1
|
Luo K, Ogawa M, Ayer A, Britton WJ, Stocker R, Kikuchi K, Oehlers SH. Zebrafish Heme Oxygenase 1a Is Necessary for Normal Development and Macrophage Migration. Zebrafish 2022; 19:7-17. [PMID: 35108124 DOI: 10.1089/zeb.2021.0058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.
Collapse
Affiliation(s)
- Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Masahito Ogawa
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Anita Ayer
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Kazu Kikuchi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,National Cerebral and Cardiovascular Center, Suita, Japan
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Sydney Institute for Infectious Diseases, The University of Sydney, Camperdown, Australia
| |
Collapse
|
2
|
Morita A, Jullienne A, Salehi A, Hamer M, Javadi E, Alsarraj Y, Tang J, Zhang JH, Pearce WJ, Obenaus A. Temporal evolution of heme oxygenase-1 expression in reactive astrocytes and microglia in response to traumatic brain injury. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2020.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
3
|
Ischemic Brain Injury Leads to Brain Edema via Hyperthermia-Induced TRPV4 Activation. J Neurosci 2018; 38:5700-5709. [PMID: 29793978 DOI: 10.1523/jneurosci.2888-17.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 01/27/2023] Open
Abstract
Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain largely unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which male mouse brain slices were treated with oxygen-glucose deprivation (OGD) to mimic ischemia. We continuously measured the cross-sectional area of the brain slice for 150 min under macroscopic microscopy, finding that OGD induces swelling of brain slices. OGD-induced swelling was prevented by pharmacologically blocking or genetically knocking out the transient receptor potential vanilloid 4 (TRPV4), a member of the thermosensitive TRP channel family. Because TRPV4 is activated at around body temperature and its activation is enhanced by heating, we next elevated the temperature of the perfusate in the recording chamber, finding that hyperthermia induces swelling via TRPV4 activation. Furthermore, using the temperature-dependent fluorescence lifetime of a fluorescent-thermosensitive probe, we confirmed that OGD treatment increases the temperature of brain slices through the activation of glutamate receptors. Finally, we found that brain edema following traumatic brain injury was suppressed in TRPV4-deficient male mice in vivo Thus, our study proposes a novel mechanism: hyperthermia activates TRPV4 and induces brain edema after ischemia.SIGNIFICANCE STATEMENT Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which mouse brain slices were treated with oxygen-glucose deprivation. Using this system, we showed that the increase in brain temperature and the following activation of the thermosensitive cation channel TRPV4 (transient receptor potential vanilloid 4) are involved in the pathology of edema. Finally, we confirmed that TRPV4 is involved in brain edema in vivo using TRPV4-deficient mice, concluding that hyperthermia activates TRPV4 and induces brain edema after ischemia.
Collapse
|
4
|
Ambegaokar SS, Kolson DL. Heme oxygenase-1 dysregulation in the brain: implications for HIV-associated neurocognitive disorders. Curr HIV Res 2015; 12:174-88. [PMID: 24862327 PMCID: PMC4155834 DOI: 10.2174/1570162x12666140526122709] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 01/20/2014] [Accepted: 01/27/2014] [Indexed: 12/17/2022]
Abstract
Heme oxygenase-1 (HO-1) is a highly inducible and ubiquitous cellular enzyme that subserves cytoprotective responses to toxic insults, including inflammation and oxidative stress. In neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease and multiple sclerosis, HO-1 expression is increased, presumably reflecting an endogenous neuroprotective response against ongoing cellular injury. In contrast, we have found that in human immunodeficiency virus (HIV) infection of the brain, which is also associated with inflammation, oxidative stress and neurodegeneration, HO-1 expression is decreased, likely reflecting a unique role for HO-1 deficiency in neurodegeneration pathways activated by HIV infection. We have also shown that HO-1 expression is significantly suppressed by HIV replication in cultured macrophages which represent the primary cellular reservoir for HIV in the brain. HO-1 deficiency is associated with release of neurotoxic levels of glutamate from both HIV-infected and immune-activated macrophages; this glutamate-mediated neurotoxicity is suppressed by pharmacological induction of HO-1 expression in the macrophages. Thus, HO-1 induction could be a therapeutic strategy for neuroprotection against HIV infection and other neuroinflammatory brain diseases. Here, we review various stimuli and signaling pathways regulating HO-1 expression in macrophages, which could promote neuronal survival through HO-1-modulation of endogenous antioxidant and immune modulatory pathways, thus limiting the oxidative stress that can promote HIV disease progression in the CNS. The use of pharmacological inducers of endogenous HO-1 expression as potential adjunctive neuroprotective therapeutics in HIV infection is also discussed.
Collapse
Affiliation(s)
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Yoneyama-Sarnecky T, Olivas AD, Azari S, Ferriero DM, Manvelyan HM, Noble-Haeusslein LJ. Heme oxygenase-2 modulates early pathogenesis after traumatic injury to the immature brain. Dev Neurosci 2010; 32:81-90. [PMID: 20389079 DOI: 10.1159/000258700] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 11/03/2009] [Indexed: 01/05/2023] Open
Abstract
We determined if heme oxygenase-2 (HO-2), an enzyme that degrades the pro-oxidant heme, confers neuroprotection in the developing brain after traumatic brain injury (TBI). Male HO-2 wild-type (WT) and homozygous knockout (KO) mice at postnatal day 21 were subjected to TBI and euthanized 1, 7, and 14 days later. Relative cerebral blood flow, measured by laser Doppler, cortical and hippocampal pathogenesis, and motor recovery were evaluated at all time points. Cerebral blood flow was found to be similar between experimental groups. Blood flow significantly decreased immediately after injury, returned to baseline by 1 day, and was significantly elevated by 7 days, post-injury. Nonheme iron preferentially accumulated in the ipsilateral cortex, hippocampus, and external capsule in both WT and KO brain-injured genotypes. There were, however, a significantly greater number of TUNEL-positive cells in the hippocampal dentate gyrus and a significantly greater cortical lesion volume in KOs relative to WTs within the first week post-injury. By 14 days post-injury, however, cortical lesion volume and cell density in the hippocampal CA3 region and dorsal thalamus were similar between the two groups. Assays of fine motor function (grip strength) over the first 2 weeks post-injury revealed a general pattern of decreased strength in the contralateral forelimbs of KOs as compared to WTs. Together, these findings demonstrate that deficiency in HO-2 alters both the kinetics of secondary damage and fine motor recovery after TBI.
