1
|
Enhancement of the transduction efficiency of a lentiviral vector for neuron-specific retrograde gene delivery through the point mutation of fusion glycoprotein type E. J Neurosci Methods 2018; 311:147-155. [PMID: 30347222 DOI: 10.1016/j.jneumeth.2018.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pseudotyping of a lentiviral vector with fusion glycoproteins composed of rabies virus glycoprotein (RVG) and vesicular stomatitis virus glycoprotein (VSVG) segments achieves high gene transfer efficiency through retrograde transport in the nervous system. In our previous study, we determined the junction of RVG/VSVG segments of glycoproteins that enhances the transduction efficiency of the neuron-specific retrograde gene transfer (NeuRet) vector (termed fusion glycoprotein type E or FuG-E). NEW METHOD We aimed to optimize the amino acid residue at position 440 in the membrane-proximal region of FuG-E to improve the efficiency of retrograde gene transfer in the brain. RESULTS We constructed variants of FuG-E with 18 kinds of single amino acid substitutions at residue 440 to generate lentiviral vectors pseudotyped with these variants, and tested in vivo gene transfer of the vectors in the mouse brain. The FuG-E (P440E) variant, in which proline was substituted by glutamate at residue 440 in FuG-E, showed the greatest retrograde gene transfer efficiency in the brain, bearing the property of the NeuRet vector. The FuG-E (P440E) pseudotype also displayed efficient retrograde gene transfer in the common marmoset brain. COMPARISON WITH EXISTING METHODS The NeuRet vector with the FuG-E (P440E) variant demonstrated higher retrograde gene transfer efficiency into different neural pathways compared with the parental FuG-E vector. CONCLUSIONS The FuG-E (P440E) pseudotype provides a powerful tool to investigate neural circuit mechanisms underlying various brain functions and for gene therapy trials of neurological and neurodegenerative diseases.
Collapse
|
2
|
Inoue KI, Miyachi S, Nishi K, Okado H, Nagai Y, Minamimoto T, Nambu A, Takada M. Recruitment of calbindin into nigral dopamine neurons protects against MPTP-Induced parkinsonism. Mov Disord 2018; 34:200-209. [PMID: 30161282 DOI: 10.1002/mds.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/06/2018] [Accepted: 06/29/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Parkinson's disease is caused by dopamine deficiency in the striatum, which is a result of loss of dopamine neurons from the substantia nigra pars compacta. There is a consensus that a subpopulation of nigral dopamine neurons that expresses the calcium-binding protein calbindin is selectively invulnerable to parkinsonian insults. The objective of the present study was to test the hypothesis that dopamine neuron degeneration might be prevented by viral vector-mediated gene delivery of calbindin into the dopamine neurons that do not normally contain it. METHODS A calbindin-expressing adenoviral vector was injected into the striatum of macaque monkeys to be conveyed to cell bodies of nigral dopamine neurons through retrograde axonal transport, or the calbindin-expressing lentiviral vector was injected into the nigra directly because of its predominant uptake from cell bodies and dendrites. The animals in which calbindin was successfully recruited into nigral dopamine neurons were administered systemically with MPTP. RESULTS In the monkeys that had received unilateral vector injections, parkinsonian motor deficits, such as muscular rigidity and akinesia/bradykinesia, appeared predominantly in the limbs corresponding to the non-calbindin-recruited hemisphere after MPTP administration. Data obtained from tyrosine hydroxylase immunostaining and PET imaging for the dopamine transporter revealed that the nigrostriatal dopamine system was preserved better on the calbindin-recruited side. Conversely, on the non-calbindin-recruited control side, many more dopamine neurons expressed α-synuclein. CONCLUSIONS The present results indicate that calbindin recruitment into nigral dopamine neurons protects against the onset of parkinsonian insults, thus providing a novel approach to PD prevention. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Shigehiro Miyachi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Cognitive Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | - Katsunori Nishi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| | - Haruo Okado
