1
|
Ashok A, Andrabi SS, Mansoor S, Kuang Y, Kwon BK, Labhasetwar V. Antioxidant Therapy in Oxidative Stress-Induced Neurodegenerative Diseases: Role of Nanoparticle-Based Drug Delivery Systems in Clinical Translation. Antioxidants (Basel) 2022; 11:antiox11020408. [PMID: 35204290 PMCID: PMC8869281 DOI: 10.3390/antiox11020408] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
Free radicals are formed as a part of normal metabolic activities but are neutralized by the endogenous antioxidants present in cells/tissue, thus maintaining the redox balance. This redox balance is disrupted in certain neuropathophysiological conditions, causing oxidative stress, which is implicated in several progressive neurodegenerative diseases. Following neuronal injury, secondary injury progression is also caused by excessive production of free radicals. Highly reactive free radicals, mainly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), damage the cell membrane, proteins, and DNA, which triggers a self-propagating inflammatory cascade of degenerative events. Dysfunctional mitochondria under oxidative stress conditions are considered a key mediator in progressive neurodegeneration. Exogenous delivery of antioxidants holds promise to alleviate oxidative stress to regain the redox balance. In this regard, natural and synthetic antioxidants have been evaluated. Despite promising results in preclinical studies, clinical translation of antioxidants as a therapy to treat neurodegenerative diseases remains elusive. The issues could be their low bioavailability, instability, limited transport to the target tissue, and/or poor antioxidant capacity, requiring repeated and high dosing, which cannot be administered to humans because of dose-limiting toxicity. Our laboratory is investigating nanoparticle-mediated delivery of antioxidant enzymes to address some of the above issues. Apart from being endogenous, the main advantage of antioxidant enzymes is their catalytic mechanism of action; hence, they are significantly more effective at lower doses in detoxifying the deleterious effects of free radicals than nonenzymatic antioxidants. This review provides a comprehensive analysis of the potential of antioxidant therapy, challenges in their clinical translation, and the role nanoparticles/drug delivery systems could play in addressing these challenges.
Collapse
Affiliation(s)
- Anushruti Ashok
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Syed Suhail Andrabi
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Saffar Mansoor
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Youzhi Kuang
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
| | - Brian K. Kwon
- Department of Orthopaedics, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Vinod Labhasetwar
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (A.A.); (S.S.A.); (S.M.); (Y.K.)
- Correspondence:
| |
Collapse
|
2
|
Andrabi SS, Yang J, Gao Y, Kuang Y, Labhasetwar V. Nanoparticles with antioxidant enzymes protect injured spinal cord from neuronal cell apoptosis by attenuating mitochondrial dysfunction. J Control Release 2019; 317:300-311. [PMID: 31805339 DOI: 10.1016/j.jconrel.2019.12.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 02/03/2023]
Abstract
In spinal cord injury (SCI), the initial damage leads to a rapidly escalating cascade of degenerative events, known as secondary injury. Loss of mitochondrial homeostasis after SCI, mediated primarily by oxidative stress, is considered to play a crucial role in the proliferation of secondary injury cascade. We hypothesized that effective exogenous delivery of antioxidant enzymes - superoxide dismutase (SOD) and catalase (CAT), encapsulated in biodegradable nanoparticles (nano-SOD/CAT) - at the lesion site would protect mitochondria from oxidative stress, and hence the spinal cord from secondary injury. Previously, in a rat contusion model of severe SCI, we demonstrated extravasation and retention of intravenously administered nanoparticles specifically at the lesion site. To test our hypothesis, a single dose of nano-SOD/CAT in saline was administered intravenously 6 h post-injury, and the spinal cords were analyzed one week post-treatment. Mitochondria isolated from the affected region of the spinal cord of nano-SOD/CAT-treated animals demonstrated significantly reduced mitochondrial reactive oxygen species (ROS) activities, increased mitochondrial membrane potential, reduced calcium levels, and also higher adenosine triphosphate (ATP) production capacity than those isolated from the spinal cords of untreated control or SOD/CAT solution treated animals. Although the treatment did not achieve the same mitochondrial function as in the spinal cords of sham control animals, it significantly attenuated mitochondrial dysfunction following SCI. Further, immunohistochemical analyses of the spinal cords of treated animals showed significantly lower ROS, cleaved caspase-3, and cytochrome c activities, leading to reduced spinal cord neuronal cell apoptosis and smaller lesion area than in untreated animals. These results imply that the treatment significantly attenuated progression of secondary injury that was also reflected from less weight loss and improved locomotive recovery of treated vs. untreated animals. In conclusion, nano-SOD/CAT mitigated activation of cascade of degenerating factors by protecting mitochondria and hence the spinal cord from secondary injury. An effective treatment during the acute phase following SCI could potentially have a positive long-term impact on neurological and functional recovery.
Collapse
Affiliation(s)
- Syed Suhail Andrabi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jun Yang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yue Gao
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Youzhi Kuang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
3
|
Binevski PV, Balabushevich NG, Uvarova VI, Vikulina AS, Volodkin D. Bio-friendly encapsulation of superoxide dismutase into vaterite CaCO3 crystals. Enzyme activity, release mechanism, and perspectives for ophthalmology. Colloids Surf B Biointerfaces 2019; 181:437-449. [DOI: 10.1016/j.colsurfb.2019.05.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022]
|
4
|
Prophylactic Zinc and Therapeutic Selenium Administration Increases the Antioxidant Enzyme Activity in the Rat Temporoparietal Cortex and Improves Memory after a Transient Hypoxia-Ischemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9416432. [PMID: 30258527 PMCID: PMC6146673 DOI: 10.1155/2018/9416432] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/31/2018] [Indexed: 11/17/2022]
Abstract
In the cerebral hypoxia-ischemia rat model, the prophylactic administration of zinc can cause either cytotoxicity or preconditioning effect, whereas the therapeutic administration of selenium decreases the ischemic damage. Herein, we aimed to explore whether supplementation of low doses of prophylactic zinc and therapeutic selenium could protect from a transient hypoxic-ischemic event. We administrated zinc (0.2 mg/kg of body weight; ip) daily for 14 days before a 10 min common carotid artery occlusion (CCAO). After CCAO, we administrated sodium selenite (6 μg/kg of body weight; ip) daily for 7 days. In the temporoparietal cerebral cortex, we determined nitrites by the Griess method and lipid peroxidation by the Gerard-Monnier assay. qPCR was used to measure mRNA of nitric oxide synthases, antioxidant enzymes, chemokines, and their receptors. We measured the enzymatic activity of SOD and GPx and protein levels of chemokines and their receptors by ELISA. We evaluated long-term memory using the Morris-Water maze test. Our results showed that prophylactic administration of zinc caused a preconditioning effect, decreasing nitrosative/oxidative stress and increasing GPx and SOD expression and activity, as well as eNOS expression. The therapeutic administration of selenium maintained this preconditioning effect up to the late phase of hypoxia-ischemia. Ccl2, Ccr2, Cxcl12, and Cxcr4 were upregulated, and long-term memory was improved. Pyknotic cells were decreased suggesting prevention of neuronal cell death. Our results show that the prophylactic zinc and therapeutic selenium administration induces effective neuroprotection in the early and late phases after CCAO.
Collapse
|
5
|
Poellmann MJ, Bu J, Hong S. Would antioxidant-loaded nanoparticles present an effective treatment for ischemic stroke? Nanomedicine (Lond) 2018; 13:2327-2340. [DOI: 10.2217/nnm-2018-0084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide and is in urgent need of new treatment options. The only approved treatment for stroke restores blood flow to the brain, but much of the tissue damage occurs during the subsequent reperfusion. Antioxidant therapies that directly address ischemia-reperfusion injury have shown promise in preclinical results. In this review, we discuss that reformulating antioxidant therapies as nanomedicine can potentially overcome the barriers that have kept these therapies from succeeding in the clinic. We begin by reviewing the pathophysiology of ischemic stroke with a focus on the effects of reperfusion injury. Next, we review nanotherapeutic systems designed to treat the disease with a focus on those addressing reperfusion injury. Mechanisms of passive and active transport required to traverse a blood–brain barrier are discussed. Finally, we conclude by outlining design parameters for potentially successful nanomedicines as front-line therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine & Public Health, University of Wisconsin, Madison, WI 53792, USA
- Yonsei Frontier Lab & Department of Pharmacy, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
6
|
Gong P, Zhang Z, Zou C, Tian Q, Chen X, Hong M, Liu X, Chen Q, Xu Z, Li M, Wang J. Hippo/YAP signaling pathway mitigates blood-brain barrier disruption after cerebral ischemia/reperfusion injury. Behav Brain Res 2018; 356:8-17. [PMID: 30092249 DOI: 10.1016/j.bbr.2018.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/23/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022]
Abstract
Ischemia/reperfusion (I/R) injuries commonly lead to breakdown of the blood-brain barrier (BBB). Restoration of the BBB can relieve neurologic damage caused by I/R injuries. The Hippo/YAP signaling pathway mediates cell proliferation, regulated cell death, and differentiation in various organisms and has been shown to participate in the restoration of the heart after I/R. In this study, we investigated whether the Hippo/YAP pathway plays a role in I/R injury in brain, especially in regard to I/R-induced BBB breakdown. The results of our study indicate that I/R injury led to an overall decrease in activity of the core proteins, YAP and TAZ, over a 24-h period. The most dramatic change was observed 1.5 h after reperfusion. In rats that underwent 1.5 h of reperfusion, intraperitoneal injection of YAP agonist dexamethasone activated YAP and TAZ and led to improved neurologic function, smaller brain infarct sizes, increased levels of tight junction proteins, decreased BBB permeability, decreased cerebral edema, and less apoptosis. Our results suggest that YAP exerts neuroprotective effects on the damaged brain that are likely related to restoration of the BBB.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhan Zhang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Changlin Zou
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, PR China
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xi Liu
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Watanabe Y, Matsuba T, Nakanishi M, Une M, Hanajima R, Nakashima K. Tetanus toxin fragments and Bcl-2 fusion proteins: cytoprotection and retrograde axonal migration. BMC Biotechnol 2018; 18:39. [PMID: 29890980 PMCID: PMC5996528 DOI: 10.1186/s12896-018-0452-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 06/06/2018] [Indexed: 12/14/2022] Open
Abstract
Background Tetanus neurotoxin (TeNT) is taken up at nerve terminals and undergoes retrograde migration. The toxic properties of TeNT reside in the toxin light chain (L), but like complete TeNT, the TeNT heavy chain (TTH) and the C-terminal domain (TTC) alone can bind and enter into neurons. Here, we explored whether atoxic fragments of TeNT could act as drug delivery vehicles in neurons. In this study, we used Bcl-2, a protein known to have anti-apoptotic properties in vivo and in vitro, as a parcel to couple to TeNT fragments. Results We expressed Bcl-2 and the TTC fragments alone, and also attempted to express fusion proteins with the Bcl-2 coupled at the N-terminus of TTH (Bcl2-TTH) and the N- and C-terminus of TTC (TTC-Bcl2 and Bcl2-TTC) in mammalian (Cos7 cells) and Escherichia coli systems. TTC and Bcl-2 were efficiently expressed in E. coli and Cos7 cells, respectively, but Bcl-2 and the fusion proteins did not express well in E. coli. The fusion proteins were also not expressed in Cos7 cells. To improve the yield and purity of the fusion protein, we genetically deleted the N-terminal half of TTC from the Bcl2-TTC fusion to yield Bcl2-hTTC. Purified Bcl2-hTTC exhibited neuronal binding and prevented cell death of neuronal PC12 cells induced by serum and NGF deprivation, as evidenced by the inhibition of cytochrome C release from the mitochondria. For in vivo assays, Bcl2-hTTC was injected into the tongues of mice and was seen to selectively migrate to hypoglossal nuclei mouse brain stems via retrograde axonal transport. Conclusions These results indicate that Bcl2-hTTC retains both Bcl-2 and TTC functions and therefore could be a potent therapeutic agent for various neurological conditions.
