125
Views
1
CrossRef citations to date
0
Altmetric
Review

The brain heme oxygenase/biliverdin reductase system as a target in drug research and development

Pages 361-374 | Received 22 Nov 2021, Accepted 09 Mar 2022, Published online: 16 Mar 2022

References

  • Tenhunen R, Marver HS, Schmid R. Microsomal heme oxygenase. Characterization of the enzyme. J Biol Chem. 1969;244:6388–6394. Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
  • Pimstone NR, Tenhunen R, Seitz PT, et al. The enzymatic degradation of hemoglobin to bile pigments by macrophages. J Exp Med. 1971;133:1264–1281.
  • Tenhunen R. Method for microassay of microsomal heme oxygenase activity. Anal Biochem. 1972;45:600–607.
  • Maines MD, Trakshel GM, Kutty RK. Characterization of two constitutive forms of rat liver microsomal heme oxygenase. Only one molecular species of the enzyme is inducible. J Biol Chem. 1986;261:411–419.
  • Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–554.
  • Mancuso C. Biliverdin reductase as a target in drug research and development: facts and hypotheses. Free Radic Biol Med. 2021;172:521–529.
  • Mancuso C. Bilirubin and brain: a pharmacological approach. Neuropharmacology. 2017;118:113–123.
  • Schipper HM, Song W, Tavitian A, et al. The sinister face of heme oxygenase-1 in brain aging and disease. Prog Neurobiol. 2019;172:40–70.
  • Mancuso C, Barone E. The heme oxygenase/biliverdin reductase pathway in drug research and development. Curr Drug Metab. 2009;10:579–594.
  • Mancuso C, Navarra P, Preziosi P. Roles of nitric oxide, carbon monoxide, and hydrogen sulfide in the regulation of the hypothalamic-pituitary-adrenal axis. J Neurochem. 2010;113:563–575.
  • Consoli V, Sorrenti V, Grosso S, et al. Heme oxygenase-1 signaling and redox homeostasis in physiopathological conditions. Biomolecules. 2021;11:589.
  • Siracusa R, Schaufler A, Calabrese V, et al. Carbon monoxide: from poison to clinical trials. Trends Pharmacol Sci. 2021;42:329–339.
  • Takahashi S, Wang J, Rousseau DL, et al. Heme-heme oxygenase complex. Structure of the catalytic site and its implication for oxygen activation. J Biol Chem. 1994;269:1010–1014.
  • Wilks A, Torpey J, Ortiz de Montellano PR. Heme oxygenase (HO-1). Evidence for electrophilic oxygen addition to the porphyrin ring in the formation of alpha-meso-hydroxyheme. J Biol Chem. 1994;269:29553–29556.
  • Vreman HJ, Ekstrand BC, Stevenson DK. Selection of metalloporphyrin heme oxygenase inhibitors based on potency and photoreactivity. Pediatr Res. 1993;33:195–200.
  • Yang G, Nguyen X, Ou J, et al. Unique effects of zinc protoporphyrin on HO-1 induction and apoptosis. Blood. 2001;97:1306–1313.
  • Maines MD, Cohn J. Bile pigment formation by skin heme oxygenase: studies on the response of the enzyme to heme compounds and tissue injury. J Exp Med. 1977;145:1054–1059.
  • Wong RJ, Vreman HJ, Schulz S, et al. In vitro inhibition of heme oxygenase isoenzymes by metalloporphyrins. J Perinatol. 2011;31 Suppl 1:S35–41.
  • Maines MD, Panahian N. The heme oxygenase system and cellular defense mechanisms. Do HO-1 and HO-2 have different functions? Adv Exp Med Biol. 2001;502:249–272.
  • Maines MD. The heme oxygenase system: update 2005. Antioxid Redox Signal. 2005;7:1761–1766.
  • Grochot-Przeczek A, Dulak J, Jozkowicz A. Haem oxygenase-1: non-canonical roles in physiology and pathology. Clin Sci (Lond). 2012;122:93–103.
  • Mancuso C. Heme oxygenase and its products in the nervous system. Antioxid Redox Signal. 2004;6:878–887.
