Skip to main content
Erschienen in: Cardiovascular Drugs and Therapy 1/2019

24.01.2019 | ORIGINAL ARTICLE

Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation

verfasst von: Jingyi Chen, Zhenzhen Jiang, Xing Zhou, Xingxing Sun, Jianwei Cao, Yongpan Liu, Xianyu Wang

Erschienen in: Cardiovascular Drugs and Therapy | Ausgabe 1/2019

Einloggen, um Zugang zu erhalten

Abstract

Myocardial ischemia/reperfusion (I/R) injury is a serious threat to the health of people around the world. Recent evidence has indicated that high-mobility group box-1 (HMGB1) is involved in I/R-induced inflammation, and inflammation can cause necroptosis of cells. Interestingly, dexmedetomidine (DEX) has anti-inflammatory properties. Therefore, we speculated that DEX preconditioning may suppress H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. We found that hypoxia/reoxygenation (H/R) significantly increased cellular damage, as measured by cell viability (100 ± 3.26% vs. 53.33 ± 3.29, p < 0.01), CK-MB (1 vs. 3.25 ± 0.26, p < 0.01), cTnI (1 vs. 2.69 ± 0.31, p < 0.01), inflammation as indicated by TNF-α (1 ± 0.09 vs. 2.57 ± 0.12, p < 0.01), IL-1β (1 ± 0.33 vs. 3.87 ± 0.41, p < 0.01) and IL-6 (1 ± 0.36 vs. 3.60 ± 0.45, p < 0.01), and necroptosis, which were accompanied by significantly increased protein levels of HMGB1. These changes [cellular damage as measured by cell viability (53.33 ± 3.29% vs. 67.59 ± 2.69%, p < 0.01), CK-MB (3.25 ± 0.26 vs. 2.27 ± 0.22, p < 0.01), cTnI (2.69 ± 0.31 vs. 1.90 ± 0.25, p < 0.01), inflammation as indicated by TNF-α (2.57 ± 0.12 vs. 1.75 ± 0.15, p < 0.01), IL-1β (3.87 ± 0.41 vs. 2.09 ± 0.36, p < 0.01) and IL-6 (3.60 ± 0.45 vs. 2.21 ± 0.39, p < 0.01), and necroptosis proteins] were inhibited by DEX preconditioning. We also found that silencing expression of HMGB1 reinforced the protective effects of DEX preconditioning and overexpression of HMGB1 counteracted the protective effects of DEX preconditioning. Thus, we concluded that DEX preconditioning inhibits H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes.
Literatur
1.
Zurück zum Zitat Koeppen M, Lee JW, Seo SW, Brodsky KS, Kreth S, Yang IV, et al. Hypoxia-inducible factor 2-alpha-dependent induction of amphiregulin dampens myocardial ischemia-reperfusion injury. Nat Commun. 2018;9:816.CrossRefPubMedPubMedCentral Koeppen M, Lee JW, Seo SW, Brodsky KS, Kreth S, Yang IV, et al. Hypoxia-inducible factor 2-alpha-dependent induction of amphiregulin dampens myocardial ischemia-reperfusion injury. Nat Commun. 2018;9:816.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Kawai H, Chaudhry F, Shekhar A, Petrov A, Nakahara T, Tanimoto T, Kim D, Chen J, Lebeche D, Blankenberg FG, et al. Molecular imaging of apoptosis in ischemia reperfusion injury with radiolabeled Duramycin targeting phosphatidylethanolamine: Effective Target Uptake and Reduced Nontarget Organ Radiation Burden. JACC Cardiovasc Imaging. 2018. Kawai H, Chaudhry F, Shekhar A, Petrov A, Nakahara T, Tanimoto T, Kim D, Chen J, Lebeche D, Blankenberg FG, et al. Molecular imaging of apoptosis in ischemia reperfusion injury with radiolabeled Duramycin targeting phosphatidylethanolamine: Effective Target Uptake and Reduced Nontarget Organ Radiation Burden. JACC Cardiovasc Imaging. 2018.
3.
Zurück zum Zitat Ferrari RS, Andrade CF. Oxidative stress and lung ischemia-reperfusion injury. Oxidative Med Cell Longev. 2015;2015:590987.CrossRef Ferrari RS, Andrade CF. Oxidative stress and lung ischemia-reperfusion injury. Oxidative Med Cell Longev. 2015;2015:590987.CrossRef
4.
