Skip to main content

Advertisement

Log in

The increased expression of the inducible Hsp70 (HSP70A1A) in serum of patients with heart failure and its protective effect against the cardiotoxic agent doxorubicin

  • Published:
Molecular and Cellular Biochemistry Aims and scope Submit manuscript

Abstract

The aim of this study was to examine the potential association between the expression of Hsp70 protein and heart failure and to investigate the possible protective effect of Hsp70 against the doxorubicin-induced toxicity. Initially, at clinical level, the expression levels of the inducible Hsp70 were quantified in serum from patients with heart failure. Our results showed that in heart failure, Hsp70 concentration appeared to be increased in blood sera of patients compared to that of healthy individuals. The enhanced expression of Hsp70 in serum of patients with heart failure seemed to be associated with various features, such as gender, age and the type of heart failure, but not with its etiology. Next, in our study at cellular level, we used primary cell cultures isolated from embryos of Hsp70-transgenic mice (Tg/Tg) overexpressing human HSP70 and wild-type mice (F1/F1). After exposure to a wide range of doxorubicin concentrations and incubation times, the dose- and time-dependent toxicity of the drug, which appeared to be reduced in Tg/Tg cells, was demonstrated. In addition, doxorubicin administration appeared to result in a dose- and time-dependent decrease in the activity of two of the major endogenous antioxidant enzymes (SOD and GPx). The increased activity of these enzymes in Tg/Tg cells compared to the control F1/F1 cells was obvious, suggesting that the presence of Hsp70 confers enhanced tolerance against DOX-induced oxidative stress. Overall, it has been indicated that Hsp70 protein exerts a very important protective action and renders cells more resistant to the harmful effects of doxorubicin.

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

Abbreviations

Hsp70:

HSP70A1A

DOX:

Doxorubicin

F1/F1 cells:

Control primary embryonic cells

Tg/Tg cells:

Primary embryonic cells overexpressing Hsp70

sHsp70:

Serum Hsp70

SOD:

Superoxide dismutase

GPx:

Glutathione peroxidase

References

  1. Kampinga HH, Hageman J, Vos MJ, Kubota H, Tanguay RM, Bruford EA, Cheetham ME, Chen B, Hightower LE (2009) Guidelines for the nomenclature of the human heat shock proteins. Cell Stress Chaperones 14(1):105–111. https://doi.org/10.1007/s12192-008-0068-7

    Article  CAS  PubMed  Google Scholar 

  2. Mayer MP, Bukau B (2005) Hsp70 chaperones: cellular functions and molecular mechanism. Cell Mol Life Sci 62(6):670–684. https://doi.org/10.1007/s00018-004-4464-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kotoglou P, Kalaitzakis A, Vezyraki P, Tzavaras T, Michalis LK, Dantzer F, Jung JU, Angelidis C (2009) Hsp70 translocates to the nuclei and nucleoli, binds to XRCC1 and PARP-1, and protects HeLa cells from single-strand DNA breaks. Cell Stress Chaperones 14(4):391–406. https://doi.org/10.1007/s12192-008-0093-6

    Article  CAS  PubMed  Google Scholar 

  4. Bozidis P, Lazaridis I, Pagoulatos GN, Angelidis CE (2002) Mydj2 as a potent partner of hsc70 in mammalian cells. Eur J Biochem 269(5):1553–1560

    Article  CAS  Google Scholar 

  5. Angelidis CE, Lazaridis I, Pagoulatos GN (1999) Aggregation of hsp70 and hsc70 in vivo is distinct and temperature-dependent and their chaperone function is directly related to non-aggregated forms. Eur J Biochem 259(1–2):505–512

    Article  CAS  Google Scholar 

  6. Angelidis CE, Lazaridis I, Pagoulatos GN (1991) Constitutive expression of heat-shock protein 70 in mammalian cells confers thermoresistance. Eur J Biochem 199(1):35–39

    Article  CAS  Google Scholar 

  7. Jaattela M, Wissing D (1993) Heat-shock proteins protect cells from monocyte cytotoxicity: possible mechanism of self-protection. J Exp Med 177(1):231–236

    Article  CAS  Google Scholar 

  8. Samali A, Cotter TG (1996) Heat shock proteins increase resistance to apoptosis. Exp Cell Res 223(1):163–170. https://doi.org/10.1006/excr.1996.0070

