Abstract

HomeCirculation ResearchVol. 89, No. 2Heme Oxygenase-1 Protects the Heart Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBHeme Oxygenase-1 Protects the Heart Augustine M.K. Choi Augustine M.K. ChoiAugustine M.K. Choi From the Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa. Search for more papers by this author Originally published3 Apr 2018https://doi.org/10.1161/res.89.2.105Circulation Research. 2001;89:105–107The seminal discovery of nitric oxide (NO) in the 1980s unraveled the novel concept that an endogenous production of a gaseous substance such as NO can impart diverse and critical functional effects on a wide spectrum of biological and pathological processes. Intense investigations in the chemistry and biology of NO have led to numerous fruitful discoveries, enhancing our understanding of many disease processes including cardiovascular disorders. Interestingly, though, we have known for a longer period of time that there exists another gaseous molecule, carbon monoxide (CO), which can be generated endogenously. The heme oxygenase (HO) enzyme system generates the majority of endogenous CO.1,2 Since the biochemical isolation of the HO enzyme in 1968, much of the focus of HO research has been in the study of HO in heme metabolism based on the known fact that the HO enzyme serves as the rate-limiting enzyme in the degradation of heme. However, in recent years, as a result of the emerging role of HO in a variety of biological processes, interest in HO has continued to grow beyond its role in heme metabolism and has expanded into many scientific disciplines. The recent characterization of the HO enzyme system in yeast, prokaryotic bacterial system, and plants further highlights the functional importance of a highly conserved enzyme throughout the evolution of living organisms.HO catalyzes the first and rate-limiting step in the degradation of heme to yield equimolar quantities of biliverdin IXa, CO, and iron1,2 (Figure). Biliverdin is subsequently converted to bilirubin via the action of biliverdin reductase, and free iron is promptly sequestered into ferritin. Three isoforms of HO exist; HO-1 is highly inducible whereas HO-2 and HO-3 are constitutively expressed.1,2 Heme, a major substrate of HO-1, and a variety of nonheme agents including heavy metals, cytokines, hormones, endotoxin, and heat shock are also strong inducers of HO-1 expression.1,2 This diversity of HO-1 inducers has provided further support for the speculation that HO-1, in addition to its role in heme degradation, may also play a vital function in maintaining cellular homeostasis. Furthermore, HO-1 is highly induced by a variety of agents causing oxidative stress including hydrogen peroxide, glutathione depletors, UV irradiation, endotoxin, hypoxia, and hyperoxia.1,2 One interpretation of this finding is that HO-1 can serve as a key biological molecule in the adaptation and/or defense against these oxidative and cellular stresses. Indeed, many laboratories have demonstrated that induction of endogenous HO-1 provides protection against oxidative stress in various in vivo and in vitro models.1,2 Furthermore, recent analysis of HO-1−/− null mice has strengthened the evolving paradigm that HO-1 is indeed an important molecule in the host’s defense against cellular stress in that HO-1−/− null mice exhibited increased susceptibility to endotoxin.3 Interestingly, the first human case of HO-1 deficiency was recently reported.4 Analysis of this patient’s HO-1 gene revealed complete loss of exon 2 of the maternal allele and a two-nucleotide deletion within exon 3 of the paternal allele. Importantly, the human patient deficient of HO-1 expression also exhibited significant phenotypic changes reflective of homeostasis imbalance, comparable to the HO-1−/− null mice. Cells derived from this HO-1–deficient patient also demonstrated increased susceptibility to oxidative