Zurück zum Zitat Chorawala MR, Prakash P, Doddapattar P, Jain M, Dhanesha N, Chauhan AK. Deletion of extra domain a of fibronectin reduces acute myocardial ischaemia/reperfusion injury in hyperlipidaemic mice by limiting thrombo-inflammation. Thromb Haemost. 2018;118:1450–60.CrossRefPubMedPubMedCentral Chorawala MR, Prakash P, Doddapattar P, Jain M, Dhanesha N, Chauhan AK. Deletion of extra domain a of fibronectin reduces acute myocardial ischaemia/reperfusion injury in hyperlipidaemic mice by limiting thrombo-inflammation. Thromb Haemost. 2018;118:1450–60.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Roberta A, Vicentino R, Carneiro VC, Carneiro VC, Allonso D, Guilherme R, et al. Emerging role of HMGB1 in the pathogenesis of schistosomiasis liver fibrosis. Front Immunol. 2018;9:1979.CrossRef Roberta A, Vicentino R, Carneiro VC, Carneiro VC, Allonso D, Guilherme R, et al. Emerging role of HMGB1 in the pathogenesis of schistosomiasis liver fibrosis. Front Immunol. 2018;9:1979.CrossRef
6.
Zurück zum Zitat Sekiguchi F, Domoto R, Nakashima K, Yamasoba D, Yamanishi H, Tsubota M, et al. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: evidence for a neuroimmune crosstalk. Neuropharmacology. 2018;141:201–13.CrossRefPubMed Sekiguchi F, Domoto R, Nakashima K, Yamasoba D, Yamanishi H, Tsubota M, et al. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: evidence for a neuroimmune crosstalk. Neuropharmacology. 2018;141:201–13.CrossRefPubMed
7.
Zurück zum Zitat Loukili N, Rosenblatt-Velin N, Li J, Clerc S, Pacher P, Feihl F, et al. Peroxynitrite induces HMGB1 release by cardiac cells in vitro and HMGB1 upregulation in the infarcted myocardium in vivo. Cardiovasc Res. 2011;89:586–94.CrossRefPubMed Loukili N, Rosenblatt-Velin N, Li J, Clerc S, Pacher P, Feihl F, et al. Peroxynitrite induces HMGB1 release by cardiac cells in vitro and HMGB1 upregulation in the infarcted myocardium in vivo. Cardiovasc Res. 2011;89:586–94.CrossRefPubMed
8.
Zurück zum Zitat Andrassy M, Volz HC, Igwe JC, Funke B, Eichberger SN, Kaya Z, et al. High-mobility group Box-1 in ischemia-reperfusion injury of the heart. Circulation. 2008;117:3216–26.CrossRefPubMed Andrassy M, Volz HC, Igwe JC, Funke B, Eichberger SN, Kaya Z, et al. High-mobility group Box-1 in ischemia-reperfusion injury of the heart. Circulation. 2008;117:3216–26.CrossRefPubMed
9.
Zurück zum Zitat In EJ, Lee Y, Koppula S, Kim TY, Han JH, Lee KH, et al. Identification and characterization of NTB451 as a potential inhibitor of necroptosis. Molecules. 2018;23. In EJ, Lee Y, Koppula S, Kim TY, Han JH, Lee KH, et al. Identification and characterization of NTB451 as a potential inhibitor of necroptosis. Molecules. 2018;23.
10.
Zurück zum Zitat Xu Z, Jin Y, Yan H, Gao Z, Xu B, Yang B, et al. High-mobility group box 1 protein-mediated necroptosis contributes to dasatinib-induced cardiotoxicity. Toxicol Lett. 2018;296:39–47.CrossRefPubMed Xu Z, Jin Y, Yan H, Gao Z, Xu B, Yang B, et al. High-mobility group box 1 protein-mediated necroptosis contributes to dasatinib-induced cardiotoxicity. Toxicol Lett. 2018;296:39–47.CrossRefPubMed
11.
Zurück zum Zitat Galluzzi L, Kepp O, Chan FK, Kroemer G. Necroptosis: mechanisms and relevance to disease. Annu Rev Pathol. 2017;12:103–30.CrossRefPubMed Galluzzi L, Kepp O, Chan FK, Kroemer G. Necroptosis: mechanisms and relevance to disease. Annu Rev Pathol. 2017;12:103–30.CrossRefPubMed
12.
Zurück zum Zitat Abdel-Ghaffar HS, Kamal SM, El Sherif FA, Mohamed SA. Comparison of nebulised dexmedetomidine, ketamine, or midazolam for premedication in preschool children undergoing bone marrow biopsy. Br J Anaesth. 2018;121:445–52.CrossRefPubMed Abdel-Ghaffar HS, Kamal SM, El Sherif FA, Mohamed SA. Comparison of nebulised dexmedetomidine, ketamine, or midazolam for premedication in preschool children undergoing bone marrow biopsy. Br J Anaesth. 2018;121:445–52.CrossRefPubMed