    Article  CAS  PubMed  Google Scholar 

  9. Jaattela M, Wissing D, Kokholm K, Kallunki T, Egeblad M (1998) Hsp70 exerts its anti-apoptotic function downstream of caspase-3-like proteases. EMBO J 17(21):6124–6134. https://doi.org/10.1093/emboj/17.21.6124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Plumier JC, Ross BM, Currie RW, Angelidis CE, Kazlaris H, Kollias G, Pagoulatos GN (1995) Transgenic mice expressing the human heat shock protein 70 have improved post-ischemic myocardial recovery. J Clin Invest 95(4):1854–1860. https://doi.org/10.1172/JCI117865

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pockley AG, Georgiades A, Thulin T, de Faire U, Frostegard J (2003) Serum heat shock protein 70 levels predict the development of atherosclerosis in subjects with established hypertension. Hypertension 42(3):235–238. https://doi.org/10.1161/01.HYP.0000086522.13672.23

    Article  CAS  PubMed  Google Scholar 

  12. Naka KK, Vezyraki P, Kalaitzakis A, Zerikiotis S, Michalis L, Angelidis C (2014) Hsp70 regulates the doxorubicin-mediated heart failure in Hsp70-transgenic mice. Cell Stress Chaperones 19(6):853–864. https://doi.org/10.1007/s12192-014-0509-4

    Article  CAS  Google Scholar 

  13. Genth-Zotz S, Bolger AP, Kalra PR, von Haehling S, Doehner W, Coats AJ, Volk HD, Anker SD (2004) Heat shock protein 70 in patients with chronic heart failure: relation to disease severity and survival. Int J Cardiol 96(3):397–401. https://doi.org/10.1016/j.ijcard.2003.08.008

    Article  PubMed  Google Scholar 

  14. Gehrig SM, van der Poel C, Sayer TA, Schertzer JD, Henstridge DC, Church JE, Lamon S, Russell AP, Davies KE, Febbraio MA, Lynch GS (2012) Hsp72 preserves muscle function and slows progression of severe muscular dystrophy. Nature 484(7394):394–398. https://doi.org/10.1038/nature10980

    Article  CAS  PubMed  Google Scholar 

  15. Turturici G, Sconzo G, Geraci F (2011) Hsp70 and its molecular role in nervous system diseases. Biochem Res Int 2011. https://doi.org/10.1155/2011/618127

  16. Jolly C, Morimoto RI (2000) Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J Natl Cancer Inst 92(19):1564–1572

    Article  CAS  Google Scholar 

  17. Benjamin IJ, McMillan DR (1998) Stress (heat shock) proteins: molecular chaperones in cardiovascular biology and disease. Circ Res 83(2):117–132

    Article  CAS  Google Scholar 

  18. Hightower LE (1991) Heat shock, stress proteins, chaperones, and proteotoxicity. Cell 66(2):191–197

    Article  CAS  Google Scholar 

  19. Morimoto RI, Sarge KD, Abravaya K (1992) Transcriptional regulation of heat shock genes. A paradigm for inducible genomic responses. J Biol Chem 267(31):21987–21990

    CAS  PubMed  Google Scholar 

  20. Guo S, Wharton W, Moseley P, Shi H (2007) Heat shock protein 70 regulates cellular redox status by modulating glutathione-related enzyme activities. Cell Stress Chaperones 12(3):245–254

    Article  CAS  Google Scholar 

  21. Jaattela M, Wissing D, Bauer PA, Li GC (1992) Major heat shock protein hsp70 protects tumor cells from tumor necrosis factor cytotoxicity. EMBO J 11(10):3507–3512

    Article  CAS  Google Scholar 

  22. Mosser DD, Caron AW, Bourget L, Denis-Larose C, Massie B (1997) Role of the human heat shock protein hsp70 in protection against stress-induced apoptosis. Mol Cell Biol 17(9):5317–5327

    Article  CAS  Google Scholar 

  23. Kelly S, Bieneman A, Horsburgh K, Hughes D, Sofroniew MV, McCulloch J, Uney JB (2001) Targeting expression of hsp70i to discrete neuronal populations using the Lmo-1 promoter: assessment of the neuroprotective effects of hsp70i in vivo and in vitro. J Cereb Blood Flow Metab 21(8):972–981. https://doi.org/10.1097/00004647-200108000-00010

