Pim-1 Kinase Regulating Dynamics Related Protein 1 Mediates Sevoflurane Postconditioning-induced Cardioprotection

Jin-Dong Liu, Hui-Juan Chen, Da-Liang Wang, Hui Wang, Qian Deng, Jin-Dong Liu, Hui-Juan Chen, Da-Liang Wang, Hui Wang, Qian Deng

Abstract

Background: It is well documented that sevoflurane postconditioning (SP) has a significant myocardial protection effect. However, the mechanisms underlying SP are still unclear. In the present study, we investigated the hypothesis that the Pim-1 kinase played a key role in SP-induced cardioprotection by regulating dynamics-related protein 1 (Drp1).

Methods: A Langendorff model was used in this study. Seventy-two rats were randomly assigned into six groups as follows: CON group, ischemia reperfusion (I/R) group, SP group , SP+proto-oncogene serine/threonine-protein kinase 1 (Pim-1) inhibitor II group, SP+dimethylsufoxide group, and Pim-1 inhibitor II group (n = 12, each). Hemodynamic parameters and infarct size were measured to reflect the extent of myocardial I/R injury. The expressions of Pim-1, B-cell leukemia/lymphoma 2 (Bcl-2) and cytochrome C (Cyt C) in cytoplasm and mitochondria, the Drp1 in mitochondria, and the total Drp1 and p-Drp1ser637 were measured by Western blotting. In addition, transmission electron microscope was used to observe mitochondrial morphology. The experiment began in October 2014 and continued until July 2016.

Results: SP improved myocardial I/R injury-induced hemodynamic parametric changes, cardiac function, and preserved mitochondrial phenotype and decreased myocardial infarct size (24.49 ± 1.72% in Sev group compared with 41.98 ± 4.37% in I/R group; P< 0.05). However, Pim-1 inhibitor II significantly (P < 0.05) abolished the protective effect of SP. Western blotting analysis demonstrated that, compared with I/R group, the expression of Pim-1 and Bcl-2 in cytoplasm and mitochondria as well as the total p-Drp1ser637 in Sev group (P < 0.05) were upregulated. Meanwhile, SP inhibited Drp1 compartmentalization to the mitochondria followed by a reduction in the release of Cyt C. Pretreatment with Pim-1 inhibitor II significantly (P < 0.05) abolished SP-induced Pim-1/p-Drp1ser637 signaling activation.

Conclusions: These findings suggested that SP could attenuate myocardial ischemia-reperfusion injury by increasing the expression of the Pim-1 kinase. Upregulation of Pim-1 might phosphorylate Drp1 and prevent extensive mitochondrial fission through Drp1 cytosolic sequestration.

Conflict of interest statement

There are no conflicts of interest.