Collapse
|
6
|
Sandrow-Feinberg HR, Zhukareva V, Santi L, Miller K, Shumsky JS, Baker DP, Houle JD. PEGylated interferon-beta modulates the acute inflammatory response and recovery when combined with forced exercise following cervical spinal contusion injury. Exp Neurol 2010; 223:439-51. [PMID: 20109445 DOI: 10.1016/j.expneurol.2010.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 12/31/2009] [Accepted: 01/14/2010] [Indexed: 12/30/2022]
Abstract
Secondary degeneration leads to an expansion of the initial tissue damage sustained during a spinal cord injury (SCI). Dampening the cellular inflammatory response that contributes to this progressive tissue damage is one possible strategy for neuroprotection after acute SCI. We initially examined whether treatment with a PEGylated form of rat interferon-beta (IFN-beta) would modulate the expression of several markers of inflammation and neuroprotection at the site of a unilateral cervical level 5 contusion injury. Adult female Sprague-Dawley rats were injured using the Infinite Horizon Impactor at a force of 200 kdyn (equivalent to a severe injury) and a mean displacement of 1600-1800 mum. A single dose (5x10(6) units) of PEGylated IFN-beta or vehicle was administered 30 min following SCI. Here we demonstrate temporal changes in pro- and anti-inflammatory cytokine levels and the expression of heat shock proteins and iNOS (involved in neuroprotection) at the lesion epicenter and one segment caudally after SCI and PEG IFN-beta treatment. The results suggested a potential therapeutic treatment strategy for modulation of secondary damage after acute SCI. Therefore, we examined whether acute treatment with PEG IFN-beta would improve forelimb function alone or when combined with forced exercise (Ex). Animals began the Ex paradigm 5 days post SCI and continued for 5 days/week over 8 weeks. Locomotion (forelimb locomotor scale [FLS], hindlimb BBB, and TreadScan) and sensorimotor function (grid walking) was tested weekly. Additional outcome measures included lesion size and glial cell reactivity. Significant FLS improvements occurred at 1 week post SCI in the PEGylated IFN-beta-treated group but not at any other time point or with any other treatment approaches. These results suggest that this acute neuroprotective treatment strategy does not translate into long term behavioral recovery even when combined with forced exercise.
Collapse
Affiliation(s)
- Harra R Sandrow-Feinberg
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Abdel Aziz MT, Mostafa T, Atta H, Wassef MA, Fouad HH, Rashed LA, Sabry D. Putative role of carbon monoxide signaling pathway in penile erectile function. J Sex Med 2009; 6:49-60. [PMID: 19170836 DOI: 10.1111/j.1743-6109.2008.01050.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Erectile response depends on nitric oxide (NO) generated by NO synthase (NOS) enzyme of the nerves and vascular endothelium in the cavernous tissue. NO activates soluble guanylate cyclase (sGC), leading to the production of cyclic guanosine monophosphate (cGMP). cGMP activates cGMP-dependent protein kinase that activates Ca(2+)/ATPase pump that activates Ca(2+)/K efflux pump extruding Ca(2+) across the plasma membrane with consequent smooth muscle cell relaxation. A role similar to that of NOS/NO signaling has been postulated for carbon monoxide (CO) produced in mammals from heme catabolism by heme oxygenase (HO) enzyme. AIM To assess CO signaling pathway for erectile function by reviewing published studies. METHODS A systematic review of published studies on this affair based on Pubmed and Medical Subject Heading databases, with search for all concerned articles. MAIN OUTCOME MEASURES Documentation of positive as well as negative criteria of CO/HO signaling focused on penile tissue. RESULTS The concept that HO-derived CO could play a role in mediating erectile function acting in synergism with, or as a potentiator for, NOS/NO signaling pathway is gaining momentum. CO/HO signaling pathway has been shown to partially mediate the actions of oral phosphodiesterase type 5 inhibitors. In addition, it was shown that the use of CO releasing molecules potentiated cavernous cGMP levels. However, increased CO production or release was reported to be associated, in some studies, with vasoconstriction. CONCLUSION This review sheds a light on the significance of cavernous tissue CO signaling pathway that may pave the way for creation of therapeutic modalities based on this pathway.
