The haptoglobin-CD163-heme oxygenase-1 pathway for hemoglobin scavenging

Jens Haugbølle Thomsen, Anders Etzerodt, Pia Svendsen, Søren K Moestrup, Jens Haugbølle Thomsen, Anders Etzerodt, Pia Svendsen, Søren K Moestrup

Abstract

The haptoglobin- (Hp-) CD163-heme oxygenase-1 (HO-1) pathway is an efficient captor-receptor-enzyme system to circumvent the hemoglobin (Hb)/heme-induced toxicity during physiological and pathological hemolyses. In this pathway, Hb tightly binds to Hp leading to CD163-mediated uptake of the complex in macrophages followed by lysosomal Hp-Hb breakdown and HO-1-catalyzed conversion of heme into the metabolites carbon monoxide (CO), biliverdin, and iron. The plasma concentration of Hp is a limiting factor as evident during accelerated hemolysis, where the Hp depletion may cause serious Hb-induced toxicity and put pressure on backup protecting systems such as the hemopexin-CD91-HO pathway. The Hp-CD163-HO-1 pathway proteins are regulated by the acute phase mediator interleukin-6 (IL-6), but other regulatory factors indicate that this upregulation is a counteracting anti-inflammatory response during inflammation. The heme metabolites including bilirubin converted from biliverdin have overall an anti-inflammatory effect and thus reinforce the anti-inflammatory efficacy of the Hp-CD163-HO-1 pathway. Future studies of animal models of inflammation should further define the importance of the pathway in the anti-inflammatory response.

Figures

Figure 1
Figure 1
The CD163 pathway for uptake of Hb-Hp complexes and the CD91 pathway for uptake of hemopexin- (Hx-) heme complexes. The endocytosis of the ligand leads to degradation in lysosomes while the receptor recycles from the endosomes back to the plasma membrane.
Figure 2
Figure 2
The intracellular pathway for heme-degradation subsequent to CD163 and CD91 mediated endocytosis in macrophages. Free heme is degraded to biliverdin, CO, and ferrous iron by the endoplasmic reticulum enzyme HO-1 facing the cytosol. Electrons are delivered by NADPH p450 cytochrome reductase. Biliverdin is reduced to bilirubin by biliverdin reductase and transported to the liver bound to albumin.

References

    1. Bratosin D, Mazurier J, Tissier JP, et al. Cellular and molecular mechanisms of senescent erythrocyte phagocytosis by macrophages. A review. Biochimie. 1998;80(2):173–195.
    1. Straat M, van Bruggen R, de Korte D, Juffermans NP. Red blood cell clearance in inflammation. Transfusion Medicine and Hemotherapy. 2012;39(5):353–360.
    1. Rother RP, Bell L, Hillmen P, Gladwin MT. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. Journal of the American Medical Association. 2005;293(13):1653–1662.
    1. Aronowski J, Zhao X. Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke. 2011;42(6):1781–1786.
    1. Levy AP, Asleh R, Blum S, et al. Haptoglobin: basic and clinical aspects. Antioxidants and Redox Signaling. 2010;12(2):293–304.
    1. Theilgaard-Mönch K, Jacobsen LC, Nielsen MJ, et al. Haptoglobin is synthesized during granulocyte differentiation, stored in specific granules, and released by neutrophils in response to activation. Blood. 2006;108(1):353–361.
    1. Andersen CB, Torvund-Jensen M, Nielsen MJ, et al. Structure of the haptoglobin-haemoglobin complex. Nature. 2012;489(7416):456–459.
    1. Wicher KB, Fries E. Evolutionary aspects of hemoglobin scavengers. Antioxidants and Redox Signaling. 2010;12(2):249–260.
    1. Nielsen MJ, Moestrup SK. Receptor targeting of hemoglobin mediated by the haptoglobins: roles beyond heme scavenging. Blood. 2009;114(4):764–771.
    1. Bode JG, Albrecht U, Haussinger D, Heinrich PC, Schaper F. Hepatic acute phase proteins—regulation by IL- and IL-1-type cytokines involving STAT3 and its crosstalk with NF-kappaB-dependent signaling. European Journal of Cell Biology. 2012;91(6-7):6496–6505.