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Atsushi Nambu
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| |
Collapse
|
3
|
Kobayashi K, Inoue KI, Tanabe S, Kato S, Takada M, Kobayashi K. Pseudotyped Lentiviral Vectors for Retrograde Gene Delivery into Target Brain Regions. Front Neuroanat 2017; 11:65. [PMID: 28824385 PMCID: PMC5539090 DOI: 10.3389/fnana.2017.00065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Gene transfer through retrograde axonal transport of viral vectors offers a substantial advantage for analyzing roles of specific neuronal pathways or cell types forming complex neural networks. This genetic approach may also be useful in gene therapy trials by enabling delivery of transgenes into a target brain region distant from the injection site of the vectors. Pseudotyping of a lentiviral vector based on human immunodeficiency virus type 1 (HIV-1) with various fusion envelope glycoproteins composed of different combinations of rabies virus glycoprotein (RV-G) and vesicular stomatitis virus glycoprotein (VSV-G) enhances the efficiency of retrograde gene transfer in both rodent and nonhuman primate brains. The most recently developed lentiviral vector is a pseudotype with fusion glycoprotein type E (FuG-E), which demonstrates highly efficient retrograde gene transfer in the brain. The FuG-E–pseudotyped vector permits powerful experimental strategies for more precisely investigating the mechanisms underlying various brain functions. It also contributes to the development of new gene therapy approaches for neurodegenerative disorders, such as Parkinson’s disease, by delivering genes required for survival and protection into specific neuronal populations. In this review article, we report the properties of the FuG-E–pseudotyped vector, and we describe the application of the vector to neural circuit analysis and the potential use of the FuG-E vector in gene therapy for Parkinson’s disease.
Collapse
Affiliation(s)
- Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological SciencesOkazaki, Japan.,SOKENDAI (The Graduate University for Advanced Studies)Hayama, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Soshi Tanabe
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of MedicineFukushima, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of MedicineFukushima, Japan
| |
Collapse
|
4
|
Qian Y, Xu S, Yang X, Xiao Q. Purinergic receptor P2Y6 contributes to 1-methyl-4-phenylpyridinium-induced oxidative stress and cell death in neuronal SH-SY5Y cells. J Neurosci Res 2017; 96:253-264. [PMID: 28752899 DOI: 10.1002/jnr.24119] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 11/08/2022]
Abstract
Oxidative stress and neural degeneration have been shown to be involved in the pathogenesis of Parkinson's disease (PD). The P2Y6 purinergic receptor (P2Y6R) has been shown to participate in the activation of microglia and the production of pro-inflammatory factors induced by lipopolysaccharide to cause neuronal loss. However, the function of P2Y6R during oxidative stress in neurons is unclear. In the present study, 1-methyl-4-phenylpyridinium (MPP+ ) treatment increased the level of UDP/P2Y6R on neuronal SH-SY5Y cells. Importantly, pharmacological inhibition of P2Y6R or knockdown of P2Y6R using a siRNA exerted an increased protective effect by preventing MPP+ -induced increases in the levels of reactive oxygen species (ROS), superoxide anion, inducible nitric oxide synthase (iNOS), and malondialdehyde (MDA) and down-regulation of superoxide dismutase 1 (SOD1) expression. UDP, an agonist of P2Y6R, enhanced the effects of MPP+ , which was also inhibited by apyrase or MRS2578. Additionally, P2Y6R knockdown also significantly reversed both the loss of cell viability and the increase in the levels of phosphorylated extracellular signal-regulated protein kinase (p-ERK1/2) and p38 (p-p38) caused by MPP+ stimulation. However, the inhibition of the ERK1/2 and p38 kinase signaling pathways had no effect on P2Y6R expression. Taken together, these results support the hypothesis that P2Y6R expressed on neuronal SH-SY5Y cell is associated with the progression of oxidative stress and cell death induced by MPP+ , suggesting that P2Y6R may play an important role in the pathogenesis of PD.