  • Dwyer BE, Nishimura RN, Lu SY. Differential expression of heme oxygenase-1 in cultured cortical neurons and astrocytes determined by the aid of a new heme oxygenase antibody. Response to oxidative stress. Brain Res Mol Brain Res. 1995;30:37–47.
  • Ewing JF, Weber CM, Maines MD. Biliverdin reductase is heat resistant and coexpressed with constitutive and heat shock forms of heme oxygenase in brain. J Neurochem. 1993;61:1015–1023.
  • Ewing JF, Maines MD. Immunohistochemical localization of biliverdin reductase in rat brain: age related expression of protein and transcript. Brain Res. 1995;672:29–41.
  • Medina MV, Sapochnik D, Garcia Solá M, et al. Regulation of the expression of heme oxygenase-1: signal transduction, gene promoter activation, and beyond. Antioxid Redox Signal. 2020;32:1033–1044.
  • Alam J, Cook JL. How many transcription factors does it take to turn on the heme oxygenase-1 gene? Am J Respir Cell Mol Biol. 2007;36:166–174.
  • Kahroba H, Ramezani B, Maadi H, et al. The role of Nrf2 in neural stem/progenitors cells: from maintaining stemness and self-renewal to promoting differentiation capability and facilitating therapeutic application in neurodegenerative disease. Ageing Res Rev. 2021;65:101211.
  • Gureev AP, Popov VN, Starkov AA. Crosstalk between the mTOR and Nrf2/ARE signaling pathways as a target in the improvement of long-term potentiation. Exp Neurol. 2020;328:113285.
  • Catino S, Paciello F, Miceli F, et al. Ferulic acid regulates the nrf2/heme oxygenase-1 system and counteracts trimethyltin-induced neuronal damage in the human neuroblastoma cell line SH-SY5Y. Front Pharmacol. 2016;6:305.
  • Sun J, Hoshino H, Takaku K, et al. Hemoprotein bach1 regulates enhancer availability of heme oxygenase-1 gene. EMBO J. 2002;21:5216–5224.
  • He F, Ru X, Wen T. NRF2, a transcription factor for stress response and beyond. Int J Mol Sci. 2020;21:4777.
  • Maines MD. New insights into biliverdin reductase functions: linking heme metabolism to cell signaling. Physiology (Bethesda). 2005;20:382–389.
  • Palozza P, Serini S, Currò D, et al. beta-Carotene and cigarette smoke condensate regulate heme oxygenase-1 and its repressor factor bach1: relationship with cell growth. Antioxid Redox Signal. 2006;8:1069–1080.
  • Shibahara S. The heme oxygenase dilemma in cellular homeostasis: new insights for the feedback regulation of heme catabolism. Tohoku J Exp Med. 2003;200:167–186.
  • Mancuso C, Preziosi P, Grossman AB, et al. The role of carbon monoxide in the regulation of neuroendocrine function. Neuroimmunomodulation. 1997;4:225–229.
  • Igarashi K, Sun J. The heme-Bach1 pathway in the regulation of oxidative stress response and erythroid differentiation. Antioxid Redox Signal. 2006;8:107–118.
  • Kitamuro T, Takahashi K, Ogawa K, et al. Bach1 functions as a hypoxia-inducible repressor for the heme oxygenase-1 gene in human cells. J Biol Chem. 2003;278:9125–9133.
  • Udono-Fujimori R, Takahashi K, Takeda K, et al. Expression of heme oxygenase-1 is repressed by interferon-gamma and induced by hypoxia in human retinal pigment epithelial cells. Eur J Biochem. 2004;271:3076–3084.
  • Oyake T, Itoh K, Motohashi H, et al. Bach proteins belong to a novel family of BTB-basic leucine zipper transcription factors that interact with mafk and regulate transcription through the NF-E2 site. Mol Cell Biol. 1996;16:6083–6095.
  • Di Domenico F, Pupo G, Mancuso C, et al. Bach1 overexpression in down syndrome correlates with the alteration of the HO-1/BVR-a system: insights for transition to Alzheimer’s disease. J Alzheimers Dis. 2015;44:1107–1120.