Figures

Figure 1
Figure 1
Hemodynamic changes at different points in rats subjected to myocardial ischemia-reperfusion injury. (a) The changes of HR at T0–T4; (b) the changes of LVDP at T0–T4; (c) the changes of LVEDP at T0–T4; (d) the changes of + dp/dtmax at T0–T4; (e) and the changes of −dp/dtmax at T0–T4. Data were reported as mean ± SD (n = 6 for each group). *P < 0.05 versus CON; †P < 0.05 versus I/R; ‡P < 0.05 versus Sev (repeated measures ANOVA). CON: Control group; I/R: Ischemia/reperfusion; Sev: Sevoflurane postconditioning; Sev+PIM-Inh: Sevoflurane postconditioning + Pim-1 inhibitor II; Sev–DMSO: Sevoflurane postconditioning + dimethyl sulfoxide; PIM-Inh: Pim-1 inhibitor II; T0: The end of equilibration; T1: 30 min of reperfusion; T2: 60 min of reperfusion; T3: 90 min of reperfusion; T4: 120 min of reperfusion; HR: Heart rate; LVDP: Left ventricular developed pressure; LVEDP: Left ventricular end-diastolic pressure; +dp/dtmax: Maximum rate of left ventricular development pressure; −dp/dtmax: Maximum rate of left ventricular fall pressure; SD: Standard deviation; ANOVA: Analysis of variance.
Figure 2
Figure 2
Western blotting analysis of cytosolic and mitochondrial fraction of Pim-1, Bcl-2, and Cyt C in myocardium. The Pim-1 (44,000 Da) (a and b) was analyzed by Western blotting with specific Pim-1 antibody at 1 h of reperfusion; the Bcl-2 (26,000 Da) (c and d) was analyzed by Western blotting with specific Bcl-2 antibody at 1 h of reperfusion; and the Cyt C (15,000 Da) (e and f) was analyzed by Western blotting with specific Cyt C antibody at 1 h of reperfusion. (n = 3 for each group). Data were reported as mean ± SD. *P < 0.05 versus CON; †P < 0.05 versus I/R; ‡P < 0.05 versus Sev (repeated measures ANOVA). CON: Control group; I/R: Ischemia/reperfusion; Sev: Sevoflurane postconditioning; Sev+PIM-Inh: Sevoflurane postconditioning + Pim-1 inhibitor II; Sev+DMSO: Sevoflurane postconditioning + dimethyl sulfoxide; PIM-Inh: Pim-1 inhibitor II; Pim-1: Proto-oncogene serine/threonine-protein kinase 1; Bcl-2: B-cell leukemia/lymphoma 2; Cyt C: Cytochrome C; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; COX-IV: Cytochrome C oxidase IV; SD: Standard deviation; ANOVA: Analysis of variance.
Figure 3
Figure 3
Western blotting analysis of total Drp1 and p-Drp1ser637 in myocardium as well as Drp1 translation to the mitochondria. The total Drp1 (80,000 Da) (a) and the Drp1 in mitochondria (c) were analyzed by Western blotting with specific Drp1 antibody at 1 h of reperfusion; The p-Drp1ser637 (80,000 Da) (b) was analyzed by Western blotting with specific p-Drp1ser637 antibody at 1 h of reperfusion (n = 3 for each group). Data were reported as mean ± SD. *P < 0.05 versus CON; †P < 0.05 versus I/R; ‡P < 0.05 versus Sev (one-way ANOVA). CON: Control group; I/R: Ischemia/reperfusion; Sev: Sevoflurane postconditioning; Sev+PIM-Inh: Sevoflurane postconditioning + Pim-1 inhibitor II; Sev+DMSO: Sevoflurane postconditioning + dimethyl sulfoxide; PIM-Inh: Pim-1 inhibitor II; Drp1: Dynamics-related protein 1; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; COX-IV: Cytochrome C oxidase IV; SD: Standard deviation; ANOVA: Analysis of variance.
Figure 4
Figure 4
Myocardial infarct size of isolated hearts in six groups. (a) Representative cross sections of rat hearts from six groups after I/R and staining with TTC to visualize the infarcted area; (b) infarct size expressed as percentage of the left ventricular area (×100%) for each group. Values are represented as mean ± SD (n = 6 for each group), *P < 0.05 versus CON; †P < 0.05 versus I/R; ‡P < 0.05 versus Sev (one-way ANOVA). CON: Control group; I/R: Ischemia/reperfusion; Sev: Sevoflurane postconditioning; Sev+PIM-Inh: Sevoflurane postconditioning + Pim-1 inhibitor II; Sev+DMSO: Sevoflurane postconditioning + dimethyl sulfoxide; PIM-Inh: Pim-1 inhibitor II; SD: Standard deviation; ANOVA: Analysis of variance.
Figure 5
Figure 5
Transmission electron microscope (uranyl acetate-lead citrate stained, original magnification ×30,000) performance of the myocardial mitochondria in six groups. (a) In the CON group, the mitochondria were normal (b) in the I/R group, the mitochondria appeared small, irregular, and less dense cristae (c) in the Sev group, the severity of mitochondrial injury decreased compared with the I/R group (d) in the Sev+PIM-Inh group, the severity of mitochondrial injury increased compared with the Sev group (e) in the Sev+DMSO group, no significant changes were found compared with the Sev group (f) in the PIM-Inh group, no significant changes were found compared with the I/R group (n = 3 for each group, scale bar = 1 μm). The black arrow shows normal mitochondria, and the red arrow shows fragmented mitochondria. CON: Control group; I/R: Ischemia/reperfusion; Sev: Sevoflurane postconditioning; Sev+PIM-Inh: Sevoflurane postconditioning + Pim-1 inhibitor II; Sev+DMSO: Sevoflurane postconditioning + dimethyl sulfoxide; PIM-Inh: Pim-1 inhibitor II.