    Article  CAS  PubMed  Google Scholar 

  24. Ferreira AL, Matsubara LS, Matsubara BB (2008) Anthracycline-induced cardiotoxicity. Cardiovasc Hematol Agents Med Chem 6(4):278–281

    Article  CAS  Google Scholar 

  25. Singal PK, Iliskovic N (1998) Doxorubicin-induced cardiomyopathy. N Engl J Med 339(13):900–905. https://doi.org/10.1056/NEJM199809243391307

    Article  CAS  PubMed  Google Scholar 

  26. Octavia Y, Tocchetti CG, Gabrielson KL, Janssens S, Crijns HJ, Moens AL (2012) Doxorubicin-induced cardiomyopathy: from molecular mechanisms to therapeutic strategies. J Mol Cell Cardiol 52(6):1213–1225. https://doi.org/10.1016/j.yjmcc.2012.03.006

    Article  CAS  PubMed  Google Scholar 

  27. Wang L, Zhang TP, Zhang Y, Bi HL, Guan XM, Wang HX, Wang X, Du J, Xia YL, Li HH (2016) Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein. Sci Rep 6:28399. https://doi.org/10.1038/srep28399

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Skladanowski A, Konopa J (1994) Interstrand DNA crosslinking induced by anthracyclines in tumour cells. Biochem Pharmacol 47(12):2269–2278

    Article  CAS  Google Scholar 

  29. Larson ED, Drummond JT (2001) Human mismatch repair and G*T mismatch binding by hMutSalpha in vitro is inhibited by adriamycin, actinomycin D, and nogalamycin. J Biol Chem 276(13):9775–9783. https://doi.org/10.1074/jbc.M006390200

    Article  CAS  PubMed  Google Scholar 

  30. Takemura G, Fujiwara H (2007) Doxorubicin-induced cardiomyopathy from the cardiotoxic mechanisms to management. Prog Cardiovasc Dis 49(5):330–352. https://doi.org/10.1016/j.pcad.2006.10.002

    Article  CAS  PubMed  Google Scholar 

  31. Das DK, Maulik N, Moraru II (1995) Gene expression in acute myocardial stress. Induction by hypoxia, ischemia, reperfusion, hyperthermia and oxidative stress. J Mol Cell Cardiol 27(1):181–193

    Article  CAS  Google Scholar 

  32. Arola OJ, Saraste A, Pulkki K, Kallajoki M, Parvinen M, Voipio-Pulkki LM (2000) Acute doxorubicin cardiotoxicity involves cardiomyocyte apoptosis. Cancer Res 60(7):1789–1792

    CAS  PubMed  Google Scholar 

  33. Wu S, Ko YS, Teng MS, Ko YL, Hsu LA, Hsueh C, Chou YY, Liew CC, Lee YS (2002) Adriamycin-induced cardiomyocyte and endothelial cell apoptosis: in vitro and in vivo studies. J Mol Cell Cardiol 34(12):1595–1607

    Article  CAS  Google Scholar 

  34. Mates JM, Perez-Gomez C, Nunez de Castro I (1999) Antioxidant enzymes and human diseases. Clin Biochem 32(8):595–603

    Article  CAS  Google Scholar 

  35. Schafer FQ, Buettner GR (2001) Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med 30(11):1191–1212

    Article  CAS  Google Scholar 

  36. Hiroi M, Ogihara T, Hirano K, Hasegawa M, Morinobu T, Tamai H, Niki E (2005) Regulation of apoptosis by glutathione redox state in PC12 cells exposed simultaneously to iron and ascorbic acid. Free Radic Biol Med 38(8):1057–1072. https://doi.org/10.1016/j.freeradbiomed.2005.01.001

    Article  CAS  PubMed  Google Scholar 

  37. Ozgocmen S, Kaya H, Fadillioglu E, Yilmaz Z (2007) Effects of calcitonin, risedronate, and raloxifene on erythrocyte antioxidant enzyme activity, lipid peroxidation, and nitric oxide in postmenopausal osteoporosis. Arch Med Res 38(2):196–205. https://doi.org/10.1016/j.arcmed.2006.09.010

    Article  CAS  PubMed  Google Scholar 

  38. Abe T, Fukamachi Y, Kanazawa Y, Furukawa H, Shimizu K, Hirano T, Kasai H, Kashimura M, Higashi K (1996) Inhibition of nucleolar function and morphological change by adriamycin associated with heat shock protein 70 accumulation. Jpn J Cancer Res 87(9):945–951

