Skip to main content

Advertisement

Log in

Inhibition of RIP1-dependent necrosis prevents adverse cardiac remodeling after myocardial ischemia–reperfusion in vivo

  • Original Contribution
  • Published:
Basic Research in Cardiology Aims and scope Submit manuscript

Abstract

Accumulating evidence indicatesthat programmed necrosis plays a critical role in cell death during ischemia–reperfusion. Necrostatin-1 (Nec-1), a small molecule capable of inhibiting a key regulator of programmed necrosis (RIP1), was shown to prevent necrotic cell death in experimental models including cardiac ischemia. However, no functional follow-up was performed and the action of Nec-1 remains unclear. Here, we studied whether Nec-1 inhibits RIP1-dependent necrosis and leads to long-term improvements after ischemia–reperfusion in vivo. Mice underwent 30 min of ischemia and received, 5 min before reperfusion, 3.3 mg/kg Nec-1 or vehicle treatment, followed by reperfusion. Nec-1 administration reduced infarct size to 26.3 ± 1.3 % (P = 0.001) compared to 38.6 ± 1.7 % in vehicle-treated animals. Furthermore, Nec-1 inhibited RIP1/RIP3 phosphorylation in vivo and significantly reduced necrotic cell death, while apoptotic cell death remained constant. By using MRI, cardiac dimensions and function were assessed before and 28 days after surgery. Nec-1-treated mice displayed less adverse remodeling (end-diastolic volume 63.5 ± 2.8 vs. 74.9 ± 2.8 μl, P = 0.031) and preserved cardiac performance (ejection fraction 45.81 ± 2.05 vs. 36.03 ± 2.37 %, P = 0.016). Nec-1 treatment significantly reduced inflammatory influx, tumor necrosis factor-α mRNA levels and oxidative stress levels. Interestingly, this was accompanied by significant changes in the expression signature of oxidative stress genes. Administration of Nec-1 at the onset of reperfusion inhibits RIP1-dependent necrosis in vivo, leading to infarct size reduction and preservation of cardiac function. The cardioprotective effect of Nec-1 highlights the importance of necrotic cell death in the ischemic heart, thereby opening a new direction for therapy in patients with myocardial infarction.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Ardanaz N, Yang XP, Cifuentes ME, Haurani MJ, Jackson KW, Liao TD, Carretero OA, Pagano PJ (2010) Lack of glutathione peroxidase 1 accelerates cardiac-specific hypertrophy and dysfunction in angiotensin II hypertension. Hypertension 55:116–123. doi:10.1161/HYPERTENSIONAHA.109.135715

    Article  PubMed  CAS  Google Scholar 

  2. Arslan F, Smeets MB, O’Neill LA, Keogh B, McGuirk P, Timmers L, Tersteeg C, Hoefer IE, Doevendans PA, Pasterkamp G, de Kleijn DP (2010) Myocardial ischemia/reperfusion injury is mediated by leukocytic toll-like receptor-2 and reduced by systemic administration of a novel anti-toll-like receptor-2 antibody. Circulation 121:80–90. doi:10.1161/CIRCULATIONAHA.109.880187

    Article  PubMed  CAS  Google Scholar 

  3. Baines CP (2009) The mitochondrial permeability transition pore and ischemia–reperfusion injury. Basic Res Cardiol 104:181–188. doi:10.1007/s00395-009-0004-8

    Article  PubMed  CAS  Google Scholar 

  4. Borchi E, Parri M, Papucci L, Becatti M, Nassi N, Nassi P, Nediani C (2009) Role of NADPH oxidase in H9c2 cardiac muscle cells exposed to simulated ischaemia–reperfusion. J Cell Mol Med 13:2724–2735. doi:10.1111/j.1582-4934.2008.00485.x

    Article  PubMed  Google Scholar 

  5. Cao C, Huang X, Han Y, Wan Y, Birnbaumer L, Feng GS, Marshall J, Jiang M, Chu WM (2009) Galpha(i1) and Galpha(i3) are required for epidermal growth factor-mediated activation of the Akt-mTORC1 pathway. Sci Signal 2:ra17. doi:10.1126/scisignal.2000118

    Article  PubMed  Google Scholar 

  6. Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M, Chan FK (2009) Phosphorylation-driven assembly of the RIP1–RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137:1112–1123. doi:10.1016/j.cell.2009.05.037

    Article  PubMed  CAS  Google Scholar 

  7. Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J (2008) Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4:313–321. doi:10.1038/nchembio.83

