Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 29.
Published in final edited form as: Ther Targets Neurol Dis. 2015 Jan 3;2(1):474. doi: 10.14800/ttnd.474

Targeting heme oxygenase after intracerebral hemorrhage

Jing Chen-Roetling 1, Xiangping Lu 1, Raymond F Regan 1
PMCID: PMC4310000  NIHMSID: NIHMS657423  PMID: 25642455

Abstract

Intracerebral hemorrhage (ICH) is the primary event in approximately 10% of strokes, and has higher rates of morbidity and mortality than ischemic stroke. Experimental evidence suggests that the toxicity of hemoglobin and its degradation products contributes to secondary injury that may be amenable to therapeutic intervention. Hemin, the oxidized form of heme, accumulates in intracranial hematomas to cytotoxic levels. The rate limiting step of its breakdown is catalyzed by the heme oxygenase (HO) enzymes, which consist of inducible HO-1 and constitutively-expressed HO-2. The effect of these enzymes on perihematomal injury and neurological outcome has been investigated in ICH models using both genetic and pharmacological approaches to alter their expression, with variable results reported. These findings are summarized and reconciled in this review; therapeutic strategies that may optimize HO expression and activity after ICH are described.

Keywords: intracerebral hemorrhage, iron, oxidative stress, stroke, subarachnoid hemorrhage

Introduction

Intracerebral hemorrhage (ICH) deposits a ~10 mM suspension of hemoglobin-bound heme into the brain parenchyma. Although initially sequestered within erythrocytes, some will be released and account for the high micromolar concentration of oxidized heme (hemin) present in one week old intracranial hematomas [1]. The mechanisms responsible for this hemin accumulation to cytotoxic levels have not been precisely defined. However, brain tissue is clearly a poor microenvironment for erythrocyte survival. Whether due to complement activation [2], heme-mediated oxidative stress [3] or other mechanisms, they begin to lyse within a few days in rodent models and in vitro [4, 5]. Extracellular hemoglobin is unstable at physiological temperatures and spontaneously oxidizes at its heme groups to ferric methemoglobin, releasing a superoxide ion in the reaction [6]. The affinity of methemoglobin for its heme moieties is relatively weak, and it readily transfers them to higher-affinity lipid and protein binding sites [7]. In the CNS, the low concentration of the heme/hemin binding protein hemopexin (~5 μg/ml [8]) increases the likelihood of hemin uptake by vulnerable cell populations such as neurons and astrocytes. Some hemoglobin in the hematoma will be scavenged by microglia and infiltrating macrophages, which phagocytose intact erythrocytes and also take up hemoglobin and hemoglobin-haptoglobin complexes via the CD163 receptor [9, 10]. The fate of hemoglobin-bound heme after uptake by these cells remains undefined. However, when intracellular free heme or hemin is present in excess, it is very likely that at least some will be ejected into the extracellular space by the exporter protein FLVCR1 [11, 12], and will then be available to participate in free radical reactions.

The accumulation of hemin to toxic concentrations within a hematoma provides an obvious therapeutic target after ICH. The goal of one approach is optimization of hemin catabolism. The latter is catalyzed by the heme oxygenase (HO) enzymes, resulting in production of equimolar iron, carbon monoxide, and biliverdin, which is then converted to bilirubin via biliverdin reductase [13]. Two HO isoforms are present in mammalian cells. HO-1 is expressed at a very low level in the healthy CNS, but it is rapidly induced by hemin and a variety of other oxidants, as well as heat shock. HO-2 is constitutively expressed and is the predominant neuronal isoform; it accounts for most HO activity under normal conditions [14]. Investigation of the effect of these enzymes on cell injury and outcome after ICH has been limited by a lack of specific and selective inhibitors, necessitating the use of genetically modified mice in mechanistic studies.

Effect of HO-2 knockout

Mice lacking the HO-2 gene were initially produced two decades ago by Poss and Tonegawa [15]. Their descendants have been used to evaluate the effect of HO-2 in studies that are directly relevant to ICH. HO-2 gene deletion per se has no significant effect on breeding, fetal viability, or postnatal growth, all of which are grossly indistinguishable from their wild-type counterparts. It has consistently altered outcome after experimental ICH, although in a direction that varies with the model used.