    Article  CAS  Google Scholar 

  39. Karlseder J, Wissing D, Holzer G, Orel L, Sliutz G, Auer H, Jaattela M, Simon MM (1996) HSP70 overexpression mediates the escape of a doxorubicin-induced G2 cell cycle arrest. Biochem Biophys Res Commun 220(1):153–159. https://doi.org/10.1006/bbrc.1996.0373

    Article  CAS  PubMed  Google Scholar 

  40. Xu L, Giffard RG (1997) HSP70 protects murine astrocytes from glucose deprivation injury. Neurosci Lett 224(1):9–12

    Article  CAS  Google Scholar 

  41. Suzuki S, Maruyama S, Sato W, Morita Y, Sato F, Miki Y, Kato S, Katsuno M, Sobue G, Yuzawa Y, Matsuo S (2005) Geranylgeranylacetone ameliorates ischemic acute renal failure via induction of Hsp70. Kidney Int 67(6):2210–2220. https://doi.org/10.1111/j.1523-1755.2005.00326.x

    Article  CAS  PubMed  Google Scholar 

  42. Ito H, Shimojo T, Fujisaki H, Tamamori M, Ishiyama S, Adachi S, Abe S, Marumo F, Hiroe M (1999) Thermal preconditioning protects rat cardiac muscle cells from doxorubicin-induced apoptosis. Life Sci 64(9):755–761

    Article  CAS  Google Scholar 

  43. Demidenko ZN, Vivo C, Halicka HD, Li CJ, Bhalla K, Broude EV, Blagosklonny MV (2006) Pharmacological induction of Hsp70 protects apoptosis-prone cells from doxorubicin: comparison with caspase-inhibitor- and cycle-arrest-mediated cytoprotection. Cell Death Differ 13(9):1434–1441. https://doi.org/10.1038/sj.cdd.4401812

    Article  CAS  PubMed  Google Scholar 

  44. Angelidis CE, Nova C, Lazaridis I, Kontoyannis D, Kollias G, Pagoulatos G (1996) Overexpression of HSP70 in transgenic mice results in increased cell thermotolerance. Transgenics 2:111–117

    CAS  Google Scholar 

  45. Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65(1–2):55–63

    Article  CAS  Google Scholar 

  46. Dhima IT, Peschos D, Simos YV, Gkiouli MI, Palatianou ME, Ragos VN, Kalfakakou V, Evangelou AM, Karkabounas SC (2017) Modulation of cisplatin cytotoxic activity against leiomyosarcoma cells by epigallocatechin-3-gallate. Nat Prod Res. https://doi.org/10.1080/14786419.2017.1343318

    Article  PubMed  Google Scholar 

  47. Liu J, Mao W, Ding B, Liang CS (2008) ERKs/p53 signal transduction pathway is involved in doxorubicin-induced apoptosis in H9c2 cells and cardiomyocytes. Am J Physiol Heart Circ Physiol 295(5):H1956-H1965. https://doi.org/10.1152/ajpheart.00407.2008

    Article  CAS  Google Scholar 

  48. Gombos T, Forhecz Z, Pozsonyi Z, Janoskuti L, Prohaszka Z (2008) Interaction of serum 70-kDa heat shock protein levels and HspA1B (+1267) gene polymorphism with disease severity in patients with chronic heart failure. Cell Stress Chaperones 13(2):199–206. https://doi.org/10.1007/s12192-007-0001-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li Z, Song Y, Xing R, Yu H, Zhang Y, Li Z, Gao W (2013) Heat shock protein 70 acts as a potential biomarker for early diagnosis of heart failure. PLoS ONE 8(7):e67964. https://doi.org/10.1371/journal.pone.0067964

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pockley AG, Shepherd J, Corton JM (1998) Detection of heat shock protein 70 (Hsp70) and anti-Hsp70 antibodies in the serum of normal individuals. Immunol Invest 27(6):367–377