  • Salinas M, Wang J, Rosa de Sagarra M, et al. Protein kinase Akt/PKB phosphorylates heme oxygenase-1 in vitro and in vivo. FEBS Lett. 2004;578:90–94.
  • Barone E, Di Domenico F, Sultana R, et al. Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med. 2012;52:3118–3127.
  • Raju VS, Wk M Jr, Maines MD. Regulation of heme oxygenase-2 by glucocorticoids in neonatal rat brain: characterization of a functional glucocorticoid response element. Biochim Biophys Acta. 1997;1351:89–104.
  • Li X, Clark JD. Spinal cord nitric oxide synthase and heme oxygenase limit morphine induced analgesia. Brain Res Mol Brain Res. 2001;95:96–102.
  • Liu L, Dumbrepatil AB, Fleischhacker AS, et al. Heme oxygenase-2 is post-translationally regulated by heme occupancy in the catalytic site. J Biol Chem. 2020;295:17227–17240.
  • Boehning D, Sedaghat L, Sedlak TW, et al. Heme oxygenase-2 is activated by calcium-calmodulin. J Biol Chem. 2004;279:1457–1461.
  • Coburn RF. Endogenous carbon monoxide production. N Engl J Med. 1970;282:207–209.
  • Wu L, Wang R. Carbon monoxide: endogenous production, physiological functions, and pharmacological applications. Pharmacol Rev. 2005;57:585–630.
  • Verma A, Hirsch DJ, Glatt CE, et al. Carbon monoxide: a putative neural messenger. Science. 1993;259:381–384.
  • Steiner AA, Branco LG. Carbon monoxide is the heme oxygenase product with a pyretic action: evidence for a cGMP signaling pathway. Am J Physiol Regul Integr Comp Physiol. 2001;280:R448–57.
  • Lamar CA, Mahesh VB, Brann DW. Regulation of gonadotrophin-releasing hormone (GnRH) secretion by heme molecules: a regulatory role for carbon monoxide? Endocrinology. 1996;137:790–793.
  • Ingi T, Ronnett GV. Direct demonstration of a physiological role for carbon monoxide in olfactory receptor neurons. J Neurosci. 1995;15:8214–8222.
  • Yamamoto T, Nozaki-Taguchi N. Zinc protoporphyrin IX, an inhibitor of the enzyme that produces carbon monoxide, blocks spinal nociceptive transmission evoked by formalin injection in the rat. Brain Res. 1995;704:256–262.
  • Fin C, Schmitz PK, Da Silva RC, et al. Intrahippocampal, but not intra-amygdala, infusion of an inhibitor of heme oxygenase causes retrograde amnesia in the rat. Eur J Pharmacol. 1994;271:227–229.
  • Glaum SR, Miller RJ. Zinc protoporphyrin-IX blocks the effects of metabotropic glutamate receptor activation in the rat nucleus tractus solitarii. Mol Pharmacol. 1993;43:965–969.
  • Faizan M, Muhammad N, Niazi KUK, et al. CO-releasing materials: an emphasis on therapeutic implications, as release and subsequent cytotoxicity are the part of therapy. Materials (Basel). 2019;12(10):1643.
  • Clark JE, Naughton P, Shurey S, et al. Cardioprotective Actions by a Water-Soluble Carbon Monoxide–Releasing Molecule. Circ Res. 2003;93(2):e2–8.
  • Jung E, Koh S-H, Yoo M, et al. Regenerative potential of carbon monoxide in adult neural circuits of the central nervous system. Int J Mol Sci. 2020;21(7):2273.
  • Johnson RA, Johnson FK. The effects of carbon monoxide as a neurotransmitter. Curr Opin Neurol. 2000;13:709–713.
  • Mhillaj E, Papi M, Paciello F, et al. Celecoxib exerts neuroprotective effects in β-amyloid-treated sh-sy5y cells through the regulation of heme oxygenase-1: novel insights for an old drug. Front Cell Dev Biol. 2020;8:561179.