Experimental models of ICH

Two ICH models are currently in use, but their relevance to clinical ICH is a source of ongoing controversy. ICH is induced by stereotactic injection of either autologous blood or clostridial collagenase, which disrupts local blood vessels. The former model allows for more precise control of hematoma volumes. However, backflow of blood through the needle entry site is a potential source of variability, particularly with injection of more than 15 μl in mice. The major criticism of this model is that it fails to simulate clinical ICH, since the hematoma is not produced by hemorrhage in situ[16]. The collagenase model addresses that concern, but raises others. Since a bacterial protein is injected, a local inflammatory response is possible, although published data quantifying inflammation in the absence of hemorrhage (e.g. by injecting an inactivated enzyme) are lacking. Another concern is that disruption of the microvascular network in the vicinity of the injection site by collagenase digestion may reduce local blood flow sufficiently to produce ischemia, which does not appear to be a prominent feature of clinical ICH and has not been observed when ICH was modeled by blood injection[17, 18]. MacLellan et al. [19] reported that the collagenase model produced more rapid and pronounced breakdown of the blood-brain barrier, greater injury to adjacent tissue, and more severe and persistent neurological deficits than the blood injection model.

HO-2 KO reduces cell injury in the blood injection ICH model

Stereotactic injection of blood into the mouse striatum is followed by reduced viability of perihematomal cells over the following four days, as detected by ex vivo MTT assay or quantification of striatal fluorescence in mice expressing the red fluorescent protein dTomato in central neurons. The validity of these methods has been established by their close agreement with cell counts of histological sections guided by design-based stereology [20]. Compared with wild-type mice, HO-2 knockouts sustained significantly less cell loss at four days, a discrepancy that remained constant when the observation period was extended to 8 days[21]. HO-2 KO also significantly reduced striatal protein and lipid oxidation, as well as striatal expression of HO-1, which is induced by oxidative stress due to the presence of antioxidant response elements in its promoter region [22]. Very similar results were observed when mice were injected with an autologous solution of stroma-free hemoglobin rather than blood, suggesting that similar mechanisms mediate cell injury in these two models [23]. However, the protection provided by HO-2 was associated with only a weak and inconsistent improvement in neurological outcome[21].

In vitro studies

HO-2 KO also protected neurons from hemoglobin or hemin in cell culture [24, 25]. This effect could be mimicked by treatment with deferoxamine or apotransferrin, which bind iron with very high affinity and in a manner that reduces or prevents its participation in free radical reactions [26, 27]. These observations support the hypothesis that HO-2 is deleterious to neurons exposed low micromolar hemoglobin or hemin concentrations due to iron release as a consequence of heme/hemin breakdown. This effect may be specific to neurons, which are very sensitive to iron due at least in part to low expression of the iron sequestering protein ferritin [28, 29]. HO-2 KO astrocytes, which are capable of rapid ferritin upregulation, are conversely more vulnerable to hemin than their wild-type counterparts [30]. Cultured HO-2 KO neurons are more vulnerable to inorganic iron, perhaps due to the protective effect of the other products of heme breakdown, i.e. biliverdin/bilirubin and carbon monoxide, when exposure to iron was equalized.

In addition to toxicity mediated by iron release, hemin can directly injure cells by oxidative and membrane destabilizing effects [31, 32]. These mechanisms will predominate if iron toxicity is mitigated by concomitant chelator administration. When Wang et al. treated cultured neurons with hemin in medium containing iron-poor transferrin, they observed that HO-2 KO increased cell death [33]. The net effect of HO-2 therefore appears to be dependent on the iron binding capacity of the cellular microenvironment. When cells are exposed to equal concentrations of redox-active iron, then HO-2 is protective.