    Article  PubMed  CAS  Google Scholar 

  8. Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J (2005) Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1:112–119. doi:10.1038/nchembio711

    Article  PubMed  CAS  Google Scholar 

  9. Gottlieb RA (2003) Mitochondrial signaling in apoptosis: mitochondrial daggers to the breaking heart. Basic Res Cardiol 98:242–249. doi:10.1007/s00395-003-0404-0

    PubMed  CAS  Google Scholar 

  10. Hausenloy DJ, Baxter G, Bell R, Botker HE, Davidson SM, Downey J, Heusch G, Kitakaze M, Lecour S, Mentzer R, Mocanu MM, Ovize M, Schulz R, Shannon R, Walker M, Walkinshaw G, Yellon DM (2010) Translating novel strategies for cardioprotection: the Hatter Workshop Recommendations. Basic Res Cardiol 105:677–686. doi:10.1007/s00395-010-0121-4

    Article  PubMed  Google Scholar 

  11. Hausenloy DJ, Ong SB, Yellon DM (2009) The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol 104:189–202. doi:10.1007/s00395-009-0010-x

    Article  PubMed  CAS  Google Scholar 

  12. Hausenloy DJ, Yellon DM (2003) The mitochondrial permeability transition pore: its fundamental role in mediating cell death during ischaemia and reperfusion. J Mol Cell Cardiol 35:339–341. doi:10.1016/S0022-2828(03)00043-9

    Article  PubMed  CAS  Google Scholar 

  13. He S, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X (2009) Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 137:1100–1111. doi:10.1016/j.cell.2009.05.021

    Article  PubMed  CAS  Google Scholar 

  14. Heusch G, Boengler K, Schulz R (2008) Cardioprotection: nitric oxide, protein kinases, and mitochondria. Circulation 118:1915–1919. doi:10.1161/CIRCULATIONAHA.108.805242

    Article  PubMed  Google Scholar 

  15. Heusch G, Boengler K, Schulz R (2010) Inhibition of mitochondrial permeability transition pore opening: the Holy Grail of cardioprotection. Basic Res Cardiol 105:151–154. doi:10.1007/s00395-009-0080-9

    Article  PubMed  Google Scholar 

  16. Heusch G, Kleinbongard P, Bose D, Levkau B, Haude M, Schulz R, Erbel R (2009) Coronary microembolization: from bedside to bench and back to bedside. Circulation 120:1822–1836. doi:10.1161/CIRCULATIONAHA.109.888784

    Article  PubMed  Google Scholar 

  17. Heymes C, Bendall JK, Ratajczak P, Cave AC, Samuel JL, Hasenfuss G, Shah AM (2003) Increased myocardial NADPH oxidase activity in human heart failure. J Am Coll Cardiol 41:2164–2171. doi:10.1016/S0735-1097(03)00471-6

    Article  PubMed  CAS  Google Scholar 

  18. Inserte J, Molla B, Aguilar R, Través PG, Barba I, Martín-Sanz P, Boscá L, Casado M, Garcia-Dorado D (2009) Constitutive COX-2 activity in cardiomyocytes confers permanent cardioprotection: Constitutive COX-2 expression and cardioprotection. J Mol Cell Cardiol 46:160–168. doi:10.1016/j.yjmcc.2008.11.011

    Article  PubMed  CAS  Google Scholar 

  19. Kleinbongard P, Heusch G, Schulz R (2010) TNFalpha in atherosclerosis, myocardial ischemia/reperfusion and heart failure. Pharmacol Ther 127:295–314. doi:10.1016/j.pharmthera.2010.05.002

    Article  PubMed  CAS  Google Scholar 

  20. Kung G, Konstantinidis K, Kitsis RN (2011) Programmed necrosis, not apoptosis, in the heart. Circ Res 108:1017–1036. doi:10.1161/CIRCRESAHA.110.225730

    Article  PubMed  CAS  Google Scholar 

  21. Li C, Jackson RM (2002) Reactive species mechanisms of cellular hypoxia–reoxygenation injury. Am J Physiol Cell Physiol 282:C227–C241. doi:10.1152/ajpcell.00112.2001

    PubMed  CAS  Google Scholar 

  22. Li Q, Guo Y, Tan W, Ou Q, Wu WJ, Sturza D, Dawn B, Hunt G, Cui C, Bolli R (2007) Cardioprotection afforded by inducible nitric oxide synthase gene therapy is mediated by cyclooxygenase-2 via a nuclear factor-kappaB dependent pathway. Circulation 116:1577–1584. doi:10.1161/CIRCULATIONAHA.107.689810