Collapse
|
8
|
Miyamoto Y, Ohshida K, Sasago K. A protein assay for heme oxygenase-1 (HO-1) induced by chemicals in HepG2 cells. J Toxicol Sci 2009; 34:709-14. [DOI: 10.2131/jts.34.709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yohei Miyamoto
- Toxicology and Pharmacokinetics Laboratories, Pharmaceutical Research Laboratories, Toray Industries, Inc
| | - Keiyu Ohshida
- Toxicology and Pharmacokinetics Laboratories, Pharmaceutical Research Laboratories, Toray Industries, Inc
| | - Kaori Sasago
- Toxicology and Pharmacokinetics Laboratories, Pharmaceutical Research Laboratories, Toray Industries, Inc
| |
Collapse
|
9
|
Toll-like receptor 2 contributes to glial cell activation and heme oxygenase-1 expression in traumatic brain injury. Neurosci Lett 2007; 431:123-8. [PMID: 18164130 DOI: 10.1016/j.neulet.2007.11.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Revised: 11/02/2007] [Accepted: 11/09/2007] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury is accompanied by glial cell activation around the site of the injury. In this study, we investigated the role of toll-like receptor 2 (TLR2) in glial cell activation using a stab-wound injury (SWI) model with TLR2 knock-out mice. Penetration of a normal mouse brain with a 26-G needle using a stereotaxic instrument resulted in an 18- and 4-fold upregulation of GFAP and CD11b mRNA, respectively, along the needle track in the injury area. However, in the TLR2 knock-out mice, the induced expression of these genes was reduced by 70% and 40%, respectively. Likewise, there was a reduction in the area of activated glial cells detected by immunohistochemistry and the glial cells had a less-activated morphology in the TLR2 knock-out mice. In addition, the expression of the heme oxygenase-1 (HO-1) gene, a glia-expressing wound-responsive gene, was reduced after SWI in TLR2 knock-out mice. Taken together, these data argue that TLR2 contributes to the glial cell activation and HO-1 gene expression associated with traumatic brain injury.
Collapse
|
10
|
Xu XH, Hua YN, Zhang HL, Wu JC, Miao YZ, Han R, Gu ZL, Qin ZH. Greater stress protein expression enhanced by combined prostaglandin A1 and lithium in a rat model of focal ischemia. Acta Pharmacol Sin 2007; 28:1097-104. [PMID: 17640469 DOI: 10.1111/j.1745-7254.2007.00624.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM To investigate the effects of lithium (Li) and prostaglandin A1 (PGA1) on the expression of heat shock factor 1 (HSF-1), heat shock proteins (HSP), and apoptosis protease activating factor-1 (Apaf-1) induced by permanent focal ischemia in rats. METHODS The rats were pretreated with a subcutaneous (sc) injection of Li for 2 d or a single intracerebral ventricle (icv) administration of PGA1 for 15 min before ischemic insult, or a combination of Li (sc, 1 mEq/kg, 2 d) and PGA1 (icv, 15 min prior to ischemic insult). Brain ischemia was induced by the permanent middle cerebral artery occlusion (pMCAO). Twenty-four hours after the occlusion, the expression of HSF-1, HSP, and Apaf-1 in the ischemic striatum were examined with Western blot analysis. RESULTS The expression of HSF-1, heme oxygenase-1 (HO-1), HSP90alpha, and Apaf-1 were significantly increased, but the expression of HSP90beta was significantly decreased 24 h after the pMCAO. PGA1 and Li and their combination significantly enhanced the ischemia-induced elevation in the levels of HSF-1, HO-1, and HSP90alpha, and recovered HSP90beta expression, but decreased Apaf-1 levels in the ischemic striatum. CONCLUSION The present study demonstrates that PGA1 and Li have synergistic effects on the enhancement of the expression of HSP, suggesting that the synergistic effects of PGA1 and Li in the rat model of permanent focal cerebral ischemia may be mediated by the enhancement expression of HSP expression and the downregulation of Apaf-1. Our studies suggest that combined PGA1 and Li may have potential clinical value for the treatment of stroke.
Collapse
Affiliation(s)
- Xi-hui Xu
- Laboratory of Aging and Nervous Disease, Soochow University School of Medicine, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
El Andaloussi A, Lesniak MS. CD4+ CD25+ FoxP3+ T-cell infiltration and heme oxygenase-1 expression correlate with tumor grade in human gliomas. J Neurooncol 2007; 83:145-52. [PMID: 17216339 DOI: 10.1007/s11060-006-9314-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
Regulatory T-cells play an important role in the regulation of the immune response and the mediation of dominant immunologic tolerance. We have previously shown that these cells are elevated in tumors and blood of patients with glioblastoma multiforme. Heme oxygenase-1, a rate-limiting enzyme in heme catabolism, has also been shown to accumulate during glioma progression and to play a critical role in FoxP3 mediated immune suppression. In this study, we investigated the correlation between FoxP3 and HO-1 expression in patients with various grades of astrocytoma (WHO grade II-IV). Using qualitative and quantitative reverse transcriptase-polymerase chain reaction and quantitative flow cytometry analyses, we analyzed FoxP3 and HO-1 expression in 19 patients with different grades of astrocytoma. We observed the highest level of FoxP3 expression in patients with grade IV tumors (11.54 +/- 1.95%) vs. grade III (6.74 +/- 0.19%) or grade II (2.53 +/- 0.11%) (P < 0.05). Moreover, in grade IV tumors, the frequency of HO-1 mRNA expression in CD4+ CD25+ cells was 11.8 +/- 2.45% vs. 7.42 +/- 0.31% in grade III and 2.33 +/- 0.12% in grade II. Tumor infiltrating Treg stained positively with anti-HO-1 antibody. The expression of HO-1 correlated with CD4+ CD25+ FoxP3+ infiltration (r = 0.966). Our results confirm that HO-1 expressing Treg accumulate during glioma progression. This study also suggests that HO-1 mRNA expression is linked to the induction of Foxp3 in CD4+ CD25+ glioma infiltrating Treg. These findings support the suppressive role played by regulatory T-cells in the growth of malignant brain tumors.