HO-2 KO worsens outcome in the collagenase ICH model

In contrast to its protective effect in the blood injection ICH model, HO-2 KO increased perihematomal lesion volume, neuroinflammation, and edema as measured by hemisphere volume after striatal collagenase injection; however, tissue water content was not altered [33, 34]. Neurological deficits were also more severe in the first three days after hemorrhage. Recent studies in our laboratory have confirmed the latter finding, although the effect was somewhat less robust [20]. Also in contrast to its effect in the blood injection ICH model, HO-2 KO had no effect on striatal neuron loss or tissue viability in the collagenase ICH model [20]. These disparate results suggest the predominance of different injury mechanisms in the collagenase and blood injection ICH models. A plausible but still speculative explanation for this discrepancy is that the protective effect of HO-2 KO on heme-mediated neurotoxicity is counteracted in the collagenase model by a deleterious effect on other injury mechanisms, such as inflammation, ischemia, or iron neurotoxicity.

Effect of HO-1 KO

In contrast the grossly healthy phenotype of HO-2 KO mice, unconditional HO-1 knockouts breed poorly and have a very high pre- and perinatal mortality. Due primarily to this limited availability, only one published study has evaluated the effect of HO-1 gene deletion on outcome after experimental ICH. Using the collagenase model, Wang and Doré reported smaller lesion volumes and reduced neuroinflammation in knockouts than in wild-type mice [35]. Early neurological deficits were also mitigated, but this effect was statistically significant only on day 1. The effect of HO-1 in the collagenase model was therefore the opposite of HO-2. Although these enzymes catalyze the same reaction, their expression differs markedly in both location and timing after ICH, which may account for their divergent effects. As noted above, neurons constitutively express HO-2 but little HO-1, with little change after ICH. Astrocytes, microglia, and endothelial cells robustly increase HO-1 expression after hemoglobin or hemin exposure. Delineation of the effect of HO-1 and HO-2 knockout or overexpression in specific cell populations after ICH seems a worthy topic for further investigation.

Pharmacological Studies

HO Inhibitors

A variety of non-iron heme analogs that competitively inhibit the activity of both HO-1 and HO-2 are commercially available. These are reactive compounds with significant off-target effects, including inhibition of nitric oxide synthase and soluble guanyl cyclase inhibition, albeit at higher concentrations than that needed for HO inhibition[36]. Another drawback to the use of these compounds as mechanistic probes is that they may induce HO-1 [37], raising the possibility that post-treatment HO activity may be increased compared with vehicle-treated controls as local drug levels decline. Furthermore, HO-1 protein may be protective even if catalytically inactive by upregulating antioxidant gene expression [38]. With the caveat that the relationship between brain or systemic administration of these compounds and perihematomal HO activity has never been established, porphyrin HO inhibitors have been consistently protective in blood injection ICH models. Wagner et al. reported that tin mesoporphyrin injected with autologous blood into pig frontal white matter reduced edema at 24 hours compared with DMSO vehicle controls [39]. A similar effect was produced in rats receiving striatal infusions of hemoglobin containing tin protoporphyrin [40]. Using a more translational approach, Gong et al. observed that zinc protoporphyrin, administered as a continuous i.p. infusion via osmotic pump, reduced brain edema, atrophy, ventricular enlargement and neurological deficits in a rat autologous blood ICH model, with a time window of 6 hours [41]. Efficacy in a collagenase ICH model has not yet been described.

Perivascular HO-1 induction

Hemin in a sodium carbonate buffer for intravenous infusion (hematin) has been in clinical use in the USA for decades to treat attacks of acute porphyria[42]; hemin arginate has been marketed in Europe for the same indication. After injection, hemin binds with extraordinary affinity to hemopexin [43], or to albumin when hemopexin binding sites are saturated [44]. In vitro, both complexes are capable of inducing HO-1 at a somewhat reduced rate than hemin per se and with markedly less toxicity [45, 46]. Systemically administered hemin has little or no access to the CNS parenchyma, but induces HO-1 in perivascular cells [47]. We have recently reported that mice treated with hemin i.p. after striatal collagenase or blood injection sustained less blood-brain barrier breakdown, perihematomal cell loss, and neurological deficits than mice receiving vehicle, with a time window of at least 3 hours [48]. Similar protection has also been provided by hemin treatment before spinal cord trauma [47], as well as focal and global brain ischemia [49, 50], although it is unknown if post-injury therapy has any benefit in these models.

