., et al. . (2015). Eosinophil cationic protein shows survival effect on H9c2 cardiac myoblast cells with enhanced phosphorylation of ERK and Akt/GSK-3beta under oxidative stress. Acta Med. Okayama 69, 145–153. 10.18926/AMO/53521
    1. Kadowaki T., Yamauchi T., Kubota N., Hara K., Ueki K., Tobe K. (2006). Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J. Clin. Invest. 116, 1784–1792. 10.1172/JCI29126
    1. Kawaguchi M., Takahashi M., Hata T., Kashima Y., Usui F., Morimoto H., et al. . (2011). Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation 123, 594–604. 10.1161/CIRCULATIONAHA.110.982777
    1. Kuznetsov A. V., Javadov S., Sickinger S., Frotschnig S., Grimm M. (2015). H9c2 and HL-1 cells demonstrate distinct features of energy metabolism, mitochondrial function and sensitivity to hypoxia-reoxygenation. Biochim. Biophys. Acta 1853, 276–284. 10.1016/j.bbamcr.2014.11.015
    1. Liu S., Yin T., Wei X., Yi W., Qu Y., Liu Y., et al. . (2011). Downregulation of adiponectin induced by tumor necrosis factor α is involved in the aggravation of posttraumatic myocardial ischemia/reperfusion injury. Crit. Care Med. 39, 1935–1943. 10.1097/CCM.0b013e31821b85db
    1. Maia-fernandes T., Roncon-Albuquerque R., Jr., Leite-moreira A. F. (2008). Cardiovascular actions of adiponectin: pathophysiologic implications. Rev. Port. Cardiol. 27, 1431–1449.
    1. Medzhitov R., Preston-Hurlburt P., Janeway C. A., Jr. (1997). A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature 388, 394–397. 10.1038/41131
    1. Miele M., Costantini S., Colonna G. (2011). Structural and functional similarities between osmotin from Nicotiana tabacum seeds and human adiponectin. PLoS ONE 6:e16690. 10.1371/journal.pone.0016690
    1. Min K., Ha S. C., Hasegawa P. M., Bressan R. A., Yun D. J., Kim K. K. (2003). Crystal structure of osmotin, a plant antifungal protein. Proteins 54, 170–173. 10.1002/prot.10571
    1. Meijing W., Yue W., Brent W., Aaron A., Jeremy H., Jiangning T., et al. (2009). Estrogen receptor β mediates increased activation of PI3K/Akt signaling and improved myocardial function in female hearts following acute ischemia. Am. J. Physiol. Regul. Integr. Comp. Physi. 296, 972–978. 10.1152/ajpregu.00045.2009
    1. Narasimhan M. L., Coca M. A., Jin J., Yamauchi T., Ito Y., Kadowaki T., et al. . (2005). Osmotin is a homolog of mammalian adiponectin and controls apoptosis in yeast through a homolog of mammalian adiponectin receptor. Mol. Cell 17, 171–180. 10.1016/j.molcel.2004.11.050
    1. Naseer M. I., Ullah I., Narasimhan M. L., Lee H. Y., Bressan R. A., Yoon G. H., et al. . (2014). Neuroprotective effect of osmotin against ethanol-induced apoptotic neurodegeneration in the developing rat brain. Cell Death Dis. 5, e1150. 10.1038/cddis.2014.53
    1. Oh D. K., Theodore C., Henry R. R. (2007). Adiponectin in health and disease. Diabetes Obes. Metab. 9, 282–289. 10.1111/j.1463-1326.2006.00610.x
    1. Ouchi N., Shibata R., Walsh K. (2006). Cardioprotection by adiponectin. Trends Cardiovasc. Med. 16, 141–146. 10.1016/j.tcm.2006.03.001
    1. Oyama J., Blais C., Jr., Liu X., Pu M., Kobzik L., Kelly R. A., et al. . (2004). Reduced myocardial ischemia-reperfusion injury in toll-like receptor 4-deficient mice. Circulation 109, 784–789. 10.1161/01.CIR.0000112575.66565.84
    1. Piñeiro R., Iglesias M. J., Gallego R., Raghay K., Eiras S., Rubio J., et al. . (2005). Adiponectin is synthesized and secreted by human and murine cardiomyocytes. FEBS Lett. 579, 5163–5169. 10.1016/j.febslet.2005.07.098