Collapse
|
12
|
Ewing JF, Maines MD. 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. J Neural Transm (Vienna) 2006; 113:439-54. [PMID: 16467964 DOI: 10.1007/s00702-005-0408-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 09/26/2005] [Indexed: 10/25/2022]
Abstract
The heme oxygenase isozymes, HO-1 and HO-2, oxidatively cleave the heme molecule to produce biliverdin and the gaseous messenger, CO. The cleavage results in the release of iron, a regulator of transferrin, ferritin, and nitric oxide (NO) synthase gene expression. Biliverdin reductase (BVR) then catalyzes the reduction of biliverdin, generating the potent intracellular antioxidant, bilirubin. We report an age-related decrease in HO-1 and HO-2 expression present in select brain regions including the hippocampus and the substantia nigra, that are involved in the high order cognitive processes of learning and memory. The age-related loss of monoxide-producing potential in select regions of the brain was not specific to the HO system but was also observed in neuronal NO-generating system. Furthermore, compared to 2-month old rats, the ability of aged brain tissue to respond to hypoxic/hyperthermia was compromised at both the protein and the transcription levels as judged by attenuated induction of HO-1 immunoreactive protein and its 1.8 Kb transcript. Neotrofin (AIT), a cognitive-enhancing and neuroprotective drug, caused a robust increase in HO-1 immunoreactive protein in select neuronal regions and increased the expression of HO-2 transcripts. The potential interplay between regulation of HO-2 gene expression and the serum levels of the adrenal steroids is discussed. We suggest the search for therapeutic agents that reverse the decline and aberrant stress response of HO enzymes may lead to effective treatment regimens for age-associated neuronal deficits.
Collapse
Affiliation(s)
- J F Ewing
- Department of Biochemistry/Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | |
Collapse
|
13
|
Cabell L, Ferguson C, Luginbill D, Kern M, Weingart A, Audesirk G. Differential induction of heme oxygenase and other stress proteins in cultured hippocampal astrocytes and neurons by inorganic lead. Toxicol Appl Pharmacol 2004; 198:49-60. [PMID: 15207648 DOI: 10.1016/j.taap.2004.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Accepted: 03/01/2004] [Indexed: 01/23/2023]
Abstract
We examined the effects of exposure to inorganic lead (Pb2+) on the induction of stress proteins in cultured hippocampal neurons and astrocytes, with particular emphasis on the induction of heme oxygenase-1 (HO-1). In radiolabeled neuronal cultures, Pb2+ exposure had no significant effect on the synthesis of any protein at any concentration (up to 250 microM) or duration of exposure (up to 4 days). In radiolabeled astrocyte cultures, however, Pb2+ exposure (100 nM to 100 microM; 1-4 days) increased synthesis of proteins with approximate molecular weights of 23, 32, 45, 57, 72, and 90 kDa. Immunoblot experiments showed that Pb2+ exposure (100 nM to 10 microM, 1-14 days) induces HO-1 synthesis in astrocytes, but not in neurons; this is probably the 32-kDa protein. The other heme oxygenase isoform, HO-2, is present in both neurons and astrocytes, but is not inducible by Pb2+ at concentrations up to 100 microM. HO-1 can be induced by a variety of stimuli. We found that HO-1 induction in astrocytes is increased by combined exposure to Pb2+ and many other stresses, including heat, nitric oxide, H2O2, and superoxide. One of the stimuli that may induce HO-1 is oxidative stress. Lead exposure causes oxidative stress in many cell types, including astrocytes. Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). Therefore, we conclude that oxidative stress is an important mechanism by which Pb2+ induces HO-1 synthesis in astrocytes.
Collapse
Affiliation(s)
- Leigh Cabell
- Biology Department, University of Colorado at Denver, Denver, CO 80217-3364, USA
| | | | | | | | | | | |
Collapse
|
14
|
Koeppen AH, Dickson AC, Smith J. Heme oxygenase in experimental intracerebral hemorrhage: the benefit of tin-mesoporphyrin. J Neuropathol Exp Neurol 2004; 63:587-97. [PMID: 15217087 DOI: 10.1093/jnen/63.6.587] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The prognosis of intracerebral hemorrhage (ICH) is unfavorable. Beyond immediate mass effect and tissue destruction, ICHs cause additional neuronal loss in a "perifocal reactive zone." Heme in ICH induces heme oxygenase-1 (HO-1), and the action of this enzyme on heme yields ferrous iron, biliverdin, and carbon monoxide. Iron is ultimately converted to ferritin and hemosiderin. Free iron is tissue-toxic, and inhibition of HO-1 should provide protection against additional damage. Experimental ICHs were made in adult rabbits by the stereotaxic injection of autologous blood, and the induction of HO-1 and increase in ferritin were followed by confocal immunofluorescence microscopy. Heme diffused rapidly through perivascular spaces, and HO-1 reaction product first occurred in perivascular cells and microglia. At this stage, HO-1 and ferritin showed extensive colocalization. As ICH resolution progressed, HO-1 immunoreactivity faded while ferritin and hemosiderin continued to accumulate. This process was accompanied by a gradient of destruction of neuronal cell bodies and dendrites in the perifocal reactive zone. In an effort to inhibit HO-1, repeated intravenous injections of tin-mesoporphyrin IX (SnMP) were given to ICH-bearing rabbits. The ICH disrupted the blood-brain barrier sufficiently to allow SnMP to enter the brain in pharmacological amounts, and the metalloporphyrin provided significant protection against neuronal loss.
Collapse
Affiliation(s)
- Arnulf H Koeppen
- Neurology Service, VA Medical Center, Albany, New York 12208, USA.
| | | | | |
Collapse
|
15
|
Wagner KR, Dwyer BE. Hematoma Removal, Heme, and Heme Oxygenase Following Hemorrhagic Stroke. Ann N Y Acad Sci 2004; 1012:237-51. [PMID: 15105270 DOI: 10.1196/annals.1306.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The hemorrhagic strokes, intracerebral (ICH) and subarachnoid hemorrhage (SAH), often have poor outcomes. Indeed, the most common hemorrhagic stroke, ICH, has the highest mortality and morbidity rates of any stroke subtype. In this report, we discuss the evidence for the staging of red blood cell removal after ICH and the significance of control of this process. The protective effects of clinically relevant metalloporphyrin heme oxygenase inhibitors in experimental models of ICH and in superficial siderosis are also discussed. We also examine literature paradoxes related to both heme and heme oxygenase in various disorders of the central nervous system. Last, new data are presented that support the concept that heme, although primarily a pro-oxidant, can also have antioxidant properties.