  • Dreyer-Andersen N, Almeida AS, Jensen P, et al. Intermittent, low dose carbon monoxide exposure enhances survival and dopaminergic differentiation of human neural stem cells. PLoS One. 2018;13(1):e0191207.
  • Vieira HLA, Queiroga CSF, Alves PM. Pre-conditioning induced by carbon monoxide provides neuronal protection against apoptosis. J Neurochem. 2008;107(2):585–630.
  • Lin -C-C, Yang -C-C, Hsiao L-D, et al. Heme oxygenase-1 induction by carbon monoxide releasing molecule-3 suppresses interleukin-1β-mediated neuroinflammation. Front Mol Neurosci. 2017;10:387.
  • Schallner N, Romão CC, Biermann J, et al. Carbon monoxide abrogates ischemic insult to neuronal cells via the soluble guanylate cyclase-cGMP pathway. PLoS One. 2013;8:e60672.
  • Jaggar JH, Leffler CW, Cheranov SY, et al. Carbon monoxide dilates cerebral arterioles by enhancing the coupling of Ca 2+ sparks to Ca 2+ -activated K + channels. Circ Res. 2002;91(7):610–617.
  • Choi YK, Park JH, Baek YY, et al. Carbon monoxide stimulates astrocytic mitochondrial biogenesis via L-type Ca2+ channel-mediated PGC-1α/ERRα activation. Biochem Biophys Res Commun. 2016;479:297–304.
  • Riego G, Redondo A, Leánez S, et al. Mechanism implicated in the anti-allodynic and anti-hyperalgesic effects induced by the activation of heme oxygenase 1/carbon monoxide signaling pathway in the central nervous system of mice with neuropathic pain. Biochem Pharmacol. 2018;148:52–63.
  • Basuroy S, Tcheranova D, Bhattacharya S, et al. Nox4 NADPH oxidase-derived reactive oxygen species, via endogenous carbon monoxide, promote survival of brain endothelial cells during TNF-α-induced apoptosis. Am J Physiol Cell Physiol. 2011;300:C256–65.
  • Ryter SW. Heme oxygenase-1/carbon monoxide as modulators of autophagy and inflammation. Arch Biochem Biophys. 2019;678:108186.
  • Queiroga CS, Vercelli A, Vieira HL. Carbon monoxide and the CNS: challenges and achievements. Br J Pharmacol. 2015;172:1533–1545.
  • Zhang J, Piantadosi CA. Mitochondrial oxidative stress after carbon monoxide hypoxia in the rat brain. J Clin Invest. 1992;90:1193–1199.
  • Mancuso C, Tringali G, Grossman A, et al. The generation of nitric oxide and carbon monoxide produces opposite effects on the release of immunoreactive interleukin-1beta from the rat hypothalamus in vitro: evidence for the involvement of different signaling pathways. Endocrinology. 1998;139:1031–1037.
  • Mancuso C, Pistritto G, Tringali G, et al. Evidence that carbon monoxide stimulates prostaglandin endoperoxide synthase activity in rat hypothalamic explants and in primary cultures of rat hypothalamic astrocytes. Brain Res Mol Brain Res. 1997;45:294–300.
  • Hopper CP, Zambrana PN, Goebel U, et al. A brief history of carbon monoxide and its therapeutic origins. Nitric Oxide. 2021;111-112:45–63.
  • Csongradi E, Juncos LA, Drummond HA, et al. Role of carbon monoxide in kidney function: is a little carbon monoxide good for the kidney? Curr Pharm Biotechnol. 2012;13:819–826.
  • Li J, Peng L, Bai W, et al. biliverdin protects against cerebral ischemia/reperfusion injury by regulating the miR-27a-3p/Rgs1 Axis. Neuropsychiatr Dis Treat. 2021;17:1165–1181.
  • Zou ZY, Liu J, Chang C, et al. Biliverdin administration regulates the microRNA-mRNA expressional network associated with neuroprotection in cerebral ischemia reperfusion injury in rats. Int J Mol Med. 2019;43:1356–1372.