stress.4Download figureDownload PowerPointCatalytic reaction of heme oxygenase. CO indicates carbon monoxide.In this issue of Circulation Research, Yet et al5 demonstrate that HO-1 provides potent cytoprotection against ischemia/reperfusion tissue injury. These investigators successfully generated cardiac-specific transgenic mice overexpressing HO-1, and by using both isolated perfused heart preparation and an in vivo myocardial infarction model, the authors provide compelling evidence that indeed HO-1 overexpression can confer marked cytoprotection against ischemia/reperfusion-induced myocardial tissue injury. This study represents an elegant extension from their previous study demonstrating that in response to chronic hypoxia, HO-1−/− null mice exhibited increased right ventricular infarcts with organized mural thrombi and increased lipid peroxidation and oxidative damage in right ventricular cardiomyocytes when compared with wild-type HO+/+ mice.6 The cytoprotective effect of HO-1 in vascular injury is further supported by a recent report by Christou et al,7 who demonstrated an important role of HO-1 in the prevention of hypoxia-induced pulmonary hypertension. By using agonists of HO-1 induction, these authors successfully showed that in vivo enhancement of HO-1 in the rat lung can prevent the development of hypoxic pulmonary hypertension and importantly inhibited the structural remodeling of the pulmonary vessel. The mechanism(s) by which HO-1 mediates these cytoprotective effects against chronic hypoxia remains to be elucidated. However, the known vasodilating and antiproliferative actions of endogenous CO, as well as indirect effect of CO, on production of vasoconstrictors and vascular growth factors such as endothelin-1 (ET-1) and platelet-derived growth factor-B (PDGF-B) may be involved in combating chronic hypoxic stress.8The mechanism(s) by which selective overexpression of HO-1 in cardiac tissue conferred protection against ischemia/reperfusion injury remains poorly understood. The central issue of how HO-1 confers cytoprotection against cellular and tissue injury remains a major enigma in the field of HO research today and will undoubtedly continue to challenge and frustrate researchers in the foreseeable future. How does HO-1 confer such potent cytoprotection against ischemia/reperfusion tissue injury? One can speculate that the three catalytic by-products of heme catalysis by HO, namely bilirubin, ferritin from sequestration of free iron, and CO (Figure), all play critical roles in mediating cytoprotection against ischemia/reperfusion tissue injury. These speculations have been increasingly supported by experimental data (direct and indirect) in recent years. For example, the known antioxidant bilirubin has been shown to protect isolated perfused rat hearts.9 Although it is not know whether ferritin can directly mediate cytoprotection against ischemia/reperfusion tissue injury, ferritin is a known cytoprotective molecule in the vascular endothelium against oxidative stress.10 Recently, intriguing data suggest that CO may serve to protect against ischemia/reperfusion injury in the lung.11 The mechanism by which CO may mediate this cytoprotection perhaps is due to the potent antifibrinolytic properties of CO.11 Interestingly, Soares and colleagues demonstrated in an in vivo vascular injury model of xenotransplantation that CO not only can confer protection as effectively as HO-1 but can also confer cytoprotection in the absence of HO-1.12,13 The beneficial effects of HO-1 have also been demonstrated in ischemia/reperfusion injury in the liver.14 It is quite plausible that all three by-products act in concert and synergize with each other to optimize cytoprotection, depending on cell type and models of injury. The additive beneficial effect of these mediators has been shown in cultured endothelial cells, with CO and ferritin each imparting