Collapse
Affiliation(s)
- Yiwei Qian
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shaoqing Xu
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaodong Yang
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin Xiao
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
5
|
Anandhan A, Jacome MS, Lei S, Hernandez-Franco P, Pappa A, Panayiotidis MI, Powers R, Franco R. Metabolic Dysfunction in Parkinson's Disease: Bioenergetics, Redox Homeostasis and Central Carbon Metabolism. Brain Res Bull 2017; 133:12-30. [PMID: 28341600 PMCID: PMC5555796 DOI: 10.1016/j.brainresbull.2017.03.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022]
Abstract
The loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of protein inclusions (Lewy bodies) are the pathological hallmarks of Parkinson's disease (PD). PD is triggered by genetic alterations, environmental/occupational exposures and aging. However, the exact molecular mechanisms linking these PD risk factors to neuronal dysfunction are still unclear. Alterations in redox homeostasis and bioenergetics (energy failure) are thought to be central components of neurodegeneration that contribute to the impairment of important homeostatic processes in dopaminergic cells such as protein quality control mechanisms, neurotransmitter release/metabolism, axonal transport of vesicles and cell survival. Importantly, both bioenergetics and redox homeostasis are coupled to neuro-glial central carbon metabolism. We and others have recently established a link between the alterations in central carbon metabolism induced by PD risk factors, redox homeostasis and bioenergetics and their contribution to the survival/death of dopaminergic cells. In this review, we focus on the link between metabolic dysfunction, energy failure and redox imbalance in PD, making an emphasis in the contribution of central carbon (glucose) metabolism. The evidence summarized here strongly supports the consideration of PD as a disorder of cell metabolism.
Collapse
Affiliation(s)
- Annadurai Anandhan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Maria S Jacome
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States
| | - Shulei Lei
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Pablo Hernandez-Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece
| | | | - Robert Powers
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States; Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Rodrigo Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States.
| |
Collapse
|
6
|
Kobayashi K, Kato S, Kobayashi K. Genetic manipulation of specific neural circuits by use of a viral vector system. J Neural Transm (Vienna) 2017; 125:67-75. [DOI: 10.1007/s00702-016-1674-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/30/2016] [Indexed: 01/05/2023]
|
7
|
Otsuki N, Homma T, Fujiwara H, Kaneko K, Hozumi Y, Shichiri M, Takashima M, Ito J, Konno T, Kurahashi T, Yoshida Y, Goto K, Fujii S, Fujii J. Trichloroethylene exposure aggravates behavioral abnormalities in mice that are deficient in superoxide dismutase. Regul Toxicol Pharmacol 2016; 79:83-90. [PMID: 27166294 DOI: 10.1016/j.yrtph.2016.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/11/2022]
Abstract
Trichloroethylene (TCE) has been implicated as a causative agent for Parkinson's disease (PD). The administration of TCE to rodents induces neurotoxicity associated with dopaminergic neuron death, and evidence suggests that oxidative stress as a major player in the progression of PD. Here we report on TCE-induced behavioral abnormality in mice that are deficient in superoxide dismutase 1 (SOD1). Wild-type (WT) and SOD1-deficient (Sod1(-/-)) mice were intraperitoneally administered TCE (500 mg/kg) over a period of 4 weeks. Although the TCE-administrated Sod1(-/-) mice showed marked abnormal motor behavior, no significant differences were observed among the experimental groups by biochemical and histopathological analyses. However, treating mouse neuroblastoma-derived NB2a cells with TCE resulted in the down regulation of the SOD1 protein and elevated oxidative stress under conditions where SOD1 production was suppressed. Taken together, these data indicate that SOD1 plays a pivotal role in protecting motor neuron function against TCE toxicity.