    Article  PubMed  CAS  Google Scholar 

  23. Lim SY, Davidson SM, Mocanu MM, Yellon DM, Smith CCT (2007) The cardioprotective effect of necrostatin requires the cyclophilin-D component of the mitochondrial permeability transition pore. Cardiovasc Drugs Ther 21:467–469. doi:10.1007/s10557-007-6067-6

    Article  PubMed  CAS  Google Scholar 

  24. Liu J, van Mil A, Vrijsen K, Zhao J, Gao L, Metz CH, Goumans MJ, Doevendans PA, Sluijter JP (2011) MicroRNA-155 prevents necrotic cell death in human cardiomyocyte progenitor cells via targeting RIP1. J Cell Mol Med 15:1474–1482. doi:10.1111/j.1582-4934.2010.01104.x

    Article  PubMed  CAS  Google Scholar 

  25. Liu X, Gai Y, Liu F, Gao W, Zhang Y, Xu M, Li Z (2010) Trimetazidine inhibits pressure overload-induced cardiac fibrosis through NADPH oxidase-ROS-CTGF pathway. Cardiovasc Res 88:150–158. doi:10.1093/cvr/cvq181

    Article  PubMed  CAS  Google Scholar 

  26. Lloyd-Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, De SG, Ferguson TB, Ford E, Furie K, Gillespie C, Go A, Greenlund K, Haase N, Hailpern S, Ho PM, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott MM, Meigs J, Mozaffarian D, Mussolino M, Nichol G, Roger VL, Rosamond W, Sacco R, Sorlie P, Roger VL, Thom T, Wasserthiel-Smoller S, Wong ND, Wylie-Rosett J (2010) Heart disease and stroke statistics—2010 update: a report from the American Heart Association. Circulation 121:e46–e215. doi:10.1161/CIRCULATIONAHA.109.192667

    Article  PubMed  Google Scholar 

  27. Morgan MJ, Kim YS, Liu ZG (2008) TNFalpha and reactive oxygen species in necrotic cell death. Cell Res 18:343–349. doi:10.1038/cr.2008.31

    Article  PubMed  CAS  Google Scholar 

  28. Nakaoka Y, Nishida K, Narimatsu M, Kamiya A, Minami T, Sawa H, Okawa K, Fujio Y, Koyama T, Maeda M, Sone M, Yamasaki S, Arai Y, Koh GY, Kodama T, Hirota H, Otsu K, Hirano T, Mochizuki N (2007) Gab family proteins are essential for postnatal maintenance of cardiac function via neuregulin-1/ErbB signaling. J Clin Invest 117:1771–1781. doi:10.1172/JCI30651

    Article  PubMed  CAS  Google Scholar 

  29. Nishida H, Matsumoto A, Tomono N, Hanakai T, Harada S, Nakaya H (2010) Biochemistry and physiology of mitochondrial ion channels involved in cardioprotection. FEBS Lett 584:2161–2166. doi:10.1016/j.febslet.2009.12.033

    Article  PubMed  CAS  Google Scholar 

  30. Noort WA, Oerlemans MI, Rozemuller H, Feyen D, Jaksani S, Stecher D, Naaijkens B, Martens AC, Buhring HJ, Doevendans PA, Sluijter JP (2011) Human versus porcine mesenchymal stromal cells: phenotype, differentiation potential, immunomodulation and cardiac improvement after transplantation. J Cell Mol Med. doi:10.1111/j.1582-4934.2011.01455.x

    PubMed  Google Scholar 

  31. Northington FJ, Chavez-Valdez R, Graham EM, Razdan S, Gauda EB, Martin LJ (2011) Necrostatin decreases oxidative damage, inflammation, and injury after neonatal HI. J Cereb Blood Flow Metab 31:178–189. doi:10.1038/jcbfm.2010.72

    Article  PubMed  CAS  Google Scholar 

  32. Oerlemans MI, Goumans MJ, van Middelaar B, Clevers H, Doevendans PA, Sluijter JP (2010) Active Wnt signaling in response to cardiac injury. Basic Res Cardiol 105:631–641. doi:10.1007/s00395-010-0100-9

    Article  PubMed  CAS  Google Scholar 

  33. Oerlemans MI, Koudstaal S, Chamuleau SA, de Kleijn DP, Doevendans PA, Sluijter JP (2012) Targeting cell death in the reperfused heart: Pharmacological approaches for cardioprotection. Int J Cardiol. doi:10.1016/j.ijcard.2012.03.055