Collapse
Affiliation(s)
- Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, 683-8504, Japan.
| | - Takashi Matsuba
- Division of Bacteriology, Department of Microbiology and immunology, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, 683-8503, Japan
| | - Mami Nakanishi
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, 683-8504, Japan
| | - Mio Une
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, 683-8504, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, 683-8504, Japan
| | - Kenji Nakashima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, 683-8504, Japan
| |
Collapse
|
8
|
L L, X W, Z Y. Ischemia-reperfusion Injury in the Brain: Mechanisms and Potential Therapeutic Strategies. ACTA ACUST UNITED AC 2016; 5. [PMID: 29888120 DOI: 10.4172/2167-0501.1000213] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ischemia-reperfusion injury is a common feature of ischemic stroke, which occurs when blood supply is restored after a period of ischemia. Reperfusion can be achieved either by thrombolysis using thrombolytic reagents such as tissue plasminogen activator (tPA), or through mechanical removal of thrombi. Spontaneous reperfusion also occurs after ischemic stroke. However, despite the beneficial effect of restored oxygen supply by reperfusion, it also causes deleterious effect compared with permanent ischemia. With the recent advancement in endovascular therapy including thrombectomy and thrombus disruption, reperfusion-injury has become an increasingly critical challenge in stroke treatment. It is therefore of extreme importance to understand the mechanisms of ischemia-reperfusion injury in the brain in order to develop effective therapeutics. Accumulating experimental evidence have suggested that the mechanisms of ischemia-reperfusion injury include oxidative stress, leukocyte infiltration, platelet adhesion and aggregation, complement activation, mitochondrial mediated mechanisms, and blood-brain-barrier (BBB) disruption, which altogether ultimately lead to edema or hemorrhagic transformation (HT) in the brain. Potential therapeutic strategies against ischemia-reperfusion injury may be developed targeting these mechanisms. In this review, we briefly discuss the pathophysiology and cellular and molecular mechanisms of cerebral ischemia-reperfusion injury, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Lin L
- Institute of Molecular Pharmacology, Wenzhou Medical University, Wenzhou 325035, PR China.,Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wang X
- Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yu Z
- Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Jiang Y, Arounleut P, Rheiner S, Bae Y, Kabanov AV, Milligan C, Manickam DS. SOD1 nanozyme with reduced toxicity and MPS accumulation. J Control Release 2016; 231:38-49. [DOI: 10.1016/j.jconrel.2016.02.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 01/15/2023]
|
10
|
Petro M, Jaffer H, Yang J, Kabu S, Morris VB, Labhasetwar V. Tissue plasminogen activator followed by antioxidant-loaded nanoparticle delivery promotes activation/mobilization of progenitor cells in infarcted rat brain. Biomaterials 2015; 81:169-180. [PMID: 26735970 DOI: 10.1016/j.biomaterials.2015.12.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 12/07/2015] [Accepted: 12/13/2015] [Indexed: 12/19/2022]
Abstract
Inherent neuronal and circulating progenitor cells play important roles in facilitating neuronal and functional recovery post stroke. However, this endogenous repair process is rather limited, primarily due to unfavorable conditions in the infarcted brain involving reactive oxygen species (ROS)-mediated oxidative stress and inflammation following ischemia/reperfusion injury. We hypothesized that during reperfusion, effective delivery of antioxidants to ischemic brain would create an environment without such oxidative stress and inflammation, thus promoting activation and mobilization of progenitor cells in the infarcted brain. We administered recombinant human tissue-type plasminogen activator (tPA) via carotid artery at 3 h post stroke in a thromboembolic rat model, followed by sequential administration of the antioxidants catalase (CAT) and superoxide dismutase (SOD), encapsulated in biodegradable nanoparticles (nano-CAT/SOD). Brains were harvested at 48 h post stroke for immunohistochemical analysis. Ipsilateral brain slices from animals that had received tPA + nano-CAT/SOD showed a widespread distribution of glial fibrillary acidic protein-positive cells (with morphology resembling radial glia-like neural precursor cells) and nestin-positive cells (indicating the presence of immature neurons); such cells were considerably fewer in untreated animals or those treated with tPA alone. Brain sections from animals receiving tPA + nano-CAT/SOD also showed much greater numbers of SOX2- and nestin-positive progenitor cells migrating from subventricular zone of the lateral ventricle and entering the rostral migratory stream than in t-PA alone treated group or untreated control. Further, animals treated with tPA + nano-CAT/SOD showed far fewer caspase-positive cells and fewer neutrophils than did other groups, as well as an inhibition of hippocampal swelling. These results suggest that the antioxidants mitigated the inflammatory response, protected neuronal cells from undergoing apoptosis, and inhibited edema formation by protecting the blood-brain barrier from ROS-mediated reperfusion injury. A longer-term study would enable us to determine if our approach would assist progenitor cells to undergo neurogenesis and to facilitate neurological and functional recovery following stroke and reperfusion injury.
Collapse
Affiliation(s)
- Marianne Petro
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hayder Jaffer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jun Yang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shushi Kabu
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Viola B Morris
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
11
|
Jiang Y, Brynskikh AM, S-Manickam D, Kabanov AV. SOD1 nanozyme salvages ischemic brain by locally protecting cerebral vasculature. J Control Release 2015; 213:36-44. [PMID: 26093094 PMCID: PMC4684498 DOI: 10.1016/j.jconrel.2015.06.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/15/2015] [Indexed: 11/30/2022]
Abstract
Copper/zinc superoxide dismutase (CuZnSOD; SOD1) is widely considered as a potential therapeutic candidate for pathologies involving oxidative stress, but its application has been greatly hindered by delivery issues. In our previous study, nanoformulated SOD1 (cl-nanozyme) was shown to decrease infarct volume and improve sensorimotor functions after a single intravenous (IV) injection in a rat middle cerebral artery occlusion (MCAO) model of ischemia/reperfusion (I/R) injury (stroke). However, it remained unclear how cl-nanozyme was able to deliver SOD1 to the brain and exert therapeutic efficacy. The present study aims to answer this question by exploring micro-distribution pattern of cl-nanozyme in the rat brain after stroke. Immunohistochemistry studies demonstrated cl-nanozyme co-localization with fibrin along damaged arteries and capillaries in the ischemic hemisphere. We further found that cl-nanozyme can be cross-linked into thrombi formed after I/R injury in the brain, and this effect is independent of animal species (rat/mouse) used for modeling I/R injury. This work is also the first report reinforcing therapeutic potential of cl-nanozyme in a well-characterized mouse MCAO model of I/R injury.
Collapse
Affiliation(s)
- Yuhang Jiang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna M Brynskikh
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Devika S-Manickam
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Alexander V Kabanov
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Laboratory for Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 117234, Russia.
| |
Collapse
|
12
|
Kabu S, Gao Y, Kwon BK, Labhasetwar V. Drug delivery, cell-based therapies, and tissue engineering approaches for spinal cord injury. J Control Release 2015; 219:141-154. [PMID: 26343846 DOI: 10.1016/j.jconrel.2015.08.060] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/23/2015] [Accepted: 08/31/2015] [Indexed: 12/28/2022]
Abstract
Spinal cord injury (SCI) results in devastating neurological and pathological consequences, causing major dysfunction to the motor, sensory, and autonomic systems. The primary traumatic injury to the spinal cord triggers a cascade of acute and chronic degenerative events, leading to further secondary injury. Many therapeutic strategies have been developed to potentially intervene in these progressive neurodegenerative events and minimize secondary damage to the spinal cord. Additionally, significant efforts have been directed toward regenerative therapies that may facilitate neuronal repair and establish connectivity across the injury site. Despite the promise that these approaches have shown in preclinical animal models of SCI, challenges with respect to successful clinical translation still remain. The factors that could have contributed to failure include important biologic and physiologic differences between the preclinical models and the human condition, study designs that do not mirror clinical reality, discrepancies in dosing and the timing of therapeutic interventions, and dose-limiting toxicity. With a better understanding of the pathobiology of events following acute SCI, developing integrated approaches aimed at preventing secondary damage and also facilitating neuroregenerative recovery is possible and hopefully will lead to effective treatments for this devastating injury. The focus of this review is to highlight the progress that has been made in drug therapies and delivery systems, and also cell-based and tissue engineering approaches for SCI.