Collapse
Affiliation(s)
- Noriyuki Otsuki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Hiroki Fujiwara
- Department of Physiology, Yamagata University School of Medicine, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Kenya Kaneko
- Department of Physiology, Yamagata University School of Medicine, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Yasukazu Hozumi
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| | - Mototada Shichiri
- Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Mizuki Takashima
- Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Junitsu Ito
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Tasuku Konno
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Toshihiro Kurahashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Yasukazu Yoshida
- Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-cho, Takamatsu, Kagawa 761-0395, Japan
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| | - Satoshi Fujii
- Department of Physiology, Yamagata University School of Medicine, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan.
| |
Collapse
|
8
|
Kobayashi K, Kato S, Inoue KI, Takada M, Kobayashi K. Altering Entry Site Preference of Lentiviral Vectors into Neuronal Cells by Pseudotyping with Envelope Glycoproteins. Methods Mol Biol 2016; 1382:175-86. [PMID: 26611586 DOI: 10.1007/978-1-4939-3271-9_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A lentiviral vector system provides a powerful strategy for gene therapy trials against a variety of neurological and neurodegenerative disorders. Pseudotyping of lentiviral vectors with different envelope glycoproteins not only confers the neurotropism to the vectors, but also alters the preference of sites of vector entry into neuronal cells. One major group of lentiviral vectors is a pseudotype with vesicular stomatitis virus glycoprotein (VSV-G) that enters preferentially cell body areas (somata/dendrites) of neurons and transduces them. Another group contains lentiviral vectors pseudotyped with fusion envelope glycoproteins composed of different sets of rabies virus glycoprotein and VSV-G segments that enter predominantly axon terminals of neurons and are transported through axons retrogradely to their cell bodies, resulting in enhanced retrograde gene transfer. This retrograde gene transfer takes a considerable advantage of delivering the transgene into neuronal cell bodies situated in regions distant from the injection site of the vectors. The rational use of these two vector groups characterized by different entry mechanisms will further extend the strategy for gene therapy of neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenta Kobayashi
- Section of Viral Vector Development, National Institute of Physiological Sciences, Okazaki, 444-8585, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, 484-8506, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, 484-8506, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
| |
Collapse
|
9
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
10
|
Kato S, Kobayashi K, Kobayashi K. Improved transduction efficiency of a lentiviral vector for neuron-specific retrograde gene transfer by optimizing the junction of fusion envelope glycoprotein. J Neurosci Methods 2014; 227:151-8. [PMID: 24613797 DOI: 10.1016/j.jneumeth.2014.02.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/17/2014] [Accepted: 02/25/2014] [Indexed: 11/13/2022]
Abstract
BACKGROUND The vector for neuron-specific retrograde gene transfer (NeuRet) is a pseudotype of human immunodeficiency virus type 1 (HIV-1)-based vector with fusion glycoprotein type C (FuG-C), which consists of the N-terminal region of the extracellular domain of rabies virus glycoprotein (RVG) and the membrane-proximal region of the extracellular domain and the transmembrane/cytoplasmic domains of vesicular stomatitis virus glycoprotein (VSVG). The NeuRet vector shows a high efficiency of gene transfer through retrograde axonal transport and transduces selectively neuronal cells around the injection site. NEW METHOD We aimed to improve the efficiency of retrograde gene transfer of the NeuRet vector by optimizing the junction of RVG and VSVG segments in fusion glycoproteins in their membrane-proximal region. RESULTS We produced various types of fusion glycoproteins, in which the junction of the two glycoprotein segments diverged in the membrane-proximal region and used for pseudotyping of HIV-1-based vector to evaluate the in vivo gene transfer efficiency after intrastriatal injection. We found a novel type of fusion glycoprotein termed type E (FuG-E) that yielded enhanced efficiency of retrograde gene delivery, showing neuron-specific transduction surrounding the injection site. COMPARISON WITH EXISTING METHODS The NeuRet vector pseudotyped with FuG-E displayed the improved efficiency of retrograde gene transfer into different neural pathways compared with the original vector pseudotyped with FuG-C. CONCLUSIONS Our vector system with FuG-E provides a powerful tool for gene therapeutic trials of neurological and neurodegenerative diseases and for the study of the mechanisms of neural networks underlying various brain functions.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute of Physiological Sciences, Okazaki 444-8585, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan.