Collapse
Affiliation(s)
- Kenneth R Wagner
- Medical Research Service, Department of Veterans Affairs Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45220, USA.
| | | |
Collapse
|
16
|
Abstract
Heme and iron metabolism are of considerable interest and importance in normal brain function as well as in neurodegeneration and neuropathologically following traumatic injury and hemorrhagic stroke. After a cerebral hemorrhage, large numbers of hemoglobin-containing red blood cells are released into the brain's parenchyma and/or subarachnoid space. After hemolysis and the subsequent release of heme from hemoglobin, several pathways are employed to transport and metabolize this heme and its iron moiety to protect the brain from potential oxidative stress. Required for these processes are various extracellular and intracellular transporters and storage proteins, the heme oxygenase isozymes and metabolic proteins with differing localizations in the various brain-cell types. In the past several years, additional new genes and proteins have been discovered that are involved in the transport and metabolism of heme and iron in brain and other tissues. These discoveries may provide new insights into neurodegenerative diseases like Alzheimer's, Parkinson's, and Friedrich's ataxia that are associated with accumulation of iron in specific brain regions or in specific organelles. The present review will examine the uptake and metabolism of heme and iron in the brain and will relate these processes to blood removal and to the potential mechanisms underlying brain injury following cerebral hemorrhage.
Collapse
Affiliation(s)
- Kenneth R Wagner
- Departments of Neurology, Pediatrics and Neuroscience Programl, Unviersity of Cincinnati College of Medicine, Cincinnati, Ohio 45220, USA.
| | | | | | | | | |
Collapse
|
17
|
Wang X, Hauptmann N, Taylor E, Foreman M, Khawli LA, Maines MD. Neotrofin increases heme oxygenase-1 selectively in neurons. Brain Res 2003; 962:1-14. [PMID: 12543450 DOI: 10.1016/s0006-8993(02)03670-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
There is evidence that heme oxygenase plays a role in cellular defense against reactive oxygen species and thereby has neuroprotective effects. We examined the interaction of Neotrofin, a cognitive-enhancing and neuroprotective drug, with the heme oxygenase system. In adult rats, both a single administration or seven daily injections of Neotrofin at 10, 30 or 100 mg/kg intraperitoneally increased HO-1 immunoreactivity in neurons of the hippocampal formation and its connections including CA1-4, fornix, septal nuclei, hippocampal commissure, septohippocampal nucleus, fimbria, anteroventral thalamic nucleus, frontal and parietal cortex. Prominent HO-1 staining of neuronal cells in the proximity of blood vessels and circumventricular organs was also observed. Increasing doses of Neotrofin resulted in an increase in the number of neuronal populations expressing HO-1 with 100 mg/kg evoking a widespread neuronal cell response in brain. Quantification by ELISA confirmed that intraperitoneal administration of 100 mg/kg Neotrofin caused a significant increase in HO-1 protein expression in the hippocampus. The increase was evident by 6 h post-injection, peaked at 24 h with a 4-fold increase, and persisted for at least 48 h. Similarly, oral administration of 100 mg/kg Neotrofin produced a 5-fold increase in HO-1 protein 24 h post-administration. The effect of Neotrofin on HO-1 appears to be at the transcriptional level, as suggested by an increase in HO-1 mRNA levels. Neotrofin treatment was also associated with a significant increase in HO-2 mRNA levels in whole brain homogenate. These data may explain the neuroprotective effects of Neotrofin in models of excitotoxic neuronal injury.
Collapse
Affiliation(s)
- Xiaojun Wang
- Department of Biochemistry and Biophysics, Box 712, University of Rochester School of Medicine, 601 Elmwood Ave., Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
18
|
Wagner KR, Packard BA, Hall CL, Smulian AG, Linke MJ, De Courten-Myers GM, Packard LM, Hall NC. Protein oxidation and heme oxygenase-1 induction in porcine white matter following intracerebral infusions of whole blood or plasma. Dev Neurosci 2003; 24:154-60. [PMID: 12401953 DOI: 10.1159/000065703] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Spontaneous or traumatic intracerebral hemorrhage (ICH) in the white matter of neonates, children and adults causes significant mortality and morbidity. The detailed biochemical mechanisms through which blood damages white matter are poorly defined. Presently, we tested the hypothesis that ICH induces rapid oxidative stress in white matter. Also, since clot-derived plasma proteins accumulate in white matter after ICH and these proteins can induce oxidative stress in microglia in vitro, we determined whether the blood's plasma component alone induces oxidative stress. Lastly, since heme oxygenase-1 (HO-1) induction is highly sensitive to oxidative stress, we also examined white matter HO-1 gene expression. We infused either whole blood or plasma (2.5 ml) into the frontal hemispheric white matter of pentobarbital-anesthetized pigs ( approximately 1 kg) over 15 min. We monitored and controlled physiologic variables and froze brains in situ between 1 and 24 h after ICH. White matter oxidative stress was determined by measuring protein carbonyl formation and HO-1 gene expression by RT-PCR. Protein carbonyl formation occurred rapidly in the white matter adjacent to both blood and plasma clots with significant elevations (3- to 4-fold) already 1 h after infusion. This increase remained through the first 24 h. HO-1 mRNA was rapidly induced in white matter with either whole blood or plasma infusions. These results demonstrate that not only whole blood but also its plasma component are capable of rapidly inducing oxidative stress in white matter. This rapid response, possibly in microglial cells, may contribute to white matter damage not only following ICH, but also in pathophysiological states in which blood-brain-barrier permeability to plasma proteins is increased.