    PubMed  Google Scholar 

  34. Prasad A, Stone GW, Holmes DR, Gersh B (2009) Reperfusion injury, microvascular dysfunction, and cardioprotection: the “dark side” of reperfusion. Circulation 120:2105–2112. doi:10.1161/CIRCULATIONAHA.108.814640

    Article  PubMed  Google Scholar 

  35. Shiomi T, Tsutsui H, Matsusaka H, Murakami K, Hayashidani S, Ikeuchi M, Wen J, Kubota T, Utsumi H, Takeshita A (2004) Overexpression of glutathione peroxidase prevents left ventricular remodeling and failure after myocardial infarction in mice. Circulation 109:544–549. doi:10.1161/01.CIR.0000109701.77059.E9

    Article  PubMed  CAS  Google Scholar 

  36. Smith CC, Davidson SM, Lim SY, Simpkin JC, Hothersall JS, Yellon DM (2007) Necrostatin: a potentially novel cardioprotective agent? Cardiovasc Drugs Ther 21:227–233. doi:10.1007/s10557-007-6035-1

    Article  PubMed  CAS  Google Scholar 

  37. Steenbergen C, Das S, Su J, Wong R, Murphy E (2009) Cardioprotection and altered mitochondrial adenine nucleotide transport. Basic Res Cardiol 104:149–156. doi:10.1007/s00395-009-0002-x

    Article  PubMed  CAS  Google Scholar 

  38. Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X, Wang X (2012) Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148:213–227. doi:10.1016/j.cell.2011.11.031

    Article  PubMed  CAS  Google Scholar 

  39. Temkin V, Huang Q, Liu H, Osada H, Pope RM (2006) Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol Cell Biol 26:2215–2225

    Article  PubMed  CAS  Google Scholar 

  40. Turrens JF (2003) Mitochondrial formation of reactive oxygen species. J Physiol 552:335–344. doi:10.1113/jphysiol.2003.049478

    Article  PubMed  CAS  Google Scholar 

  41. Vandenabeele P, Declercq W, Van Herreweghe F, Vanden Berghe T (2010) The role of the kinases RIP1 and RIP3 in TNF-induced necrosis. Sci Signal 3:re4. doi:10.1126/scisignal.3115re4

    Article  PubMed  Google Scholar 

  42. Vandenabeele P, Galluzzi L, Vanden BT, Kroemer G (2010) Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11:700–714. doi:10.1038/nrm2970

    Article  PubMed  CAS  Google Scholar 

  43. Vinten-Johansen J (2004) Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc Res 61:481–497. doi:10.1016/j.cardiores.2003.10.011

    Article  PubMed  CAS  Google Scholar 

  44. Whelan RS, Kaplinskiy V, Kitsis RN (2010) Cell death in the pathogenesis of heart disease: mechanisms and significance. Annu Rev Physiol 72:19–44. doi:10.1146/annurev.physiol.010908.163111

    Article  PubMed  CAS  Google Scholar 

  45. Xiang G, Seki T, Schuster MD, Witkowski P, Boyle AJ, See F, Martens TP, Kocher A, Sondermeijer H, Krum H, Itescu S (2005) Catalytic degradation of vitamin D up-regulated protein 1 mRNA enhances cardiomyocyte survival and prevents left ventricular remodeling after myocardial ischemia. J Biol Chem 280:39394–39402. doi:10.1074/jbc.M502966200

    Article  PubMed  CAS  Google Scholar 

  46. Yellon DM, Hausenloy DJ (2007) Myocardial reperfusion injury. N Engl J Med 357:1121–1135. doi:10.1056/NEJMra071667

    Article  PubMed  CAS  Google Scholar 

  47. Yoshioka J, Imahashi K, Gabel SA, Chutkow WA, Burds AA, Gannon J, Schulze PC, MacGillivray C, London RE, Murphy E, Lee RT (2007) Targeted deletion of thioredoxin-interacting protein regulates cardiac dysfunction in response to pressure overload. Circ Res 101:1328–1338. doi:10.1161/CIRCRESAHA.106.160515

    Article  PubMed  CAS  Google Scholar 

  48. You Z, Savitz SI, Yang J, Degterev A, Yuan J, Cuny GD, Moskowitz MA, Whalen MJ (2008) Necrostatin-1 reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 28:1564–1573. doi:10.1038/jcbfm.2008.44