    1. Alayash AI, Andersen CB, Moestrup SK, Bülow L. Haptoglobin: the hemoglobin detoxifier in plasma. Trends in Biotechnology. 2013;31(1):2–3.
    1. Wobeto VPDA, Zaccariotto TR, Sonati MDF. Polymorphism of human haptoglobin and its clinical importance. Genetics and Molecular Biology. 2008;31(3):602–620.
    1. Etzerodt A, Moestrup SK. CD163 and inflammation: biological, diagnostic and therapeutic aspects. Antioxid Redox Signal. 2012;18(17):2352–2363.
    1. Banerjee S, Jia Y, Siburt CJP, et al. Haptoglobin alters oxygenation and oxidation of hemoglobin and decreases propagation of peroxide-induced oxidative reactions. Free Radical Biology and Medicine. 2012;53(6):1317–1326.
    1. Buehler PW, Abraham B, Vallelian F, et al. Haptoglobin preserves the CD163 hemoglobin scavenger pathway by shielding hemoglobin from peroxidative modification. Blood. 2009;113(11):2578–2586.
    1. Tolosano E, Fagoonee S, Morello N, Vinchi F, Fiorito V. Heme scavenging and the other facets of hemopexin. Antioxidants and Redox Signaling. 2010;12(2):305–320.
    1. Azarov I, He X, Jeffers A, et al. Rate of nitric oxide scavenging by hemoglobin bound to haptoglobin. Nitric Oxide. 2008;18(4):296–302.
    1. Kristiansen M, Graversen JH, Jacobsen C, et al. Identification of the haemoglobin scavenger receptor. Nature. 2001;409(6817):198–201.
    1. Nielsen MJ, Petersen SV, Jacobsen C, et al. A unique loop extension in the serine protease domain of haptoglobin is essential for CD163 recognition of the haptoglobin-hemoglobin complex. Journal of Biological Chemistry. 2007;282(2):1072–1079.
    1. Nielsen MJ, Andersen CBF, Moestrup SK. CD163 binding to haptoglobin-hemoglobin complexes involves a dual-point electrostatic receptor-ligand pairing. Journal of Biological Chemistry. 2013
    1. Levy AP, Hochberg I, Jablonski K, et al. Haptoglobin phenotype is an independent risk factor for cardiovascular disease in individuals with diabetes: the strong heart study. Journal of the American College of Cardiology. 2002;40(11):1984–1990.
    1. Levy AP, Blum S, Vardi M, et al. Vitamin e reduces cardiovascular disease in individuals with diabetes mellitus and the haptoglobin 2-2 genotype. Pharmacogenomics. 2010;11(5):675–684.
    1. Elagib AA, Kider AO, Åkerström B, Elbashir MI. Association of the haptoglobin phenotype (1-1) with falciparum malaria in Sudan. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1998;92(3):309–311.
    1. Quaye IK, Ekuban FA, Goka BQ, et al. Haptoglobin 1-1 is associated with susceptibility to severe Plasmodium falciparum malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2000;94(2):216–219.
    1. Cunnington AJ, Njie M, Correa S, Takem EN, Riley EM, Walther M. Prolonged neutrophil dysfunction after Plasmodium falciparum malaria is related to hemolysis and heme oxygenase-1 induction. Journal of Immunology. 2012;189(11):5336–5346.
    1. Mendonca VR, Luz NF, Santos NJG, et al. Association between the haptoglobin and heme oxygenase 1 genetic profiles and soluble CD163 in susceptibility to and severity of human malaria. Infection and Immunity. 2012;80(4):1445–1454.
    1. Quaye I. Haptoglobin genotypes are not associated with resistance to severe malaria in The Gambia. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2003;97(1):p. 121.
    1. Beiguelman B, Alves FP, Moura MM, et al. The association of genetic markers and malaria infection in the Brazilian Western Amazonian region. Memorias do Instituto Oswaldo Cruz. 2003;98(4):455–460.
    1. Freeman M, Ashkenas J, Rees DJG, et al. An ancient, highly conserved family of cysteine-rich protein domains revealed by cloning type I and type II murine macrophage scavenger receptors. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(22):8810–8814.