Collapse
Affiliation(s)
- Kenneth R Wagner
- Department of Neurology, University of Cincinnati College of Medicine, Department of Veterans Affairs Medical Center, Cincinnati, Ohio 45220, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Ghattas MH, Chuang LT, Kappas A, Abraham NG. Protective effect of HO-1 against oxidative stress in human hepatoma cell line (HepG2) is independent of telomerase enzyme activity. Int J Biochem Cell Biol 2002; 34:1619-28. [PMID: 12379283 DOI: 10.1016/s1357-2725(02)00097-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Heme oxygenase-1 (HO-1) is a stress response protein and its induction is associated with protection against oxidative stress. Cell survival during exposure to environmental stresses is associated with elevation of HO-1. Telomerase plays an important role in cell proliferation and immortalization. Our objective was to determine whether the adaptive cellular response to survive exposure to environmental stresses is dependent on expression of HO-1 and telomerase activity in hepatoma cell line (HepG2). Exposure of HepG2 to oxidants, H(2)O(2) (100 microM), as well as HO-1 inducers, heme (10 microM) and stannic chloride (SnCl(2)) (10 microM), resulted in an increased HO-1 mRNA, protein and total HO activity. On the other hand, HO activity was inhibited by addition of stannic mesoporphyrin (SnMP) (10 microM). These effects were brought about without altering endogenous HO-2 protein levels. Telomerase activity was not affected by oxidants, inducers of HO-1 or inhibitors of HO activity. Similarly, the catalytic subunit of telomerase enzyme human telomerase reverse transcriptase (hTERT), which is considered as the major regulator of telomerase activity, was not affected by oxidants, heme and H(2)O(2), or downregulation of HO gene activity by SnMP. This study demonstrates, for the first time, that induction of HO-1 gene mediates protection against oxidants and increases cell survival by a mechanism independent of telomerase enzyme activity. Suppression of HO activity by SnMP decreased cell resistance to oxidant stressors without altering telomerase activity.
Collapse
Affiliation(s)
- Maivel H Ghattas
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
20
|
Sharp FR, Bernaudin M, Bartels M, Wagner KR. Glial expression of heat shock proteins (HSPs) and oxygen-regulated proteins (ORPs). PROGRESS IN BRAIN RESEARCH 2001; 132:427-40. [PMID: 11545009 DOI: 10.1016/s0079-6123(01)32093-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- F R Sharp
- Department of Neurology, University of Cincinnati, Vontz Center for Molecular Studies, Room 2327, 3125 Eden Avenue, Cincinnati, OH 45267-0536, USA.
| | | | | | | |
Collapse
|
21
|
Goldbaum O, Richter-Landsberg C. Stress proteins in oligodendrocytes: differential effects of heat shock and oxidative stress. J Neurochem 2001; 78:1233-42. [PMID: 11579132 DOI: 10.1046/j.1471-4159.2001.00507.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heat shock proteins (HSP) or stress proteins serve as biomarkers to identify the contribution of stress situations underlying the pathogenesis of degenerative diseases of the CNS. We have analyzed by immunoblot technique the constitutive and inducible occurrence of stress proteins in cultured rat brain oligodendrocytes subjected to heat shock or oxidative stress exerted by hydrogen peroxide, or a combination of both. The data demonstrate that oligodendrocytes constitutively express HSP32, HSP60 and the cognate form of the HSP70 family of proteins, HSC70. After heat shock, HSP25, alpha B-crystallin and HSP70 were up-regulated, while after oxidative stress the specific induction of HSP32 and alpha B-crystallin was observed. HSP32 represents heme oxygenase 1 (HO-1), a small stress protein with enzymatic activity involved in the oxidative degradation of heme which participates in iron metabolism. The presence of the iron chelators phenanthroline or deferoxamine (DFO), which previously has been shown to protect oligodendrocytes from oxidative stress-induced onset of apoptosis, caused a marked stimulation of HSP32 without affecting HSP70. This indicates that DFO possibly exerts its protective role by directly influencing the antioxidant capacity of HO-1. In summary, HSP in oligodendrocytes are differentially stimulated by heat stress and oxidative stress. Heme oxygenase-1 has been linked to inflammatory processes and oxidative stress, its specific up-regulation after oxidative stress in oligodendrocytes suggests that it is an ideal candidate to investigate the involvement of oxidative stress in demyelinating diseases.
Collapse
Affiliation(s)
- O Goldbaum
- Department of Biology, Molecular Neurobiology, University of Oldenburg, Oldenburg, Germany
| | | |
Collapse
|
22
|
Kämpfer H, Kolb N, Manderscheid M, Wetzler C, Pfeilschifter J, Frank S. Macrophage-Derived Heme-Oxygenase-1: Expression, Regulation, and Possible Functions in Skin Repair. Mol Med 2001. [DOI: 10.1007/bf03401854] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
23
|
Mautes AE, Bergeron M, Sharp FR, Panter SS, Weinzierl M, Guenther K, Noble LJ. Sustained induction of heme oxygenase-1 in the traumatized spinal cord. Exp Neurol 2000; 166:254-65. [PMID: 11085891 DOI: 10.1006/exnr.2000.7520] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Oxidative stress contributes to secondary injury after spinal cord trauma. Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Here we examine the induction of HO-1 in the hemisected spinal cord, a model that results in reproducible degeneration in the ipsilateral white matter. HO-1 was induced in microglia and macrophages from 24 h to at least 42 days after injury. Within the first week after injury, HO-1 was induced in both the gray and the white matter. Thereafter, HO-1 expression was limited to degenerating fiber tracts. HSP70, a heat shock protein induced mainly by the presence of denatured proteins, was consistently colocalized with HO-1 in the microglia and macrophages. This study to demonstrates long-term induction of HO-1 and HSP70 in microglia and macrophages after traumatic injury and an association between induction of HO-1 and Wallerian degeneration. White matter degeneration is characterized by phagocytosis of cellular debris and remodeling of surviving tissue. This results in the metabolism, synthesis, and turnover of heme and heme proteins. Thus, sustained induction of HO-1 and HSP70 in microglia and macrophages suggests that tissue degeneration is an ongoing process, lasting 6 weeks and perhaps even longer.
