1
|
Gao MZ, Zeng JY, Chen XJ, Shi L, Hong FY, Lin M, Luo JW, Chen H. Dimethyl fumarate ameliorates oxidative stress-induced acute kidney injury after traumatic brain injury by activating Keap1-Nrf2/HO-1 signaling pathway. Heliyon 2024; 10:e32377. [PMID: 38947486 PMCID: PMC11214498 DOI: 10.1016/j.heliyon.2024.e32377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Acute kidney injury (AKI) frequently emerges as a consequential non-neurological sequel to traumatic brain injury (TBI), significantly contributing to heightened mortality risks. The intricate interplay of oxidative stress in the pathophysiology of TBI underscores the centrality of the Keap1-Nrf2/HO-1 signaling pathway as a pivotal regulator in this context. This study endeavors to elucidate the involvement of the Keap1-Nrf2/HO-1 pathway in modulating oxidative stress in AKI subsequent to TBI and concurrently explore the therapeutic efficacy of dimethyl fumarate (DMF). A rat model of TBI was established via the Feeney free-fall method, incorporating interventions with varying concentrations of DMF. Assessment of renal function ensued through measurements of serum creatinine and neutrophil gelatinase-associated lipocalin. Morphological evaluation of renal pathology was conducted employing quantitative hematoxylin and eosin staining. The inflammatory response was scrutinized by quantifying interleukin (IL)-6, IL-1β, and tumor necrosis factor-α levels. Oxidative stress levels were discerned through quantification of malondialdehyde and superoxide dismutase. The apoptotic cascade was examined via the terminal deoxynucleotidyl transferase dUTP deletion labeling assay. Western blotting provided insights into the expression dynamics of proteins affiliated with the Keap1-Nrf2/HO-1 pathway and apoptosis. The findings revealed severe kidney injury, heightened oxidative stress, inflammation, and apoptosis in the traumatic brain injury model. Treatment with DMF effectively reversed these changes, alleviating oxidative stress by activating the Keap1-Nrf2/HO-1 signaling pathway, ultimately conferring protection against AKI. Activating Keap1-Nrf2/HO-1 signaling pathway may be a potential therapeutic strategy for attenuating oxidative stress-induced AKI after TBI.
Collapse
Affiliation(s)
- Mei-zhu Gao
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Jing-yi Zeng
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xue-jing Chen
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Lan Shi
- Department of Intensive Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fu-yuan Hong
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Miao Lin
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Jie-wei Luo
- Department of Traditional Chinese Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Han Chen
- The Fourth Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, 350001, China
| |
Collapse
|
2
|
Yu M, Wang Z, Wang D, Aierxi M, Ma Z, Wang Y. Oxidative stress following spinal cord injury: From molecular mechanisms to therapeutic targets. J Neurosci Res 2023; 101:1538-1554. [PMID: 37272728 DOI: 10.1002/jnr.25221] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023]
Abstract
Spinal cord injury (SCI) is a medical condition that results from severe trauma to the central nervous system; it imposes great psychological and economic burdens on affected patients and their families. The dynamic balance between reactive oxygen species (ROS) and antioxidants is essential for maintaining normal cellular physiological functions. As important intracellular signaling molecules, ROS regulate numerous physiological activities, including vascular reactivity and neuronal function. However, excessive ROS can cause damage to cellular macromolecules, including DNA, lipids, and proteins; this damage eventually leads to cell death. This review discusses the mechanisms of oxidative stress in SCI and describes some signaling pathways that regulate oxidative injury after injury, with the aim of providing guidance for the development of novel SCI treatment strategies.
Collapse
Affiliation(s)
- Mengsi Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhiying Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongmin Wang
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Milikemu Aierxi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhanjun Ma
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, Brussels, Belgium
| | - Yonggang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
3
|
Fesharaki-Zadeh A. Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms232113000. [PMID: 36361792 PMCID: PMC9657447 DOI: 10.3390/ijms232113000] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic Brain Injury (TBI) remains a major cause of disability worldwide. It involves a complex neurometabolic cascade, including oxidative stress. The products of this manuscript is examining the underlying pathophysiological mechanism, including reactive oxygen species (ROS) and reactive nitrogen species (RNS). This process in turn leads to secondary injury cascade, which includes lipid peroxidation products. These reactions ultimately play a key role in chronic inflammation and synaptic dysfunction in a synergistic fashion. Although there are no FDA approved antioxidant therapy for TBI, there is a number of antioxidant therapies that have been tested and include free radical scavengers, activators of antioxidant systems, inhibitors of free radical generating enzymes, and antioxidant enzymes. Antioxidant therapies have led to cognitive and functional recovery post TBI, and they offer a promising treatment option for patients recovering from TBI. Current major challenges in treatment of TBI symptoms include heterogenous nature of injury, as well as access to timely treatment post injury. The inherent benefits of antioxidant therapies include minimally reported side effects, and relative ease of use in the clinical setting. The current review also provides a highlight of the more studied anti-oxidant regimen with applicability for TBI treatment with potential use in the real clinical setting.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Yale School of Medicine, Department of Neurology, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
4
|
Hakiminia B, Alikiaii B, Khorvash F, Mousavi S. Oxidative stress and mitochondrial dysfunction following traumatic brain injury: From mechanistic view to targeted therapeutic opportunities. Fundam Clin Pharmacol 2022; 36:612-662. [PMID: 35118714 DOI: 10.1111/fcp.12767] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/15/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is one of the most prevalent causes of permanent physical and cognitive disabilities. TBI pathology results from primary insults and a multi-mechanistic biochemical process, termed as secondary brain injury. Currently, there are no pharmacological agents for definitive treatment of patients with TBI. This article is presented with the purpose of reviewing molecular mechanisms of TBI pathology, as well as potential strategies and agents against pathological pathways. In this review article, materials were obtained by searching PubMed, Scopus, Elsevier, Web of Science, and Google Scholar. This search was considered without time limitation. Evidence indicates that oxidative stress and mitochondrial dysfunction are two key mediators of the secondary injury cascade in TBI pathology. TBI-induced oxidative damage results in the structural and functional impairments of cellular and subcellular components, such as mitochondria. Impairments of mitochondrial electron transfer chain and mitochondrial membrane potential result in a vicious cycle of free radical formation and cell apoptosis. The results of some preclinical and clinical studies, evaluating mitochondria-targeted therapies, such as mitochondria-targeted antioxidants and compounds with pleiotropic effects after TBI, are promising. As a proposed strategy in recent years, mitochondria-targeted multipotential therapy is a new hope, waiting to be confirmed. Moreover, based on the available findings, biologics, such as stem cell-based therapy and transplantation of mitochondria are novel potential strategies for the treatment of TBI; however, more studies are needed to clearly confirm the safety and efficacy of these strategies.
Collapse
Affiliation(s)
- Bahareh Hakiminia
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Babak Alikiaii
- Department of Anesthesiology and Intensive Care, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fariborz Khorvash
- Department of Neurology, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sarah Mousavi
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Revisiting Traumatic Brain Injury: From Molecular Mechanisms to Therapeutic Interventions. Biomedicines 2020; 8:biomedicines8100389. [PMID: 33003373 PMCID: PMC7601301 DOI: 10.3390/biomedicines8100389] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
Studying the complex molecular mechanisms involved in traumatic brain injury (TBI) is crucial for developing new therapies for TBI. Current treatments for TBI are primarily focused on patient stabilization and symptom mitigation. However, the field lacks defined therapies to prevent cell death, oxidative stress, and inflammatory cascades which lead to chronic pathology. Little can be done to treat the mechanical damage that occurs during the primary insult of a TBI; however, secondary injury mechanisms, such as inflammation, blood-brain barrier (BBB) breakdown, edema formation, excitotoxicity, oxidative stress, and cell death, can be targeted by therapeutic interventions. Elucidating the many mechanisms underlying secondary injury and studying targets of neuroprotective therapeutic agents is critical for developing new treatments. Therefore, we present a review on the molecular events following TBI from inflammation to programmed cell death and discuss current research and the latest therapeutic strategies to help understand TBI-mediated secondary injury.
Collapse
|
6
|
Abstract
Epilepsy is a significant worldwide public health problem that leads to reduced quality of life and negative psychosocial consequences and significantly increases mortality rates in those who are affected. The development of epilepsy from subarachnoid hemorrhage (SAH) has an important negative impact on long-term survival, functional status, and cognitive recovery in patients following aneurysmal rupture. Anticonvulsant medication (AED) administration to prevent the development of epilepsy following SAH is controversial, and studies to date have not shown effectiveness of AED use as prophylaxis. This paper reviews the pathophysiology of SAH in the development of epilepsy, the scope of the problem of epilepsy related to SAH, and the studies that have evaluated AED administration as prophylaxis for seizures and epilepsy.