References

    1. Gong KZ, Zhang ZG, Li AH, Huang YF, Bu P, Dong F, et al. ROS-mediated ERK activation in delayed protection from anoxic preconditioning in neonatal rat cardiomyocytes. Chin Med J. 2004;117:395–400.
    1. Aldakkak M, Stowe DF, Chen Q, Lesnefsky EJ, Camara AK. Inhibited mitochondrial respiration by amobarbital during cardiac ischaemia improves redox state and reduces matrix Ca2+ overload and ROS release. Cardiovasc Res. 2008;77:406–15. doi: 10.1016/j.cardiores.2007.08.008.
    1. Gateau-Roesch O, Argaud L, Ovize M. Mitochondrial permeability transition pore and postconditioning. Cardiovasc Res. 2006;70:264–73. doi: 10.1016/j.cardiores.2006.02.024.
    1. Xu FF, Liu XH. Calreticulin translocation aggravates endoplasmic reticulum stress-associated apoptosis during cardiomyocyte hypoxia/reoxygenation. Chin Med J. 2015;128:353–60. doi: 10.4103/0366-6999.150103.
    1. Liesa M, Palacín M, Zorzano A. Mitochondrial dynamics in mammalian health and disease. Physiol Rev. 2009;89:799–845. doi: 10.1152/physrev.00030.2008.
    1. Hall AR, Burke N, Dongworth RK, Hausenloy DJ. Mitochondrial fusion and fission proteins: Novel therapeutic targets for combating cardiovascular disease. Br J Pharmacol. 2014;171:1890–906. doi: 10.1111/bph.12516.
    1. Disatnik MH, Ferreira JC, Campos JC, Gomes KS, Dourado PM, Qi X, et al. Acute inhibition of excessive mitochondrial fission after myocardial infarction prevents long-term cardiac dysfunction. J Am Heart Assoc. 2013;2:e000461. doi: 10.1161/JAHA.113.000461.
    1. Kamga Pride C, Mo L, Quesnelle K, Dagda RK, Murillo D, Geary L, et al. Nitrite activates protein kinase A in normoxia to mediate mitochondrial fusion and tolerance to ischaemia/reperfusion. Cardiovasc Res. 2014;101:57–68. doi: 10.1093/cvr/cvt224.
    1. Sharp WW. Dynamin-related protein 1 as a therapeutic target in cardiac arrest. J Mol Med (Berl) 2015;93:243–52. doi: 10.1007/s00109-015-1257-3.
    1. Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121:2012–22. doi: 10.1161/CIRCULATIONAHA.109.906610.
    1. Gao Y, Li T, Wu C, Bittle GJ, Chen S, Wu ZJ, et al. Pim-1 mediated signaling during the process of cardiac remodeling following myocardial infarction in ovine hearts. J Mol Cell Cardiol. 2013;63:89–97. doi: 10.1016/j.yjmcc.2013.07.012.
    1. Din S, Mason M, Völkers M, Johnson B, Cottage CT, Wang Z, et al. Pim-1 preserves mitochondrial morphology by inhibiting dynamin-related protein 1 translocation. Proc Natl Acad Sci U S A. 2013;110:5969–74. doi: 10.1073/pnas.1213294110.
    1. Chen HT, Yang CX, Li H, Zhang CJ, Wen XJ, Zhou J, et al. Cardioprotection of sevoflurane postconditioning by activating extracellular signal-regulated kinase 1/2 in isolated rat hearts. Acta Pharmacol Sin. 2008;29:931–41. doi: 10.1111/j.1745-7254.2008.00824.x.
    1. Gong JS, Yao YT, Fang NX, Huang J, Li LH. Sevoflurane postconditioning alleviates action potential duration shortening and L-type calcium current suppression induced by ischemia/reperfusion injury in rat epicardial myocytes. Chin Med J. 2012;125:3485–91. doi: 10.3760/cma.j.issn.0366-6999.2012.19.022.