additive cytoprotection against tumor necrosis factor-α (TNF-α)–induced apoptosis.15Whether it is ferritin, bilirubin, or CO, or all three of these mediators, the challenge in front of us is delineating the signaling pathways by which these molecules confer cytoprotection. The anti-inflammatory5,12,16 and antiapoptotic effects of HO-115,17 lend some clues as to how the catalytic by-products may transduce signals to provide downstream cytoprotection against cellular stresses. For example, recent observations suggest that CO may impart potent anti-inflammatory and antiapoptotic effects via the mitogen kinase pathway in macrophages and endothelial cells, respectively.15,16 Importantly, these cytoprotective effects involve neither the guanylyl cyclase–cGMP nor the NO pathway. The cGMP-independent pathway of CO perhaps reminds us that we no longer can assume that CO has to “compete” with NO for guanylyl cyclase and subsequent cGMP production for physiological function. Nevertheless, the complexity of the signaling pathways by which CO imparts cytoprotection is further highlighted by recent reports demonstrating the critical role of cGMP in mediating antifibrinolytic and antiproliferative effect of CO in ischemia/reperfusion and vascular injury models.11,18As observed in many physiological, biological, and toxicological systems, overzealous production of effector molecules may be counterproductive for the maintenance of cellular and organismal homeostasis during both normal and pathophysiological conditions. Hence, the levels of cytoprotective genes are usually induced by cellular stresses, rather than constitutively expressed. In the case of the HO enzyme system, ironically, at one time or another in the past (still is), each of three catalytic by-products of heme degradation by HO has been a known cellular toxin.19 For example, the released iron from breakdown of heme can serve as a potent oxidant molecule facilitating the production of hydroxyl radicals via the Fenton reaction. Bilirubin plays an important role in the development of kernicterus. The lethality of CO is well known by its ability to bind to the oxygen-carrying heme moiety of hemoglobin, dissociating oxygen and depriving tissues of their oxygen supply; CO remains the most common cause of poison-related deaths in the United States. Thus, the HO enzyme system, which possesses both constitutive and inducible isoforms, is poised and ready to “switch” the signal, when stressed, with an appropriate level of HO-1 induction to combat cellular stresses. Once induced, the HO-1 molecule is uniquely qualified to thwart deleterious toxins by the liberation of bilirubin, ferritin, or CO for cellular protection.Ample evidence now exists that HO-1 plays a critical role in the adaptive response of cells and tissues to a variety of stresses. There is hard work in front of us: we need to delineate more precisely the mediators of cytoprotection of HO-1 and the biochemical and cellular mechanism(s) by which HO-1 or its catalytic by-products confer cytoprotection.The opinions expressed in this editorial are not necessarily those of the editors or of the American Heart Association.The author is supported by NIH grants HL-55330, HL-60234, and AI-42365 and an AHA Established Investigator Award.FootnotesCorrespondence to Augustine M.K. Choi, MD, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, MUH 628 NW, Pittsburgh, PA 15213. E-mail [email protected] References 1 Otterbein LE, Choi AMK. Heme oxygenase: colors of defense against cellular stress. Am J Physiol. 2000; 279: L1029–L1037.CrossrefMedlineGoogle Scholar2 Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997; 37: 517–554.CrossrefMedlineGoogle Scholar3 Poss KD, Tonegawa S. Reduced stress defense in heme oxygenase-1 deficient cells. Proc Natl Acad Sci U S A. 1997; 94: 10925–10930.CrossrefMedlineGoogle Scholar4 Yachie A, Niida Y, Wada T, Igarashi N, Kaneda H. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. J Clin Invest. 1999; 103: 129–135.CrossrefMedlineGoogle Scholar5 Yet S-F, Tian R, Layne MD, Wang ZY, Maemura K, Solovyeva M, Ith B, Melo LG, Zhang L, Ingwall JS, Dzau VJ, Lee M-E, Perrella MA. Cardiac-specific expression of heme oxygenase-1 protects against ischemia and reperfusion injury in transgenic mice. Circ Res. 2001; 89: 168–173.CrossrefMedlineGoogle Scholar6 Yet S-F, Perrella MA, Layne MD, Hsieh C-M, Maemura K, Kobzik L, Wiesel P, Christou H, Kourembanas S, Lee M-E. Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. J Clin Invest. 1999; 103: 23-29.CrossrefMedlineGoogle Scholar7 Christou H, Morita T, Hsieh CM, Koike H, Arkonac B, Perrella M, Kourembanas S. Prevention of hypoxia-induced pulmonary hypertension by enhancement of endogenous heme oxygenase-1 in the rat. Circ Res. 2001; 86: 1224–1229.Google Scholar8 Morita T, Kourembanas S. Endothelial cell expression of vasoconstrictors and growth factors is regulated by smooth muscle cell-derived carbon monoxide. J Clin Invest. 1995; 96: 2676–2682.CrossrefMedlineGoogle Scholar9 Clark JE, Foresti R, Sarathchandra P, Kaur H, Green CJ, Motterlini R. Heme oxygenase-1 derived bilirubin ameliorates postischemic myocardial dysfunction. Am J Physiol. 2000; 278: H643–H651.CrossrefMedlineGoogle Scholar10 Balla G, Jacob HS, Balla J, Rosenberg M, Nath KA, Apple F, Eaton JW, Vercelotti GM. Ferritin: a cytoprotective antioxidant strategem of endothelium. J Biol Chem. 1992; 267: 18148–18153.CrossrefMedlineGoogle Scholar11 Fujita T, Toda K, Karimova A, Yan S-F, Naka Y, Yet S-F, Pinsky DJ. Paradoxical rescue from ischemic lung injury by inhaled carbon monoxide driven by derepression of fibrinolysis. Nat Med. 2001; 7: 598–604.CrossrefMedlineGoogle Scholar12 Soares MP, Lin Y, Anrather J, Csizmadia E, Takigami K, Sato K, Grey ST, Colvin RB, Choi AMK, Poss KD, Bach FH. Expression of heme oxygenase-1 can determine cardiac xenograft survival. Nat Med. 1998; 4: 1073–1077.CrossrefMedlineGoogle Scholar13 Sato K, Balla J, Otterbein L, Smith RN, Brouard S, Lin Y, Csizmadia E, Sevigny J, Robson SC, Vercelloti G, Choi AMK, Bach FH, Soares MP. Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol. 2001; 166: 4185–4194.CrossrefMedlineGoogle Scholar14 Amersi F, Buelow R, Kato H, Ke B, Coito AJ, Shen XD, Zhao D, Zaky J, Melinek J, Lassman CR, Kolls JK, Alam J, Ritter T, Volk HD, Farmer DG, Ghobrial RM, Busuttil RW, Kupiec-Weglinski JW. Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest. 1999; 104: 1631–1639.CrossrefMedlineGoogle Scholar15 Brouard S, Otterbein LE, Anrather J, Tobiasch E, Bach FH, Choi AMK, Soares MP. Carbon monoxide generated by heme oxygenase-1 suppresses endothelial cell apoptosis. J Exp Med. 2001; 92: 1015–1026.Google Scholar16 Otterbein LE, Bach FH, Alam J, Soares M, Lu HT, Wysk M, Davis RJ, Flavell RA, Choi AMK. Carbon monoxide mediates anti-inflammatory effects via the p38 mitogen activated protein kinase pathway. Nat Med. 2000; 6: 422–428.CrossrefMedlineGoogle Scholar17 Petrache I, Wiegand G, Choi AMK. Heme oxygenase-1 inhibits tumor necrosis factor-α–induced apoptosis in L929 fibroblasts. Am J Physiol. 2000; 278: L312–L319.Google Scholar18 Duckers HJ, Boehm M, True AL, Yet SF, San H, Park JL, Clinton Webb R, Lee ME, Nabel GJ, Nabel EG. Heme oxygenase-1 protects against vascular constriction and proliferation. Nat Med. 2001; 7: 693–699.CrossrefMedlineGoogle Scholar19 Platt JL, Nath KA. Heme oxygenase: protective gene or Trojan horse. Nat Med. 1998; 4: 1364–1365.