Collapse
|
7
|
Seizures Do Not Affect Disability and Mortality Outcomes of Stroke: A Population-Based Study. J Clin Med 2019; 8:jcm8112006. [PMID: 31744217 PMCID: PMC6912525 DOI: 10.3390/jcm8112006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
Although seizures are frequently seen after cerebrovascular accidents, their effects on long-term outcome in stroke patients are still unknown. Therefore, the aim of this study was to investigate the relationship between post-stroke seizures and the risk of long-term disability and mortality in stroke patients. This study is part of a larger population-based study. All patients were prospectively followed up by a face-to-face interview or a structured telephone interview. We enrolled 635 patients with first-ever stroke and without a history of seizures. Prevalence of ischemic stroke (IS) was 85.2%, while the remaining 14.8% of patients were affected by intracerebral hemorrhage (ICH). During the study period, 51 subjects (8%) developed post-stroke seizures. Patients with post-stroke seizures were younger, had a higher prevalence of ICH, had a more severe stroke at admission, were more likely to have an IS involving the total anterior circulation, and were more likely to have a lobar ICH than patients without seizures. Moreover, subjects with seizures had more frequently hemorrhagic transformation after IS and cortical strokes. At 24 months, the risk of disability in patients with seizures was almost twice than in those without seizures. However, the negative effect of seizures disappeared in multivariate analysis. Kaplan-Meier survival curves at 12 years were not significantly different between patients with and without post-stroke seizures. Using the Cox multivariate analysis, age, NIHSS at admission, and pre-stroke mRS were independently associated with all-cause long-term mortality. In our sample, seizures did not impair long-term outcome in patients affected by cerebrovascular accidents. The not significant, slight difference in favor of a better survival for patients with seizures may be attributed to the slight age difference between the two groups.
Collapse
|
8
|
Vawda R, Badner A, Hong J, Mikhail M, Lakhani A, Dragas R, Xhima K, Barretto T, Librach CL, Fehlings MG. Early Intravenous Infusion of Mesenchymal Stromal Cells Exerts a Tissue Source Age-Dependent Beneficial Effect on Neurovascular Integrity and Neurobehavioral Recovery After Traumatic Cervical Spinal Cord Injury. Stem Cells Transl Med 2019; 8:639-649. [PMID: 30912623 PMCID: PMC6591557 DOI: 10.1002/sctm.18-0192] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/04/2019] [Indexed: 12/16/2022] Open
Abstract
Localized vascular disruption after traumatic spinal cord injury (SCI) triggers a cascade of secondary events, including inflammation, gliosis, and scarring, that can further impact recovery. In addition to immunomodulatory and neurotrophic properties, mesenchymal stromal cells (MSCs) possess pericytic characteristics. These features make MSCs an ideal candidate for acute cell therapy targeting vascular disruption, which could reduce the severity of secondary injury, enhance tissue preservation and repair, and ultimately promote functional recovery. A moderately severe cervical clip compression/contusion injury was induced at C7‐T1 in adult female rats, followed by an intravenous tail vein infusion 1 hour post‐SCI of (a) term‐birth human umbilical cord perivascular cells (HUCPVCs); (b) first‐trimester human umbilical cord perivascular cells (FTM HUCPVCs); (c) adult bone marrow mesenchymal stem cells; or (d) vehicle control. Weekly behavioral testing was performed. Rats were sacrificed at 24 hours or 10 weeks post‐SCI and immunohistochemistry and ultrasound imaging were performed. Both term and FTM HUCPVC‐infused rats displayed improved (p < .05) grip strength compared with vehicle controls. However, only FTM HUCPVC‐infusion led to significant weight gain. All cell infusion treatments resulted in reduced glial scarring (p < .05). Cell infusion also led to increased axonal, myelin, and vascular densities (p < .05). Although post‐traumatic cavity volume was reduced with cell infusion, this did not reach significance. Taken together, we demonstrate selective long‐term functional recovery alongside histological improvements with HUCPVC infusion in a clinically relevant model of cervical SCI. Our findings highlight the potential of these cells for acute therapeutic intervention after SCI.
Collapse
Affiliation(s)
- Reaz Vawda
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Anna Badner
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - James Hong
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alam Lakhani
- CReATe Fertility Centre, Toronto, Ontario, Canada
| | - Rachel Dragas
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kristiana Xhima
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery and Spinal Program, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Green Tea and Red Tea from Camellia sinensis Partially Prevented the Motor Deficits and Striatal Oxidative Damage Induced by Hemorrhagic Stroke in Rats. Neural Plast 2018; 2018:5158724. [PMID: 30174686 PMCID: PMC6098885 DOI: 10.1155/2018/5158724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/17/2018] [Accepted: 07/16/2018] [Indexed: 12/27/2022] Open
Abstract
Green tea from Camellia sinensis plays a well-established neuroprotective role in several neurodegenerative diseases, including intracerebral hemorrhage (ICH). However, the other teas of the same plant do not have their properties well understood; but they can be as effective as green tea as an alternative therapy. In this study, we investigated the effects of supplementation with green tea and red tea from Camellia sinensis on motor deficits and striatum oxidative damage in rats submitted to hemorrhagic stroke (ICH). Male Wistar rats were supplemented with green tea, red tea, or vehicle for 10 days prior to ICH induction. After injury, the rats were submitted to motor tests (open field for locomotion, rotarod for balance, and neurological deficit scale (NDS)) 1, 3, and 7 days after ICH induction, while the tea supplementation was maintained. Subsequently, the rats were euthanized to striatal tissue dissection for biochemical analyzes (lipid peroxidation, reactive oxygen species, glutathione levels, and total antioxidant capacity). ICH caused locomotor and balance deficits, as well as increased the neurological deficit (NDS). Only red tea prevented locomotor deficits after injury. Green tea and red tea prevented balance deficits on the seventh day after ICH. On NDS evaluation, green tea presented a better neuroprotection than red tea (until day 3 after ICH injury). In addition, ICH increased reactive oxygen species and lipid peroxidation levels, without altering antioxidant markers. Green and red teas were effective in decreasing the lipid peroxidation levels. Therefore, green and red teas partially prevented the motor deficits and striatal oxidative damage induced by ICH. Based on our results, we can consider that the two teas seem to be equally effective to prevent motor deficits and striatal oxidative damage induced by hemorrhagic stroke in rats.
Collapse
|
10
|
Righy C, Bozza MT, Oliveira MF, Bozza FA. Molecular, Cellular and Clinical Aspects of Intracerebral Hemorrhage: Are the Enemies Within? Curr Neuropharmacol 2016; 14:392-402. [PMID: 26714583 PMCID: PMC4876594 DOI: 10.2174/1570159x14666151230110058] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 11/28/2015] [Accepted: 12/29/2015] [Indexed: 12/13/2022] Open
Abstract
Hemorrhagic stroke is a disease with high incidence and mortality rates. In addition to the mass lesions that result from hemorrhagic stroke, substances such as the blood-derived products (BDP) (hemoglobin (Hb), heme and iron) induce a potent inflammatory response and exert direct toxic effects on neurons, astrocytes, and microglia. In the present review, we discuss the mechanisms of brain injury secondary to hemorrhagic stroke, focusing on the involvement of BDP as major players of cellular redox imbalance, inflammation, and glutamate excitotoxicity. Potential natural mechanisms of protection against free Hb and heme such as haptoglobin and hemopexin, respectively, are highlighted. We finally discuss the experimental and clinical trials targeting free iron and heme scavenging as well as inflammation, as potential new therapies to minimize the devastating effects of hemorrhagic stroke on brain structure and function.
Collapse
Affiliation(s)
- Cássia Righy
- Avenida Brasil 4.365, Manguinhos, Rio de Janeiro-RJ, CEP 21.040-900, Pavilhão Gaspar Viana.
| | | | | | | |
Collapse
|
11
|
Rifkind JM, Mohanty JG, Nagababu E. The pathophysiology of extracellular hemoglobin associated with enhanced oxidative reactions. Front Physiol 2015; 5:500. [PMID: 25642190 PMCID: PMC4294139 DOI: 10.3389/fphys.2014.00500] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/02/2014] [Indexed: 11/16/2022] Open
Abstract
Hemoglobin (Hb) continuously undergoes autoxidation producing superoxide which dismutates into hydrogen peroxide (H2O2) and is a potential source for subsequent oxidative reactions. Autoxidation is most pronounced under hypoxic conditions in the microcirculation and for unstable dimers formed at reduced Hb concentrations. In the red blood cell (RBC), oxidative reactions are inhibited by an extensive antioxidant system. For extracellular Hb, whether from hemolysis of RBCs and/or the infusion of Hb-based blood substitutes, the oxidative reactions are not completely neutralized by the available antioxidant system. Un-neutralized H2O2 oxidizes ferrous and ferric Hbs to Fe(IV)-ferrylHb and OxyferrylHb, respectively. FerrylHb further reacts with H2O2 producing heme degradation products and free iron. OxyferrylHb, in addition to Fe(IV) contains a free radical that can undergo additional oxidative reactions. Fe(III)Hb produced during Hb autoxidation also readily releases heme, an additional source for oxidative stress. These oxidation products are a potential source for oxidative reactions in the plasma, but to a greater extent when the lower molecular weight Hb dimers are taken up into cells and tissues. Heme and oxyferryl have been shown to have a proinflammatory effect further increasing their potential for oxidative stress. These oxidative reactions contribute to a number of pathological situations including atherosclerosis, kidney malfunction, sickle cell disease, and malaria. The toxic effects of extracellular Hb are of particular concern with hemolytic anemia where there is an increase in hemolysis. Hemolysis is further exacerbated in various diseases and their treatments. Blood transfusions are required whenever there is an appreciable decrease in RBCs due to hemolysis or blood loss. It is, therefore, essential that the transfused blood, whether stored RBCs or the blood obtained by an Autologous Blood Recovery System from the patient, do not further increase extracellular Hb.