    1. Liu JD, Deng Q, Tian HH, Pang YT, Deng GL. Wnt/Glycogen synthase kinase 3ß/ß-catenin signaling activation mediated sevoflurane preconditioning-induced cardioprotection. Chin Med J. 2015;128:2346–53. doi: 10.4103/0366-6999.163375.
    1. Apaijai N, Chinda K, Palee S, Chattipakorn S, Chattipakorn N. Combined vildagliptin and metformin exert better cardioprotection than monotherapy against ischemia-reperfusion injury in obese-insulin resistant rats. PLoS One. 2014;9:e102374. doi: 10.1371/journal.pone.0102374.
    1. Shao ZH, Sharp WW, Wojcik KR, Li CQ, Han M, Chang WT, et al. Therapeutic hypothermia cardioprotection via Akt- and nitric oxide-mediated attenuation of mitochondrial oxidants. Am J Physiol Heart Circ Physiol. 2010;298:H2164–73. doi: 10.1152/ajpheart.00994.2009.
    1. Allahyari S, Delazar A, Najafi M. Evaluation of general toxicity, anti-oxidant activity and effects of Ficus carica leaves extract on ischemia/reperfusion injuries in isolated heart of rat. Adv Pharm Bull. 2014;4(Suppl 2):577–82. doi: 10.5681/apb.2014.085.
    1. Marsboom G, Toth PT, Ryan JJ, Hong Z, Wu X, Fang YH, et al. Dynamin-related protein 1-mediated mitochondrial mitotic fission permits hyperproliferation of vascular smooth muscle cells and offers a novel therapeutic target in pulmonary hypertension. Circ Res. 2012;110:1484–97. doi: 10.1161/CIRCRESAHA.111.263848.
    1. Huhn R, Heinen A, Weber NC, Hollmann MW, Schlack W, Preckel B. Hyperglycaemia blocks sevoflurane-induced postconditioning in the rat heart in vivo: Cardioprotection can be restored by blocking the mitochondrial permeability transition pore. Br J Anaesth. 2008;100:465–71. doi: 10.1093/bja/aen022.
    1. Inamura Y, Miyamae M, Sugioka S, Kaneda K, Okusa C, Onishi A, et al. Aprotinin abolishes sevoflurane postconditioning by inhibiting nitric oxide production and phosphorylation of protein kinase C-delta and glycogen synthase kinase 3beta. Anesthesiology. 2009;111:1036–43. doi: 10.1097/ALN.0b013e3181bbbf9b.
    1. Yao YY, Zhu MH, Zhang FJ, Wen CY, Ma LL, Wang WN, et al. Activation of Akt and cardioprotection against reperfusion injury are maximal with only five minutes of sevoflurane postconditioning in isolated rat hearts. J Zhejiang Univ Sci B. 2013;14:511–7. doi: 10.1631/jzus.B1200195.
    1. Welchen E, Gonzalez DH. Cytochrome c, a hub linking energy, redox, stress and signaling pathways in mitochondria and other cell compartments. Physiol Plant. 2016;157:310–21. doi: 10.1111/ppl.12449.
    1. Stumpner J, Redel A, Kellermann A, Lotz CA, Blomeyer CA, Smul TM, et al. Differential role of Pim-1 kinase in anesthetic-induced and ischemic preconditioning against myocardial infarction. Anesthesiology. 2009;111:1257–64. doi: 10.1097/ALN.0b013e3181bdf9f4.
    1. Stumpner J, Smul TM, Redel A, Hilz T, Tischer-Zeitz T, Eisenbarth H, et al. Desflurane-induced and ischaemic postconditioning against myocardial infarction are mediated by Pim-1 kinase. Acta Anaesthesiol Scand. 2012;56:904–13. doi: 10.1111/j.1399-6576.2012.02657.x.