    1. Rodamilans B, Muñoz IG, Bragado-Nilsson E, et al. Crystal structure of the third extracellular domain of CD5 reveals the fold of a group B scavenger cysteine-rich receptor domain. Journal of Biological Chemistry. 2007;282(17):12669–12677.
    1. Hohenester E, Sasaki T, Timpl R. Crystal structure of a scavenger receptor cysteine-rich domain sheds light on an ancient superfamily. Nature Structural Biology. 1999;6(3):228–232.
    1. Somoza JR, Ho JD, Luong C, et al. The structure of the extracellular region of human hepsin reveals a serine protease domain and a novel scavenger receptor cysteine-rich (SRCR) domain. Structure. 2003;11(9):1123–1131.
    1. Martínez VG, Moestrup SK, Holmskov U, Mollenhauer J, Lozano F. The conserved scavenger receptor cysteine-rich super family in therapy and diagnosis. Pharmacological Reviews. 2011;63(4):967–1000.
    1. Van Gorp H, Delputte PL, Nauwynck HJ. Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy. Molecular Immunology. 2010;47(7-8):1650–1660.
    1. Ritter M, Buechler C, Langmann T, Orso E, Klucken J, Schmitz G. The scavenger receptor CD163: regulation, promoter structure and genomic organization. Pathobiology. 2000;67(5-6):257–261.
    1. Ritter M, Buechler C, Kapinsky M, Schmitz G. Interaction of CD163 with the regulatory subunit of casein kinase II (CKII) and dependence of CD163 signaling on CKII and protein kinase C. European Journal of Immunology. 2001;31(4):999–1009.
    1. Nielsen MJ, Madsen M, Møller HJ, Moestrup SK. The macrophage scavenger receptor CD163: endocytic properties of cytoplasmic tail variants. Journal of Leukocyte Biology. 2006;79(4):837–845.
    1. Maniecki MB, Møller HJ, Moestrup SK, Møller BK. CD163 positive subsets of blood dendritic cells: the scavenging macrophage receptors CD163 and CD91 are coexpressed on human dendritic cells and monocytes. Immunobiology. 2006;211(6-8):407–417.
    1. Kodelja V, Goerdt S. Dissection of macrophage differentiation pathways in cutaneous macrophage disorders and in vitro. Experimental Dermatology. 1994;3(6):257–268.
    1. Zwadlo-Klarwasser G, Bent S, Haubeck HD, Sorg C, Schmutzler W. Glucocorticoid-induced appearance of the macrophage subtype RM 3/1 in peripheral blood of man. International Archives of Allergy and Applied Immunology. 1990;91(2):175–180.
    1. Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Reviews of Physiology, Biochemistry and Pharmacology. 2003;149:1–38.
    1. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593–604.
    1. Zwadlo G, Voegeli R, Schulze Osthoff K, Sorg C. A monoclonal antibody to a novel differentiation antigen on human macrophages associated with the down-regulatory phase of the inflammatory process. Experimental Cell Biology. 1987;55(6):295–304.
    1. Finn AV, Nakano M, Polavarapu R, et al. Hemoglobin directs macrophage differentiation and prevents foam cell formation in human atherosclerotic plaques. Journal of the American College of Cardiology. 2012;59(2):166–177.
    1. Boyle JJ. Heme and haemoglobin direct macrophage Mhem phenotype and counter foam cell formation in areas of intraplaque haemorrhage. Current Opinion in Lipidology. 2012;23(5):453–461.
    1. Moller HJ. Soluble CD163. Scandinavian Journal of Clinical & Laboratory Investigation. 2012;72(1):1–13.
    1. Etzerodt A, Maniecki MB, Møller K, Møller HJ, Moestrup SK. Tumor necrosis factor α-converting enzyme (TACE/ADAM17) mediates ectodomain shedding of the scavenger receptor CD163. Journal of Leukocyte Biology. 2010;88(6):1201–1205.
    1. Kneidl J, Löffler B, Erat MC, et al. Soluble CD163 promotes recognition, phagocytosis and killing of Staphylococcus aureus via binding of specific fibronectin peptides. Cellular Microbiology. 2012;14(6):914–936.