Collapse
Affiliation(s)
- Joseph M Rifkind
- Molecular Dynamics Section, Laboratory of Molecular Gerontology, National Institute on Aging Baltimore, MD, USA ; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions Baltimore, MD, USA
| | - Joy G Mohanty
- Molecular Dynamics Section, Laboratory of Molecular Gerontology, National Institute on Aging Baltimore, MD, USA
| | - Enika Nagababu
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions Baltimore, MD, USA
| |
Collapse
|
12
|
Piloni NE, Fermandez V, Videla LA, Puntarulo S. Acute iron overload and oxidative stress in brain. Toxicology 2013; 314:174-82. [PMID: 24120471 DOI: 10.1016/j.tox.2013.09.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/09/2013] [Accepted: 09/30/2013] [Indexed: 12/11/2022]
Abstract
An in vivo model in rat was developed by intraperitoneally administration of Fe-dextran to study oxidative stress triggered by Fe-overload in rat brain. Total Fe levels, as well as the labile iron pool (LIP) concentration, in brain from rats subjected to Fe-overload were markedly increased over control values, 6h after Fe administration. In this in vivo Fe overload model, the ascorbyl (A)/ascorbate (AH(-)) ratio, taken as oxidative stress index, was assessed. The A/AH(-) ratio in brain was significantly higher in Fe-dextran group, in relation to values in control rats. Brain lipid peroxidation indexes, thiobarbituric acid reactive substances (TBARS) generation rate and lipid radical (LR) content detected by Electron Paramagnetic Resonance (EPR), in Fe-dextran supplemented rats were similar to control values. However, values of nuclear factor-kappaB deoxyribonucleic acid (NFκB DNA) binding activity were significantly increased (30%) after 8h of Fe administration, and catalase (CAT) activity was significantly enhanced (62%) 21h after Fe administration. Significant enhancements in Fe content in cortex (2.4 fold), hippocampus (1.6 fold) and striatum (2.9 fold), were found at 6h after Fe administration. CAT activity was significantly increased after 8h of Fe administration in cortex, hippocampus and striatum (1.4 fold, 86, and 47%, respectively). Fe response in the whole brain seems to lead to enhanced NF-κB DNA binding activity, which may contribute to limit oxygen reactive species-dependent damage by effects on the antioxidant enzyme CAT activity. Moreover, data shown here clearly indicate that even though Fe increased in several isolated brain areas, this parameter was more drastically enhanced in striatum than in cortex and hippocampus. However, comparison among the net increase in LR generation rate, in different brain areas, showed enhancements in cortex lipid peroxidation, without changes in striatum and hippocampus LR generation rate after 6h of Fe overload. This information has potential clinical relevance, as it could be the key to understand specific brain damage occurring in conditions of Fe overload.
Collapse
Affiliation(s)
- Natacha E Piloni
- Physical Chemistry-Institute of Biochemistry and Molecular Medicine (IBIMOL), School of Pharmacy and Biochemistry, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
13
|
Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, Radak Z, Calabrese EJ, Cuzzocrea S. Traumatic brain injury: oxidative stress and neuroprotection. Antioxid Redox Signal 2013; 19:836-53. [PMID: 23547621 DOI: 10.1089/ars.2012.4981] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE A vast amount of circumstantial evidence implicates high energy oxidants and oxidative stress as mediators of secondary damage associated with traumatic brain injury. The excessive production of reactive oxygen species due to excitotoxicity and exhaustion of the endogenous antioxidant system induces peroxidation of cellular and vascular structures, protein oxidation, cleavage of DNA, and inhibition of the mitochondrial electron transport chain. RECENT ADVANCES Different integrated responses exist in the brain to detect oxidative stress, which is controlled by several genes termed vitagens. Vitagens encode for cytoprotective heat shock proteins, and thioredoxin and sirtuins. CRITICAL ISSUES AND FUTURE DIRECTIONS This article discusses selected aspects of secondary brain injury after trauma and outlines key mechanisms associated with toxicity, oxidative stress, inflammation, and necrosis. Finally, this review discusses the role of different oxidants and presents potential clinically relevant molecular targets that could be harnessed to treat secondary injury associated with brain trauma.
Collapse
Affiliation(s)
- Carolin Cornelius
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Molecular and cellular mechanisms underlying the role of blood vessels in spinal cord injury and repair. Cell Tissue Res 2012; 349:269-88. [PMID: 22592628 DOI: 10.1007/s00441-012-1440-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/24/2012] [Indexed: 02/07/2023]
Abstract
Spinal cord injury causes immediate damage of nervous tissue accompanied by the loss of motor and sensory function. The limited self-repair ability of damaged nervous tissue underlies the need for reparative interventions to restore function after spinal cord injury. Blood vessels play a crucial role in spinal cord injury and repair. Injury-induced loss of local blood vessels and a compromised blood-brain barrier contribute to inflammation and ischemia and thus to the overall damage to the nervous tissue of the spinal cord. Lack of vasculature and leaking blood vessels impede endogenous tissue repair and limit prospective repair approaches. A reduction of blood vessel loss and the restoration of blood vessels so that they no longer leak might support recovery from spinal cord injury. The promotion of new blood vessel formation (i.e., angio- and vasculogenesis) might aid repair but also incorporates the danger of exacerbating tissue loss and thus functional impairment. The delicate interplay between cells and molecules that govern blood vessel repair and formation determines the extent of damage and the success of reparative interventions. This review deals with the cellular and molecular mechanisms underlying the role of blood vessels in spinal cord injury and repair.
Collapse
|
15
|
Lapchak PA. Identifying Vascular Targets to Treat Hemorrhagic Stroke. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Shin EJ, Jeong JH, Chung YH, Kim WK, Ko KH, Bach JH, Hong JS, Yoneda Y, Kim HC. Role of oxidative stress in epileptic seizures. Neurochem Int 2011; 59:122-37. [PMID: 21672578 PMCID: PMC3606551 DOI: 10.1016/j.neuint.2011.03.025] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 03/27/2011] [Accepted: 03/28/2011] [Indexed: 11/16/2022]
Abstract
Oxidative stress resulting from excessive free-radical release is likely implicated in the initiation and progression of epilepsy. Therefore, antioxidant therapies aimed at reducing oxidative stress have received considerable attention in epilepsy treatment. However, much evidence suggests that oxidative stress does not always have the same pattern in all seizures models. Thus, this review provides an overview aimed at achieving a better understanding of this issue. We summarize work regarding seizure models (i.e., genetic rat models, kainic acid, pilocarpine, pentylenetetrazol, and trimethyltin), oxidative stress as an etiologic factor in epileptic seizures (i.e., impairment of antioxidant systems, mitochondrial dysfunction, involvement of redox-active metals, arachidonic acid pathway activation, and aging), and antioxidant strategies for seizure treatment. Combined, this review highlights pharmacological mechanisms associated with oxidative stress in epileptic seizures and the potential for neuroprotection in epilepsy that targets oxidative stress and is supported by effective antioxidant treatment.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul 136-705, South Korea
| | - Kwang-Ho Ko
- Pharmacology Laboratory, College of Pharmacy, Seoul National University, Seoul 143-701, South Korea
| | - Jae-Hyung Bach
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Ishikawa 920-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharamcology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| |
Collapse
|
17
|
Vitamin C deficiency: more than just a nutritional disorder. GENES AND NUTRITION 2011; 6:341-6. [PMID: 21614623 DOI: 10.1007/s12263-011-0237-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/10/2011] [Indexed: 10/18/2022]
Abstract
Although vitamin C deficiency and scurvy are generally considered as pure nutritional disorders, only a minority of the vitamin C concentration is determined by food intake. In the presence of transition metals (iron and copper), the antiscorbutic factor shifts from an antioxidant to a pro-oxidant function. Haptoglobin (Hp) is a plasma α-2 glycoprotein characterized by 3 common phenotypes (Hp 1-1, Hp 2-1 and Hp 2-2). Its free hemoglobin (Hb)-binding capacity prevents Hb-driven oxidative damage. When the antioxidant capacity of Hp is insufficient, its role is taken over by hemopexin (heme-binding protein) and by vitamin C (free radical scavenger). The Hp 2-2 phenotype has a lower capacity to inhibit oxidation and vitamin C depletion. In this article, two consequences of this major finding are tackled. The Hp polymorphism is an important non-nutritional modifying factor in the pathogenesis of vitamin C deficiency and scurvy, which may explain the success of long-range human migration by the natural selection of some populations characterized by high Hp 1 allele frequencies. Moreover, we propose tailoring the recommended dietary allowance (RDA) values of vitamin C, taking into consideration the Hp phenotype dependency.
Collapse
|
18
|
Rink C, Khanna S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid Redox Signal 2011; 14:1889-903. [PMID: 20673202 PMCID: PMC3078506 DOI: 10.1089/ars.2010.3474] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The significance of the hypoxia component of stroke injury is highlighted by hypermetabolic brain tissue enriched with arachidonic acid (AA), a 22:6n-3 polyunsaturated fatty acid. In an ischemic stroke environment in which cerebral blood flow is arrested, oxygen-starved brain tissue initiates the rapid cleavage of AA from the membrane phospholipid bilayer. Once free, AA undergoes both enzyme-independent and enzyme-mediated oxidative metabolism, resulting in the formation of number of biologically active metabolites which themselves contribute to pathological stroke outcomes. This review is intended to examine two divergent roles of molecular dioxygen in brain tissue as (1) a substrate for life-sustaining homeostatic metabolism of glucose and (2) a substrate for pathogenic metabolism of AA under conditions of stroke. Recent developments in research concerning supplemental oxygen therapy as an intervention to correct the hypoxic component of stroke injury are discussed.