Collapse
Affiliation(s)
- A E Mautes
- Department of Neurosurgery, University of California at San Francisco, San Francisco, California, 94143, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Schluesener HJ, Seid K. Heme oxygenase-1 in lesions of rat experimental autoimmune encephalomyelitis and neuritis. J Neuroimmunol 2000; 110:114-20. [PMID: 11024540 DOI: 10.1016/s0165-5728(00)00352-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The enzyme heme oxygenase-1 (HO-1) is reducing heme to the gaseous mediator carbon monoxide, to iron and the antioxidant biliverdin. The inducible expression of HO-1 is considered a protective cellular mechanism against reactive oxygen intermediates. Further, carbon monoxide (CO) is a regulator of cGMP synthesis, of NO-synthetases and cyclooxygenases, thereby indirectly modulating reactive processes. Here we report expression of HO-1 in rat experimental autoimmune encephalomyelitis (EAE) and neuritis (EAN). With both models, similar results were obtained: HO-1 was localized predominantly to infiltrating, monocytic, but only rarely to ramified microglial cells or astrocytes surrounding the inflammatory lesions. Prominent expression by monocytic cells was seen from day 11 after immunization correlating with the development of neurologic disease. Further, local expression is persistent for long after cessation of neurologic signs. Thus, HO-1 could be considered a factor in the formation and resolution of inflammatory autoimmune lesions of the nervous system.
Collapse
Affiliation(s)
- H J Schluesener
- Institute of Brain Research, University of Tübingen, Calwer Str. 3, D-72076, Tübingen, Germany.
| | | |
Collapse
|
25
|
Torisu-Itakura H, Furue M, Kuwano M, Ono M. Co-expression of thymidine phosphorylase and heme oxygenase-1 in macrophages in human malignant vertical growth melanomas. Jpn J Cancer Res 2000; 91:906-10. [PMID: 11011118 PMCID: PMC5926440 DOI: 10.1111/j.1349-7006.2000.tb01033.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Expression of thymidine phosphorylase (TP) is often associated with tumor angiogenesis and / or prognosis in patients. Further, infiltration of macrophages is closely correlated with the depth of tumor and angiogenesis in melanomas. In this study, we examined the expression of TP and an activated macrophage-specific enzyme, heme oxygenase-1 (HO-1), involved in malignancy in 22 cases with melanomas. TP was strongly expressed not only in CD68-positive macrophages in and around tumors, but also in S100 protein-positive melanoma cells, fibroblasts and keratinocytes. By contrast, HO-1 was specifically expressed in macrophages, but only slightly in melanoma cells and other cell types in the stroma of melanomas. We thus observed apparent co-expression of TP and HO-1 in macrophages infiltrating in the late stage of malignant melanomas. There appeared increasing numbers of TP-positive cells in Clark level IV and V melanoma compared with Clark level I (in situ) melanoma, and there was also a close correlation between numbers of TP-positive cells and HO-1-positive cells. Both TP- and HO-1-positive macrophages could be observed in the stroma in and around tumors in vertical growth melanomas.
Collapse
Affiliation(s)
- H Torisu-Itakura
- Department of Medical Biochemistry, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | |
Collapse
|
26
|
Aizawa T, Ishizaka N, Taguchi J, Kimura S, Kurokawa K, Ohno M. Balloon injury does not induce heme oxygenase-1 expression, but administration of hemin inhibits neointimal formation in balloon-injured rat carotid artery. Biochem Biophys Res Commun 1999; 261:302-7. [PMID: 10425182 DOI: 10.1006/bbrc.1999.1020] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been shown that antioxidant agents act inhibitorily against neointimal formation after balloon injury, suggesting the role of oxidative stress as a promotor of intimal cell proliferation. Heme oxygenase-1 (HO-1) is an inducible form of heme catabolizing enzyme that is induced by and acts against oxidative tissue injury. In this set of experiments, we showed that HO-1 was present in newly formed neointima; however, arterial HO-1 expression did not increase in response to balloon injury in rat carotid artery. Intraperitoneal administration of hemin, a HO-1 inducer, for 5 consecutive days resulted in about a 4-fold increase of serum bilirubin concentration. In addition, hemin injection increased HO-1 protein expression in the carotid artery, the heart, the kidney, and the liver. In this condition, balloon injury-induced neointimal formation was markedly inhibited. Local application of tin protoporphyrin, a HO inhibitor, blocked this effect, suggesting that induced HO-1 in the carotid artery was responsible for the inhibition of neointimal formation after balloon injury. This study suggests that induction of the endogenous antioxidant gene can suppress neointimal formation after balloon injury.