  • Bisht K, Wegiel B, Tampe J, et al. Biliverdin modulates the expression of C5aR in response to endotoxin in part via mTOR signaling. Biochem Biophys Res Commun. 2014;449:94–99.
  • Tang LM, Wang YP, Wang K, et al. Exogenous biliverdin ameliorates ischemia-reperfusion injury in small-for-size rat liver grafts. Transplant Proc. 2007;39:1338–1344.
  • Nakao A, Neto JS, Kanno S, et al. Protection against ischemia/reperfusion injury in cardiac and renal transplantation with carbon monoxide, biliverdin and both. Am J Transplant. 2005;5:282–291.
  • Mölzer C, Wallner M, Kern C, et al. Characteristics of the heme catabolic pathway in mild unconjugated hyperbilirubinemia and their associations with inflammation and disease prevention. Sci Rep. 2017;7:755.
  • Schwertner HA, Vítek L. Gilbert syndrome, UGT1A1*28 allele, and cardiovascular disease risk: possible protective effects and therapeutic applications of bilirubin. Atherosclerosis. 2008;198:1–11.
  • Tsiftsoglou AS, Tsamadou AI, Papadopoulou LC. Heme as key regulator of major mammalian cellular functions: molecular, cellular, and pharmacological aspects. Pharmacol Ther. 2006;111:327–345.
  • DeGregorio-Rocasolano N, Martí-Sistac O, Gasull T. Deciphering the iron side of stroke: neurodegeneration at the crossroads between iron dyshomeostasis, excitotoxicity, and ferroptosis. Front Neurosci. 2019;13:85.
  • Chiueh CC. Iron overload, oxidative stress, and axonal dystrophy in brain disorders. Pediatr Neurol. 2001;25:138–147.
  • Song W, Cressatti M, Zukor H, et al. Parkinsonian features in aging GFAP.HMOX1 transgenic mice overexpressing human HO-1 in the astroglial compartment. Neurobiol Aging. 2017;58:163–179.
  • Hascalovici JR, Song W, Liberman A, et al. Neural HO-1/sterol interactions in vivo: implications for Alzheimer’s disease. Neuroscience. 2014;280:40–49.
  • Gáll T, Balla G, Balla JH. Heme oxygenase, and endoplasmic reticulum stress-a new insight into the pathophysiology of vascular diseases. Int J Mol Sci. 2019;20:3675.
  • Ono S, Zhang ZD, Marton LS, et al. Heme oxygenase-1 and ferritin are increased in cerebral arteries after subarachnoid hemorrhage in monkeys. J Cereb Blood Flow Metab. 2000;20:1066–1076.
  • Panahian N, Yoshiura M, Maines MD. Overexpression of heme oxygenase-1 is neuroprotective in a model of permanent middle cerebral artery occlusion in transgenic mice. J Neurochem. 1999;72:1187–1203.
  • Chakrabarty A, Emerson MR, LeVine SM. Heme oxygenase-1 in SJL mice with experimental allergic encephalomyelitis. Mult Scler. 2003;9:372–381.
  • Gonzales S, Erario MA, Tomaro ML. Heme oxygenase-1 induction and dependent increase in ferritin. A protective antioxidant stratagem in hemin-treated rat brain. Dev Neurosci. 2002;24:161–168.
  • Vanderah TW, Pappano AJ. Cholinoceptor-activating & cholinesterase-inhibiting drugs. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 111–128.
  • Nakao A, Kaczorowski DJ, Zuckerbraun BS, et al. Galantamine and carbon monoxide protect brain microvascular endothelial cells by heme oxygenase-1 induction. Biochem Biophys Res Commun. 2008;367:674–679.
  • Ma KG, Qian YH. Alpha 7 nicotinic acetylcholine receptor and its effects on Alzheimer’s disease. Neuropeptides. 2019;73:96–106.
  • Navarro E, Gonzalez-Lafuente L, Pérez-Liébana I, et al. Heme-oxygenase i and pcg-1α regulate mitochondrial biogenesis via microglial activation of alpha7 nicotinic acetylcholine receptors using PNU282987. Antioxid Redox Signal. 2017;27:93–105.