    1. Baeten D, Møller HJ, Delanghe J, Veys EM, Moestrup SK, De Keyser F. Association of CD163+ macrophages and local production of soluble CD163 with decreased lymphocyte activation in spondylarthropathy synovitis. Arthritis and Rheumatism. 2004;50(5):1611–1623.
    1. Moreno JA, Muñoz-García B, Martín-Ventura JL, et al. The CD163-expressing macrophages recognize and internalize TWEAK. Potential consequences in atherosclerosis. Atherosclerosis. 2009;207(1):103–110.
    1. Madsen M, Møller HJ, Nielsen MJ, et al. Molecular characterization of the haptoglobin-hemoglobin receptor CD163: ligand binding properties of the scavenger receptor cysteine-rich domain region. Journal of Biological Chemistry. 2004;279(49):51561–51567.
    1. Schaer CA, Schoedon G, Imhof A, Kurrer MO, Schaer DJ. Constitutive endocytosis of CD163 mediates hemoglobin-heme uptake and determines the noninflammatory and protective transcriptional response of macrophages to hemoglobin. Circulation Research. 2006;99(9):943–950.
    1. Schaer DJ, Schaer CA, Buehler PW, et al. CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin. Blood. 2006;107(1):373–380.
    1. Etzerodt A, Kjolby M, Nielsen MJ, Maniecki M, Svendsen P, Moestrup SK. Plasma clearance of hemoglobin and haptoglobin in mice and effect of CD163 gene targeting disruption. Antioxidants & Redox Signaling. 2012;18(17):2254–2263.
    1. Philippidis P, Mason JC, Evans BJ, et al. Hemoglobin scavenger receptor CD163 mediates interleukin-10 release and heme oxygenase-1 synthesis: antiinflammatory monocyte-macrophage responses in vitro, in resolving skin blisters in vivo, and after cardiopulmonary bypass surgery. Circulation Research. 2004;94(1):119–126.
    1. Fabriek BO, Polfliet MMJ, Vloet RPM, et al. The macrophage CD163 surface glycoprotein is an erythroblast adhesion receptor. Blood. 2007;109(12):5223–5229.
    1. Fabriek BO, Bruggen RV, Deng DM, et al. The macrophage scavenger receptor CD163 functions as an innate immune sensor for bacteria. Blood. 2009;113(4):887–892.
    1. Bover LC, Cardó-Vila M, Kuniyasu A, et al. A previously unrecognized protein-protein interaction between TWEAK and CD163: potential biological implications. Journal of Immunology. 2007;178(12):8183–8194.
    1. Llaurado G, González-Clemente JM, Maymó-Masip E, Subías D, Vendrell J, Chacón MR. Serum levels of TWEAK and scavenger receptor CD163 in type 1 diabetes mellitus: relationship with cardiovascular risk factors. a case-control study. PLoS ONE. 2012;7(8)e43919
    1. Urbonaviciene G, Martin-Ventura JL, Lindholt JS, et al. Impact of soluble TWEAK and CD163/TWEAK ratio on long-term cardiovascular mortality in patients with peripheral arterial disease. Atherosclerosis. 2011;219(2):892–899.
    1. Sánchez-Torres C, Gómez-Puertas P, Gómez-Del-Moral M, et al. Expression of porcine CD163 on monocytes/macrophages correlates with permissiveness to African swine fever infection. Archives of Virology. 2003;148(12):2307–2323.
    1. Welch SKW, Calvert JG. A brief review of CD163 and its role in PRRSV infection. Virus Research. 2010;154(1-2):98–103.
    1. Belcher JD, Beckman JD, Balla G, Balla J, Vercellotti G. Heme degradation and vascular injury. Antioxidants and Redox Signaling. 2010;12(2):233–248.
    1. Hvidberg V, Maniecki MB, Jacobsen C, Højrup P, Møller HJ, Moestrup SK. Identification of the receptor scavenging hemopexin-heme complexes. Blood. 2005;106(7):2572–2579.
    1. Moestrup SK, Gliemann J, Pallesen G. Distribution of the α2-macroglobulin receptor/low density lipoprotein receptor-related protein in human tissues. Cell and Tissue Research. 1992;269(3):375–382.