Conclusions

The variable effects of HO-1 and HO-2 in models relevant to ICH suggest that they are challenging therapeutic targets. Given the multiple injury cascades initiated by clinical ICH [51], demonstration of efficacy in only one ICH model is an insufficient recommendation for further development of drug or genetic therapies. Administration of HO inhibitors, for example, may attenuate neuron loss due to iron release as heme/hemin is catabolized, while also increasing the vulnerability of neurons and other cell populations to the toxicity of hemin itself and enhancing the inflammatory response. Maximal benefit may require a combinatorial approach using two of more agents to increase HO activity while detoxifying iron with chelators or antioxidants. Therapies focused on increasing HO-1 expression in perivascular cells appear to be particularly promising given the consistent benefit of systemic hemin after striatal blood or collagenase injection, and the protective effect that has already been demonstrated in other acute CNS injury models.

Acknowledgments

Supported by a grant from the NIH to RFR (RO1NS079500).

Footnotes

Conflict of interest statement

The authors declare that they have no conflict of interest.

References

  • 1.Letarte PB, Lieberman K, Nagatani K, Haworth RA, Odell GB, Duff TA. Hemin: levels in experimental subarachnoid hematoma and effects on dissociated vascular smooth muscle cells. J Neurosurg. 1993;79:252–255. doi: 10.3171/jns.1993.79.2.0252. [DOI] [PubMed] [Google Scholar]
  • 2.Hua Y, Xi G, Keep RF, Hoff JT. Complement activation in the brain after experimental intracerebral hemorrhage. J Neurosurg. 2000;92:1016–1022. doi: 10.3171/jns.2000.92.6.1016. [DOI] [PubMed] [Google Scholar]
  • 3.Baysal E, Monteiro HP, Sullivan SG, Stern A. Desferrioxamine protects human red blood cells from hemin-induced hemolysis. Free Radic Biol Med. 1990;9:5–10. doi: 10.1016/0891-5849(90)90043-i. [DOI] [PubMed] [Google Scholar]
  • 4.Wagner KR, Dwyer BE. Hematoma removal, heme, and heme oxygenase following hemorrhagic stroke. Ann NY Acad Sci. 2004;1012:237–251. doi: 10.1196/annals.1306.020. [DOI] [PubMed] [Google Scholar]
  • 5.Jaremko KM, Chen-Roetling J, Chen L, Regan RF. Accelerated Hemolysis and Neurotoxicity in Neuron-Glia-Blood Clot Co-cultures. J Neurochem. 2010;114:1063–73. doi: 10.1111/j.1471-4159.2010.06826.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Alyash A. Redox and radical reactions of hemoglobin solutions: toxicities and protective strategies. In: Winslow R, editor. Blood Substitutes. Academic Press; London: 2006. pp. 197–205. [Google Scholar]
  • 7.Bunn HF, Jandl JH. Exchange of heme along hemoglobins and between hemoglobin and albumin. J Biol Chem. 1968;243:465–475. [PubMed] [Google Scholar]
  • 8.Roher AE, Maarouf CL, Sue LI, Hu Y, Wilson J, Beach TG. Proteomics-derived cerebrospinal fluid markers of autopsy-confirmed Alzheimer’s disease. Biomarkers. 2009;14:493–501. doi: 10.3109/13547500903108423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kristiansen M, Graversen JH, Jacobsen C, Sonne O, Hoffman HJ, Law SK, et al. Identification of the haemoglobin scavenger receptor. Nature. 2001;409:198–201. doi: 10.1038/35051594. [DOI] [PubMed] [Google Scholar]