Collapse
Affiliation(s)
- T Aizawa
- Faculty of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Yang L, Quan S, Abraham NG. Retrovirus-mediated HO gene transfer into endothelial cells protects against oxidant-induced injury. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:L127-33. [PMID: 10409239 DOI: 10.1152/ajplung.1999.277.1.l127] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heme oxygenase (HO)-1 is a stress protein that has been implicated in defense mechanisms against agents that may induce oxidative injury, such as endotoxins, heme, and cytokines. Overexpression of HO-1 in cells might, therefore, protect against oxidative stress produced by certain agents, specifically heme, by catalyzing its degradation to bilirubin, which by itself has antioxidant properties. We report for the first time the successful transduction of human HO-1 gene into rat lung microvessel endothelium using replication-defective retroviral vector. Cells transduced with human HO-1 gene exhibited a 2.1-fold increase in HO-1 protein level, which was associated with a 2.3-fold elevation in enzyme activity compared with that in nontransduced cells. The cGMP content in transduced endothelial cells was increased by 2.9-fold relative to that in nontransduced cells. Moreover, human HO-1 gene-transduced endothelial cells acquired substantial resistance to toxicity produced by exposure to heme and H(2)O(2) compared with that in nontransduced cells. The protective effect of enhancement of HO-1 activity against heme and H(2)O(2) was reversed by pretreatment with stannic mesoporphyrin, a competitive inhibitor of HO. These data demonstrate that the induction of HO-1 in response to injurious stimuli represents an important mechanism for moderating the severity of cell damage. Regulation of HO activity in this manner may have clinical applications.
Collapse
Affiliation(s)
- L Yang
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | | | | |
Collapse
|
28
|
Abstract
Heme oxygenase-1 (HO-1) is a stress protein inducible in some cells by oxidative stress. The status of heme oxygenase was investigated in a transgenic mouse model of amyotrophic lateral sclerosis (ALS) since oxidative mechanisms are postulated in neuronal injury. Three ALS mice [(SOD1-G93A)1Gur] and three controls [(SOD-1)2Gur] were obtained from The Jackson Laboratory. Behavioral differences suggestive of neurodegeneration in ALS mice developed at 4-5 months of age. All mice were killed at 7-8 months of age. Tissue vacuolation, cell loss, and the presence of GFAP+ cells were noted in the spinal cords of ALS mice. Spinal cord motor neurons in both control and ALS mice stained positive for heme oxygenase-2 (HO-2). While not precluding the presence of low levels of HO-1 neither immunohistochemical staining nor Western blot analysis provided evidence for significant HO-1 induction in degenerating spinal cord.
Collapse
Affiliation(s)
- B E Dwyer
- Molecular Neurobiology Laboratory (151), The Department of Veterans Affairs Medical Center, White River Junction, Vermont 05009-0001, USA
| | | | | |
Collapse
|
29
|
Richmon JD, Fukuda K, Maida N, Sato M, Bergeron M, Sharp FR, Panter SS, Noble LJ. Induction of heme oxygenase-1 after hyperosmotic opening of the blood-brain barrier. Brain Res 1998; 780:108-18. [PMID: 9473615 DOI: 10.1016/s0006-8993(97)01314-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The induction of the stress protein heme oxygenase-1 (HO-1) was studied in the rat brain after intracarotid administration of hyperosmolar mannitol. HO-1 was immunolocalized in fixed sections of brain 24 h to 7 days after injection. Immunoglobulin G (IgG) was immunolocalized in adjacent sections to demonstrate areas of breakdown of the blood-brain barrier. Induction of HO-1 was also evaluated by Western immunoblots, performed at 24 h after the insult. Immunofluorescent double labelling with monoclonal antibodies to HO-1 and either glial fibrillary acidic protein or the complement C3bi receptor was used to determine if glia/macrophages expressed HO-1. There was pronounced, widespread induction of HO-1 in the ipsilateral hemisphere and cerebellum by 24 h both by immunocytochemistry and by Western blots. This induction was markedly attenuated at later times. HO-1 was induced in astrocytes and microglia/macrophages in the ipsilateral hemisphere. In addition, the protein was induced in Bergmann glia and scattered microglia/macrophages in the cerebellum. The mechanism of induction of HO-1 in glia after opening of the blood-brain barrier could include exposure to heme proteins, denatured proteins and other plasma constituents known to induce HO-1. This glial induction may reflect a protective response of these cells.
Collapse
Affiliation(s)
- J D Richmon
- Department of Neurosurgery, University of California, San Francisco 94143-0520, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gissel C, Doutheil J, Paschen W. Activation of heme oxygenase-1 expression by disturbance of endoplasmic reticulum calcium homeostasis in rat neuronal cell culture. Neurosci Lett 1997; 231:75-8. [PMID: 9291144 DOI: 10.1016/s0304-3940(97)00528-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Evidence has been presented that disturbances of endoplasmic reticulum (ER) calcium homeostasis contribute to neuronal injury induced by transient cerebral ischemia. The present series of experiments was designed to study whether the expression of heme oxygenase-1 (HO-1), which is markedly increased after transient cerebral ischemia, is also activated by a disturbance of ER calcium homeostasis. ER calcium pools were depleted by a 30 min exposure of primary cortical and hippocampal neurons to thapsigargin (Tg), an irreversible inhibitor of ER Ca2+-ATPase. In cortical neurons, HO-1 mRNA levels (analysed by quantitative polymerase chain reaction (PCR)) were significantly increased (22-fold) 12 h after exposure to Tg but had decreased again to only nine times control levels by 24 h after treatment. In hippocampal neurons, a significant increase in HO-1 mRNA levels was already apparent 4 h after treatment (8.3-fold over controls), levels rose further to 27-fold over controls after 6 h, and stayed high for up to 24 h after treatment (34-fold over controls). The similarity between the pattern of changes in HO-1 mRNA levels induced by transient ischemia and depletion of ER calcium stores suggests common underlying mechanisms.
Collapse
Affiliation(s)
- C Gissel
- Max-Planck-Institute for Neurological Research, Department of Experimental Neurology, Cologne, Germany
| | | | | |
Collapse
|