    1. Tolosano E, Fagoonee S, Hirsch E, et al. Enhanced splenomegaly and severe liver inflammation in haptoglobin/hemopexin double-null mice after acute hemolysis. Blood. 2002;100(12):4201–4208.
    1. Fruitier I, Garreau I, Lacroix A, Cupo A, Piot JM. Proteolytic degradation of hemoglobin by endogenous lysosomal proteases gives rise to bioactive peptides: hemorphins. The FEBS Letters. 1999;447(1):81–86.
    1. Wang CY, Chau LY. Heme oxygenase-1 in cardiovascular diseases: molecular mechanisms and clinical perspectives. Chang Gung Medical Journal. 2010;33(1):13–24.
    1. Wu ML, Ho YC, Lin CY, Yet SF. Heme oxygenase-1 in inflammation and cardiovascular disease. American Journal of Cardiovascular Disease. 2011;1(2):150–158.
    1. Hayashi S, Omata Y, Sakamoto H, et al. Characterization of rat heme oxygenase-3 gene. Implication of processed pseudogenes derived from heme oxygenase-2 gene. Gene. 2004;336(2):241–250.
    1. Mccoubrey WK, Huang TJ, Maines MD. Isolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. European Journal of Biochemistry. 1997;247(2):725–732.
    1. Yoshida T, Sato M. Posttranslational and direct integration of heme oxygenase into microsomes. Biochemical and Biophysical Research Communications. 1989;163(2):1086–1092.
    1. Jung NH, Kim HP, Kim BR, et al. Evidence for heme oxygenase-1 association with caveolin-1 and -2 in mouse mesangial cells. IUBMB Life. 2003;55(9):525–532.
    1. Delaby C, Rondeau C, Pouzet C, et al. Subcellular localization of iron and heme metabolism related proteins at early stages of erythrophagocytosis. PLoS ONE. 2012;7(7)e42199
    1. Lin Q, Weis S, Yang G, et al. Heme oxygenase-1 protein localizes to the nucleus and activates transcription factors important in oxidative stress. Journal of Biological Chemistry. 2007;282(28):20621–20633.
    1. Unno M, Matsui T, Ikeda-Saito M. Structure and catalytic mechanism of heme oxygenase. Natural Product Reports. 2007;24(3):553–570.
    1. Unno M, Matsui T, Ikeda-Saito M. Crystallographic studies of heme oxygenase complexed with an unstable reaction intermediate, verdoheme. Journal of Inorganic Biochemistry. 2012;113:102–109.
    1. Matsui T, Nakajima A, Fujii H, et al. O2- and H2O2-dependent verdoheme degradation by heme oxygenase: reaction mechanisms and potential physiological roles of the dual pathway degradation. Journal of Biological Chemistry. 2005;280(44):36833–36840.
    1. Linnenbaum M, Busker M, Kraehling JR, Behrends S. Heme oxygenase isoforms differ in their subcellular trafficking during hypoxia and are differentially modulated by cytochrome P450 reductase. PLoS ONE. 2012;7(4)e35483
    1. Ryter SW, Alam J, Choi AMK. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiological Reviews. 2006;86(2):583–650.
    1. Hartsfield CL, Alam J, Choi AMK. Differential signaling pathways of HO-1 gene expression in pulmonary and systemic vascular cells. American Journal of Physiology. 1999;277(6):L1133–L1141.
    1. Alam J, Shibahara S, Smith A. Transcriptional activation of the heme oxygenase gene by heme and cadmium in mouse hepatoma cells. Journal of Biological Chemistry. 1989;264(11):6371–6375.
    1. Applegate LA, Noel A, Vile G, Frenk E, Tyrrell RM. Two genes contribute to different extents to the heme oxygenase enzyme activity measured in cultured human skin fibroblasts and keratinocytes: implications for protection against oxidant stress. Photochemistry and Photobiology. 1995;61(3):285–291.
    1. Rizzardini M, Carelli M, Cabello Porras MR, Cantoni L. Mechanisms of endotoxin-induced haem oxygenase mRNA accumulation in mouse liver: synergism by glutathione depletion and protection by N-acetylcysteine. Biochemical Journal. 1994;304(2):477–483.