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Bayo Jimenez M, Frenis K, Kröller-Schön S, Kuntic M, Stamm P, Kvandová M, Oelze M, Li H, Steven S, Münzel T and Daiber A (2021) Noise-Induced Vascular Dysfunction, Oxidative Stress, and Inflammation Are Improved by Pharmacological Modulation of the NRF2/HO-1 Axis, Antioxidants, 10.3390/antiox10040625, 10:4, (625) Kawashiri M, Nakanishi C, Tsubokawa T, Shimojima M, Yoshida S, Yoshimuta T, Konno T, Yamagishi M and Hayashi K (2015) Impact of Enhanced Production of Endogenous Heme Oxygenase-1 by Pitavastatin on Survival and Functional Activities of Bone Marrow–derived Mesenchymal Stem Cells, Journal of Cardiovascular Pharmacology, 10.1097/FJC.0000000000000231, 65:6, (601-606), Online publication date: 1-Jun-2015. Pósa A, Szabó R, Csonka A, Veszelka M, Berkó A, Baráth Z, Ménesi R, Pávó I, Gyöngyösi M, László F, Kupai K and Varga C (2015) Endogenous Estrogen-Mediated Heme Oxygenase Regulation in Experimental Menopause, Oxidative Medicine and Cellular Longevity, 10.1155/2015/429713, 2015, (1-7), . He X, Wang Y, Yan X, Wang Y, Wang C, Zhang Z, Li H and Jiang H (2013) Transduction of PEP-1–Heme Oxygenase-1 Fusion Protein Reduces Myocardial Ischemia/Reperfusion Injury in Rats, Journal of Cardiovascular Pharmacology, 10.1097/FJC.0b013e3182a0b638, 62:5, (436-442), Online publication date: 1-Nov-2013. Pósa A, Kupai K, Ménesi R, Szalai Z, Szabó R, Pintér Z, Pálfi G, Gyöngyösi M, Berkó A, Pávó I and Varga C (2013) Sexual Dimorphism of Cardiovascular Ischemia Susceptibility Is Mediated by Heme Oxygenase, Oxidative Medicine and Cellular Longevity, 10.1155/2013/521563, 2013, (1-11), . Ho C, Cheng Y, Chau C and Yen G (2011) Effect of Diallyl Sulfide on in Vitro and in Vivo Nrf2-Mediated Pulmonic Antioxidant Enzyme Expression via Activation ERK/p38 Signaling Pathway, Journal of Agricultural and Food Chemistry, 10.1021/jf203800d, 60:1, (100-107), Online publication date: 11-Jan-2012. Xu S, Hu B, He Z, Ma F, Feng J, Shen W and Yang J (2011) Enhancement of Salinity Tolerance during Rice Seed Germination by Presoaking with Hemoglobin, International Journal of Molecular Sciences, 10.3390/ijms12042488, 12:4, (2488-2501) Ding B, Gibbs P, Brookes P and Maines M (2010) The coordinated increased expression of biliverdin reductase and heme oxygenase‐2 promotes cardiomyocyte survival: a reductase‐based peptide counters β‐adrenergic receptor ligand‐mediated cardiac dysfunction, The FASEB Journal, 10.1096/fj.10-166454, 25:1, (301-313), Online publication date: 1-Jan-2011. Mitchell L, Channell M, Royer C, Ryter S, Choi A and McDonald J (2010) Evaluation of inhaled carbon monoxide as an anti-inflammatory therapy in a nonhuman primate model of lung inflammation, American Journal of Physiology-Lung Cellular and Molecular Physiology, 10.1152/ajplung.00366.2009, 299:6, (L891-L897), Online publication date: 1-Dec-2010. Chakravarti R, Aulak K, Fox P and Stuehr D (2010) GAPDH regulates cellular heme insertion into inducible nitric oxide synthase, Proceedings of the National Academy of Sciences, 10.1073/pnas.1008133107, 107:42, (18004-18009), Online publication date: 19-Oct-2010. Tsubokawa T, Yagi K, Nakanishi C, Zuka M, Nohara A, Ino H, Fujino N, Konno T, Kawashiri M, Ishibashi-Ueda H, Nagaya N and Yamagishi M (2010) Impact of anti-apoptotic and anti-oxidative effects of bone marrow mesenchymal stem cells with transient overexpression of heme oxygenase-1 on myocardial ischemia, American Journal of Physiology-Heart and Circulatory Physiology, 10.1152/ajpheart.01330.2008, 298:5, (H1320-H1329), Online publication date: 1-May-2010. Li Q, Guo Y, Ou Q, Cui C, Wu W, Tan W, Zhu X, Lanceta L, Sanganalmath S, Dawn B, Shinmura K, Rokosh G, Wang S and Bolli R (2009) Gene Transfer of Inducible Nitric Oxide Synthase Affords Cardioprotection by Upregulating Heme Oxygenase-1 Via a Nuclear Factor-κB-Dependent Pathway, Circulation, 120:13, (1222-1230), Online publication date: 29-Sep-2009. Guan L, Wen T, Zhang Y, Wang X and Zhao J (2009) Induction of heme oxygenase-1 with hemin attenuates hippocampal injury in rats after acute carbon monoxide poisoning, Toxicology, 10.1016/j.tox.2009.06.001, 262:2, (146-152), Online publication date: 1-Aug-2009. Yeh C, Ching L and Yen G (2009) Inducing gene expression of cardiac antioxidant enzymes by dietary