  • Navarro E, Buendia I, Parada E, et al. Alpha7 nicotinic receptor activation protects against oxidative stress via heme-oxygenase I induction. Biochem Pharmacol. 2015;97:473–481.
  • Foucault-Fruchard L, Tronel C, Bodard S, et al. Alpha-7 nicotinic acetylcholine receptor agonist treatment in a rat model of Huntington’s disease and involvement of heme oxygenase-1. Neural Regen Res. 2018;13:737–741.
  • Biaggioni I. Adrenoceptor agonists & sympathomimetic drugs. In: Katzung BG, Vanderah TW, editors. Basic & Clinical Pharmacology. New York (NY): McGraw Hill; 2021. p. 142–160.
  • Khorchid A, Fragoso G, Shore G, et al. Catecholamine-induced oligodendrocyte cell death in culture is developmentally regulated and involves free radical generation and differential activation of caspase-3. Glia. 2002;40:283–299.
  • Schipper HM, Bernier L, Mehindate K, et al. Mitochondrial iron sequestration in dopamine-challenged astroglia: role of heme oxygenase-1 and the permeability transition pore. J Neurochem. 1999;72:1802–1811.
  • Huang YN, Wu CH, Lin TC, et al. Methamphetamine induces heme oxygenase-1 expression in cortical neurons and glia to prevent its toxicity. Toxicol Appl Pharmacol. 2009;240:315–326.
  • Trevor AJ. Sedative-hypnotic drugs. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 395–409.
  • Carvalho-Costa PG, Branco LG, Leite-Panissi CR. Activation of locus coeruleus heme oxygenase-carbon monoxide pathway promoted an anxiolytic-like effect in rats. Braz J Med Biol Res. 2016;49:e5135.
  • Rogawski MA. Antiseizure drugs. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 422–455.
  • Park CW, Ahn JH, Lee TK, et al. Post-treatment with oxcarbazepine confers potent neuroprotection against transient global cerebral ischemic injury by activating Nrf2 defense pathway. Biomed Pharmacother. 2020;124:109850.
  • Liao G, Li R, Chen X, et al. Sodium valproate prevents radiation-induced injury in hippocampal neurons via activation of the Nrf2/HO-1 pathway. Neuroscience. 2016;331:40–51.
  • DeBattista C. Antidepressant agents. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 550–572.
  • Omar NN, Tash RF. Fluoxetine coupled with zinc in a chronic mild stress model of depression: providing a reservoir for optimum zinc signaling and neuronal remodeling. Pharmacol Biochem Behav. 2017;160:30–38.
  • Engel DF, de Oliveira J, Lieberknecht V, et al. Duloxetine protects human neuroblastoma cells from oxidative stress-induced cell death through Akt/Nrf-2/HO-1 pathway. Neurochem Res. 2018;43:387–396.
  • Chen Z, Xu H, Haimano S, et al. Quetiapine and venlafaxine synergically regulate heme oxygenase-2 protein expression in the hippocampus of stressed rats. Neurosci Lett. 2005;389:173–177.
  • Tran NQV, Nguyen AN, Takabe K, et al. Pre-treatment with amitriptyline causes epigenetic up-regulation of neuroprotection-associated genes and has anti-apoptotic effects in mouse neuronal cells. Neurotoxicol Teratol. 2017;62:1–12.
  • Lin HY, Yeh WL, Huang BR, et al. Desipramine protects neuronal cell death and induces heme oxygenase-1 expression in Mes23.5 dopaminergic neurons. PLoS One. 2012;7:e50138.
  • De Battista C. Antipsychotic agents & lithium. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 529–549.
  • Terada K, Murata A, Toki E, et al. Atypical antipsychotic drug ziprasidone protects against rotenone-induced neurotoxicity: an in vitro study. Molecules. 2020;25:4206.
  • Lee YS, Park SY, Heo HJ, et al. Multitarget-directed cotreatment with cilostazol and aripiprazole for augmented neuroprotection against oxidative stress-induced toxicity in HT22 mouse hippocampal cells. Eur J Pharmacol. 2019;857:172454.