| |
Collapse
|
11
|
Hennis MR, Seamans KW, Marvin MA, Casey BH, Goldberg MS. Behavioral and neurotransmitter abnormalities in mice deficient for Parkin, DJ-1 and superoxide dismutase. PLoS One 2013; 8:e84894. [PMID: 24386432 PMCID: PMC3873453 DOI: 10.1371/journal.pone.0084894] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/27/2013] [Indexed: 01/10/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease characterized by loss of neurons in the substantia nigra that project to the striatum and release dopamine. The cause of PD remains uncertain, however, evidence implicates mitochondrial dysfunction and oxidative stress. Although most cases of PD are sporadic, 5-10% of cases are caused by inherited mutations. Loss-of-function mutations in Parkin and DJ-1 were the first to be linked to recessively inherited Parkinsonism. Surprisingly, mice bearing similar loss-of-function mutations in Parkin and DJ-1 do not show age-dependent loss of nigral dopaminergic neurons or depletion of dopamine in the striatum. Although the normal cellular functions of Parkin and DJ-1 are not fully understood, we hypothesized that loss-of-function mutations in Parkin and DJ-1 render cells more sensitive to mitochondrial dysfunction and oxidative stress. To test this hypothesis, we crossed mice deficient for Parkin and DJ-1 with mice deficient for the mitochondrial antioxidant protein Mn-superoxide dismutase (SOD2) or the cytosolic antioxidant protein Cu-Zn-superoxide dismutase (SOD1). Aged Parkin-/-DJ-1-/- and Mn-superoxide dismutase triple deficient mice have enhanced performance on the rotorod behavior test. Cu/Zn-superoxide dismutase triple deficient mice have elevated levels of dopamine in the striatum in the absence of nigral cell loss. Our studies demonstrate that on a Parkin/DJ-1 null background, mice that are also deficient for major antioxidant proteins do not have progressive loss of dopaminergic neurons but have behavioral and striatal dopamine abnormalities.
Collapse
Affiliation(s)
- Meghan R. Hennis
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Katherine W. Seamans
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Marian A. Marvin
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Bradford H. Casey
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Matthew S. Goldberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
12
|
Abstract
Although severe stress can elicit toxicity, mild stress often elicits adaptations. Here we review the literature on stress-induced adaptations versus stress sensitization in models of neurodegenerative diseases. We also describe our recent findings that chronic proteotoxic stress can elicit adaptations if the dose is low but that high-dose proteotoxic stress sensitizes cells to subsequent challenges. In these experiments, long-term, low-dose proteasome inhibition elicited protection in a superoxide dismutase-dependent manner. In contrast, acute, high-dose proteotoxic stress sensitized cells to subsequent proteotoxic challenges by eliciting catastrophic loss of glutathione. However, even in the latter model of synergistic toxicity, several defensive proteins were upregulated by severe proteotoxicity. This led us to wonder whether high-dose proteotoxic stress can elicit protection against subsequent challenges in astrocytes, a cell type well known for their resilience. In support of this new hypothesis, we found that the astrocytes that survived severe proteotoxicity became harder to kill. The adaptive mechanism was glutathione dependent. If these findings can be generalized to the human brain, similar endogenous adaptations may help explain why neurodegenerative diseases are so delayed in appearance and so slow to progress. In contrast, sensitization to severe stress may explain why defenses eventually collapse in vulnerable neurons.