  • 10.Schaer DJ, Schaer CA, Buehler PW, Boykins RA, Schoedon G, Alayash AI, et al. CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin. Blood. 2006;107:373–80. doi: 10.1182/blood-2005-03-1014. [DOI] [PubMed] [Google Scholar]
  • 11.Yang Z, Philips JD, Doty RT, Giraudi P, Ostrow JD, Tiribelli C, et al. Kinetics and specificity of feline leukemia virus subgroup C receptor (FLVCR) export function and its dependence on hemopexin. J Biol Chem. 2010;285:28874–82. doi: 10.1074/jbc.M110.119131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen-Roetling J, Cai Y, Lu X, Regan RF. Hemin uptake and release by neurons and glia. Free Radic Res. 2014;48:200–5. doi: 10.3109/10715762.2013.859386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Abraham NG, Kappas A. Pharmacological and Clinical Aspects of Heme Oxygenase. Pharmacol Rev. 2008;60:79–127. doi: 10.1124/pr.107.07104. [DOI] [PubMed] [Google Scholar]
  • 14.Schipper HM, Song W, Zukor H, Hascalovici JR, Zeligman D. Heme oxygenase-1 and neurodegeneration: expanding frontiers of engagement. J Neurochem. 2009;110:469–85. doi: 10.1111/j.1471-4159.2009.06160.x. [DOI] [PubMed] [Google Scholar]
  • 15.Poss KD, Thomas MJ, Ebralidze AK, TJOD, Tonegawa S. Hippocampal long-term potentiation is normal in heme oxygenase-2 mutant mice. Neuron. 1995;15:867–873. doi: 10.1016/0896-6273(95)90177-9. [DOI] [PubMed] [Google Scholar]
  • 16.James ML, Warner DS, Laskowitz DT. Preclinical models of intracerebral hemorrhage: a translational perspective. Neurocrit Care. 2008;9:139–52. doi: 10.1007/s12028-007-9030-2. [DOI] [PubMed] [Google Scholar]
  • 17.Qureshi AI, Wilson DA, Hanley DF, Traystman RJ. No evidence for an ischemic penumbra in massive experimental intracerebral hemorrhage. Neurology. 1999;52:266–72. doi: 10.1212/wnl.52.2.266. [DOI] [PubMed] [Google Scholar]
  • 18.Zazulia AR, Diringer MN, Videen TO, Adams RE, Yundt K, Aiyagari V, et al. Hypoperfusion without ischemia surrounding acute intracerebral hemorrhage. J Cereb Blood Flow Metab. 2001;21:804–10. doi: 10.1097/00004647-200107000-00005. [DOI] [PubMed] [Google Scholar]
  • 19.MacLellan CL, Silasi G, Poon CC, Edmundson CL, Buist R, Peeling J, et al. Intracerebral hemorrhage models in rat: comparing collagenase to blood infusion. J Cereb Blood Flow Metab. 2008;28:516–25. doi: 10.1038/sj.jcbfm.9600548. [DOI] [PubMed] [Google Scholar]
  • 20.Chen-Roetling J, Lu X, Regan KA, Regan RF. A rapid fluorescent method to quantify neuronal loss after experimental intracerebral hemorrhage. J Neurosci Methods. 2013;216:128–36. doi: 10.1016/j.jneumeth.2013.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen-Roetling J, Cai Y, Regan RF. Neuroprotective effect of heme oxygenase-2 knockout in the blood injection model of intracerebral hemorrhage. BMC Res Notes. 2014;7:561. doi: 10.1186/1756-0500-7-561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Qu Y, Chen-Roetling J, Benvenisti-Zarom L, Regan RF. Attenuation of oxidative injury after induction of experimental intracerebral hemorrhage in heme oxygenase-2 knockout mice. J Neurosurg. 2007;106:428–35. doi: 10.3171/jns.2007.106.3.428. [DOI] [PubMed] [Google Scholar]
  • 23.Qu Y, Chen J, Benvenisti-Zarom L, Ma X, Regan RF. Effect of targeted deletion of the heme oxygenase-2 gene on hemoglobin toxicity in the striatum. J Cereb Blood Flow Metab. 2005;25:1466–1475. doi: 10.1038/sj.jcbfm.9600143. [DOI] [PubMed] [Google Scholar]