    Article  CAS  Google Scholar 

  51. Jenei ZM, Gombos T, Forhecz Z, Pozsonyi Z, Karadi I, Janoskuti L, Prohaszka Z (2013) Elevated extracellular HSP70 (HSPA1A) level as an independent prognostic marker of mortality in patients with heart failure. Cell Stress Chaperones 18(6):809–813. https://doi.org/10.1007/s12192-013-0425-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Knowlton AA, Eberli FR, Brecher P, Romo GM, Owen A, Apstein CS (1991) A single myocardial stretch or decreased systolic fiber shortening stimulates the expression of heat shock protein 70 in the isolated, erythrocyte-perfused rabbit heart. J Clin Invest 88(6):2018–2025. https://doi.org/10.1172/JCI115529

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wright BH, Corton JM, El-Nahas AM, Wood RF, Pockley AG (2000) Elevated levels of circulating heat shock protein 70 (Hsp70) in peripheral and renal vascular disease. Heart Vessels 15(1):18–22

    Article  CAS  Google Scholar 

  54. Flohe SB, Bangen JM, Flohe S, Agrawal H, Bergmann K, Schade FU (2007) Origin of immunomodulation after soft tissue trauma: potential involvement of extracellular heat-shock proteins. Shock 27(5):494–502. https://doi.org/10.1097/shk.0b013e31802dec51

    Article  CAS  PubMed  Google Scholar 

  55. Dybdahl B, Wahba A, Lien E, Flo TH, Waage A, Qureshi N, Sellevold OF, Espevik T, Sundan A (2002) Inflammatory response after open heart surgery: release of heat-shock protein 70 and signaling through toll-like receptor-4. Circulation 105(6):685–690

    Article  CAS  Google Scholar 

  56. Dybdahl B, Slordahl SA, Waage A, Kierulf P, Espevik T, Sundan A (2005) Myocardial ischaemia and the inflammatory response: release of heat shock protein 70 after myocardial infarction. Heart 91(3):299–304. https://doi.org/10.1136/hrt.2003.028092

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Satoh M, Shimoda Y, Akatsu T, Ishikawa Y, Minami Y, Nakamura M (2006) Elevated circulating levels of heat shock protein 70 are related to systemic inflammatory reaction through monocyte Toll signal in patients with heart failure after acute myocardial infarction. Eur J Heart Fail 8(8):810–815. https://doi.org/10.1016/j.ejheart.2006.03.004

    Article  CAS  PubMed  Google Scholar 

  58. Bausero MA, Gastpar R, Multhoff G, Asea A (2005) Alternative mechanism by which IFN-gamma enhances tumor recognition: active release of heat shock protein 72. J Immunol 175(5):2900–2912

    Article  CAS  Google Scholar 

  59. Zhan R, Leng X, Liu X, Wang X, Gong J, Yan L, Wang L, Wang Y, Wang X, Qian LJ (2009) Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes. Biochem Biophys Res Commun 387(2):229–233. https://doi.org/10.1016/j.bbrc.2009.06.095

    Article  CAS  PubMed  Google Scholar 

  60. St Rammos K, Koullias GJ, Hassan MO, Argyrakis NP, Voucharas CG, Scarupa SJ, Cowte TG (2002) Low preoperative HSP70 atrial myocardial levels correlate significantly with high incidence of postoperative atrial fibrillation after cardiac surgery. Cardiovasc Surg 10(3):228–232

    Article  Google Scholar 

  61. Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK (2000) Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol 12(11):1539–1546

    Article  CAS  Google Scholar 

  62. Febbraio MA, Ott P, Nielsen HB, Steensberg A, Keller C, Krustrup P, Secher NH, Pedersen BK (2002) Exercise induces hepatosplanchnic release of heat shock protein 72 in humans. J Physiol 544(Pt 3):957–962

    Article  CAS  Google Scholar 

  63. Contreras-Sesvold C, Revenis BD, O’Connor FG, Deuster PA (2015) Association of plasma heat shock protein 70, interleukin 6, and creatine kinase concentrations in a healthy, young adult population. J Biomark. https://doi.org/10.1155/2015/967120

    Article  PubMed  PubMed Central  Google Scholar 

  64. Njemini R, Bautmans I, Onyema OO, Van Puyvelde K, Demanet C, Mets T (2011) Circulating heat shock protein 70 in health, aging and disease. BMC Immunol 12:24. https://doi.org/10.1186/1471-2172-12-24

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhu J, Quyyumi AA, Wu H, Csako G, Rott D, Zalles-Ganley A, Ogunmakinwa J, Halcox J, Epstein SE (2003) Increased serum levels of heat shock protein 70 are associated with low risk of coronary artery disease. Arterioscler Thromb Vasc Biol 23(6):1055–1059. https://doi.org/10.1161/01.ATV.0000074899.60898.FD