13.
Zurück zum Zitat Peng K, Qiu Y, Li J, Zhang ZC, Ji FH. Dexmedetomidine attenuates hypoxia/reoxygenation injury in primary neonatal rat cardiomyocytes. Exp Ther Med. 2017;14:689–95.CrossRefPubMedPubMedCentral Peng K, Qiu Y, Li J, Zhang ZC, Ji FH. Dexmedetomidine attenuates hypoxia/reoxygenation injury in primary neonatal rat cardiomyocytes. Exp Ther Med. 2017;14:689–95.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Zhang JJ, Peng K, Zhang J, Meng XW, Ji FH. Dexmedetomidine preconditioning may attenuate myocardial ischemia/reperfusion injury by down-regulating the HMGB1-TLR4-MyD88-NF-small ka, CyrillicB signaling pathway. PLoS One. 2017;12:e0172006.CrossRefPubMedPubMedCentral Zhang JJ, Peng K, Zhang J, Meng XW, Ji FH. Dexmedetomidine preconditioning may attenuate myocardial ischemia/reperfusion injury by down-regulating the HMGB1-TLR4-MyD88-NF-small ka, CyrillicB signaling pathway. PLoS One. 2017;12:e0172006.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Meng L, Li L, Lu S, Li K, Su Z, Wang Y, et al. The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-kappaB and PI3K/Akt/mTOR pathways. Mol Immunol. 2018;94:7–17.CrossRefPubMed Meng L, Li L, Lu S, Li K, Su Z, Wang Y, et al. The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-kappaB and PI3K/Akt/mTOR pathways. Mol Immunol. 2018;94:7–17.CrossRefPubMed
16.
Zurück zum Zitat Deng F, Wang S, Cai S, Hu Z, Xu R, Wang J, et al. Inhibition of caveolae contributes to propofol preconditioning-suppressed microvesicles release and cell injury by hypoxia-reoxygenation. Oxidative Med Cell Longev. 2017;2017:3542149.CrossRef Deng F, Wang S, Cai S, Hu Z, Xu R, Wang J, et al. Inhibition of caveolae contributes to propofol preconditioning-suppressed microvesicles release and cell injury by hypoxia-reoxygenation. Oxidative Med Cell Longev. 2017;2017:3542149.CrossRef
17.
Zurück zum Zitat Deng F, Wang S, Zhang L, Xie X, Cai S, Li H, et al. Propofol through upregulating caveolin-3 attenuates post-hypoxic mitochondrial damage and cell death in H9C2 cardiomyocytes during hyperglycemia. Cell Physiol Biochem. 2017;44:279–92.CrossRefPubMed Deng F, Wang S, Zhang L, Xie X, Cai S, Li H, et al. Propofol through upregulating caveolin-3 attenuates post-hypoxic mitochondrial damage and cell death in H9C2 cardiomyocytes during hyperglycemia. Cell Physiol Biochem. 2017;44:279–92.CrossRefPubMed
18.
Zurück zum Zitat Muller T, Dewitz C, Schmitz J, Schroder AS, Brasen JH, Stockwell BR, et al. Necroptosis and ferroptosis are alternative cell death pathways that operate in acute kidney failure. Cell Mol Life Sci. 2017;74:3631–45.CrossRefPubMedPubMedCentral Muller T, Dewitz C, Schmitz J, Schroder AS, Brasen JH, Stockwell BR, et al. Necroptosis and ferroptosis are alternative cell death pathways that operate in acute kidney failure. Cell Mol Life Sci. 2017;74:3631–45.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat McDonald KA, Huang H, Tohme S, Loughran P, Ferrero K, Billiar T, et al. Toll-like receptor 4 (TLR4) antagonist eritoran tetrasodium attenuates liver ischemia and reperfusion injury through inhibition of high-mobility group box protein B1 (HMGB1) signaling. Mol Med. 2015;20:639–48.CrossRefPubMedPubMedCentral McDonald KA, Huang H, Tohme S, Loughran P, Ferrero K, Billiar T, et al. Toll-like receptor 4 (TLR4) antagonist eritoran tetrasodium attenuates liver ischemia and reperfusion injury through inhibition of high-mobility group box protein B1 (HMGB1) signaling. Mol Med. 2015;20:639–48.