Collapse
Affiliation(s)
- Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University
| |
Collapse
|
13
|
Rodriguez-Rocha H, Garcia-Garcia A, Pickett C, Li S, Jones J, Chen H, Webb B, Choi J, Zhou Y, Zimmerman MC, Franco R. Compartmentalized oxidative stress in dopaminergic cell death induced by pesticides and complex I inhibitors: distinct roles of superoxide anion and superoxide dismutases. Free Radic Biol Med 2013; 61:370-83. [PMID: 23602909 PMCID: PMC3883883 DOI: 10.1016/j.freeradbiomed.2013.04.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 04/11/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022]
Abstract
The loss of dopaminergic neurons induced by the parkinsonian toxins paraquat, rotenone, and 1-methyl-4-phenylpyridinium (MPP(+)) is associated with oxidative stress. However, controversial reports exist regarding the source/compartmentalization of reactive oxygen species (ROS) generation and its exact role in cell death. We aimed to determine in detail the role of superoxide anion (O2(•-)), oxidative stress, and their subcellular compartmentalization in dopaminergic cell death induced by parkinsonian toxins. Oxidative stress and ROS formation were determined in the cytosol, intermembrane (IMS), and mitochondrial matrix compartments, using dihydroethidine derivatives and the redox sensor roGFP, as well as electron paramagnetic resonance spectroscopy. Paraquat induced an increase in ROS and oxidative stress in both the cytosol and the mitochondrial matrix prior to cell death. MPP(+) and rotenone primarily induced an increase in ROS and oxidative stress in the mitochondrial matrix. No oxidative stress was detected at the level of the IMS. In contrast to previous studies, overexpression of manganese superoxide dismutase (MnSOD) or copper/zinc SOD (CuZnSOD) had no effect on alterations in ROS steady-state levels, lipid peroxidation, loss of mitochondrial membrane potential (ΔΨm), and dopaminergic cell death induced by MPP(+) or rotenone. In contrast, paraquat-induced oxidative stress and cell death were selectively reduced by MnSOD overexpression, but not by CuZnSOD or manganese-porphyrins. However, MnSOD also failed to prevent ΔΨm loss. Finally, paraquat, but not MPP(+) or rotenone, induced the transcriptional activation of the redox-sensitive antioxidant response elements (ARE) and nuclear factor kappa-B (NF-κB). These results demonstrate a selective role of mitochondrial O2(•-) in dopaminergic cell death induced by paraquat, and show that toxicity induced by the complex I inhibitors rotenone and MPP(+) does not depend directly on mitochondrial O2(•-) formation.
Collapse
Affiliation(s)
- Humberto Rodriguez-Rocha
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Aracely Garcia-Garcia
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Chillian Pickett
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Sumin Li
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Jocelyn Jones
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Han Chen
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Brian Webb
- Thermo Scientific, Research and Development, Rockford, IL 61105, USA
| | - Jae Choi
- Thermo Scientific, Research and Development, Rockford, IL 61105, USA
| | - You Zhou
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Matthew C Zimmerman
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA.
| |
Collapse
|
14
|
Kato S, Kuramochi M, Takasumi K, Kobayashi K, Inoue KI, Takahara D, Hitoshi S, Ikenaka K, Shimada T, Takada M, Kobayashi K. Neuron-specific gene transfer through retrograde transport of lentiviral vector pseudotyped with a novel type of fusion envelope glycoprotein. Hum Gene Ther 2011; 22:1511-23. [PMID: 21806473 DOI: 10.1089/hum.2011.111] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The lentiviral vector system is used extensively in gene therapy trials for various neurological and neurodegenerative disorders. The vector system permits efficient and sustained gene expression in many cell types through integration of the transgene into the host cell genome. However, there is a significant issue concerning the therapeutic use of lentiviral vectors, that transgene insertion may lead to tumorigenesis by altering the expression of proto-oncogenes adjacent to the integration sites. One useful approach for improving safety is to restrict vector transduction to neuronal cells. We have reported the use of human immunodeficiency virus type 1 (HIV-1)-based vectors for efficient retrograde transport by