  • Schumacher MA, Basbaum AI, Naidu RK. Opioid agonists & antagonists. In: Katzung BG, Vanderah TW, editors. Basic & Clinical Pharmacology. New York (NY): McGraw Hill; 2021. p. 573–595.
  • Liang D, Li X, Lighthall G, et al. Heme oxygenase type 2 modulates behavioral and molecular changes during chronic exposure to morphine. Neuroscience. 2003;121:999–1005.
  • Li X, Angst MS, Clark JD. A murine model of opioid-induced hyperalgesia. Brain Res Mol Brain Res. 2001;86:56–62.
  • Rabbani M, Hajhashemi V, Mesripour A. Increase in brain corticosterone concentration and recognition memory impairment following morphine withdrawal in mice. Stress. 2009;12:451–456.
  • Núñez C, Földes A, Pérez-Flores D, et al. Elevated glucocorticoid levels are responsible for induction of tyrosine hydroxylase mRNA expression, phosphorylation, and enzyme activity in the nucleus of the solitary tract during morphine withdrawal. Endocrinology. 2009;150:3118–3127.
  • Castany S, Carcolé M, Leánez S, et al. the induction of heme oxygenase 1 decreases painful diabetic neuropathy and enhances the antinociceptive effects of morphine in diabetic mice. PLoS One. 2016;11:e0146427.
  • Castany S, Carcolé M, Leánez S, et al. The antinociceptive effects of a δ-opioid receptor agonist in mice with painful diabetic neuropathy: involvement of heme oxygenase 1. Neurosci Lett. 2016;614:49–54.
  • de Ávila Ma P, Giusti-Paiva A, de Oliveira Nascimento C G. The peripheral antinociceptive effect induced by the heme oxygenase/carbon monoxide pathway is associated with ATP-sensitive K+ channels. Eur J Pharmacol. 2014;726:41–48.
  • Abdel Aziz MT, Atta HM, Samer H, et al. Heme oxygenase effect on mesenchymal stem cells action on experimental Alzheimer’s disease. EXCLI J. 2013;12:778–792.
  • Bhardwaj M, Deshmukh R, Kaundal M, et al. Pharmacological induction of hemeoxygenase-1 activity attenuates intracerebroventricular streptozotocin induced neurocognitive deficit and oxidative stress in rats. Eur J Pharmacol. 2016;772:43–50.
  • Howlett D, Cutler P, Heales S, et al. Hemin and related porphyrins inhibit beta-amyloid aggregation. FEBS Lett. 1997;417:249–251.
  • Delmastro-Greenwood MM, Tse HM, Piganelli JD. Effects of metalloporphyrins on reducing inflammation and autoimmunity. Antioxid Redox Signal. 2014;20:2465–2477.
  • Sheng H, Chaparro RE, Sasaki T, et al. Metalloporphyrins as therapeutic catalytic oxidoreductants in central nervous system disorders. Antioxid Redox Signal. 2014;20:2437–2464.
  • Salerno L, Floresta G, Ciaffaglione V, et al. Progress in the development of selective heme oxygenase-1 inhibitors and their potential therapeutic application. Eur J Med Chem. 2019;167:439–453.
  • Vlahakis JZ, Kinobe RT, Bowers RJ, et al. Synthesis and evaluation of azalanstat analogues as heme oxygenase inhibitors. Bioorg Med Chem Lett. 2005;15:1457–1461.
  • Fernández-Fierro A, Funes SC, Rios M, et al. Immune modulation by inhibitors of the HO system. Int J Mol Sci. 2020;22:294.
  • Mucha O, Podkalicka P, Mikulski M, et al. Development and characterization of a new inhibitor of heme oxygenase activity for cancer treatment. Arch Biochem Biophys. 2019;671:130–142.
  • Gupta A, Lacoste B, Pistell PJ, et al. Neurotherapeutic effects of novel HO-1 inhibitors in vitro and in a transgenic mouse model of Alzheimer’s disease. J Neurochem. 2014;131(6):778–790.