    Article  CAS  PubMed  Google Scholar 

  66. Paroo Z, Tiidus PM, Noble EG (1999) Estrogen attenuates HSP 72 expression in acutely exercised male rodents. Eur J Appl Physiol Occup Physiol 80(3):180–184. https://doi.org/10.1007/s004210050579

    Article  CAS  PubMed  Google Scholar 

  67. Shinohara T, Takahashi N, Ooie T, Ichinose M, Hara M, Yonemochi H, Saikawa T, Yoshimatsu H (2004) Estrogen inhibits hyperthermia-induced expression of heat-shock protein 72 and cardioprotection against ischemia/reperfusion injury in female rat heart. J Mol Cell Cardiol 37(5):1053–1061. https://doi.org/10.1016/j.yjmcc.2004.09.006

    Article  CAS  PubMed  Google Scholar 

  68. Mallat Z, Fornes P, Costagliola R, Esposito B, Belmin J, Lecomte D, Tedgui A (2001) Age and gender effects on cardiomyocyte apoptosis in the normal human heart. J Gerontol A Biol Sci Med Sci 56(11):M719–M723

    Article  CAS  Google Scholar 

  69. Jin X, Wang R, Xiao C, Cheng L, Wang F, Yang L, Feng T, Chen M, Chen S, Fu X, Deng J, Wang R, Tang F, Wei Q, Tanguay RM, Wu T (2004) Serum and lymphocyte levels of heat shock protein 70 in aging: a study in the normal Chinese population. Cell Stress Chaperones 9(1):69–75

    Article  CAS  Google Scholar 

  70. Rao DV, Watson K, Jones GL (1999) Age-related attenuation in the expression of the major heat shock proteins in human peripheral lymphocytes. Mech Ageing Dev 107(1):105–118

    Article  CAS  Google Scholar 

  71. Vasilaki A, Jackson MJ, McArdle A (2002) Attenuated HSP70 response in skeletal muscle of aged rats following contractile activity. Muscle Nerve 25(6):902–905. https://doi.org/10.1002/mus.10094

    Article  CAS  PubMed  Google Scholar 

  72. Wei YJ, Huang YX, Shen Y, Cui CJ, Zhang XL, Zhang H, Hu SS (2009) Proteomic analysis reveals significant elevation of heat shock protein 70 in patients with chronic heart failure due to arrhythmogenic right ventricular cardiomyopathy. Mol Cell Biochem 332(1–2):103–111. https://doi.org/10.1007/s11010-009-0179-1

    Article  CAS  PubMed  Google Scholar 

  73. Zhang DX, Ma DY, Yao ZQ, Fu CY, Shi YX, Wang QL, Tang QQ (2016) ERK1/2/p53 and NF-kappaB dependent-PUMA activation involves in doxorubicin-induced cardiomyocyte apoptosis. Eur Rev Med Pharmacol Sci 20(11):2435–2442

    PubMed  Google Scholar 

  74. Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S, Kalyanaraman B (2004) Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J Biol Chem 279(24):25535–25543. https://doi.org/10.1074/jbc.M400944200

    Article  CAS  PubMed  Google Scholar 

  75. Fang X, Jiang Y, Feng L, Chen H, Zhen C, Ding M, Wang X (2013) Blockade of PI3K/AKT pathway enhances sensitivity of Raji cells to chemotherapy through down-regulation of HSP70. Cancer Cell Int 13(1):48. https://doi.org/10.1186/1475-2867-13-48

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Stankiewicz AR, Lachapelle G, Foo CP, Radicioni SM, Mosser DD (2005) Hsp70 inhibits heat-induced apoptosis upstream of mitochondria by preventing Bax translocation. J Biol Chem 280(46):38729–38739. https://doi.org/10.1074/jbc.M509497200

    Article  CAS  PubMed  Google Scholar 

  77. Beere HM, Wolf BB, Cain K, Mosser DD, Mahboubi A, Kuwana T, Tailor P, Morimoto RI, Cohen GM, Green DR (2000) Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat Cell Biol 2(8):469–475. https://doi.org/10.1038/35019501