Collapse
Affiliation(s)
- Shushi Kabu
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yue Gao
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Brian K Kwon
- Department of Orthopaedics, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada V5Z 1M9
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
13
|
Wang B, Tian S, Wang J, Han F, Zhao L, Wang R, Ning W, Chen W, Qu Y. Intraperitoneal administration of thioredoxin decreases brain damage from ischemic stroke. Brain Res 2015; 1615:89-97. [DOI: 10.1016/j.brainres.2015.04.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/15/2015] [Accepted: 04/17/2015] [Indexed: 11/26/2022]
|
14
|
Abstract
Stroke is a leading cause of death, long-term disability, and socioeconomic costs, highlighting the urgent need for more effective treatments. Intravenous administration of tissue plasminogen activator (t-PA) is the only FDA-approved therapy to re-establish cerebral blood flow. However, because of increased risk of hemorrhage beyond 3 h post stroke, few stroke patients (1-2%) benefit from t-PA; t-PA, which has neurotoxic effects, can also aggravate the extent of reperfusion injury by increasing blood-brain barrier permeability. An alternative strategy is needed to extend the window of intervention, minimize damage from reperfusion injury, and promote brain repair leading to neurological recovery. Reactive oxygen species (ROS), generated soon after ischemia and during reperfusion and thereafter, are considered the main mediators of ischemic injury. Antioxidant enzymes such as catalase, superoxide dismutase, etc. can neutralize ROS-mediated injury but their effective delivery to the brain remains a challenge. In this article, we review various therapeutic approaches including surgical interventions, and discuss the potential of nanoparticle-mediated delivery of antioxidants for stroke therapy.
Collapse
Affiliation(s)
- Hayder Jaffer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | | | | | | |
Collapse
|
15
|
Ovsepian SV, Ovespian SV, Bodeker M, O'Leary VB, Lawrence GW, Oliver Dolly J. Internalization and retrograde axonal trafficking of tetanus toxin in motor neurons and trans-synaptic propagation at central synapses exceed those of its C-terminal-binding fragments. Brain Struct Funct 2015; 220:1825-38. [PMID: 25665801 DOI: 10.1007/s00429-015-1004-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/04/2015] [Indexed: 12/22/2022]
Abstract
The prominent tropism of tetanus toxin (TeTx) towards peripheral nerves with retrograde transport and transfer to central neurons render it an invaluable probe for exploring fundamental neuronal processes such as endocytosis, retrograde trafficking and trans-synaptic transport to central neurons. While the specificity of TeTx to nerve cells has been attributed to its binding domains (HC and HCC), molecular determinants of the long-range trafficking that ensure its central delivery and induction of spastic paralysis remain elusive. Here, we report that a protease-inactive TeTx mutant (TeTIM) fused to core streptavidin (CS) proved superior to CS-HC and CS-HCC fragments in antagonizing the internalization of the active toxin in cultured spinal cord neurons. Also, in comparison to CS-HC and CS-HCC, CS-TeTIM undergoes faster clearance from motor nerve terminals after peripheral injection, and is detected in a greater number of neurons in the spinal cord and brain stem ipsi-lateral to the administration site. Consistent with trans-synaptic transfer from motor neurons to inter-neurons, CS-TeTIM infiltrated non-cholinergic cells in the spinal cord; in contrast, the retrograde spread of CS-HC was largely restricted to neurons stained for choline acetyltransferase. Peripheral injection of CS-TeTIM conjugated to a lentivirus encoding mutated SNAP-25, resistant to cleavage by botulinum neurotoxin A, E and C1, rendered spontaneous excitatory postsynaptic currents in motor neurons resilient to challenge by type A toxin in vitro, whereas the same virus conjugated to CS-HC proved ineffective. These findings indicate that full-length inactive TeTx greatly exceeds HC and HCC in targeting and invading motor nerve terminals at the periphery and exploits more efficiently the retrograde transport and trans-synaptic transfer mechanisms of motor neurons to arrive at central neurons. Such qualities render TeTIM a more suitable research probe and neuron-targeting vehicle for retro-axonal delivery of viral vectors to the CNS.
Collapse
Affiliation(s)
| | - Saak V Ovespian
- International Centre for Neurotherapeutics, Dublin City University, Dublin 9, Ireland,
| | | | | | | | | |
Collapse
|
16
|
Blood-brain barrier breakdown involves four distinct stages of vascular damage in various models of experimental focal cerebral ischemia. J Cereb Blood Flow Metab 2015; 35:292-303. [PMID: 25425076 PMCID: PMC4426746 DOI: 10.1038/jcbfm.2014.199] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022]
Abstract
Ischemic stroke not only impairs neuronal function but also affects the cerebral vasculature as indicated by loss of blood-brain barrier (BBB) integrity. Therefore, therapeutical recanalization includes an enhanced risk for hemorrhagic transformation and bleeding, traditionally attributed to a 'reperfusion injury'. To investigate the mechanisms underlying ischemia-/reperfusion-related BBB opening, we applied multiple immunofluorescence labeling and electron microscopy in a rat model of thromboembolic stroke as well as mouse models of permanent and transient focal cerebral ischemia. In these models, areas exhibiting BBB breakdown were identified by extravasation of intravenously administered fluorescein isothiocyanate (FITC)-albumin. After 24 hours, expression of markers for tight and adherens junctions in areas of FITC-albumin leakage consistently remained unaltered in the applied models. However, lectin staining with isolectin B4 indicated structural alterations in the endothelium, which were confirmed by electron microscopy. While ultrastructural alterations in endothelial cells did not differ between the applied models including the reperfusion scenario, we regularly identified vascular alterations, which we propose to reflect four distinct stages of BBB breakdown with ultimate loss of endothelial cells. Therefore, our data strongly suggest that ischemia-related BBB failure is predominantly caused by endothelial degeneration. Thus, protecting endothelial cells may represent a promising therapeutical approach in addition to the established recanalizing strategies.
Collapse
|
17
|
Singhal A, Morris VB, Labhasetwar V, Ghorpade A. Nanoparticle-mediated catalase delivery protects human neurons from oxidative stress. Cell Death Dis 2013; 4:e903. [PMID: 24201802 PMCID: PMC3847304 DOI: 10.1038/cddis.2013.362] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/13/2013] [Accepted: 08/14/2013] [Indexed: 12/28/2022]
Abstract
Several neurodegenerative diseases and brain injury involve reactive oxygen species and implicate oxidative stress in disease mechanisms. Hydrogen peroxide (H2O2) formation due to mitochondrial superoxide leakage perpetuates oxidative stress in neuronal injury. Catalase, an H2O2-degrading enzyme, thus remains an important antioxidant therapy target. However, catalase therapy is restricted by its labile nature and inadequate delivery. Here, a nanotechnology approach was evaluated using catalase-loaded, poly(lactic co-glycolic acid) nanoparticles (NPs) in human neuronal protection against oxidative damage. This study showed highly efficient catalase encapsulation capable of retaining∼99% enzymatic activity. NPs released catalase rapidly, and antioxidant activity was sustained for over a month. NP uptake in human neurons was rapid and nontoxic. Although human neurons were highly sensitive to H2O2, NP-mediated catalase delivery successfully protected cultured neurons from H2O2-induced oxidative stress. Catalase-loaded NPs significantly reduced H2O2-induced protein oxidation, DNA damage, mitochondrial membrane transition pore opening and loss of cell membrane integrity and restored neuronal morphology, neurite network and microtubule-associated protein-2 levels. Further, catalase-loaded NPs improved neuronal recovery from H2O2 pre-exposure better than free catalase, suggesting possible applications in ameliorating stroke-relevant oxidative stress. Brain targeting of catalase-loaded NPs may find wide therapeutic applications for oxidative stress-associated acute and chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- A Singhal
- Department of Cell Biology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | | | | |
Collapse
|
18
|
O'Leary VB, Ovsepian SV, Bodeker M, Dolly JO. Improved lentiviral transduction of ALS motoneurons in vivo via dual targeting. Mol Pharm 2013; 10:4195-206. [PMID: 24066863 DOI: 10.1021/mp400247t] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Treatment of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease, is hampered by its complex etiology and lack of efficient means for targeted transfer of therapeutics into motoneurons. The objective of this research was engineering of a versatile motoneuron targeting adapter--a full-length atoxic tetanus toxin fused to core-streptavidin (CS-TeTIM)--for retro-axonal transduction of viral vectors; validation of the targeting efficiency of CS-TeTIM in vivo, by expression of green fluorescence protein (GFP) reporter in motoneurons of presymptomatic and symptomatic ALS-like SOD1(G93A) mice, and comparison with age-matched controls; and appraisal of lentiviral transduction with CS-TeTIM relative to (1) a HC binding fragment of tetanus toxin CS-TeTx(HC), (2) rabies glycoprotein (RG), and (3) a CS-TeTIM-RG dual targeting approach. CS-TeTIM and CS-TeTx(HC) were engineered using recombinant technology and site-directed mutagenesis. Biotinylated vectors, pseudotyped with vesicular stomatitis virus glycoprotein (VSV-G) or RG, were linked to these adaptors and injected intraperitoneally (ip) into presymptomatic (12 weeks old), symptomatic SOD1(G93A) (22 weeks old) or wild type control mice, followed by monitoring of GFP expression in the spinal cord and supraspinal motor structures with quantitative PCR and immuno-histochemistry. Transcripts were detected in the spinal cord and supraspinal motor structures of all mice 2 weeks after receiving a single ip injection, although in symptomatic SOD1(G93A) animals reporter RNA levels were lower compared to presymptomatic and wild-type controls irrespective of the targeting approach. GFP transduction with CS-TeTIM proved more efficient than CS-TeTx(HC) across all groups while CS-TeTIM-RG dual-targeted vectors yielded the highest transcript numbers. Importantly, in both wild-type and presymptomatic SOD1(G93A) mice strong colabeling of choline-acetyltransferase (ChAT) and GFP was visualized in neurons of the brain stem and spinal cord. CS-TeTIM, a versatile adaptor protein for targeted lentiviral transduction of motoneurons, has been engineered and its competence assessed relative to CS-TeTx(HC) and RG. Evidence has been provided that highlights the potential usefulness of this novel recombinant tool for basic research with implications for improved transfer of therapeutic candidates into motoneurons for the amelioration of ALS and related diseases.