    1. Santos-Gallego C. G., Picatoste B., Badimon J. J. (2014). Pathophysiology of acute coronary syndrome. Curr. Atheroscler. Rep. 16:401. 10.1007/s11883-014-0401-9
    1. Santos-Gallego C. G., Vahl T. P., Goliasch G., Picatoste B., Arias T., Ishikawa K., et al. . (2016). Sphingosine-1-phosphate receptor agonist fingolimod increases myocardial salvage and decreases adverse postinfarction left ventricular remodeling in a porcine model of ischemia/reperfusion. Circulation 133, 954–966. 10.1161/CIRCULATIONAHA.115.012427
    1. Shah S. A., Lee H. Y., Bressan R. A., Yun D. J., Kim M. O. (2014). Novel osmotin attenuates glutamate-induced synaptic dysfunction and neurodegeneration via the JNK/PI3K/Akt pathway in postnatal rat brain. Cell Death Dis. 5:e1026. 10.1038/cddis.2013.538
    1. Shah S. A., Yoon G. H., Chung S. S., Abid M. N., Kim T. H., Lee H. Y., et al. . (2016). Novel osmotin inhibits SREBP2 via the AdipoR1/AMPK/SIRT1 pathway to improve Alzheimer's disease neuropathological deficits. Mol. Psychiatry 22, 407–416. 10.1038/mp.2016.23
    1. Shibata R., Sato K., Pimentel D. R., Takemura Y., Kihara S., Ohashi K., et al. . (2005). Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat. Med. 11, 1096–1103. 10.1038/nm1295
    1. Tao L., Gao E., Jiao X., Yuan Y., Li S., Christopher T. A., et al. . (2007). Adiponectin cardioprotection after myocardial ischemia/reperfusion involves the reduction of oxidative/nitrative stress. Circulation 115, 1408–1416. 10.1161/CIRCULATIONAHA.106.666941
    1. Ullah N., Naseer M. I., Ullah I., Lee H. Y., Koh P. O., Kim M. O. (2014). Protective effect of pyruvate against ethanol-induced apoptotic neurodegeneration in the developing rat brain. Neuropharmacology 61, 1248–1255. 10.1016/j.neuropharm.2011.06.031
    1. Villarreal-Molina M. T., Antuna-Puente B. (2012). Adiponectin: anti-inflammatory and cardioprotective effects. Biochimie 94, 2143–2149. 10.1016/j.biochi.2012.06.030
    1. Wang Y., Lau W. B., Gao E., Tao L., Yuan Y., Li R., et al. . (2010). Cardiomyocyte-derived adiponectin is biologically active in protecting against myocardial ischemia-reperfusion injury. Am. J. Physiol. Endocrinol. Metab. 298, E663–E670. 10.1152/ajpendo.00663.2009
    1. Wang Y., Tao L., Yuan Y., Lau W. B., Li R., Lopez B. L., et al. . (2009). Cardioprotective effect of adiponectin is partially mediated by its AMPK-independent antinitrative action. Am. J. Physiol. Endocrinol. Metab. 297, E384–E391. 10.1152/ajpendo.90975.2008
    1. Wu W. Y., Wang W. Y., Ma Y. L., Yan H., Wang X. B., Qin Y. L., et al. . (2013). Sodium tanshinone IIA silate inhibits oxygen-glucose deprivation/recovery-induced cardiomyocyte apoptosis via suppression of the NF-κB/TNF-α pathway. Br. J. Pharmacol. 169, 1058–1071. 10.1111/bph.12185
    1. Yamauchi T., Kadowaki T. (2008). Physiological and pathophysiological roles of adiponectin and adiponectin receptors in the integrated regulation of metabolic and cardiovascular diseases. Int. J. Obes. 32(Suppl. 7), S13–S18. 10.1038/ijo.2008.233
    1. Yun D. J., Ibeas J. I., Lee H., Coca M. A., Narasimhan M. L., Uesono Y., et al. . (1998). Osmotin, a plant antifungal protein, subverts signal transduction to enhance fungal cell susceptibility. Mol. Cell 1, 807–817. 10.1016/S1097-2765(00)80080-5
    1. Zhang J., Feng Z., Wang C., Zhou H., Liu W., Kanchana K., et al. . (2017). Curcumin derivative WZ35 efficiently suppresses colon cancer progression through inducing ROS production and ER stress-dependent apoptosis. Am. J. Cancer Res. 7, 275–288.

Source: PubMed

3
Abonnieren