    1. Stuhlmeier KM. Activation and regulation of Hsp32 and Hsp70. European Journal of Biochemistry. 2000;267(4):1161–1167.
    1. Callahan KS. Effect of tumor necrosis factor-a and interleukin-la on heme oxygenase-1 expression in human endothelial cells. American Journal of Physiology. 1998;274(3, part 2):H883–H891.
    1. Foresti R, Clark JE, Green CJ, Motterlini R. Thiol compounds interact with nitric oxide in regulating heme oxygenase-1 induction in endothelial cells: involvement of superoxide and peroxynitrite anions. Journal of Biological Chemistry. 1997;272(29):18411–18417.
    1. Surh YJ, Kundu JK, Na HK. Nrf2 as a master redox switch in turning on the cellular signaling involved in the induction of cytoprotective genes by some chemopreventive phytochemicals. Planta Medica. 2008;74(13):1526–1539.
    1. Lee TS, Chau LY. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nature Medicine. 2002;8(3):240–246.
    1. Kyriakis JM, Avruch J. Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Physiological Reviews. 2012;92(2):689–737.
    1. Hill-Kapturczak N, Voakes C, Garcia J, Visner G, Nick HS, Agarwal A. A cis-acting region regulates oxidized lipid-mediated induction of the human heme oxygenase-1 gene in endothelial cells. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23(8):1416–1422.
    1. Sikorski EM, Hock T, Hill-Kapturczak N, Agarwal A. The story so far: molecular regulation of the heme oxygenase-1 gene in renal injury. American Journal of Physiology. 2004;286(3):F425–F441.
    1. Choi AMK, Alam J. Heme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. American Journal of Respiratory Cell and Molecular Biology. 1996;15(1):9–19.
    1. Chapple SJ, Siow RC, Mann GE. Crosstalk between Nrf2 and the proteasome: therapeutic potential of Nrf2 inducers in vascular disease and aging. International Journal of Biochemistry & Cell Biology. 2012;44(8):1315–1320.
    1. Itoh K, Mimura J, Yamamoto M. Discovery of the negative regulator of Nrf2, keap1: a historical overview. Antioxidants and Redox Signaling. 2010;13(11):1665–1678.
    1. Yu R, Chen C, Mo YY, et al. Activation of mitogen-activated protein kinase pathways induces antioxidant response element-mediated gene expression via a Nrf2-dependent mechanism. Journal of Biological Chemistry. 2000;275(51):39907–39913.
    1. Alam J, Wicks C, Stewart D, et al. Mechanism of heme oxygenase-1 gene activation by cadmium in MCF-7 mammary epithelial cells: role of p38 kinase and Nrf2 transcription factor. Journal of Biological Chemistry. 2000;275(36):27694–27702.
    1. Yet SF, Perrella MA, Layne MD, et al. Hypoxia induces severe right ventricular dilatation and infarction in heine oxygenase-1 null mice. Journal of Clinical Investigation. 1999;103(8):R23–R29.
    1. Yet SF, Layne MD, Liu X, et al. Absence of heme oxygenase-1 exacerbates atherosclerotic lesion formation and vascular remodeling. The FASEB Journal. 2003;17(12):1759–1761.
    1. Ishikawa K, Sugawara D, Wang XP, et al. Heme oxygenase-1 inhibits atherosclerotic lesion formation in LDL-receptor knockout mice. Circulation Research. 2001;88(5):506–512.
    1. Liu X, Wei J, Peng DH, Layne MD, Yet SF. Absence of heme oxygenase-1 exacerbates myocardial ischemia/reperfusion injury in diabetic mice. Diabetes. 2005;54(3):778–784.
    1. Zuckerbraun BS, Billiar TR, Otterbein SL, et al. Carbon monoxide protects against liver failure through nitric oxide-induced heme oxygenase 1. Journal of Experimental Medicine. 2003;198(11):1707–1716.
    1. Ke B, Kupiec-Weglinski JW, Buelow R, et al. Heme oxygenase 1 gene transfer prevents CD95/Fas ligand-mediated apoptosis and improves liver allograft survival via carbon monoxide signaling pathway. Human Gene Therapy. 2002;13(10):1189–1199.