    Article  CAS  PubMed  Google Scholar 

  78. Xia L, Jaafar L, Cashikar A, Flores-Rozas H (2007) Identification of genes required for protection from doxorubicin by a genome-wide screen in Saccharomyces cerevisiae. Cancer Res 67(23):11411–11418. https://doi.org/10.1158/0008-5472.CAN-07-2399

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kang YJ, Zhou ZX, Wang GW, Buridi A, Klein JB (2000) Suppression by metallothionein of doxorubicin-induced cardiomyocyte apoptosis through inhibition of p38 mitogen-activated protein kinases. J Biol Chem 275(18):13690–13698

    Article  CAS  Google Scholar 

  80. Poornima P, Kumar VB, Weng CF, Padma VV (2014) Doxorubicin induced apoptosis was potentiated by neferine in human lung adenocarcima, A549 cells. Food Chem Toxicol 68:87–98. https://doi.org/10.1016/j.fct.2014.03.008

    Article  CAS  PubMed  Google Scholar 

  81. Berthiaume JM, Wallace KB (2007) Adriamycin-induced oxidative mitochondrial cardiotoxicity. Cell Biol Toxicol 23(1):15–25. https://doi.org/10.1007/s10565-006-0140-y

    Article  CAS  PubMed  Google Scholar 

  82. Fogli S, Nieri P, Breschi MC (2004) The role of nitric oxide in anthracycline toxicity and prospects for pharmacologic prevention of cardiac damage. FASEB J 18(6):664–675. https://doi.org/10.1096/fj.03-0724rev

    Article  CAS  PubMed  Google Scholar 

  83. Doroshow JH (1983) Effect of anthracycline antibiotics on oxygen radical formation in rat heart. Cancer Res 43(2):460–472

    CAS  PubMed  Google Scholar 

  84. Xing W, Mao Y (2012) Effect of catalpol on doxorubicin-induced cytotoxicity in H9c2 cells. J Med Plants Res 6:849–854. https://doi.org/10.5897/JMPR11.1493

    Article  CAS  Google Scholar 

  85. Mohan IK, Kumar KV, Naidu MU, Khan M, Sundaram C (2006) Protective effect of CardiPro against doxorubicin-induced cardiotoxicity in mice. Phytomedicine 13(4):222–229. https://doi.org/10.1016/j.phymed.2004.09.003

    Article  CAS  PubMed  Google Scholar 

  86. Hozayen WG, Seif HSA, Amin S (2014) Protective effects of rutin and/or hesperidin against doxorubicin-induced hepatotoxicity. Int J Clin Nutr 2(1):11–17

    Google Scholar 

  87. Zhang HY, Lu NH, Xie Y, Guo GH, Zhan JH, Chen J (2008) Influence of heat shock preconditioning on structure and function of mitochondria in gastric mucosa of severely burned animals: experiment with rats. Zhonghua Yi Xue Za Zhi 88(8):564–567

    CAS  PubMed  Google Scholar 

  88. Broome CS, Kayani AC, Palomero J, Dillmann WH, Mestril R, Jackson MJ, McArdle A (2006) Effect of lifelong overexpression of HSP70 in skeletal muscle on age-related oxidative stress and adaptation after nondamaging contractile activity. FASEB J 20(9):1549–1551. https://doi.org/10.1096/fj.05-4935fje

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stelios Zerikiotis.

Ethics declarations

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

All procedures performed in this study involving human participants were approved by the Ethics Committee of the “Michaelidion” Cardiac Center, University of Ioannina, Greece, and were in accordance with the Helsinki Declaration. All applicable international, national and institutional guidelines for the care and use of animals were followed. All procedures performed in this study involving laboratory animals were in accordance with the guidelines of the European Community for the Protection of Vertebrate Animals used for Experimental and other Scientific Purposes. All conducted experimental protocols were approved by the Veterinarian Services of the Prefecture of Ioannina in compliance with the Greek legislation for breeding and handling animals subject to the European Directives.

Informed consent

Written informed consent was obtained from all individual participants included in the study.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zerikiotis, S., Angelidis, C., Dhima, I. et al. The increased expression of the inducible Hsp70 (HSP70A1A) in serum of patients with heart failure and its protective effect against the cardiotoxic agent doxorubicin. Mol Cell Biochem 455, 41–59 (2019). https://doi.org/10.1007/s11010-018-3469-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11010-018-3469-7

Keywords

Navigation