  • Katzung BG. Introduction: the nature of drugs and drug development and regulation. In: Katzung BG, Vanderah TW, editors. Basic & clinical pharmacology. New York (NY): McGraw Hill; 2021. p. 1–20.
  • Syapin PJ. Regulation of haeme oxygenase-1 for treatment of neuroinflammation and brain disorders. Br J Pharmacol. 2008;155(5):623–640.
  • Kaizaki A, Tanaka S, Ishige K, et al. The neuroprotective effect of heme oxygenase (HO) on oxidative stress in HO-1 siRNA-transfected HT22 cells. Brain Res. 2006;1108:39–44.
  • Fredenburgh LE, Perrella MA, Barragan-Bradford D, et al. A phase I trial of low-dose inhaled carbon monoxide in sepsis-induced ARDS. JCI Insight. 2018;3:e124039.
  • Rosas IO, Goldberg HJ, Collard HR, et al. A phase II clinical trial of low-dose inhaled carbon monoxide in idiopathic pulmonary fibrosis. Chest. 2018;153:94–104.
  • Goodrich JA, Frisco DJ, Ryan SPP, et al. Intermittent low dose carbon monoxide inhalation does not influence glucose regulation in overweight adults: a randomized controlled crossover trial. Exp Physiol. 2020;105:460–467.
  • Schmidt WFJ, Hoffmeister T, Haupt S, et al. chronic exposure to low-dose carbon monoxide alters hemoglobin mass and V˙O2max. Med Sci Sports Exerc. 2020;52:1879–1887.
  • Ghanizada H, Arngrim N, Schytz HW, et al. Carbon monoxide inhalation induces headache but no migraine in patients with migraine without aura. Cephalalgia. 2018;38:1940–1949.
  • Abu Jawdeh BG, Woodle ES, Leino AD, et al. A phase Ib, open-label, single arm study to assess the safety, pharmacokinetics, and impact on humoral sensitization of SANGUINATE infusion in patients with end-stage renal disease. Clin Transplant. 2018;32:e13155.
  • Misra H, Lickliter J, Kazo F, et al. PEGylated carboxyhemoglobin bovine (SANGUINATE): results of a phase I clinical trial. Artif Organs. 2014;38:702–707.
  • Coburn RF. The partial pressure of carbon monoxide in human tissues calculated using a parallel capillary-tissue cylinder model. J Appl Physiol. 2018;124:761–768. (1985).
  • Rennie J, Burman-Roy S, Murphy MS; Guideline Development Group. Neonatal jaundice: summary of NICE guidance. BMJ. 2010;340:c2409.
  • Suresh GK, Martin CL, Soll RF. Metalloporphyrins for treatment of unconjugated hyperbilirubinemia in neonates. Cochrane Database Syst Rev. 2003;2. CD004207.
  • Dennery PA. Metalloporphyrins for the treatment of neonatal jaundice. Curr Opin Pediatr. 2005;17:167–169.
  • Böni RE, Huch Böni RA, Galbraith RA, et al. Tin-mesoporphyrin inhibits heme oxygenase activity and heme-iron absorption in the intestine. Pharmacology. 1993;47:318–329.
  • Cooke RW. New approach to prevention of kernicterus. Lancet. 1999;353:1814–1815.
  • Fasae KD, Abolaji AO, Faloye TR, et al. Metallobiology and therapeutic chelation of biometals (copper, zinc and iron) in Alzheimer’s disease: limitations, and current and future perspectives. J Trace Elem Med Biol. 2021;67:126779.
  • Ward RJ, Dexter DT, Martin-Bastida A, et al. Is chelation therapy a potential treatment for Parkinson’s disease? Int J Mol Sci. 2021;22:3338.
  • Devos D, Moreau C, Devedjian JC, et al. Targeting chelatable iron as a therapeutic modality in Parkinson’s disease. Antioxid Redox Signal. 2014;21:195–210.
  • Martin-Bastida A, Ward RJ, Newbould R, et al. Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease. Sci Rep. 2017;7:1398.
  • McLachlan DR C, Dalton AJ, Kruck TP, et al. Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet. 1991;337:1304–1308.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.