  • 24.Rogers B, Yakopson V, Teng ZP, Guo Y, Regan RF. Heme oxygenase-2 knockout neurons are less vulnerable to hemoglobin toxicity. Free Rad Biol Med. 2003;35:872–881. doi: 10.1016/s0891-5849(03)00431-3. [DOI] [PubMed] [Google Scholar]
  • 25.Regan RF, Chen J, Benvenisti-Zarom L. Heme oxygenase-2 gene deletion attenuates oxidative stress in neurons exposed to extracellular hemin. BMC Neurosci. 2004;5:34. doi: 10.1186/1471-2202-5-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Regan RF, Rogers B. Delayed treatment of hemoglobin neurotoxicity. J Neurotrauma. 2003;20:111–120. doi: 10.1089/08977150360517236. [DOI] [PubMed] [Google Scholar]
  • 27.Chen-Roetling J, Chen L, Regan RF. Apotransferrin protects cortical neurons from hemoglobin toxicity. Neuropharmacology. 2011;60:423–31. doi: 10.1016/j.neuropharm.2010.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kress GJ, Dineley KE, Reynolds IJ. The relationship between intracellular free iron and cell injury in cultured neurons, astrocytes, and oligodendrocytes. J Neurosci. 2002;22:5848–55. doi: 10.1523/JNEUROSCI.22-14-05848.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Moos T, Morgan EH. The metabolism of neuronal iron and its pathogenic role in neurological disease: review. Ann N Y Acad Sci. 2004;1012:14–26. doi: 10.1196/annals.1306.002. [DOI] [PubMed] [Google Scholar]
  • 30.Chen J, Regan RF. Heme oxygenase-2 gene deletion increases astrocyte vulnerability to hemin. Biochem Biophys Res Commun. 2004;318:88–94. doi: 10.1016/j.bbrc.2004.03.187. [DOI] [PubMed] [Google Scholar]
  • 31.Chou AC, Fitch CD. Mechanism of hemolysis induced by ferriprotoporphyrin IX. J Clin Invest. 1981;68:672–677. doi: 10.1172/JCI110302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Robinson SR, Dang TN, Dringen R, Bishop GM. Hemin toxicity: a preventable source of brain damage following hemorrhagic stroke. Redox Rep. 2009;14:228–35. doi: 10.1179/135100009X12525712409931. [DOI] [PubMed] [Google Scholar]
  • 33.Wang J, Zhuang H, Doré S. Heme oxygenase 2 is neuroprotective against intracerebral hemorrhage. Neurobiol Dis. 2006;22:473–476. doi: 10.1016/j.nbd.2005.12.009. [DOI] [PubMed] [Google Scholar]
  • 34.Wang J, Dore S. Heme oxygenase 2 deficiency increases brain swelling and inflammation after intracerebral hemorrhage. Neuroscience. 2008;155:1133–41. doi: 10.1016/j.neuroscience.2008.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang J, Doré S. Heme oxygenase-1 exacerbates early brain injury after intracerebral haemorrhage. Brain. 2007;130:1643–52. doi: 10.1093/brain/awm095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Appleton SD, Chretien ML, McLaughlin BE, Vreman HJ, Stevenson DK, Brien JF, et al. Selective inhibition of heme oxygenase, without inhibition of nitric oxide synthase or soluble guanylyl cyclase, by metalloporphyrins at low concentrations. Drug Metab Disp. 1999;27:1214–1219. [PubMed] [Google Scholar]
  • 37.Yang G, Nguyen X, Ou J, Rekulapelli P, Stevenson DK, Dennery PA. Unique effects of zinc protoporphyrin on HO-1 induction and apoptosis. Blood. 2001;97:1306–13. doi: 10.1182/blood.v97.5.1306. [DOI] [PubMed] [Google Scholar]
  • 38.Lin Q, Weis S, Yang G, Weng YH, Helston R, Rish K, et al. Heme oxygenase-1 protein localizes to the nucleus and activates transcription factors important in oxidative stress. J Biol Chem. 2007;282:20621–33. doi: 10.1074/jbc.M607954200. [DOI] [PubMed] [Google Scholar]