Collapse
Affiliation(s)
- Valerie B O'Leary
- International Centre for Neurotherapeutics, Dublin City University , Dublin 9, Ireland
| | | | | | | |
Collapse
|
19
|
Liu J, Hou J, Xia ZY, Zeng W, Wang X, Li R, Ke C, Xu J, Lei S, Xia Z. Recombinant PTD-Cu/Zn SOD attenuates hypoxia-reoxygenation injury in cardiomyocytes. Free Radic Res 2013; 47:386-93. [PMID: 23445361 DOI: 10.3109/10715762.2013.780286] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Oxidative stress plays a pivotal role in myocardial ischemia-reperfusion injury. Increasing the protein expression of intracellular Cu/Zn SOD, which is the major endogenous antioxidant enzyme, may attenuate or prevent hypoxia-reoxygenation injury (HRI) in cultured cardiomyocytes. However, ectogenic Cu/Zn-SOD can hardly be transferred into cells to exert biological effects. In this study, we constructed PTD-Cu/Zn SOD plasmid with a kind of translocation structure-Protein transduction domain (PTD) and detected its transmembrane ability and antioxidant effects in H9c2 rat cardiomyocytes subjected to hypoxia/reoxygenation injury (HRI). METHODS We constructed the pET-PTD-Cu/Zn SOD (CDs) prokaryotic expression vectors in plasmid that were inserted into E. coli BL21 to induce the protein expression of PTD-Cu/Zn SOD. H9c2 cardiomyocyte HRI was achieved by exposing cardiomyocytes to 12 h hypoxia followed by 2 h reoxygenation. Protein expression of PTD-Cu/Zn SOD in cardiomyocytes was assayed by Western blot and their enzyme activities were investigated by immunohistochemistry and flow cytometry. RESULTS In cultured cardiomyocytes hypoxia-reoxygenation injury model, exogenous PTD-Cu/Zn SOD could penetrate cell membrane to clear superoxide anion and decrease hydrogen peroxide level in H9c2 cardiomyocytes subjected to HRI. The level of mitochondrial membrane potential was restored to normal, and the cell apoptosis was reduced in cardiomyocytes with PTD-Cu/Zn SOD treatment during HRI. CONCLUSION Recombinant PTD-Cu/Zn SOD could scavenge intracellular-free superoxide anion, protect mitochondria from damages, and attenuate the hypoxia-reoxygenation injury in cultured cardiomyocytes.
Collapse
Affiliation(s)
- J Liu
- Department of Anesthesiology, Remin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nanoparticles for targeted delivery of antioxidant enzymes to the brain after cerebral ischemia and reperfusion injury. J Cereb Blood Flow Metab 2013; 33:583-92. [PMID: 23385198 PMCID: PMC3618396 DOI: 10.1038/jcbfm.2012.209] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Stroke is one of the major causes of death and disability in the United States. After cerebral ischemia and reperfusion injury, the generation of reactive oxygen species (ROS) and reactive nitrogen species may contribute to the disease process through alterations in the structure of DNA, RNA, proteins, and lipids. We generated various nanoparticles (liposomes, polybutylcyanoacrylate (PBCA), or poly(lactide-co-glycolide) (PLGA)) that contained active superoxide dismutase (SOD) enzyme (4,000 to 20,000 U/kg) in the mouse model of cerebral ischemia and reperfusion injury to determine the impact of these molecules. In addition, the nanoparticles were untagged or tagged with nonselective antibodies or antibodies directed against the N-methyl-D-aspartate (NMDA) receptor 1. The nanoparticles containing SOD protected primary neurons in vitro from oxygen-glucose deprivation (OGD) and limited the extent of apoptosis. The nanoparticles showed protection against ischemia and reperfusion injury when applied after injury with a 50% to 60% reduction in infarct volume, reduced inflammatory markers, and improved behavior in vivo. The targeted nanoparticles not only showed enhanced protection but also showed localization to the CA regions of the hippocampus. Nanoparticles alone were not effective in reducing infarct volume. These studies show that targeted nanoparticles containing protective factors may be viable candidates for the treatment of stroke.
Collapse
|
21
|
Manickam DS, Brynskikh AM, Kopanic JL, Sorgen PL, Klyachko NL, Batrakova EV, Bronich TK, Kabanov AV. Well-defined cross-linked antioxidant nanozymes for treatment of ischemic brain injury. J Control Release 2012; 162:636-45. [PMID: 22902590 DOI: 10.1016/j.jconrel.2012.07.044] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 06/09/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
Abstract
Development of well-defined nanomedicines is critical for their successful clinical translation. A simple synthesis and purification procedure is established for chemically cross-linked polyion complexes of Cu/Zn superoxide dismutase (SOD1) or catalase with a cationic block copolymer, methoxy-poly(ethylene glycol)-block-poly(L-lysine hydrochloride) (PEG-pLL₅₀). Such complexes, termed cross-linked nanozymes (cl-nanozymes) retain catalytic activity and have narrow size distribution. Moreover, their cytotoxicity is decreased compared to non-cross-linked complexes due to suppression of release of the free block copolymer. SOD1 cl-nanozymes exhibit prolonged ability to scavenge experimentally induced reactive oxygen species (ROS) in cultured brain microvessel endothelial cells and central neurons. In vivo they decrease ischemia/reperfusion-induced tissue injury and improve sensorimotor functions in a rat middle cerebral artery occlusion (MCAO) model after a single intravenous (i.v.) injection. Altogether, well-defined cl-nanozymes are promising modalities for attenuation of oxidative stress after brain injury.
Collapse
Affiliation(s)
- Devika S Manickam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center-UNMC, Omaha, NE 68198, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Han CG, Han JK, Park KB, Kwak KH, Park SS, Lim DG. Effect of superoxide on the development and maintenance of mechanical allodynia in a rat model of chronic post-ischemia pain. Korean J Anesthesiol 2012; 63:149-56. [PMID: 22949983 PMCID: PMC3427808 DOI: 10.4097/kjae.2012.63.2.149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 07/05/2012] [Accepted: 07/12/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Reactive oxygen species and inflammatory responses contribute to the development of neuropathic pain. Superoxide serves to mediate cell signaling processes and tissue injury during inflammation. We examined the effects of superoxide on the development and maintenance of mechanical allodynia, as well as its contribution to central sensitization in a superoxide-rich animal model of neuropathic pain. METHODS Chronic post-ischemia pain (CPIP) was induced via the left hindpaw ischemia for 3 h, followed by reperfusion. Superoxide dismutase (4,000 U/kg, i.p.) was administered either 5 min before ischemia (BI), 5 min before reperfusion (BR), or 3 days after reperfusion (3AR). Withdrawal thresholds of the four paws were measured to assess the mechanical allodynia and the effects of circulating xanthine oxidase (XO)-mediated superoxide production. In addition, we measured the levels of N-methyl D-aspartate receptor subunit 1 phosphorylation (p-NR1) in the ipsilateral and contralateral spinal cord (L4-6), by Western blotting, to examine the superoxide-mediated central sensitization. Superoxide production was assessed by allopurinol-sensitive, XO-mediated lipid peroxidation of the spinal cord and gastrocnemius muscles. RESULTS Withdrawal thresholds of forepaws did not vary across the 7 days of testing. In the hindpaws, both ipsilateral and contralateral mechanical allodynia was most attenuated in the BR group, followed by the BI and 3AR groups. The degree of NR1 activation was in contrast to the changes in the withdrawal thresholds. CONCLUSIONS These data suggest that superoxide is involved in the development and maintenance of mechanical allodynia, particularly via central sensitization in the spinal cord.
Collapse
Affiliation(s)
- Chang Gyu Han
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jae Kyung Han
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ki Bum Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Kyung Hwa Kwak
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Sung Sik Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Dong Gun Lim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
23
|
Zhao ST, Huang XT, Zhang C, Ke Y. Humanin protects cortical neurons from ischemia and reperfusion injury by the increased activity of superoxide dismutase. Neurochem Res 2011; 37:153-60. [PMID: 21935731 DOI: 10.1007/s11064-011-0593-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/17/2011] [Accepted: 08/29/2011] [Indexed: 11/28/2022]
Abstract
The neuroprotective effects of superoxide dismutase (SOD) against hypoxia/reperfusion (I/R) injury and of humanin (HN) against toxicity by familial amyotrophic lateral sclerosis (ALS)-related mutant SOD led us to hypothesize that HN might have a role to increase the activity of SOD, which might be involved in the protective effects of HN on neuron against Alzheimer's disease-unrelated neurotoxicities. In the present study, we found that 4 h ischemia and 24 h reperfusion induced a significant increase in lactate dehydrogenase (LDH) release, malondialdehyde (MDA) formation and the number of karyopyknotic nuclei (4',6-diamidino-2-phenylindole dihydrochloride nuclear dyeing) and a decrease in the number of Calcein-AM-positive living cells and cell viability. Pretreatment of the cells with HN led to a significant decrease in LDH release, MDA formation and the number of karyopyknotic nuclei, and an increase in the number of Calcein-AM-positive living cells and cell viability in neurons treated with I/R. We also found a significant decrease in SOD activity in neurons treated with I/R only, while pre-treatment with HN before I/R induced a significant increase in the activity of SOD as compared with the I/R group. Our findings implied that HN protects cortical neurons from I/R injury by the increased SOD activity and that the protective effect of HN on neurons against I/R is concentration-dependent.
Collapse
Affiliation(s)
- Shen-Ting Zhao
- Department of Physiology, Guangzhou Medical School, Guangzhou 510182, People's Republic of China
| | | | | | | |
Collapse
|
24
|
Wang C, Li S, Shang DJ, Wang XL, You ZL, Li HB. Antihyperglycemic and neuroprotective effects of one novel Cu-Zn SOD mimetic. Bioorg Med Chem Lett 2011; 21:4320-4. [PMID: 21669524 DOI: 10.1016/j.bmcl.2011.05.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 05/05/2011] [Accepted: 05/17/2011] [Indexed: 11/24/2022]
Abstract
Increasing evidence supports that OS plays important roles in diabetes mellitus and cerebral ischemia. This suggests that recovering an impaired endogenous superoxide dismutase (SOD) enzyme system induced by OS with a mimetic would be beneficial and protective for these diseases. In present study, one nonpeptidyl small molecular weight compound (D34) was synthesized. Its SOD mimetic activity and the potential therapeutic actions were also evaluated both in vivo and in vitro. The in vitro nitro blue tetrazolium (NBT) assay indicated that D34 presents an SOD mimetic activity. D34 (20μmol/kg) exhibited significant antihyperglycemic activity in alloxan-diabetic mice. D34 could also ameliorate the cerebral neuronal death in hippocampus of global cerebral ischemia mice. Furthermore, the D34 treatment significantly decreased malondialdehyde (MDA) contents and increased SOD activities in brains or livers of diabetes mice or cerebral ischemic mice. In conclusion, these preliminary findings support that D34 exhibits SOD mimetic activity and possesses significant antihyperglycemic and neuroprotective effects.