Collapse
Affiliation(s)
- Cameron Rink
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | |
Collapse
|
19
|
Abstract
One of the most investigated molecular mechanisms involved in the secondary pathophysiology of acute spinal cord injury (SCI) is free radical-induced, iron-catalyzed lipid peroxidation (LP) and protein oxidative/nitrative damage to spinal neurons, glia, and microvascular cells. The reactive nitrogen species peroxynitrite and its highly reactive free radicals are key initiators of LP and protein nitration in the injured spinal cord, the biochemistry, and pathophysiology of which are first of all reviewed in this article. This is followed by a presentation of the antioxidant mechanistic approaches and pharmacological compounds that have been shown to have neuroprotective properties in preclinical SCI models. Two of these, which act by inhibition of LP, are high-dose treatment with the glucocorticoid steroid methylprednisolone (MP) and the nonglucocorticoid 21-aminosteroid tirilazad, have been demonstrated in the multicenter NASCIS clinical trials to produce at least a modest improvement in neurological recovery when administered within the first 8 hours after SCI. Although these results have provided considerable validation of oxidative damage as a clinically practical neuroprotective target, there is a need for the discovery of safer and more effective antioxidant compounds for acute SCI.
Collapse
Affiliation(s)
- Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40506, USA.
| |
Collapse
|
20
|
García-Macia M, Vega-Naredo I, De Gonzalo-Calvo D, Rodríguez-González SM, Camello PJ, Camello-Almaraz C, Martín-Cano FE, Rodríguez-Colunga MJ, Pozo MJ, Coto-Montes AM. Melatonin induces neural SOD2 expression independent of the NF-kappaB pathway and improves the mitochondrial population and function in old mice. J Pineal Res 2011; 50:54-63. [PMID: 21062349 DOI: 10.1111/j.1600-079x.2010.00809.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aging is commonly defined as a physiological phenomenon associated with morphological and functional deleterious changes in which oxidative stress has a fundamental impact; therefore, readjusting the oxidative balance should have beneficial effects. In our study, we tested the antioxidant melatonin in old mouse brains and showed positive effects at the cellular and mitochondrial levels. Melatonin attenuated β-amyloid protein expression and α-synuclein deposits in the brain compared to aged group. Furthermore, oxidative stress was increased by aging and induced the nuclear translocation of nuclear factor-kappa B (NF-κB), which was suppressed by melatonin treatment. The antioxidant mitochondrial expression, superoxide dismutase 2 (SOD2), was increased in both control and melatonin-treated old mice, despite the different activation states of the NF-κB pathway. The NF-κB pathway was activated in the old mice, which may be explained by this group's response to the increased oxidative insult; this insult was inhibited in melatonin-treated animals, showing this group an increase in active mitochondria population that was not observed in old group. We also report that melatonin is capable of restoring the mitochondrial potential of age-damaged neurons. In conclusion, melatonin's beneficial effects on brain aging are linked to the increase in mitochondrial membrane potential and SOD2 expression, which probably reduces the mitochondrial contribution to the oxidative stress imbalance.
Collapse
Affiliation(s)
- Marina García-Macia
- Department of Morphology and Cellular Biology, Medicine Faculty, Oviedo University, Oviedo, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lapchak PA, Wu Q. Vascular Dysfunction in Brain Hemorrhage: Translational Pathways to Developing New Treatments from Old Targets. JOURNAL OF NEUROLOGY & NEUROPHYSIOLOGY 2011; 2011:S1-e001. [PMID: 22400125 PMCID: PMC3293216 DOI: 10.4172/2155-9562.s1-e001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hemorrhagic stroke which is a form of stroke that affects 20% of all stroke patients is a devastating condition for which new treatments must be developed. Current treatment methods are quite insufficient to reduce long term morbidity and high mortality rate, up to 50%, associated with bleeding into critical brain structures, into ventricular spaces and within the subarachnoid space. During the last 10-15 years, significant advances in the understanding of important mechanisms that contribute to cell death and clinical deficits have been made. The most important observations revolve around a key set of basic mechanisms that are altered in brain bleeding models, including activation of membrane metalloproteinases, oxidative stress and both inflammatory and coagulation pathways. Moreover, it is now becoming apparent that brain hemorrhage can activate the ischemic stroke cascade in neurons, glial cells and the vascular compartment. The activation of multiple pathways allows comes the opportunity to intervene pharmacologically using monotherapy or combination therapy. Ultimately, combination therapy or pleiotropic compounds with multi-target activities should prove to be more efficacious than any single therapy alone. This article provides a comprehensive look at possible targets for small molecule intervention as well as some new approaches that result in metabolic down-regulation or inhibition of multiple pathways simultaneously.
Collapse
Affiliation(s)
- Paul A. Lapchak
- Director of Translational Research, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D- 2091, 110 N, George Burns Road, Los Angeles, CA 90048 USA
| | - Qiang Wu
- Project Scientist, Cedars-Sinai Medical Center, Department of Neurology, Davis Research Building, D-2094E, 110 N. George Burns Road, Los Angeles, CA 90048 USA
| |
Collapse
|
22
|
Abstract
Heme is an essential molecule in aerobic organisms. Heme consists of protoporphyrin IX and a ferrous (Fe(2+)) iron atom, which has high affinity for oxygen (O(2)). Hemoglobin, the major oxygen-carrying protein in blood, is the most abundant heme-protein in animals and humans. Hemoglobin consists of four globin subunits (alpha(2)beta(2)), with each subunit carrying a heme group. Ferrous (Fe(2+)) hemoglobin is easily oxidized in circulation to ferric (Fe(3+)) hemoglobin, which readily releases free hemin. Hemin is hydrophobic and intercalates into cell membranes. Hydrogen peroxide can split the heme ring and release "free" redox-active iron, which catalytically amplifies the production of reactive oxygen species. These oxidants can oxidize lipids, proteins, and DNA; activate cell-signaling pathways and oxidant-sensitive, proinflammatory transcription factors; alter protein expression; perturb membrane channels; and induce apoptosis and cell death. Heme-derived oxidants induce recruitment of leukocytes, platelets, and red blood cells to the vessel wall; oxidize low-density lipoproteins; and consume nitric oxide. Heme metabolism, extracellular and intracellular defenses against heme, and cellular cytoprotective adaptations are emphasized. Sickle cell disease, an archetypal example of hemolysis, heme-induced oxidative stress, and cytoprotective adaptation, is reviewed.
Collapse
Affiliation(s)
- John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
23
|
Abstract
OBJECTIVE The potential relation between metabolic activity within the central nervous system and retention of cognitive functioning capacity was assessed. METHODS A detailed literature review was conducted and summarized. RESULTS A large body of scientific evidence describes the interactions among cognitive activity, oxidative stress, neurodegeneration, neuroprotection, cognitive aging, and retention of cognitive functioning ability. CONCLUSION Maintenance of redox balance within the central nervous system can forestall cognitive decline and promote cognitive longevity.
Collapse
|
24
|
Hall ED, Vaishnav RA, Mustafa AG. Antioxidant therapies for traumatic brain injury. Neurotherapeutics 2010; 7:51-61. [PMID: 20129497 PMCID: PMC2818465 DOI: 10.1016/j.nurt.2009.10.021] [Citation(s) in RCA: 272] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/19/2009] [Indexed: 12/31/2022] Open
Abstract
Free radical-induced oxidative damage reactions, and membrane lipid peroxidation (LP), in particular, are among the best validated secondary injury mechanisms in preclinical traumatic brain injury (TBI) models. In addition to the disruption of the membrane phospholipid architecture, LP results in the formation of cytotoxic aldehyde-containing products that bind to cellular proteins and impair their normal functions. This article reviews the progress of the past three decades in regard to the preclinical discovery and attempted clinical development of antioxidant drugs designed to inhibit free radical-induced LP and its neurotoxic consequences via different mechanisms including the O(2)(*-) scavenger superoxide dismutase and the lipid peroxidation inhibitor tirilazad. In addition, various other antioxidant agents that have been shown to have efficacy in preclinical TBI models are briefly presented, such as the LP inhibitors U83836E, resveratrol, curcumin, OPC-14177, and lipoic acid; the iron chelator deferoxamine and the nitroxide-containing antioxidants, such as alpha-phenyl-tert-butyl nitrone and tempol. A relatively new antioxidant mechanistic strategy for acute TBI is aimed at the scavenging of aldehydic LP byproducts that are highly neurotoxic with "carbonyl scavenging" compounds. Finally, it is proposed that the most effective approach to interrupt posttraumatic oxidative brain damage after TBI might involve the combined treatment with mechanistically complementary antioxidants that simultaneously scavenge LP-initiating free radicals, inhibit LP propagation, and lastly remove neurotoxic LP byproducts.