phenolic acids in rats, The Journal of Nutritional Biochemistry, 10.1016/j.jnutbio.2008.01.005, 20:3, (163-171), Online publication date: 1-Mar-2009. Idriss N, Blann A and Lip G (2008) Hemoxygenase-1 in Cardiovascular Disease, Journal of the American College of Cardiology, 10.1016/j.jacc.2008.06.019, 52:12, (971-978), Online publication date: 1-Sep-2008. Zhen-Wei X, Jian-Le S, Qi Q, Wen-Wei Z, Xue-Hong Z and Zi-Li Z (2007) Heme oxygenase-1 improves the survival of discordant cardiac xenograft through its anti-inflammatory and anti-apoptotic effects, Pediatric Transplantation, 10.1111/j.1399-3046.2007.00701.x, 11:8, (850-859), Online publication date: 1-Dec-2007. KOIZUMI S (2007) Human heme oxygenase-1 deficiency: A lesson on serendipity in the discovery of the novel disease, Pediatrics International, 10.1111/j.1442-200X.2007.02353.x, 49:2, (125-132), Online publication date: 1-Apr-2007. Wijpkema J, van Haelst P, Monraats P, Bruinenberg M, Zwinderman A, Zijlstra F, van der Steege G, de Winter R, Doevendans P, Waltenberger J, Wouter Jukema J and Tio R (2006) Restenosis after percutaneous coronary intervention is associated with the angiotensin-II type-1 receptor 1166A/C polymorphism but not with polymorphisms of angiotensin-converting enzyme, angiotensin-II receptor, angiotensinogen or heme oxygenase-1, Pharmacogenetics and Genomics, 10.1097/01.fpc.0000205001.07054.fa, 16:5, (331-337), Online publication date: 1-May-2006. Ewing J and Maines M (2006) Regulation and expression of heme oxygenase enzymes in aged-rat brain: age related depression in HO-1 and HO-2 expression and altered stress-response, Journal of Neural Transmission, 10.1007/s00702-005-0408-z, 113:4, (439-454), Online publication date: 1-Apr-2006. Tang Y, Tang Y, Zhang Y, Qian K, Shen L and Phillips M (2005) Improved Graft Mesenchymal Stem Cell Survival in Ischemic Heart With a Hypoxia-Regulated Heme Oxygenase-1 Vector, Journal of the American College of Cardiology, 10.1016/j.jacc.2005.05.079, 46:7, (1339-1350), Online publication date: 1-Oct-2005. Ameriso S, Villamil A, Zedda C, Parodi J, Garrido S, Sarchi M, Schultz M, Boczkowski J and Sevlever G (2005) Heme Oxygenase-1 Is Expressed in Carotid Atherosclerotic Plaques Infected by Helicobacter pylori and Is More Prevalent in Asymptomatic Subjects, Stroke, 36:9, (1896-1900), Online publication date: 1-Sep-2005. Jürgensen J, Rosenberger C, Wiesener M, Warnecke C, Hörstrup J, Gräfe M, Philipp S, Griethe W, Maxwell P, Frei U, Bachmann S, Willenbrock R and Eckardt K (2004) Persistent induction of HIF‐1α and ‐2α in cardiomyocytes and stromal cells of ischemic myocardium, The FASEB Journal, 10.1096/fj.04-1605fje, 18:12, (1415-1417), Online publication date: 1-Sep-2004. Bak I, Szendrei L, Turoczi T, Papp G, Joo F, Das D, Leiris J, Der P, Juhasz B, Varga E, Bacskay I, Balla J, Kovacs P and Tosaki A (2003) Heme oxygenase‐1 related carbon monoxide production and ventricular fibrillation in isolated ischemic/reperfused mouse myocardium, The FASEB Journal, 10.1096/fj.03-0032fje, 17:14, (1-21), Online publication date: 1-Nov-2003. de Perrot M, Liu M, Waddell T and Keshavjee S (2003) Ischemia–Reperfusion–induced Lung Injury, American Journal of Respiratory and Critical Care Medicine, 10.1164/rccm.200207-670SO, 167:4, (490-511), Online publication date: 15-Feb-2003. Bak I, Papp G, Turoczi T, Varga E, Szendrei L, Vecsernyes M, Joo F and Tosaki A (2002) The role of heme oxygenase-related carbon monoxide and ventricular fibrillation in ischemic/reperfused hearts, Free Radical Biology and Medicine, 10.1016/S0891-5849(02)00913-9, 33:5, (639-648), Online publication date: 1-Sep-2002. July 20, 2001Vol 89, Issue 2 Advertisement Article InformationMetrics https://doi.org/10.1161/res.89.2.105PMID: 11463713 Originally publishedApril 3, 2018 Keywordsbilirubinferritincarbon monoxidePDF download Advertisement

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call