Collapse
Affiliation(s)
- Che Wang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | | | | | | | | | | |
Collapse
|
25
|
Iranzo O. Manganese complexes displaying superoxide dismutase activity: A balance between different factors. Bioorg Chem 2011; 39:73-87. [DOI: 10.1016/j.bioorg.2011.02.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/14/2011] [Accepted: 02/14/2011] [Indexed: 01/05/2023]
|
26
|
Doeppner TR, Hermann DM. Free radical scavengers and spin traps – therapeutic implications for ischemic stroke. Best Pract Res Clin Anaesthesiol 2010; 24:511-20. [DOI: 10.1016/j.bpa.2010.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 10/11/2010] [Indexed: 01/03/2023]
|
27
|
Imai N, Okabe T. Kinetics differ between copper-zinc and manganese superoxide dismutase activity with acute ischemic stroke. J Stroke Cerebrovasc Dis 2010; 20:75-78. [PMID: 20598578 DOI: 10.1016/j.jstrokecerebrovasdis.2009.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 11/10/2009] [Accepted: 11/15/2009] [Indexed: 11/19/2022] Open
Abstract
This study aimed to clarify the kinetics of copper-zinc (CuZn) and manganese (Mn) superoxide dismutase (SOD) activity in acute ischemic stroke victims. Using the nitrite method, we investigated sequential changes in CuZn and Mn SOD activity in the cerebrospinal fluid (CSF) of 8 patients with acute ischemic stroke. SOD activity in each patient was measured at 36 hours and 3, 7, 14, and 28 days after stroke. CuZn SOD activity in CSF peaked 3 days after stroke, with values gradually decreasing after 7 days. In contrast, Mn SOD activity remained significantly lower in the stroke group than in controls throughout the study. These findings may reflect differences between the 2 isoenzymes in terms of the distribution, role, and method of synthesis in brain tissue.
Collapse
Affiliation(s)
- Noboru Imai
- Department of Neurology, Shizuoka Red Cross Hospital, Shizuoka, Japan.
| | - Takashi Okabe
- Department of Neurology, Shizuoka Red Cross Hospital, Shizuoka, Japan
| |
Collapse
|
28
|
Ferrer EG, Baeza N, Naso LG, Castellano EE, Piro OE, Williams PAM. Superoxidedismutase-mimetic copper(II) complexes containing saccharinate and 4-aminopyridine/4-cyanopyridine. J Trace Elem Med Biol 2010; 24:20-6. [PMID: 20122575 DOI: 10.1016/j.jtemb.2009.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 09/23/2009] [Accepted: 09/28/2009] [Indexed: 11/17/2022]
Abstract
Two copper(II) complexes, [Cu(sac)(2)(4-cypy)(2)(H(2)O)], 1 and [Cu(sac)(2)(4-Ampy)(2)(H(2)O)], 2 (4-cypy: 4-cyanopyridine; 4-Ampy: 4-aminopyridine) were prepared. Physicochemical properties of the complexes were studied by spectroscopic (solution UV-vis, diffuse reflectance and IR) techniques. Structural X-ray diffraction data could be obtained only for [Cu(sac)(2)(4-cypy)(2)(H(2)O)] that it crystallized in the tetragonal space group P4cc with a=b=15.313(1), c=13.240(1)A, and Z=4 molecules per unit cell. The complex was cited on a crystallographic C(2)-axis with the Cu(II) ion in a square-pyramidal environment, coordinated at the pyramid basis to the nitrogen atom of two saccharine anions [d(Cu-N)=2.011(3)A] and the pyridine N-atom of two 4-cyanopyridine ligands [d(Cu-N)=2.038(4)A]. The coordination was completed by a water molecule at the pyramid apex [d(Cu-Ow)=2.189(5)A]. Elemental and spectroscopic analyses revealed an O-saccharinate coordination mode for complex 2 and a square-pyramidal structure. Only complex 2 retained its structure in methanolic solution. However, both complexes were able to catalyze the dismutation of superoxide anion (O(2)(-)) (pH 7.5) at micromolar concentrations. Therefore, these complexes behaved as useful SOD-mimetic compounds.
Collapse
Affiliation(s)
- Evelina G Ferrer
- Centro de Química Inorgánica (CEQUINOR/CONICET, UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, C. Correo 962, 1900 La Plata, Argentina
| | | | | | | | | | | |
Collapse
|
29
|
Valdivia A, Pérez-Álvarez S, Aroca-Aguilar JD, Ikuta I, Jordán J. Superoxide dismutases: a physiopharmacological update. J Physiol Biochem 2009; 65:195-208. [DOI: 10.1007/bf03179070] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Kwak KH, Han CG, Lee SH, Jeon Y, Park SS, Kim SO, Baek WY, Hong JG, Lim DG. Reactive oxygen species in rats with chronic post-ischemia pain. Acta Anaesthesiol Scand 2009; 53:648-56. [PMID: 19419360 DOI: 10.1111/j.1399-6576.2009.01937.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND An emerging theme in the study of the pathophysiology of persistent pain is the role of reactive oxygen species (ROS). In the present study, we examined the hypothesis that the exogenous supply of antioxidant drugs during peri-reperfusion would attenuate pain induced by ischemia/reperfusion (IR) injury. We investigated the analgesic effects of three antioxidants administered during peri-reperfusion using an animal model of complex regional pain syndrome-type I consisting of chronic post-ischemia pain (CPIP) of the hind paw. METHODS Application of a tight-fitting tourniquet for a period of 3 h produced CPIP in male Sprague-Dawley rats. Low-dose allopurinol (4 mg/kg), high-dose allopurinol (40 mg/kg), superoxide dismutase (SOD, 4000 U/kg), N-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg), or SOD (4000 U/kg)+L-NAME (10 mg/kg) was administered intraperitoneally just after tourniquet application and at 1 and 2 days after reperfusion for 3 days. The effects of antioxidants in rats were investigated using mechanical and cold stimuli. Each group consisted of seven rats. RESULTS Allopurinol caused significant alleviation in mechanical and cold allodynia for a period of 4 weeks in rats with CPIP. Both SOD and L-NAME, which were used to investigate the roles of superoxide (O2(-)) and nitric oxide (NO) in pain, also attenuated neuropathic-like pain symptoms in rats for 4 weeks. CONCLUSIONS Our findings suggest that O2(-) and NO mediate IR injury-induced chronic pain, and that ROS scavengers administered during the peri-reperfusion period have long-term analgesic effects.
Collapse
Affiliation(s)
- K H Kwak
- Department of Anesthesiology and Pain medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Qi J, Li Y, Zhang H, Cheng Y, Sun Y, Cao J, Zhao Y, Wang F. A novel conjugate of low-molecular-weight heparin and Cu,Zn-superoxide dismutase: Study on its mechanism in preventing brain reperfusion injury after ischemia in gerbils. Brain Res 2009; 1260:76-83. [DOI: 10.1016/j.brainres.2008.12.079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 12/31/2008] [Accepted: 12/31/2008] [Indexed: 10/21/2022]
|
32
|
Reddy MK, Labhasetwar V. Nanoparticle-mediated delivery of superoxide dismutase to the brain: an effective strategy to reduce ischemia-reperfusion injury. FASEB J 2009; 23:1384-95. [PMID: 19124559 DOI: 10.1096/fj.08-116947] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Excessive production of reactive oxygen species (ROS) after cerebral ischemia and reperfusion is implicated in brain damage through different cellular and molecular mechanisms, and it is further aggravated by impaired cellular antioxidant defense systems under ischemic conditions. Therapeutic strategies based on exogenous delivery of the native form of superoxide dismutase (SOD), a free radical scavenger, are limited because of its short half-life (approximately 6 min) in vivo and poor permeability across the blood-brain-barrier (BBB). We encapsulated SOD in biodegradable poly(D,L-lactide co-glycolide) nanoparticles (SOD-NPs) and tested their efficacy in a rat focal cerebral ischemia-reperfusion injury model. We hypothesized that localized brain delivery of SOD-NPs would sustain the protective effect of SOD by neutralizing the deleterious effects of ROS formed following ischemia-reperfusion. SOD-NPs were administered at the time of reperfusion via the intracarotid route to maximize their localization in the brain. Animals receiving SOD-NPs (10,000 U of SOD/kg) demonstrated a 65% reduction in infarct volume, whereas an equivalent dose of SOD in solution (SOD-Sol) increased it by 25% over saline control (P<0.001; data at 6 h following reperfusion). Control NPs alone or mixed with SOD-Sol were ineffective in reducing infract volume, with results similar to saline control, indicating the protective effect of the encapsulated enzyme. SOD-NPs maintained BBB integrity, thereby preventing edema, reduced the level of ROS formed following reperfusion, and protected neurons from undergoing apoptosis. Animals treated with SOD-NPs demonstrated greater survival than those with saline control (75% vs. 0% at 28 days) and later regained most vital neurological functions. SOD-NPs may be an effective treatment option in conjunction with a thrombolytic agent for stroke patients.