    1. Fischer KM, Cottage CT, Konstandin MH, Völkers M, Khan M, Sussman MA. Pim-1 kinase inhibits pathological injury by promoting cardioprotective signaling. J Mol Cell Cardiol. 2011;51:554–8. doi: 10.1016/j.yjmcc.2011.01.004.
    1. Gu X, Yao Y, Cheng R, Zhang Y, Dai Z, Wan G, et al. Plasminogen K5 activates mitochondrial apoptosis pathway in endothelial cells by regulating Bak and Bcl-x(L) subcellular distribution. Apoptosis. 2011;16:846–55. doi: 10.1007/s10495-011-0618-9.
    1. Aon MA, Cortassa S, Akar FG, O’Rourke B. Mitochondrial criticality: A new concept at the turning point of life or death. Biochim Biophys Acta. 2006;1762:232–40. doi: 10.1016/j.bbadis.2005.06.008.
    1. Hwang SJ, Kim W. Mitochondrial dynamics in the heart as a novel therapeutic target for cardioprotection. Chonnam Med J. 2013;49:101–7. doi: 10.4068/cmj.2013.49.3.101.
    1. Cellier L, Tamareille S, Kalakech H, Guillou S, Lenaers G, Prunier F, et al. Remote ischemic conditioning influences mitochondrial dynamics. Shock. 2016;45:192–7. doi: 10.1097/SHK.0000000000000500.
    1. Losón OC, Song Z, Chen H, Chan DC. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol Biol Cell. 2013;24:659–67. doi: 10.1091/mbc.E12-10-0721.
    1. Palmer CS, Elgass KD, Parton RG, Osellame LD, Stojanovski D, Ryan MT. Adaptor proteins MiD49 and MiD51 can act independently of Mff and Fis1 in Drp1 recruitment and are specific for mitochondrial fission. J Biol Chem. 2013;288:27584–93. doi: 10.1074/jbc.M113.479873.
    1. Otera H, Miyata N, Kuge O, Mihara K. Drp1-dependent mitochondrial fission via MiD49/51 is essential for apoptotic cristae remodeling. J Cell Biol. 2016;212:531–44. doi: 10.1083/jcb.201508099.
    1. Zepeda R, Kuzmicic J, Parra V, Troncoso R, Pennanen C, Riquelme JA, et al. Drp1 loss-of-function reduces cardiomyocyte oxygen dependence protecting the heart from ischemia-reperfusion injury. J Cardiovasc Pharmacol. 2014;63:477–87. doi: 10.1097/FJC.0000000000000071.
    1. Santel A, Frank S. Shaping mitochondria: The complex posttranslational regulation of the mitochondrial fission protein DRP1. IUBMB Life. 2008;60:448–55. doi: 10.1002/iub.71.
    1. Chang CR, Blackstone C. Dynamic regulation of mitochondrial fission through modification of the dynamin-related protein Drp1. Ann N Y Acad Sci. 2010;1201:34–9. doi: 10.1111/j.1749-6632.2010.05629.x.
    1. Sharp WW, Fang YH, Han M, Zhang HJ, Hong Z, Banathy A, et al. Dynamin-related protein 1 (Drp1)-mediated diastolic dysfunction in myocardial ischemia-reperfusion injury: Therapeutic benefits of Drp1 inhibition to reduce mitochondrial fission. FASEB J. 2014;28:316–26. doi: 10.1096/fj.12-226225.
    1. Karbowski M, Lee YJ, Gaume B, Jeong SY, Frank S, Nechushtan A, et al. Spatial and temporal association of Bax with mitochondrial fission sites, Drp1, and Mfn2 during apoptosis. J Cell Biol. 2002;159:931–8. doi: 10.1083/jcb.200209124.
    1. Arnoult D, Rismanchi N, Grodet A, Roberts RG, Seeburg DP, Estaquier J, et al. Bax/Bak-dependent release of DDP/TIMM8a promotes Drp1-mediated mitochondrial fission and mitoptosis during programmed cell death. Curr Biol. 2005;15:2112–8. doi: 10.1016/j.cub.2005.10.041.

Source: PubMed

3
Subskrybuj