Collapse
Affiliation(s)
- Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA.
| | | | | |
Collapse
|
25
|
Vecchione C, Frati A, Di Pardo A, Cifelli G, Carnevale D, Gentile MT, Carangi R, Landolfi A, Carullo P, Bettarini U, Antenucci G, Mascio G, Busceti CL, Notte A, Maffei A, Cantore GP, Lembo G. Tumor Necrosis Factor-α Mediates Hemolysis-Induced Vasoconstriction and the Cerebral Vasospasm Evoked by Subarachnoid Hemorrhage. Hypertension 2009; 54:150-6. [DOI: 10.1161/hypertensionaha.108.128124] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Carmine Vecchione
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alessandro Frati
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alba Di Pardo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giuseppe Cifelli
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Daniela Carnevale
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Maria Teresa Gentile
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Rosa Carangi
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alessandro Landolfi
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Pierluigi Carullo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Umberto Bettarini
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giovanna Antenucci
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giada Mascio
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Carla Letizia Busceti
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Antonella Notte
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Angelo Maffei
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Gian Paolo Cantore
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giuseppe Lembo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| |
Collapse
|
26
|
Langlois MR, De Buyzere ML, Delanghe JR. Plasma vitamin C for predicting cardiovascular disease: more than a nutritional biomarker. Acta Clin Belg 2009; 64:341-3. [PMID: 19810422 DOI: 10.1179/acb.2009.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- M R Langlois
- Department of Clinical Chemistry, Ghent University, De Pintelaan 185, B-9000 Ghent, Belgium
| | | | | |
Collapse
|
27
|
Lara FA, Kahn SA, da Fonseca AC, Bahia CP, Pinho JP, Graca-Souza AV, Houzel JC, de Oliveira PL, Moura-Neto V, Oliveira MF. On the fate of extracellular hemoglobin and heme in brain. J Cereb Blood Flow Metab 2009; 29:1109-20. [PMID: 19337276 DOI: 10.1038/jcbfm.2009.34] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major cause of disability in adults worldwide. The pathophysiology of this syndrome is complex, involving both inflammatory and redox components triggered by the extravasation of blood into the cerebral parenchyma. Hemoglobin, heme, and iron released therein seem be important in the brain damage observed in ICH. However, there is a lack of information concerning hemoglobin traffic and metabolism in brain cells. Here, we investigated the fate of hemoglobin and heme in cultured neurons and astrocytes, as well as in the cortex of adult rats. Hemoglobin was made traceable by conjugation to Alexa 488, whereas a fluorescent heme analogue (tin-protoporphyrin IX) was prepared to allow heme tracking. Using fluorescence microscopy we observed that neurons were more efficient in uptake hemoglobin and heme than astrocytes. Exposure of cortical neurons to hemoglobin or heme resulted in an oxidative stress condition. Viability assays showed that neurons were more susceptible to both hemoglobin and heme toxicity than astrocytes. Together, these results show that neurons, rather than astrocytes, preferentially take up hemoglobin-derived products, indicating that these cells are actively involved in the ICH-associated brain damage.
Collapse
Affiliation(s)
- Flavio A Lara
- Laboratório de Morfogênese Celular, Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Simoni J, Villanueva-Meyer J, Simoni G, Moeller JF, Wesson DE. Control of oxidative reactions of hemoglobin in the design of blood substitutes: role of the ascorbate-glutathione antioxidant system. Artif Organs 2009; 33:115-26. [PMID: 19178455 DOI: 10.1111/j.1525-1594.2008.00695.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Uncontrolled oxidative reactions of hemoglobin (Hb) are still the main unresolved problem for Hb-based blood substitute developers. Spontaneous oxidation of acellular ferrous Hb into a nonfunctional ferric Hb generates superoxide anion. Hydrogen peroxide, formed after superoxide anion dismutation, may react with ferrous/ferric Hb to produce toxic ferryl Hb, fluorescent heme degradation products, and/or protein-based free radicals. In the presence of free iron released from heme, superoxide anion and hydrogen peroxide might react via the Haber-Weiss and Fenton reactions to generate the hydroxyl radical. These highly reactive oxygen and heme species may not only be involved in shifting the cellular redox balance to the oxidized state that facilitates signal transduction and pro-inflammatory gene expression, but could also be involved in cellular and organ injury, and generation of vasoactive compounds such as isoprostanes and angiotensins. It is believed that these toxic species may be formed after administration of Hb-based blood substitutes, particularly in ischemic patients with a diminished ability to control oxidative reactions. Although varieties of antioxidant strategies have been suggested, this in vitro study examined the ability of the ascorbate-glutathione antioxidant system in preventing Hb oxidation and formation of its ferryl intermediate. The results suggest that although ascorbate is effective in reducing the formation of ferryl Hb, glutathione protects heme against excessive oxidation. Ascorbate without glutathione failed to protect the red blood cell membranes against Hb/hydrogen peroxide-mediated peroxidation. This study provides evidence that the ascorbate-glutathione antioxidant system is essential in attenuation of the pro-oxidant potential of redox active acellular Hbs, and superior to either ascorbate or glutathione alone.
Collapse
Affiliation(s)
- Jan Simoni
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.
| | | | | | | | | |
Collapse
|
29
|
Suprameatal approach for cochlear implantation in 45 Chinese children. Int J Pediatr Otorhinolaryngol 2008; 72:397-403. [PMID: 18221796 DOI: 10.1016/j.ijporl.2007.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 12/05/2007] [Accepted: 12/05/2007] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To investigate the feasibility of applying the suprameatal approach (SMA) for cochlear implantation in Chinese children with profound sensory hearing loss, and to demonstrate a technical modification incorporated in the procedure due to an observed racial difference. STUDY DESIGN Retrospective study. SETTING University hospital. PATIENTS Forty-five Chinese children (total 47 ears) with profound sensory hearing loss were surgically treated from May 2005 to May 2006. The patients were followed anywhere from 1 month to 20 months post-surgery, with 30 patients being followed for more than 6 months. INTERVENTIONS All patients received cochlear implantation through the suprameatal approach. In this procedure, the cochleostomy was performed in one stage after the suprameatal tunnel was finished, rather than the two-stage approach described by Kronenberg (who firstly introduced the suprameatal approach). Three patients with low-lying dura (which is considered to be the contraindication for cochlear implantation with SMA) were treated with a further modified surgical approach. RESULTS Among the 47 ears, full electrode pairs were completely inserted in 45 ears without surgical difficulties, but 1 ear was only fitted with 9 pairs of electrodes because of an ossified cochlea, and another with just 8 pairs of electrodes due to serious cochlear dysplasia. An intraoperative "gusher" occurred in the dysplasia case, and a small piece of temporalis muscle was used, along with biology glue, to seal the cochleostomy and prevent further leakage. In 1 case, the electrode was inserted into the cochlea through the tunnel lateral to the chorda tympani because adhesion had occurred between the incus and chorda tympani. There were no postoperative complications in any case. Thirty cases exhibited better hearing or speech development from cochlear implantation after more than 6 months of follow-up. CONCLUSIONS The SMA was found to be a simple and safe technique for cochlear implantation in Chinese children. It enables wide exposure of the middle ear, and is especially suitable for cases with a narrow facial recess, an anteriorly located facial nerve, or an ossified cochlea. It is almost impossible to injure the facial nerve or the chorda tympani nerve. The cochleostomy can be performed in one stage in those patients with a normal cochlea. With some modifications, a low-lying dura will not be the absolute contraindication of SMA.
Collapse
|
30
|
Balla J, Vercellotti GM, Jeney V, Yachie A, Varga Z, Jacob HS, Eaton JW, Balla G. Heme, heme oxygenase, and ferritin: how the vascular endothelium survives (and dies) in an iron-rich environment. Antioxid Redox Signal 2007; 9:2119-37. [PMID: 17767398 DOI: 10.1089/ars.2007.1787] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Iron-derived reactive oxygen species are involved in the pathogenesis of numerous vascular disorders. One abundant source of redox active iron is heme, which is inherently dangerous when it escapes from its physiologic sites. Here, we present a review of the nature of heme-mediated cytotoxicity and of the strategies by which endothelium manages to protect itself from this clear and present danger. Of all sites in the body, the endothelium may be at greatest risk of exposure to heme. Heme greatly potentiates endothelial cell killing mediated by leukocytes and other sources of reactive oxygen. Heme also promotes the conversion of low-density lipoprotein to cytotoxic oxidized products. Hemoglobin in plasma, when oxidized, transfers heme to endothelium and lipoprotein, thereby enhancing susceptibility to oxidant-mediated injury. As a defense against such stress, endothelial cells upregulate heme oxygenase-1 and ferritin. Heme oxygenase opens the porphyrin ring, producing biliverdin, carbon monoxide, and a most dangerous product-redox active iron. The latter can be effectively controlled by ferritin via sequestration and ferroxidase activity. These homeostatic adjustments have been shown to be effective in the protection of endothelium against the damaging effects of heme and oxidants; lack of adaptation in an iron-rich environment led to extensive endothelial damage in humans.
Collapse
Affiliation(s)
- József Balla
- Department of Medicine, University of Debrecen, Debrecen, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ayyildiz M, Coskun S, Yildirim M, Agar E. The Effects of Ascorbic Acid on Penicillin-induced Epileptiform Activity in Rats. Epilepsia 2007; 48:1388-95. [PMID: 17433052 DOI: 10.1111/j.1528-1167.2007.01080.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Epileptic seizure results from excessive discharge in a population of hyperexcitable neurons. A number of studies help to document the effects of active oxygen free radical scavengers such as alpha-tocopherol or ascorbic acid (vitamin C). In the present study, we examined the effects of ascorbic acid, at the six different doses, on penicillin-induced epileptiform activity. METHODS A single microinjection of penicillin (2.5 microl, 500 units, intracortically) into the left sensorimotor cortex induced epileptiform activity within 2-5 min, progressing to full seizure activity lasting approximately 3-5 h. In the first set of experiments, 30 min after penicillin injection, six different doses of ascorbic acid (25, 50, 100, 200, 400, or 800 mg/kg) were administered intraperitoneally (IP). The other group of animals received the effective dose of ascorbic acid (100 mg/kg, IP) for 7 days. Ascorbic acid administration was stopped 24 h before penicillin treatment. Another group of rats received the effective dose of ascorbic acid (100 mg/kg, IP) 30 min before penicillin treatment. In the second set of experiments, the lipid peroxidation (MDA) and reduced glutathione (GSH) levels of brain were measured in the control, control + ascorbic acid, penicillin, and penicillin + ascorbic acid groups. RESULTS Ascorbic acid, at the low dose (50, 100 mg/kg, 30 min after penicillin injection), decreased both the frequency and amplitude of penicillin-induced epileptiform activity in rats. Ascorbic acid, at intermediate doses (200, 400 mg/kg, 30 min after penicillin injection), decreased the frequency of epileptiform activity without changing the amplitude. Ascorbic acid, at the lowest dose (25 mg/kg) and highest dose (800 mg/kg) (30 min after penicillin injection), did not change either the frequency or amplitude of epileptiform activity. Ascorbic acid, at the low dose (100 mg/kg) was the most effective dose in changing the frequency and amplitude of penicillin-induced epileptiform activity. Pretreatment with ascorbic acid (100 mg/kg) 30 min before penicillin treatment caused a significant delay in the onset of penicillin-induced epileptiform activity. Pretreatment with ascorbic acid (100 mg/kg) for 7 days did not change the latency of epileptiform activity. The most effective dose of ascorbic acid (100 mg/kg) prevented both the decrease in GSH level and the increase in lipid peroxidation level (MDA) occurring after penicillin-induced epileptiform activity. CONCLUSIONS These data indicate that ascorbic acid has neuroprotective activity against penicillin-induced epileptiform electrocorticogram activity.