Collapse
Affiliation(s)
- Maram K Reddy
- Department of Biomedical Engineering/ND-20, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
33
|
Reddy MK, Wu L, Kou W, Ghorpade A, Labhasetwar V. Superoxide dismutase-loaded PLGA nanoparticles protect cultured human neurons under oxidative stress. Appl Biochem Biotechnol 2008; 151:565-77. [PMID: 18509606 PMCID: PMC2670226 DOI: 10.1007/s12010-008-8232-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2007] [Accepted: 04/09/2008] [Indexed: 02/07/2023]
Abstract
The objective of our study was to investigate the neuroprotective efficacy of superoxide dismutase (SOD), loaded in poly(D,L-lactide co-glycolide; PLGA) nanoparticles (NPs), in cultured human neurons challenged with hydrogen peroxide (H(2)O(2))-induced oxidative stress. We hypothesized that the protected and sustained intracellular delivery of SOD encapsulated in NPs would demonstrate better neuroprotection from oxidative stress than either SOD or pegylated SOD (PEG-SOD) in solution. SOD-NPs (approximately 81 +/- 4 nm in diameter, 0.9% w/w SOD loading) released the encapsulated SOD in an active form with 8.2% cumulative release during the first 24 h, followed by a slower release thereafter. The results demonstrated that PLGA-NPs are compatible with human neurons, and the neuroprotective effect of SOD-NPs is dose-dependent, with efficacy seen at >100 U SOD, and less significant effects at lower doses. Neither SOD (25-200 U) nor PEG-SOD (100 U) in solution demonstrated the neuroprotective effect under similar conditions. The neuroprotective effect of SOD-NPs was seen up to 6 h after H(2)O(2)-induced oxidative stress, but the effect diminished thereafter. Confocal microscopic studies demonstrated better intracellular neuronal uptake of the encapsulated model protein (fluorescein isothiocyanate-labeled BSA) than the protein in solution. Thus, the mechanism of efficacy of SOD-NPs appears to be due to the stability of the encapsulated enzyme and its better neuronal uptake after encapsulation.
Collapse
Affiliation(s)
- Maram K. Reddy
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Li Wu
- Department of Pharmacology and Experimental Neuroscience and Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198–5215
| | - Wei Kou
- Department of Pharmacology and Experimental Neuroscience and Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198–5215
| | - Anuja Ghorpade
- Department of Pharmacology and Experimental Neuroscience and Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198–5215
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198–6025
| |
Collapse
|
34
|
Hafeez F, Razzaq MA, Levine RL, Ramirez MAN. Reperfusion Seizures: A Manifestation of Cerebral Reperfusion Injury After Administration of Recombinant Tissue Plasminogen Activator for Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2007; 16:273-7. [DOI: 10.1016/j.jstrokecerebrovasdis.2007.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 07/19/2007] [Accepted: 07/26/2007] [Indexed: 11/24/2022] Open
|
35
|
Hussein G, Sankawa U, Goto H, Matsumoto K, Watanabe H. Astaxanthin, a carotenoid with potential in human health and nutrition. JOURNAL OF NATURAL PRODUCTS 2006; 69:443-9. [PMID: 16562856 DOI: 10.1021/np050354+] [Citation(s) in RCA: 377] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Astaxanthin (1), a red-orange carotenoid pigment, is a powerful biological antioxidant that occurs naturally in a wide variety of living organisms. The potent antioxidant property of 1 has been implicated in its various biological activities demonstrated in both experimental animals and clinical studies. Compound 1 has considerable potential and promising applications in human health and nutrition. In this review, the recent scientific literature (from 2002 to 2005) is covered on the most significant activities of 1, including its antioxidative and anti-inflammatory properties, its effects on cancer, diabetes, the immune system, and ocular health, and other related aspects. We also discuss the green microalga Haematococcus pluvialis, the richest source of natural 1, and its utilization in the promotion of human health, including the antihypertensive and neuroprotective potentials of 1, emphasizing our experimental data on the effects of dietary astaxanthin on blood pressure, stroke, and vascular dementia in animal models, is described.
Collapse
Affiliation(s)
- Ghazi Hussein
- International Research Center for Traditional Medicine, Toyama Prefecture, Japan.
| | | | | | | | | |
Collapse
|
36
|
Benn SC, Ay I, Bastia E, Chian RJ, Celia SA, Pepinsky RB, Fishman PS, Brown RH, Francis JW. Tetanus toxin fragment C fusion facilitates protein delivery to CNS neurons from cerebrospinal fluid in mice. J Neurochem 2005; 95:1118-31. [PMID: 16271047 DOI: 10.1111/j.1471-4159.2005.03459.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To improve protein delivery to the CNS following intracerebroventricular administration, we compared the distribution of a human Cu/Zn superoxide dismutase:tetanus toxin fragment C fusion protein (SOD1:TTC) in mouse brain and spinal cord with that of tetanus toxin fragment C (TTC) or human SOD1 (hSOD1) alone, following continuous infusion into the lateral ventricle. Mice infused with TTC or SOD1:TTC showed intense anti-TTC or anti-hSOD1 labeling, respectively, throughout the CNS. In contrast, animals treated with hSOD1 revealed moderate staining in periventricular tissues. In spinal cord sections from animals infused with SOD1:TTC, the fusion protein was found in neuron nuclear antigen-positive (NeuN+) neurons and not glial fibrillary acidic protein-positive (GFAP+) astrocytes. The percentage of NeuN+ ventral horn cells that were co-labeled with hSOD1 antibody was greater in mice treated with SOD1:TTC (cervical cord = 73 +/- 8.5%; lumbar cord = 62 +/- 7.7%) than in mice treated with hSOD1 alone (cervical cord = 15 +/- 3.9%; lumbar cord = 27 +/-4.7%). Enzyme-linked immunosorbent assay for hSOD1 further demonstrated that SOD1:TTC-infused mice had higher levels of immunoreactive hSOD1 in CNS tissue extracts than hSOD1-infused mice. Following 24 h of drug washout, tissue extracts from SOD1:TTC-treated mice still contained substantial amounts of hSOD1, while extracts from hSOD1-treated mice lacked detectable hSOD1. Immunoprecipitation of SOD1:TTC from these extracts using anti-TTC antibody revealed that the recovered fusion protein was structurally intact and enzymatically active. These results indicate that TTC may serve as a useful prototype for development as a non-viral vehicle for improving delivery of therapeutic proteins to the CNS.
Collapse
Affiliation(s)
- Susanna C Benn
- Cecil B. Day Laboratory for Neuromuscular Research, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Margaill I, Plotkine M, Lerouet D. Antioxidant strategies in the treatment of stroke. Free Radic Biol Med 2005; 39:429-43. [PMID: 16043015 DOI: 10.1016/j.freeradbiomed.2005.05.003] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 04/19/2005] [Accepted: 05/04/2005] [Indexed: 11/22/2022]
Abstract
Excessive production of free radicals is known to lead to cell injury in a variety of diseases, such as cerebral ischemia. In this review, we describe some of the numerous studies that have examined this oxidative stress and the efficiency of antioxidant strategies in focal cerebral ischemia. Besides using genetically modified mice, these strategies can be divided into three groups: (1) inhibition of free radical production, (2) scavenging of free radicals, and (3) increase of free radical degradation by using agents mimicking the enzymatic activity of endogenous antioxidants. Finally, the clinical trials that have tested or are currently testing the efficiency of antioxidants in patients suffering from stroke are reviewed. The results presented here lead us to consider that antioxidants are very promising drugs for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Isabelle Margaill
- Faculté des Sciences Pharmaceutiques et Biologiques, Laboratoire de Pharmacologie (UPRES EA 2510), Université René Descartes, 4 avenue de l'Observatoire, 75006 Paris, France.
| | | | | |
Collapse
|
38
|
Mahakunakorn P, Tohda M, Murakami Y, Watanabe H, Matsumoto K. Effects of Choto-san and its related constituents on endogenous antioxidant systems. Biol Pharm Bull 2005; 28:53-7. [PMID: 15635163 DOI: 10.1248/bpb.28.53] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that Choto-san acts as an antioxidant and cytoprotective agents against H2O2-induced oxidative damage in NG108-15 cells, and the effect is due at least partly to the phenolic compounds. To further investigate the detail mechanisms of this cytoprotection effects of Choto-san and related compounds on enzyme activities of antioxidant systems were examined. Choto-san (5-100 microg/ml) and Chotoko (5-100 mg/ml) stimulated the activity of superoxide dismutase (SOD), catalase and glutathione peroxidase (GPX). These also increased the level of glutathione. Although Choto-san without Chotoko (w/o CKO) did not show the effects on SOD and catalase, GPX activity and glutathion content also, but weakly, stimulated by w/o CKO. The effects of phenolic compounds, epicatechin, caffeic acid and quercetin were also investigated. Epicatechin stimulated catalase, GPX and glutathion content, but not SOD. On the other hand, caffeic acid stimulated SOD activity but had no effects on others. Quercetin stimulated all, although intensities were different among. These results suggest that simultaneous induction of cellular antioxidant defense systems by Choto-sam and its related constituents may be an important mechanisms underlying the protective effects of Choto-san on ischemia-induced neuronal cells injury, and the characteristics of the stimulative effects of phenolic compounds were depend on enzymes.
Collapse
Affiliation(s)
- Pramote Mahakunakorn
- Division of Medicinal Pharmacology, Institute of Natural Medicine, Toyama Medical and Pharmaceutical University, Japan
| | | | | | | | | |
Collapse
|
39
|
Dousset E, Carrega L, Steinberg JG, Clot-Faybesse O, Jouirou B, Sauze N, Devaux C, Autier Y, Jammes Y, Martin-Eauclaire MF, Guieu R. Evidence that free radical generation occurs during scorpion envenomation. Comp Biochem Physiol C Toxicol Pharmacol 2005; 140:221-6. [PMID: 15907767 DOI: 10.1016/j.cca.2005.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 02/07/2005] [Accepted: 02/07/2005] [Indexed: 10/25/2022]
Abstract
Although it is well established that symptomatology, morbidity and death following scorpion envenomation are due to increases in neurotransmitter release secondary to toxins binding to voltage-sensitive sodium channels, the mechanism by which venom action is involved in damaging heart, liver, lungs and kidneys remains unclear. We hypothesized that scorpion toxins could induce the generation of high levels of free radicals responsible for membrane damage in organs targeted by venom action. We have investigated lipid peroxidation in different organs, through the evaluation of thiobarbituric acid reactive substances (TBARS), after experimental envenomation of rats by toxic fractions of Androctonus australis Hector venom. We have shown that scorpion toxins cause considerable lipid peroxidation in most vital organs. We also evaluated the protective effects of antioxidants in mice injected with lethal doses of toxins. Among the drugs tested, N-acetylcysteine (NAC) was effective in protecting the mice when injected prior to toxin application. However, the free radical scavenging properties of NAC seem less implicated in these protective effects than its ability to increase the fluidity of bronchial secretions. We therefore conclude that free radical generation only plays a minor role in the toxicity of scorpion venom.