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Mersmann J, Iskandar F, Latsch K, Habeck K, Sprunck V, Zimmermann R, et al. Attenuation of myocardial injury by HMGB1 blockade during ischemia/reperfusion is toll-like receptor 2-dependent. Mediat Inflamm. 2013;2013:174168.CrossRef Mersmann J, Iskandar F, Latsch K, Habeck K, Sprunck V, Zimmermann R, et al. Attenuation of myocardial injury by HMGB1 blockade during ischemia/reperfusion is toll-like receptor 2-dependent. Mediat Inflamm. 2013;2013:174168.CrossRef
21.
Zurück zum Zitat Tong S, Zhang L, Joseph J, Jiang X. Celastrol pretreatment attenuates rat myocardial ischemia/ reperfusion injury by inhibiting high mobility group box 1 protein expression via the PI3K/Akt pathway. Biochem Biophys Res Commun. 2018;497:843–9.CrossRefPubMed Tong S, Zhang L, Joseph J, Jiang X. Celastrol pretreatment attenuates rat myocardial ischemia/ reperfusion injury by inhibiting high mobility group box 1 protein expression via the PI3K/Akt pathway. Biochem Biophys Res Commun. 2018;497:843–9.CrossRefPubMed
22.
Zurück zum Zitat Sun HJ, Lu Y, Wang HW, Zhang H, Wang SR, Xu WY, et al. Activation of endocannabinoid receptor 2 as a mechanism of propofol pretreatment-induced cardioprotection against ischemia-reperfusion injury in rats. Oxidative Med Cell Longev. 2017;2017:2186383. Sun HJ, Lu Y, Wang HW, Zhang H, Wang SR, Xu WY, et al. Activation of endocannabinoid receptor 2 as a mechanism of propofol pretreatment-induced cardioprotection against ischemia-reperfusion injury in rats. Oxidative Med Cell Longev. 2017;2017:2186383.
23.
Zurück zum Zitat Sun Y, Jiang C, Jiang J, Qiu L. Dexmedetomidine protects mice against myocardium ischaemic/reperfusion injury by activating an AMPK/PI3K/Akt/eNOS pathway. Clin Exp Pharmacol Physiol. 2017;44:946–53.CrossRefPubMed Sun Y, Jiang C, Jiang J, Qiu L. Dexmedetomidine protects mice against myocardium ischaemic/reperfusion injury by activating an AMPK/PI3K/Akt/eNOS pathway. Clin Exp Pharmacol Physiol. 2017;44:946–53.CrossRefPubMed
24.
Zurück zum Zitat Chen Z, Ding T, Ma CG. Dexmedetomidine (DEX) protects against hepatic ischemia/reperfusion (I/R) injury by suppressing inflammation and oxidative stress in NLRC5 deficient mice. Biochem Biophys Res Commun. 2017;493:1143–50.CrossRefPubMed Chen Z, Ding T, Ma CG. Dexmedetomidine (DEX) protects against hepatic ischemia/reperfusion (I/R) injury by suppressing inflammation and oxidative stress in NLRC5 deficient mice. Biochem Biophys Res Commun. 2017;493:1143–50.CrossRefPubMed
25.
26.
Zurück zum Zitat Frank D, Vince JE. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 2018. Frank D, Vince JE. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 2018.
Metadaten
Titel
Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation
verfasst von
Jingyi Chen
Zhenzhen Jiang
Xing Zhou
Xingxing Sun
Jianwei Cao
Yongpan Liu
Xianyu Wang
Publikationsdatum
24.01.2019
Verlag
Springer US
Erschienen in
Cardiovascular Drugs and Therapy / Ausgabe 1/2019
Print ISSN: 0920-3206
Elektronische ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-019-06857-1

Weitere Artikel der Ausgabe 1/2019

Cardiovascular Drugs and Therapy 1/2019 Zur Ausgabe

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Shunt-Therapie bei Herzinsuffizienz: Kein Anzug, der allen passt

13.05.2024 Chronische Herzinsuffizienz Nachrichten

Die Anlage eines interatrialen Shunts zur Reduktion des linksatrialen Drucks ist ein neuer Therapieansatz bei Herzinsuffizienz. Viele Patienten sprechen darauf an, andere jedoch nicht. 

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.