Collapse
Affiliation(s)
- Mustafa Ayyildiz
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | | | | | | |
Collapse
|
32
|
Abstract
Because heme oxygenase (HO) is the rate limiting enzyme in the degradation of the pro-oxidant hemin/heme from blood, here we investigated the contribution of the inducible HO-1 to early brain injury produced by intracerebral haemorrhage (ICH). We found that after induction of ICH, HO-1 proteins were highly detectable in the peri-ICH region predominantly in microglia/macrophages and endothelial cells. Remarkably, the injury volume was significantly smaller in HO-1 knockout (HO-1-/-) mice than in wild-type controls 24 and 72 h after ICH. Although the brain water content did not appear to be significantly different, the protection in HO-1-/- mice was associated with a marked reduction in ICH-induced leucocyte infiltration, microglia/macrophage activation and free radical levels. These data reveal a previously unrecognized role of HO-1 in early brain injury after ICH. Thus, modulation of HO-1 signalling should be assessed further in clinical settings, especially for haemorrhagic states.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sylvain Doré
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
33
|
Abstract
Intracerebral hemorrhage (ICH) is a devastating clinical event without effective therapies. Increasing evidence suggests that inflammatory mechanisms are involved in the progression of ICH-induced brain injury. Inflammation is mediated by cellular components, such as leukocytes and microglia, and molecular components, including prostaglandins, chemokines, cytokines, extracellular proteases, and reactive oxygen species. Better understanding of the role of the ICH-induced inflammatory response and its potential for modulation might have profound implications for patient treatment. In this review, a summary of the available literature on the inflammatory responses after ICH is presented along with discussion of some of the emerging opportunities for potential therapeutic strategies. In the near future, additional strategies that target inflammation could offer exciting new promise in the therapeutic approach to ICH.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
34
|
Radeloff A, Unkelbach MH, Tillein J, Braun S, Helbig S, Gstöttner W, Adunka OF. Impact of Intrascalar Blood on Hearing. Laryngoscope 2007; 117:58-62. [PMID: 17202931 DOI: 10.1097/01.mlg.0000242073.02488.f4] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE/HYPOTHESIS The objective of this controlled animal study was to evaluate the effects of intrascalar blood on hearing. MATERIAL AND METHODS Eight guinea pigs underwent intrascalar administration of their own blood in one ear and control solution in the contralateral ear. Solutions were applied through cochleostomy to the scala tympani. Compound action potential (CAP) thresholds were determined before administration and at different intervals for 2 months thereafter. RESULTS Immediate deterioration of thresholds was seen mainly in the high-frequency range, averaging 27 dB and 20 dB in the study and control groups, respectively. At day 3, threshold shifts recovered in the control group but remained in the low-frequency range in the study group. An extensive recovery was seen in both groups. However, permanent threshold shifts persisted. There was an enhanced shift of thresholds of up to 7 dB in the study group. CONCLUSIONS Even small amounts of intrascalar blood seem to cause transient and permanent detrimental effects on cochlear function. In procedures involving opening of the otic capsule-like stapes surgery and cochlear implantation with hearing preservation-minimizing surgical blood admixture to intracochlear compartments seems therefore fundamental.
Collapse
Affiliation(s)
- Andreas Radeloff
- Department of Otolaryngology-Head and Neck Surgery, JW Goethe-University, Frankfurt am Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Aydin S. Antioxidant Status, α-Amylase Production, Growth, and Survival of Hemoglobin Bearing Escherichia coli Exposed to Hypochlorous Acid. BIOCHEMISTRY (MOSCOW) 2005; 70:1369-76. [PMID: 16417460 DOI: 10.1007/s10541-005-0271-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present work, two matched strains of E. coli that bear a recombinant R-amylase gene (MK57) or the R-amylase gene and vgb (MK79-hemoglobin expressing strain) were exposed to HOCl. In these cells, glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), alpha-amylase production, growth and lethality were assessed in the presence and absence of HOCl. It was observed that the hemoglobin makes cells highly susceptible to killing by HOCl. The maximum survival for both strains was with stationary phase cells at any concentration of HOCl. Both strains grown in the presence of 0.0125-0.075 mg/liter HOCl showed a substantial increase in SOD activity and GSH level, with MK79 being the most increased strain in this respect, while the level of CAT activity was decreased in a dose depended manner. Growth of MK57 and MK79 strains decreased as HOCl concentration increased. However, HOCl at concentration above zero enhanced alpha-amylase production (about 2-fold) in both MK79 and MK57. Furthermore, total amylase production (at all HOCl concentrations) by MK79 was always greater than that by MK57. The results indicate that except for survival, the hemoglobin helps cells to grow better and produces more recombinant products and activates general defense systems more in response to oxidative stress when compared with the non-hemoglobin-containing counterpart.
Collapse
Affiliation(s)
- S Aydin
- Department of Biochemistry and Clinical Biochemistry, Firat University, Faculty of Medicine, Elazig, 23119, Turkey.
| |
Collapse
|
37
|
Balla J, Vercellotti GM, Jeney V, Yachie A, Varga Z, Eaton JW, Balla G. Heme, heme oxygenase and ferritin in vascular endothelial cell injury. Mol Nutr Food Res 2005; 49:1030-43. [PMID: 16208635 DOI: 10.1002/mnfr.200500076] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Iron-derived reactive oxygen species are implicated in the pathogenesis of numerous vascular disorders including atherosclerosis, microangiopathic hemolytic anemia, vasculitis, and reperfusion injury. One abundant source of redox active iron is heme, which is inherently dangerous when released from intracellular heme proteins. The present review concerns the involvement of heme in vascular endothelial cell damage and the strategies used by endothelium to minimize such damage. Exposure of endothelium to heme greatly potentiates cell killing mediated by polymorphonuclear leukocytes and other sources of reactive oxygen. Free heme also promotes the conversion of low-density lipoprotein (LDL) into cytotoxic oxidized products. Only because of its abundance, hemoglobin probably represents the most important potential source of heme within the vascular endothelium; hemoglobin in plasma, when oxidized, transfers heme to endothelium and LDL, thereby enhancing cellular susceptibility to oxidant-mediated injury. As a defense against such toxicity, upon exposure to heme or hemoglobin, endothelial cells up-regulate heme oxygenase-1 and ferritin. Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. The latter can be effectively controlled by ferritin via sequestration and ferroxidase activity. Ferritin serves as a protective gene by virtue of antioxidant, antiapoptotic, and antiproliferative actions. These homeostatic adjustments have been shown effective in the protection of endothelium against the damaging effects of exogenous heme and oxidants. The central importance of this protective system was recently highlighted by a child diagnosed with heme oxygenase-1 deficiency, who exhibited extensive endothelial damage.
Collapse
Affiliation(s)
- József Balla
- Department of Medicine, University of Debrecen, Debrecen, Hungary.
| | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Sayed M H Sadrzadeh
- Department of Laboratory Medicine, University of Washington, Harborview Medical Center, Seattle, WA 98104, USA.
| | | | | |
Collapse
|
39
|
Jeney V, Balla J, Yachie A, Varga Z, Vercellotti GM, Eaton JW, Balla G. Pro-oxidant and cytotoxic effects of circulating heme. Blood 2002; 100:879-87. [PMID: 12130498 DOI: 10.1182/blood.v100.3.879] [Citation(s) in RCA: 474] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Numerous pathologies may involve toxic side effects of free heme and heme-derived iron. Deficiency of the heme-catabolizing enzyme, heme oxygenase-1 (HO-1), in both a human patient and transgenic knockout mice leads to an abundance of circulating heme and damage to vascular endothelium. Although heme can be directly cytotoxic, the present investigations examine the possibility that hemoglobin-derived heme and iron might be indirectly toxic through the generation of oxidized forms of low-density lipoprotein (LDL). In support, hemoglobin in plasma, when oxidized to methemoglobin by oxidants such as leukocyte-derived reactive oxygen, causes oxidative modification of LDL. Heme, released from methemoglobin, catalyzes the oxidation of LDL, which in turn induces endothelial cytolysis primarily caused by lipid hydroperoxides. Exposure of endothelium to sublethal concentrations of this oxidized LDL leads to induction of both HO-1 and ferritin. Similar endothelial cytotoxicity was caused by LDL isolated from plasma of an HO-1-deficient child. Spectral analysis of the child's plasma revealed a substantial oxidation of plasma hemoglobin to methemoglobin. Iron accumulated in the HO-1-deficient child's LDL and several independent assays revealed oxidative modification of the LDL. We conclude that hemoglobin, when oxidized in plasma, can be indirectly cytotoxic through the generation of oxidized LDL by released heme and that, in response, the intracellular defense-HO-1 and ferritin-is induced. These results may be relevant to a variety of disorders-such as renal failure associated with intravascular hemolysis, hemorrhagic injury to the central nervous system, and, perhaps, atherogenesis-in which hemoglobin-derived heme may promote the formation of fatty acid hydroperoxides.