Collapse
Affiliation(s)
- E Dousset
- Laboratoire des Déterminants Physiologiques de l'Activité Physique (UPRES EA 3285), Institut Fédératif de Recherche E-J Marey (IFR 107), Faculté des Sciences du Sport, Université de la Méditerranée (Aix-Marseille II), Marseille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sheldon RA, Jiang X, Francisco C, Christen S, Vexler ZS, Täuber MG, Ferriero DM. Manipulation of antioxidant pathways in neonatal murine brain. Pediatr Res 2004; 56:656-62. [PMID: 15295091 DOI: 10.1203/01.pdr.0000139413.27864.50] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
To assess the role of brain antioxidant capacity in the pathogenesis of neonatal hypoxic-ischemic brain injury, we measured the activity of glutathione peroxidase (GPX) in both human-superoxide dismutase-1 (hSOD1) and human-GPX1 overexpressing transgenic (Tg) mice after neonatal hypoxia-ischemia (HI). We have previously shown that mice that overexpress the hSOD1 gene are more injured than their wild-type (WT) littermates after HI, and that H(2)O(2) accumulates in HI hSOD1-Tg hippocampus. We hypothesized that lower GPX activity is responsible for the accumulation of H(2)O(2). Therefore, increasing the activity of this enzyme through gene manipulation should be protective. We show that brains of hGPX1-Tg mice, in contrast to those of hSOD-Tg, have less injury after HI than WT littermates: hGPX1-Tg, median injury score = 8 (range, 0-24) versus WT, median injury score = 17 (range, 2-24), p < 0.01. GPX activity in hSOD1-Tg mice, 2 h and 24 h after HI, showed a delayed and bilateral decline in the cortex 24 h after HI (36.0 +/- 1.2 U/mg in naive hSOD1-Tg versus 29.1 +/- 1.7 U/mg in HI cortex and 29.2 +/- 2.0 for hypoxic cortex, p < 0.006). On the other hand, GPX activity in hGPX1-Tg after HI showed a significant increase by 24 h in the cortex ipsilateral to the injury (48.5 +/- 5.2 U/mg, compared with 37.2 +/- 1.5 U/mg in naive hGPX1-Tg cortex, p < 0.008). These findings support the hypothesis that the immature brain has limited GPX activity and is more susceptible to oxidative damage and may explain the paradoxical effect seen in ischemic neonatal brain when SOD1 is overexpressed.
Collapse
Affiliation(s)
- R Ann Sheldon
- Department of Neurology, University of California San Francisco, San Francisco, California 94143.
| | | | | | | | | | | | | |
Collapse
|
41
|
Siu FKW, Lo SCL, Leung MCP. Electroacupuncture reduces the extent of lipid peroxidation by increasing superoxide dismutase and glutathione peroxidase activities in ischemic-reperfused rat brains. Neurosci Lett 2004; 354:158-62. [PMID: 14698462 DOI: 10.1016/j.neulet.2003.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Reactive oxygen species can be scavenged by superoxide dismutase (SOD) and glutathione peroxidase (GPx). During ischemia-reperfusion, the normal functioning of these antioxidant enzymes may be insufficient for the prevention of oxidant-induced peroxidation of membrane lipids and hence cerebral infarction. We therefore investigated whether electroacupuncture (EA) treatment at Fengchi points in post-ischemic rats could increase the antioxidant enzyme activities and thereby reduce the extent of lipid peroxidation. The results indicated that while EA did not alter the antioxidant enzyme activities in non-ischemic normal rat brains, ischemia-reperfusion caused significant increases in SOD and GPx activities. EA treatment further increased the antioxidant enzyme activities in ischemic-reperfused brain tissues, with a concomitant decrease in the extent of lipid peroxidation. Our finding suggests that EA treatment at Fengchi reduced the extent of lipid peroxidation in ischemic-reperfused rat brains, possibly by increasing the activities of SOD and GPx.
Collapse
Affiliation(s)
- Flora K W Siu
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China.
| | | | | |
Collapse
|
42
|
Hattori I, Takagi Y, Nakamura H, Nozaki K, Bai J, Kondo N, Sugino T, Nishimura M, Hashimoto N, Yodoi J. Intravenous administration of thioredoxin decreases brain damage following transient focal cerebral ischemia in mice. Antioxid Redox Signal 2004; 6:81-7. [PMID: 14713338 DOI: 10.1089/152308604771978372] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Thioredoxin (TRX) is induced by a variety of oxidative stimuli and shows cytoprotective roles against oxidative stress. To clarify the possibility of clinical application, we examined the effects of intravenously administered TRX in a model of transient focal cerebral ischemia in this study. Mature male C57BL/6j mice received either continuous intravenous infusion of recombinant human TRX (rhTRX) over a range of 1-10 mg/kg, bovine serum albumin, or vehicle alone for 2 h after 90-min transient middle cerebral artery occlusion (MCAO). Twenty-four hours after the transient MCAO, the animals were evaluated neurologically and the infarct volumes were assessed. Infarct volume, neurological deficit, and protein carbonyl contents, a marker of protein oxidation, in the brain were significantly ameliorated in rhTRX-treated mice at the dose of 3 and 10 mg/kg versus these parameters in control animals. Moreover, activation of p38 mitogen-activated protein kinase, whose pathway is involved in ischemic neuronal death, was suppressed in the rhTRX-treated mice. Further, rhTRX was detected in the ischemic hemisphere by western blot analysis, suggesting that rhTRX was able to permeate the blood-brain barrier in the ischemic hemisphere. These data indicate that exogenous TRX exerts distinct cytoprotective effects on cerebral ischemia/reperfusion injury in mice by means of its redox-regulating activity.
Collapse
|
43
|
Solås AB, Kalous P, Davis JM, Saugstad OD. Effects of recombinant human superoxide dismutase during reoxygenation with 21% or 100% oxygen after cerebral asphyxia in newborn piglets. J Matern Fetal Neonatal Med 2004; 14:96-101. [PMID: 14629089 DOI: 10.1080/jmf.14.2.96.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Superoxide radicals (O2-) are generated during reoxygenation following asphyxia, possibly more when higher concentrations of O2 are used during resuscitation. Superoxide dismutase (SOD) is an antioxidant enzyme, which scavenges O2-. We tested the hypothesis that a single intravenous dose of recombinant human Cu,Zn SOD (rhSOD) could influence microcirculation and biochemical markers of asphyxia in piglets reoxygenated with 21% or 100% O2 after combined cerebral hypoxemia-ischemia-hypercapnia. METHODS Anesthetized newborn piglets were randomized to asphyxia (n = 40) or control (n = 3). Asphyxia was induced by ventilation with 8% O2, adding CO2, and temporary occlusion of both common carotid arteries. After 20 min, 16 piglets received rhSOD 5 mg/kg intravenously and reoxygenation with 21% O2 (rhSOD, 21%; n = 8) or 100% O2 (rhSOD, 100%; n = 8), and 24 piglets received saline and reoxygenation with 21% O2 (21%, n = 13) or 100% O2 (100%, n = 11). The cortical microcirculation was assessed by laser Doppler flowmetry, and glutamate in the striatum and hypoxanthine in the cortex were measured by in vivo microdialysis. RESULTS AND CONCLUSION rhSOD peaked in plasma after 5 min. No rhSOD was detected in brain tissue. There were no significant differences between rhSOD and non-rhSOD groups in any measured variable.
Collapse
Affiliation(s)
- A B Solås
- Department of Pediatric Research, The National Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
44
|
Mollace V, Iannone M, Muscoli C, Palma E, Granato T, Modesti A, Nisticò R, Rotiroti D, Salvemini D. The protective effect of M40401, a superoxide dismutase mimetic, on post-ischemic brain damage in Mongolian gerbils. BMC Pharmacol 2003; 3:8. [PMID: 12809567 PMCID: PMC165580 DOI: 10.1186/1471-2210-3-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2003] [Accepted: 06/16/2003] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Overproduction of free radical species has been shown to occur in brain tissues after ischemia-reperfusion injury. However, most of free radical scavengers known to antagonize oxidative damage (e.g. superoxide dismutase, catalase), are unable to protect against ischemia-reperfusion brain injury when given in vivo, an effect mainly due to their difficulty to gain access to brain tissues. Here we studied the effect of a low molecular weight superoxide dismutase mimetic (M40401) in brain damage subsequent to ischemia-reperfusion injury in Mongolian gerbils. RESULTS In animals undergoing ischemia-reperfusion injury, neuropathological and ultrastructural changes were monitored for 1-7 days either in the presence or in the absence of M40401 after bilateral common carotid artery occlusion (BCCO). Administration of M40401 (1-40 mg/kg, given i.p. 1 h after BCCO) protected against post-ischemic, ultrastructural and neuropathological changes occurring within the hippocampal CA1 area. The protective effect of M40401 was associated with a significant reduction of the levels of malondialdehyde (MDA; a marker of lipid peroxidation) in ischemic brain tissues after ischemia-reperfusion. CONCLUSION Taken together, these results demonstrate that M40401 provides protective effects when given early after the induction of ischemia-reperfusion of brain tissues and suggest the possible use of such compounds in the treatment of neurological dysfunction subsequent to cerebral flow disturbances.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Faculty of Pharmacy, University of Catanzaro "Magna Graecia", Roccelletta di Borgia, Catanzaro Italy
| | - Michelangelo Iannone
- Institute of Neurological Science ISN – Section of Pharmacology, CNR, Roccelletta di Borgia, Catanzaro, Italy
| | - Carolina Muscoli
- Faculty of Pharmacy, University of Catanzaro "Magna Graecia", Roccelletta di Borgia, Catanzaro Italy
- Metaphore Pharmaceuticals, 1910 Innerbelt Business Center Dr, St Louis MO 63114, USA
| | - Ernesto Palma
- Faculty of Pharmacy, University of Catanzaro "Magna Graecia", Roccelletta di Borgia, Catanzaro Italy
| | - Teresa Granato
- Institute of Neurological Science ISN – Section of Pharmacology, CNR, Roccelletta di Borgia, Catanzaro, Italy
| | - Andrea Modesti
- Department of Experimental Medicine and Biochemical Science, University of Rome "Tor Vergata", Rome 00161, Italy
| | - Robert Nisticò
- Faculty of Pharmacy, University of Calabria – Arcavacata di Rende (CS), Italy
| | - Domenicantonio Rotiroti
- Faculty of Pharmacy, University of Catanzaro "Magna Graecia", Roccelletta di Borgia, Catanzaro Italy
| | - Daniela Salvemini
- Metaphore Pharmaceuticals, 1910 Innerbelt Business Center Dr, St Louis MO 63114, USA
| |
Collapse
|
45
|
Shimizu K, Rajapakse N, Horiguchi T, Payne RM, Busija DW. Protective effect of a new nonpeptidyl mimetic of SOD, M40401, against focal cerebral ischemia in the rat. Brain Res 2003; 963:8-14. [PMID: 12560107 DOI: 10.1016/s0006-8993(02)03796-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We tested the neuroprotective effects of M40401, a new, low molecular weight (511.4 Da) maganese superoxide dismutase mimetic, against 90 min of middle cerebral artery occlusion (MCAO) in male Wistar rats. Animals received a single injection of vehicle (n=8), 1 mg/kg (n=6), or 3 mg/kg (n=7) 30 min before MCAO. Total lesion volume was reduced only in the group receiving 3 mg/kg M40401 (163.5+/-18.7 versus 43.4+/-7.0 mm(3), for vehicle and M40401, respectively; P<0.05), with almost complete reduction of lesion volume in the cortex but little protection in the basal ganglia. Neurological score was also improved in this group. The dose of 1 mg/kg M40401 had smaller and inconsistent effects on lesion parameters. Administration of a single dose of 3 mg/kg M40401 at 60 min of MCAO or at the end of MCAO (90 min) failed to significantly reduce lesion volume. A single dose of M40401 plus prolonged infusion into the post-MCAO period also failed to decrease lesion volume significantly. These data indicate that M40401 protects cerebral tissue from ischemic insult when administered before MCAO, probably by limiting damage mediated by detrimental actions of superoxide anion.