    Article  PubMed  CAS  Google Scholar 

  49. Zhang C, Wu J, Xu X, Potter BJ, Gao X (2010) Direct relationship between levels of TNF-alpha expression and endothelial dysfunction in reperfusion injury. Basic Res Cardiol 105:453–464. doi:10.1007/s00395-010-0083-6

    Article  PubMed  CAS  Google Scholar 

  50. Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, Dong MQ, Han J (2009) RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science 325:332–336. doi:10.1126/science.1172308

    Article  PubMed  CAS  Google Scholar 

  51. Zhao J, Jitkaew S, Cai Z, Choksi S, Li Q, Luo J, Liu ZG (2012) Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc Natl Acad Sci USA. doi:10.1073/pnas.1200012109

    Google Scholar 

  52. Zorov DB, Juhaszova M, Sollott SJ (2006) Mitochondrial ROS-induced ROS release: an update and review. Biochim Biophys Acta 1757:509–517. doi:10.1016/j.bbabio.2006.04.029

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank the following persons for their excellent assistance: Maringa Emons, Sebastian Baars, Jerry Oduro, Esther van Eeuwijk, Sridevi Jaksani, Corina Metz, Chaylendra Strijder and Arjan Schoneveld (all at the University Medical Center Utrecht, The Netherlands). This work was supported by the Novartis Foundation for Cardiovascular Excellence (JS), a Bekalis price (PD), the “Wijnand M. Pon Stichting” (MO) and “Stichting Swaeneborgh” (MO).

Conflict of interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joost P. G. Sluijter.

Electronic supplementary material

Below is the link to the electronic supplementary material.

395_2012_270_MOESM1_ESM.pdf

Supplementary Fig. 1 Overview of the experimental design and tissue collection obtained at different times of reperfusion, including MRI measurements. For both vehicle- and Nec-1-treated animals, a similar design was used. (PDF 362 kb)

395_2012_270_MOESM2_ESM.pdf

Supplementary Fig. 2 Cardiac mRNA levels of RIP3 and RIP1 at 1 day after I/R. (a) RIP3 mRNA levels increased significantly after I/R in both groups when compared to baseline. (b) RIP1 mRNA levels increased slightly after I/R, which was similar in both groups. N = 6/group, *P < 0.05 vs. baseline. (c) Control western blot showing a strong depletion of RIP1 after immunoprecipitation (post IP), while RIP1 was not present in the negative control (IgG). n = 5/group. (PDF 299 kb)

395_2012_270_MOESM3_ESM.pdf

Supplementary Fig. 3 TUNEL staining in vivo after I/R. Representative fluorescent pictures of TUNEL (green) and nuclear (Hoechst, blue) staining 1d after I/R of vehicle-treated (a) and Nec-1–treated (b) animals (scale bar 20 μm). (c) Quantification of TUNEL-positive cells representing apoptotic cell death 1 day after I/R (n = 6/group), showing no difference between treatments. (PDF 1033 kb)

395_2012_270_MOESM4_ESM.pdf

Supplementary Fig. 4 Nec-1 reduces reactive oxygen species in vivo after I/R. Representative fluorescent pictures of ROS detection using dihydroethidium (DHE) of vehicle-treated (a) and Nec-1–treated (b) animals (scale bar 20 μm). (c) Quantification of DHE intensity, showing a significant decrease after Nec-1 treatment 1 day after reperfusion (n = 6/group, *P < 0.05). (d) Cardiac mRNA levels of PYGL and GLUL increased significantly in both groups after I/R, while GLUD1 mRNA levels decreased significantly when compared to baseline (n = 6/group, *P < 0.05 vs. baseline). (PDF 3084 kb)

395_2012_270_MOESM5_ESM.pdf

Supplementary Fig. 5 Nec-1 changes the expression of oxidative stress genes after I/R. (a) Scatter plot showing the relative gene expression of 84 genes involved in oxidative stress in Nec-1 vs. vehicle treatment after 1 day of reperfusion. Lines on both sides mark a twofold difference. (b) List of differentially expressed genes (> twofold difference when compared to vehicle treatment) related to cardiac injury with representative references. (PDF 366 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Oerlemans, M.I.F.J., Liu, J., Arslan, F. et al. Inhibition of RIP1-dependent necrosis prevents adverse cardiac remodeling after myocardial ischemia–reperfusion in vivo. Basic Res Cardiol 107, 270 (2012). https://doi.org/10.1007/s00395-012-0270-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00395-012-0270-8

Keywords

Navigation