Collapse
|
40
|
Abstract
In humans the iron status is influenced by environmental and genetic factors. Among them, the genetic polymorphism of the hemoglobin (Hb)-binding plasma protein haptoglobin (Hp) has been shown to affect iron turnover. The best known biological function of Hp is capture of free Hb in plasma to allow hepatic recycling of heme iron and to prevent kidney damage during hemolysis. In healthy males, but not in females, the Hp 2-2 phenotype is associated with higher serum iron, higher transferrin saturation, and higher ferritin than Hp 1-1 and 2-1. Moreover, serum ferritin correlates with monocyte L-ferritin content, which is also highest in Hp 2-2 subjects due to endocytosis of multimeric Hb-Hp 2-2 complexes by the recently identified Hb scavenger receptor CD163 in macrophages. This iron delocalization pathway, occurring selectively in Hp 2-2 subjects, has important biological and clinical consequences. The Hp polymorphism is related to the prevalence and the outcome of various pathological conditions with altered iron metabolism such as hemochromatosis, infections, and atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Joris R Delanghe
- Department of Clinical Chemistry, Ghent University Hospital, Belgium.
| | | |
Collapse
|
41
|
Van Vlierberghe H, Langlois M, Delanghe J, Horsmans Y, Michielsen P, Henrion J, Cartuyvels R, Billiet J, De Vos M, Leroux-Roels G. Haptoglobin phenotype 2-2 overrepresentation in Cys282Tyr hemochromatotic patients. J Hepatol 2001; 35:707-11. [PMID: 11738096 DOI: 10.1016/s0168-8278(01)00203-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIMS Patients with genotypic Cys282Tyr homozygous hemochromatosis differ largely in phenotypic presentation. The HFE mutation on itself does not explain the different manifestations of hemochromatosis. We hypothesized that the genetic haptoglobin (Hp) polymorphism, because of its effect on iron metabolism, could be a modifying factor that influences the clinical presentation of hereditary hemochromatosis. METHODS In 167 Cys282Tyr homozygous hemochromatotic patients, the frequencies of Hp types (1-1, 2-1 and 2-2) and alleles (Hp1, Hp2) were compared with those in 918 healthy subjects. Clinical and laboratory indices of iron overload were incorporated in the analysis. RESULTS The Hp 2-2 type was overrepresented in the patient group (P<0.01). Male patients carrying Hp 2-2 had higher serum iron (P=0.003) and ferritin levels (P=0.03) than those with a Hp 1-1 or 2-1 type. The amount of iron removed with phlebotomy was also higher in Hp 2-2 patients (P=0.03). CONCLUSIONS The Hp 2-2 type is overrepresented among Cys282Tyr homozygous hemochromatotic patients. At diagnosis, iron overload was more pronounced in male patients carrying Hp 2-2. Our data suggest that Hp polymorphism affects iron metabolism in hereditary hemochromatosis.
Collapse
Affiliation(s)
- H Van Vlierberghe
- Department of Gastroenterology, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Riess JG. Oxygen carriers ("blood substitutes")--raison d'etre, chemistry, and some physiology. Chem Rev 2001; 101:2797-920. [PMID: 11749396 DOI: 10.1021/cr970143c] [Citation(s) in RCA: 544] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- J G Riess
- MRI Institute, University of California at San Diego, San Diego, CA 92103, USA.
| |
Collapse
|
43
|
Yusa T. Increased extracellular ascorbate release reflects glutamate re-uptake during the early stage of reperfusion after forebrain ischemia in rats. Brain Res 2001; 897:104-13. [PMID: 11282363 DOI: 10.1016/s0006-8993(01)02099-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ascorbate is highly concentrated in neuropils, and its extracellular release is closely related to that of the excitatory neurotransmitters. Thus, the extracellular release of ascorbate and glutamate was measured during the early stage of forebrain ischemia-reperfusion in the rat hippocampus using a microdialysis biosensor system. Male Wistar rats were anesthetized with halothane under mechanical ventilation and normothermia. Two probes of the microdialysis biosensor electrode were inserted in the hippocampus bilaterally. One probe was perfused with phosphate-buffered saline (PBS) and the oxidation signal of dialyzed ascorbate was recorded. A second electropolymerized probe was perfused with PBS containing glutamate oxidase for glutamate measurement. Forebrain ischemia-reperfusion was performed by bilateral carotid artery occlusion with hemorrhagic hypotension (MAP=30 mmHg) for 10 min (Group 10, n=10) or 15 min (Group 15, n=10), followed by reperfusion for 60 min. The release of glutamate increased significantly to 294% (Group 10) and 334% (Group 15) during ischemia, and then decreased rapidly. In Group 15, however, it remained significantly higher after reperfusion than in Group 10. The release of ascorbate increased significantly to 504% (Group 10) and 334% (Group 15) after reperfusion. In Group 10, it was significantly higher for 5-15 min after reperfusion than in Group 15. The marked increase of ascorbate during reperfusion was associated with the rapid decrease in glutamate. The extended time of ischemia significantly inhibited glutamate re-uptake and ascorbate release during reperfusion. These findings suggest the extracellular ascorbate release during reperfusion after global ischemia as a marker of glutamate re-uptake.
Collapse
Affiliation(s)
- T Yusa
- Department of Anesthesiology, University of the Ryukyus, Faculty of Medicine, 207 Uehara, Nishihara-cho, Nakagami-gun, 903-0215, Okinawa, Japan.
| |
Collapse
|
44
|
Langlois MR, Martin ME, Boelaert JR, Beaumont C, Taes YE, De Buyzere ML, Bernard DR, Neels HM, Delanghe JR. The Haptoglobin 2-2 Phenotype Affects Serum Markers of Iron Status in Healthy Males. Clin Chem 2000. [DOI: 10.1093/clinchem/46.10.1619] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AbstractBackground: Human iron status is influenced by environmental and genetic factors. We hypothesized that the genetic polymorphism of haptoglobin (Hp), a hemoglobin-binding plasma protein, could affect iron status.Methods: Reference values of serum iron status markers were compared according to Hp phenotypes (Hp 1-1, Hp 2-1, Hp 2-2; determined by starch gel electrophoresis) in 717 healthy adults. Iron storage was investigated in peripheral blood monocyte-macrophages by measuring cytosolic L- and H-ferritins and by in vitro uptake of radiolabeled (125I) hemoglobin-haptoglobin complexes.Results: In males but not in females, the Hp 2-2 phenotype was associated with higher serum iron (P <0.05), transferrin saturation (P <0.05), and ferritin (P <0.01) concentrations than Hp 1-1 and 2-1, whereas soluble transferrin receptor concentrations were lower (P <0.05). Moreover, serum ferritin correlated with monocyte L-ferritin content (r = 0.699), which was also highest in the male Hp 2-2 subgroup (P <0.01). In vitro, monocyte-macrophages took up a small fraction of 125I-labeled hemoglobin complexed to Hp 2-2 but not to Hp 1-1 or 2-1.Conclusions: The Hp 2-2 phenotype affects serum iron status markers in healthy males and is associated with higher L-ferritin concentrations in monocyte-macrophages because of a yet undescribed iron delocalization pathway, selectively occurring in Hp 2-2 subjects.