Collapse
Affiliation(s)
- Katsuyoshi Shimizu
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1083, USA.
| | | | | | | | | |
Collapse
|
46
|
Ostrowski RP. Effect of coenzyme Q(10) on biochemical and morphological changes in experimental ischemia in the rat brain. Brain Res Bull 2000; 53:399-407. [PMID: 11136995 DOI: 10.1016/s0361-9230(00)00406-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The aim of the work was to evaluate an influence of CoQ(10) on lactate acidosis, adenosine-5'-triphosphate (ATP) concentrations, oxidized to reduced glutathione ratio and on superoxide dismutase activity in endothelin model of cerebral ischemia in the rat. Light microscopic studies in the central nervous system and morphometric analysis of pyramidal cells in the hippocampus were also performed. Endothelins (ET-1 or ET-3; 20 pmoles) were injected into the right lateral cerebral ventricle (intracerebroventricularly). CoQ(10) was given intraperitoneally (i.p.) just before the operation (i.p. 10 mgkg b. wt.). More severe changes of investigated biochemical parameters were observed in the animals treated with ET-1 in comparison with ET-3. Recovery was noted earlier in the group subjected to ET-3 and CoQ(10) administration, than in the animals subjected to ET-1 and CoQ(10) treatment. Histopathological observations showed sparse foci of a neuronal loss in the cerebral cortex and in the hippocampus only in the ET-1 model of ischemia. Additionally more numerous dark neurons were present in above brain structures following ET-1 administration comparing with ET-3 one. Morphometrical studies demonstrated that CoQ(10) diminished neuronal injury in the hippocampal CA1, CA2 and CA3 zones. Above data indicate on neuroprotective effect of CoQ(10) as a potent antioxidant and oxygen derived free radicals scavenger in the cerebral ischemia.
Collapse
Affiliation(s)
- R P Ostrowski
- Laboratory of Clinical Neurochemistry, Department of Neuropathology, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
47
|
Matthews CC, Figueiredo DM, Wollack JB, Fairweather NF, Dougan G, Hallewell RA, Cadet JL, Fishman PS. Protective effect of supplemental superoxide dismutase on survival of neuronal cells during starvation. Requirement for cytosolic distribution. J Mol Neurosci 2000; 14:155-66. [PMID: 10984191 DOI: 10.1385/jmn:14:3:155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2000] [Accepted: 02/20/2000] [Indexed: 01/22/2023]
Abstract
There is evidence that raising cellular levels of Cu2+/Zn2+ superoxide dismutase (SOD1) can protect neurons from oxidative injury. We compared a novel method of elevating neuronal SOD activity using a recombinant hybrid protein composed of the atoxic neuronal binding domain of tetanus toxin (C fragment or TTC) and human SOD1 (hSOD1) with increasing cellular SOD levels through overexpression. Fetal murine cortical neurons or N18-RE-105 cells were incubated with the TTC-hSOD1 hybrid protein and compared to cells constitutively expressing hSOD1 for level of SOD activity, cellular localization of hSOD1, and capacity to survive glucose and pyruvate starvation. Cells incubated with TTC-hSOD1 showed a threefold increase in cellular SOD activity over control cells. This level of increase was comparable to fetal cortical neurons from transgenic mice constitutively expressing hSOD1 and transfected N18-RE-105 cells expressing a green fluorescent protein-hSOD1 fusion protein (GFP-hSOD1). Human SOD1 was distributed diffusely throughout the cytoplasm of the transgenic murine neurons and transfected N18-RE-105 cells. In contrast, cells incubated with TTC-hSOD1 showed hSOD1 localized to the cell surface and intra-cytoplasmic vesicles. The cells expressing hSOD1 showed enhanced survival in glucose- and pyruvate-free medium. Neither cortical neurons nor N18-RE-105 cells incubated in TTC-hSOD1 showed increased survival during starvation. Access to the site where toxic superoxides are generated or their targets may be necessary for the protective function of SOD1.
Collapse
Affiliation(s)
- C C Matthews
- Department of Neurology, University of Maryland, School of Medicine, and Veteran's Affairs Medical Center, Baltimore, MD, 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Francis JW, Brown RH, Figueiredo D, Remington MP, Castillo O, Schwarzschild MA, Fishman PS, Murphy JR, vanderSpek JC. Enhancement of diphtheria toxin potency by replacement of the receptor binding domain with tetanus toxin C-fragment: a potential vector for delivering heterologous proteins to neurons. J Neurochem 2000; 74:2528-36. [PMID: 10820215 DOI: 10.1046/j.1471-4159.2000.0742528.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This study describes the expression, purification, and characterization of a recombinant fusion toxin, DAB(389)TTC, composed of the catalytic and membrane translocation domains of diphtheria toxin (DAB(389)) linked to the receptor binding fragment of tetanus toxin (C-fragment). As determined by its ability to inhibit cellular protein synthesis in primary neuron cultures, DAB(389)TTC was approximately 1,000-fold more cytotoxic than native diphtheria toxin or the previously described fusion toxin, DAB(389)MSH. The cytotoxic effect of DAB(389)TTC on cultured cells was specific toward neuronal-type cells and was blocked by coincubation of the chimeric toxin with tetanus antitoxin. The toxicity of DAB(389)TTC, like that of diphtheria toxin, was dependent on passage through an acidic compartment and ADP-ribosyltransferase activity of the DAB(389) catalytic fragment. These results suggest that a catalytically inactive form of DAB(389)TTC may be useful as a nonviral vehicle to deliver exogenous proteins to the cytosolic compartment of neurons.
Collapse
Affiliation(s)
- J W Francis
- Cecil B. Day Center for Neuromuscular Research, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hill IE, Murray C, Richard J, Rasquinha I, MacManus JP. Despite the internucleosomal cleavage of DNA, reactive oxygen species do not produce other markers of apoptosis in cultured neurons. Exp Neurol 2000; 162:73-88. [PMID: 10716890 DOI: 10.1006/exnr.2000.7322] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The cell death induced by hydroxyl radicals generated by Cu-phenanthroline and peroxynitrite generated by 3-morpholinosydnonimine hydrochloride (SIN-1) in rat primary cortical neuronal cultures was compared with the apoptotic death induced by staurosporine and the necrotic death induced by glutamate. Both SIN-1 and Cu-phenanthroline were capable of generating internucleosomal cleavage of DNA-a hallmark of apoptosis. Other characteristics of this cell death, such as nuclear morphology by light microscopy; DNA breaks by single-cell gel electrophoresis; the effects of the apoptotic inhibitors cycloheximide, aurintricarboxylic acid, and tosyl-l-lysine chloromethyl ketone; the measurement of caspase activity; and the effects of antioxidants, were then analyzed. The conclusion from these hallmarks of apoptosis is that the cell death induced by these reactive oxygen species is not apoptosis.
Collapse
Affiliation(s)
- I E Hill
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, K1A 0R6, Canada
| | | | | | | | | |
Collapse
|
50
|
Abstract
Roles proposed for nitric oxide (NO) in CNS pathophysiology are increasingly diverse and range from intercellular signaling, through necrotic killing of cells and invading pathogens, to the involvement of NO in apoptosis and tissue remodeling. In vitro evidence and observations from experimental animal models of a variety of human neuropathologies, including stroke, indicate that glial cells can produce NO. Regulation of at least one of the NO synthase genes (NOS-2) in glia has been well described; however, apart from hints emerging out of co-culture studies and extrapolation based upon the reactivity of NO, we are a long way from identifying functions for glial-derived NO in the CNS. Although the assumption is that NO is very often cytotoxic, it is evident that NO production does not always equate with tissue damage, and that both the cellular source of NO and the timing of NO production are important factors in terms of its effects. With the development of strategies to transfer or manipulate expression of the NOS genes in specific cells in situ, the ability to deliver NO into the CNS via long-lived chemical donors, and the emergence of more selective NOS inhibitors, an appreciation of the significance of glial-derived NO will change.
Collapse
Affiliation(s)
- S Murphy
- Department of Pharmacology and the Neuroscience Program, University of Iowa College of Medicine, Iowa City, USA.
| |
Collapse
|