  • 39.Wagner KR, Hua Y, de Courten-Myers GM, Broderick JP, Nishimura RN, Lu SY, et al. Tin-mesoporphyrin, a potent heme oxygenase inhibitor, for treatment of intracerebral hemorrhage: in vivo and in vitro studies. Cell Mol Biol. 2000;46:597–608. [PubMed] [Google Scholar]
  • 40.Huang FP, Xi G, Keep RF, Hua Y, Nemoianu A, Hoff JT. Brain edema after experimental intracerebral hemorrhage: role of hemoglobin degradation products. J Neurosurg. 2002;96:287–293. doi: 10.3171/jns.2002.96.2.0287. [DOI] [PubMed] [Google Scholar]
  • 41.Gong Y, Tian H, Xi G, Keep RF, Hoff JT, Hua Y. Systemic zinc protoporphyrin administration reduces intracerebral hemorrhage-induced brain injury. Acta Neurochir Suppl. 2006;96:232–6. doi: 10.1007/3-211-30714-1_50. [DOI] [PubMed] [Google Scholar]
  • 42.Anderson KE, Bloomer JR, Bonkovsky HL, Kushner JP, Pierach CA, Pimstone NR, et al. Recommendations for the diagnosis and treatment of the acute porphyrias. Ann Intern Med. 2005;142:439–50. doi: 10.7326/0003-4819-142-6-200503150-00010. [DOI] [PubMed] [Google Scholar]
  • 43.Hrkal Z, Vodrazka Z, Kalousek I. Transfer of heme from ferrihemoglobin and ferrihemoglobin isolated chains to hemopexin. Eur J Biochem. 1974;43:73–8. doi: 10.1111/j.1432-1033.1974.tb03386.x. [DOI] [PubMed] [Google Scholar]
  • 44.Siegert SW, Holt RJ. Physicochemical properties, pharmacokinetics, and pharmacodynamics of intravenous hematin: a literature review. Adv Ther. 2008;25:842–57. doi: 10.1007/s12325-008-0094-y. [DOI] [PubMed] [Google Scholar]
  • 45.Li RC, Saleem S, Zhen G, Cao W, Zhuang H, Lee J, et al. Heme-hemopexin complex attenuates neuronal cell death and stroke damage. J Cereb Blood Flow Metab. 2009;29:953–64. doi: 10.1038/jcbfm.2009.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chen-Roetling J, Liu W, Regan RF. Hemopexin decreases hemin accumulation and catabolism by neural cells. Neurochem Int. 2012;60:488–94. doi: 10.1016/j.neuint.2012.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yamauchi T, Lin Y, Sharp FR, Noble-Haeusslein LJ. Hemin induces heme oxygenase-1 in spinal cord vasculature and attenuates barrier disruption and neutrophil infiltration in the injured murine spinal cord. J Neurotrauma. 2004;21:1017–30. doi: 10.1089/0897715041651042. [DOI] [PubMed] [Google Scholar]
  • 48.Lu X, Chen-Roetling J, Regan RF. Systemic hemin therapy attenuates blood-brain barrier disruption after intracerebral hemorrhage. Neurobiol Dis. 2014;70C:245–251. doi: 10.1016/j.nbd.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Takizawa S, Hirabayashi H, Matsushima K, Tokuoka K, Shinohara Y. Induction of heme oxygenase protein protects neurons in cortex and striatum, but not hippocampus, against transient forebrain ischemia. J Cereb Blood Flow Metab. 1998;18:559–569. doi: 10.1097/00004647-199805000-00011. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang B, Wei X, Cui X, Kobayashi T, Li W. Effects of heme oxygenase 1 on brain edema and neurologic outcome after cardiopulmonary resuscitation in rats. Anesthesiology. 2008;109:260–8. doi: 10.1097/ALN.0b013e31817f5c2e. [DOI] [PubMed] [Google Scholar]
  • 51.Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol. 2006;5:53–63. doi: 10.1016/S1474-4422(05)70283-0. [DOI] [PubMed] [Google Scholar]

RESOURCES