Collapse
Affiliation(s)
- Michel R Langlois
- Department of Clinical Chemistry, Immunology and Microbiology, University Hospital Gent, De Pintelaan 185, B-9000 Gent, Belgium
| | - Marie-Elise Martin
- Génétique et Pathologie Moléculaire de l’Hématopoièse, INSERM U409, Faculté Xavier Bichat, 16 rue Henri Huchard, BP416, 75870 Paris Cedex 18, France
| | - Johan R Boelaert
- Unit for Renal and Infectious Diseases and Laboratory of Clinical Chemistry, General Hospital St.-Jan, Ruddershove 10, B-8000 Brugge, Belgium
| | - Carole Beaumont
- Génétique et Pathologie Moléculaire de l’Hématopoièse, INSERM U409, Faculté Xavier Bichat, 16 rue Henri Huchard, BP416, 75870 Paris Cedex 18, France
| | - Youri E Taes
- Department of Clinical Chemistry, Immunology and Microbiology, University Hospital Gent, De Pintelaan 185, B-9000 Gent, Belgium
| | - Marc L De Buyzere
- Department of Clinical Chemistry, Immunology and Microbiology, University Hospital Gent, De Pintelaan 185, B-9000 Gent, Belgium
| | - Dirk R Bernard
- Unit for Renal and Infectious Diseases and Laboratory of Clinical Chemistry, General Hospital St.-Jan, Ruddershove 10, B-8000 Brugge, Belgium
| | - Hugo M Neels
- Laboratory of Clinical Chemistry, General Hospital Middelheim, Lindendreef 1, B-2020 Antwerpen, Belgium
| | - Joris R Delanghe
- Department of Clinical Chemistry, Immunology and Microbiology, University Hospital Gent, De Pintelaan 185, B-9000 Gent, Belgium
| |
Collapse
|
45
|
Fornai F, Saviozzi M, Piaggi S, Gesi M, Corsini GU, Malvaldi G, Casini AF. Localization of a glutathione-dependent dehydroascorbate reductase within the central nervous system of the rat. Neuroscience 1999; 94:937-48. [PMID: 10579586 DOI: 10.1016/s0306-4522(99)00349-8] [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: 11/16/2022]
Abstract
In this study, we describe for the first time the occurrence, within the central nervous system of the rat, of a dehydroascorbate reductase analogous to the one we recently described in the liver. Dehydroascorbate reductase plays a pivotal role in regenerating ascorbic acid from its oxidation product, dehydroascorbate. In a first set of experiments, we showed that a dehydroascorbate reductase activity is present in brain cytosol; immunoblotting analysis confirmed the presence of an immunoreactive cytosolic protein in selected brain areas. Immunotitration showed that approximately 65% of dehydroascorbate reductase activity of brain cytosol which was recovered in the ammonium sulphate fraction can be attributed to this enzyme. Using immunohistochemistry, we found that a variety of brain areas expresses the enzyme. Immunoreactivity was confined to the gray matter. Amongst the several brain regions, the cerebellum appears to be the most densely stained. The enzyme was also abundant in the hippocampus and the olfactory cortex. The lesion of norepinephrine terminals following systemic administration of DSP-4 markedly decreased immunoreactivity in the cerebellum. Apart from the possible co-localization of the enzyme with norepinephrine, the relative content of dehydroascorbate reductase in different brain regions might be crucial in conditioning regional sensitivity to free radical-induced brain damage. Given the scarcity of protective mechanisms demonstrated in the brain, the discovery of a new enzyme with antioxidant properties might represent a starting-point to increase our knowledge about the antioxidant mechanisms operating in several central nervous system disorders.
Collapse
Affiliation(s)
- F Fornai
- Department of Human Morphology and Applied Biology, School of Medicine, University of Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
46
|
Yoo YM, Kim KM, Kim SS, Han JA, Lea HZ, Kim YM. Hemoglobin toxicity in experimental bacterial peritonitis is due to production of reactive oxygen species. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 1999; 6:938-45. [PMID: 10548590 PMCID: PMC95802 DOI: 10.1128/cdli.6.6.938-945.1999] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hemoglobin (Hb) is a toxic molecule responsible for the extreme lethality associated with experimental Escherichia coli peritonitis, but the mechanism has yet to be elucidated. Hb, but not globin, showed toxic effects in a live E. coli model but not in a model using killed E. coli. Methemoglobin, hematin, and the well-known Fenton reagents iron and iron-EDTA demonstrated the same lethal effect in E. coli peritonitis as Hb, while the addition of the Fenton inhibitors desferrioxamine (DF) and diethylenetriamine pentaacetate removed most of the cytotoxic activity of iron. Administration of a combined dose of superoxide dismutase and catalase minimized the action of Hb and iron-EDTA, suggesting that both O(2)(.-) and H(2)O(2) are involved in the toxic action of Hb in this rat model. The combination of the antioxidative enzymes and DF further suppressed iron-mediated lethality. An electron spin resonance technique with the spin-trapping reagent 5, 5-dimethyl-1-pyroline-N-oxide (DMPO) showed O(2)(.-) generation in the peritoneal fluid of rats injected with E. coli alone or E. coli plus iron-DF, and (.)OH generation was detected in the peritoneal fluid of the rats injected with iron-EDTA. Hb did not show any spin adduct of oxygen radicals, suggesting that Hb produces non-spin-trapping radical ferryl ion, which decayed the spin adduct of DMPO. In the presence of Hb or iron-EDTA, O(2)(-)-generating activity and viability of phagocytes decreased, whereas lipid peroxidation of peritoneal phagocytes increased. Generation of oxygen radicals and lipid peroxidation did not differ in the live and dead bacterial models. Bacterial numbers in the peritoneal cavity and blood were markedly increased in the live bacterial model with Hb and iron-EDTA. The Fenton inhibitor iron-DF prevented the loss of phagocyte function, lipid peroxidation, and bacterial proliferation. These results led us to conclude that the lethal toxicity of Hb in bacterial peritonitis is associated with a Fenton-type reaction, the products of which decrease phagocyte viability, through the induction of lipid peroxidation, allowing bacterial proliferation and resulting in mortality.
Collapse
Affiliation(s)
- Y M Yoo
- Departments of Molecular and Cellular Biochemistry, Kangwon National University, Chunchon, Kangwon-do, Korea
| | | | | | | | | | | |
Collapse
|
47
|
Gaté L, Paul J, Ba GN, Tew KD, Tapiero H. Oxidative stress induced in pathologies: the role of antioxidants. Biomed Pharmacother 1999; 53:169-80. [PMID: 10392289 DOI: 10.1016/s0753-3322(99)80086-9] [Citation(s) in RCA: 253] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Exposure to oxidant molecules issued from the environment (pollution, radiation), nutrition, or pathologies can generate reactive oxygen species (ROS for example, H2O2, O2-, OH). These free radicals can alter DNA, proteins and/or membrane phospholipids. Depletion of intracellular antioxidants in acute oxidative stress or in various diseases increases intracellular ROS accumulation. This in turn is responsible for several chronic pathologies including cancer, neurodegenerative or cardiovascular pathologies. Thus, to prevent against cellular damages associated with oxidative stress it is important to balance the ratio of antioxidants to oxidants by supplementation or by cell induction of antioxidants.
Collapse
Affiliation(s)
- L Gaté
- Laboratoire de Pharmacologie Cellulaire et Moleculaire, UMR CNRS 8612, Université de Paris XI, Faculté de Pharmacie, France
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Jourd'heuil D, Mills L, Miles AM, Grisham MB. Effect of nitric oxide on hemoprotein-catalyzed oxidative reactions. Nitric Oxide 1998; 2:37-44. [PMID: 9706741 DOI: 10.1006/niox.1998.0167] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemoglobin or myoglobin-catalyzed oxidation reactions have been suggested to initiate and/or exacerbate tissue injury associated with a variety of pathological conditions including post-ischemic tissue injury, hemorrhagic disorders, and chronic inflammation. In the present study, we investigated what effect different fluxes of nitric oxide (NO) have on hemoprotein-catalyzed oxidation reactions in vitro. The hypoxanthine/xanthine oxidase system was used to generate both O2- and H2O2, whereas the spontaneous decomposition of the spermine/NO adduct was used to generate NO at a known and constant rate. We assessed the ability of myoglobin (Mb) or hemoglobin (Hb) to oxidize dihydrorhodamine (DHR) to rhodamine (RH) in the presence of O2-/H2O2 and/or NO. In the presence of a constant flux of O2- and H2O2 (1 nmol/min each), 500 nM MetMb (Fe3+) stimulated DHR oxidation from normally undetectable levels to approximately 35 microM. This oxidation reaction was inhibited by catalase but not SOD, suggesting the formation of the ferryl-hemoprotein adduct (Fe4+). Equimolar fluxes of O2-, H2O2, and NO increased further DHR oxidation to approximately 50 microM. The 15 microM increase in DHR oxidation was independent of heme concentration and was inhibited by SOD. This suggested that equal fluxes of O2- and NO interact to yield a potent oxidant such as peroxinitrite (OONO-) which together with Mb-Fe4+ oxidizes DHR. Further increases in NO fluxes significantly inhibited DHR oxidation (80%) via the NO-dependent inhibition of Mb-Fe4+ formation. Additional studies using methemoglobin (Hb-Fe3+)-catalyzed oxidative reactions yielded virtually identical results. We conclude that in the presence of a hemoprotein such as myoglobin or hemoglobin, NO may promote or inhibit oxidation reactions depending upon the relative fluxes of O2-, H2O2, and NO.
Collapse
Affiliation(s)
- D Jourd'heuil
- Department of Molecular and Cellular Physiology, Louisiana State University Medical Center, Shreveport 71130, USA.
| | | | | | | |
Collapse
|
50
|
Layton ME, Samson FE, Pazdernik TL. Kainic acid causes redox changes in cerebral cortex extracellular fluid: NMDA receptor activity increases ascorbic acid whereas seizure activity increases uric acid. Neuropharmacology 1998; 37:149-57. [PMID: 9680239 DOI: 10.1016/s0028-3908(98)00002-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Kainic acid (KA) causes seizures and extensive brain damage in rats. To study the effects of KA on the redox state in cerebral cortex extracellular fluid (ECF), ascorbic and uric acid concentrations were measured in intracerebral microdialysis samples before and after systemic KA administration (ip). During seizures, concentrations of ascorbic and uric acid increased 500 and 100%, respectively. When midazolam was given with KA to prevent seizures, ascorbic acid still increased 400%, but uric acid increased only transiently. When the NMDA receptor antagonist aminophosphonovaleric acid (APV) was included in the microdialysis perfusion media, ascorbic acid levels decreased during baseline perfusion in a concentration-dependent manner. APV then suppressed the KA-induced increase in ascorbic acid levels, without blocking seizure activity. In summary, increased uric acid levels in brain ECF activity after KA administration are related to the induced seizure, but ascorbic acid levels are associated with NMDA receptor activity.
Collapse
Affiliation(s)
- M E Layton
- Department of Psychiatry and Behavioral Sciences, University of Washington Medical Center, Seattle, USA
| | | | | |
Collapse
|