    1. Otterbein LE, Kolls JK, Mantell LL, Cook JL, Alam J, Choi AMK. Exogenous administration of heme oxygenase-1 by gene transfer provides protection against hyperoxia-induced lung injury. Journal of Clinical Investigation. 1999;103(7):1047–1054.
    1. Johnson RA, Teran FJ, Durante W, Peyton KJ, Johnson FK. Enhanced heme oxygenase-mediated coronary vasodilation in Dahl salt-sensitive hypertension. American Journal of Hypertension. 2004;17(1):25–30.
    1. Gullotta F, di Masi A, Coletta M, Ascenzi P. CO metabolism, sensing, and signaling. Biofactors. 2012;38(1):1–13.
    1. Ryter SW, Otterbein LE. Carbon monoxide in biology and medicine. BioEssays. 2004;26(3):270–280.
    1. Pae HO, Son Y, Kim NH, Jeong HJ, Chang KC, Chung HT. Role of heme oxygenase in preserving vascular bioactive NO. Nitric Oxide. 2010;23(4):251–257.
    1. Williams SE, Brazier SP, Baban N, et al. A structural motif in the C-terminal tail of slo1 confers carbon monoxide sensitivity to human BKCa channels. Pflugers Archiv European Journal of Physiology. 2008;456(3):561–572.
    1. Otterbein LE, Bach FH, Alam J, et al. Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nature Medicine. 2000;6(4):422–428.
    1. Hoetzel A, Dolinay T, Vallbracht S, et al. Carbon monoxide protects against ventilator-induced lung injury via PPAR-γ and inhibition of Egr-1. American Journal of Respiratory and Critical Care Medicine. 2008;177(11):1223–1232.
    1. Dolinay T, Wu W, Kaminski N, et al. Mitogen-activated protein kinases regulate susceptibility to ventilator-induced lung injury. PLoS ONE. 2008;3(2)e1601
    1. Gajdócsy R, Horváth I. Exhaled carbon monoxide in airway diseases: from research findings to clinical relevance. Journal of Breath Research. 2010;4(4)047102
    1. Baranano DE, Wolosker H, Bae BI, Barrow RK, Snyder SH, Ferris CD. A mammalian iron ATPase induced by iron. Journal of Biological Chemistry. 2000;275(20):15166–15173.
    1. Yachie A, Niida Y, Wada T, et al. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. Journal of Clinical Investigation. 1999;103(1):129–135.
    1. Poss KD, Tonegawa S. Heme oxygenase 1 is required for mammalian iron reutilization. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(20):10919–10924.
    1. Eisenstein RS, Munro HN. Translational regulation of ferritin synthesis by iron. Enzyme. 1990;44(1–4):42–58.
    1. Vile GF, Tyrrell RM. Oxidative stress resulting from ultraviolet A irradiation of human skin fibroblasts leads to a heme oxygenase-dependent increase in ferritin. Journal of Biological Chemistry. 1993;268(20):14678–14681.
    1. Berberat PO, Katori M, Kaczmarek E, et al. Heavy chain ferritin acts as an antiapoptotic gene that protects livers from ischemia reperfusion injury. The FASEB Journal. 2003;17(12):1724–1726.
    1. Vile GF, Basu-Modak S, Waltner C, Tyrrell RM. Heme oxygenase 1 mediates an adaptive response to oxidative stress in human skin fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(7):2607–2610.
    1. Kapitulnik J, Maines MD. The role of bile pigments in health and disease: effects on cell signaling, cytotoxicity, and cytoprotection. Frontiers in Pharmacology. 2012;3:p. 136.
    1. Vitek L. The role of bilirubin in diabetes, metabolic syndrome, and cardiovascular diseases. Frontiers in Pharmacology. 2012;3:p. 55.
    1. Stocker R. Antioxidant activities of bile pigments. Antioxidants and Redox Signaling. 2004;6(5):841–849.
    1. Jansen T, Daiber A. Direct antioxidant properties of bilirubin and biliverdin. Is there a role for biliverdin reductase? Frontiers in Pharmacology. 2012;3:p. 30.
    1. Wegiel B, Otterbein LE. Go green: the anti-inflammatory effects of biliverdin reductase. Frontiers in Pharmacology. 2012;3:p. 47.

Source: PubMed

3
Tilaa