pseudotyping with rabies virus glycoprotein (RV-G) or fusion glycoprotein B type, in which the cytoplasmic domain of RV-G was substituted with the counterpart of vesicular stomatitis virus glycoprotein (VSV-G). Here we developed a novel vector system for neuron-specific retrograde gene transfer (termed NeuRet) by pseudotyping the HIV-1 vector with fusion glycoprotein C type (FuG-C), in which a short C-terminal segment of the extracellular domain and the transmembrane/cytoplasmic domains of RV-G were replaced with the corresponding regions of VSV-G. FuG-C pseudotyping caused efficient gene transfer, mainly through retrograde transport, into neuronal cells in diverse brain regions, whereas the pseudotyping resulted in less efficiency for the transduction of glial and neural stem/progenitor cells. Our NeuRet vector system achieves efficient retrograde gene delivery for therapeutic trials and improves their safety by greatly reducing the risk of gene transduction of dividing cells in the brain.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Barone MC, Sykiotis GP, Bohmann D. Genetic activation of Nrf2 signaling is sufficient to ameliorate neurodegenerative phenotypes in a Drosophila model of Parkinson's disease. Dis Model Mech 2011; 4:701-7. [PMID: 21719443 PMCID: PMC3180234 DOI: 10.1242/dmm.007575] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder. Oxidative stress has been associated with the etiology of both sporadic and monogenic forms of PD. The transcription factor Nrf2, a conserved global regulator of cellular antioxidant responses, has been implicated in neuroprotection against PD pathology. However, direct evidence that upregulation of the Nrf2 pathway is sufficient to confer neuroprotection in genetic models of PD is lacking. Expression of the PD-linked gene encoding α-synuclein in dopaminergic neurons of Drosophila results in decreased locomotor activity and selective neuron loss in a progressive age-dependent manner, providing a genetically accessible model of PD. Here we show that upregulation of the Nrf2 pathway by overexpressing Nrf2 or its DNA-binding dimerization partner, Maf-S, restores the locomotor activity of α-synuclein-expressing flies. Similar benefits are observed upon RNA-interference-mediated downregulation of the prime Nrf2 inhibitor, Keap1, as well as in conditions of keap1 heterozygosity. Consistently, the α-synuclein-induced dopaminergic neuron loss is suppressed by Maf-S overexpression or keap1 heterozygosity. Our data validate the sustained upregulation of the Nrf2 pathway as a neuroprotective strategy against PD. This model provides a genetically accessible in vivo system in which to evaluate the potential of additional Nrf2 pathway components and regulators as therapeutic targets.
Collapse
Affiliation(s)
- Maria Cecilia Barone
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | |
Collapse
|
16
|
Kato S, Kobayashi K, Inoue KI, Kuramochi M, Okada T, Yaginuma H, Morimoto K, Shimada T, Takada M, Kobayashi K. A lentiviral strategy for highly efficient retrograde gene transfer by pseudotyping with fusion envelope glycoprotein. Hum Gene Ther 2011; 22:197-206. [PMID: 20954846 DOI: 10.1089/hum.2009.179] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The lentiviral vector system based on human immunodeficiency virus type 1 (HIV-1) is used extensively in gene therapy trials of neurological and neurodegenerative diseases. Retrograde axonal transport of viral vectors offers a great advantage to the delivery of genes into neuronal cell bodies that are situated in regions distant from the injection site. Pseudotyping of HIV-1-based vectors with selective variants of rabies virus glycoprotein (RV-G) increases gene transfer via retrograde transport into the central nervous system. Because large-scale application for gene therapy trials requires high titer stocks of the vector, pseudotyping of a lentiviral vector that produces more efficient retrograde transport is needed. In the present study, we developed a novel vector system for highly efficient retrograde gene transfer by pseudotyping an HIV-1 vector with a fusion envelope glycoprotein (termed FuG-B) in which the cytoplasmic domain of RV-G was substituted by the corresponding part of vesicular stomatitis virus glycoprotein. The FuG-B pseudotype shifted the transducing property of the lentiviral vector and enhanced the retrograde transport-mediated gene transfer into different brain regions innervating the striatum with greater efficiency than that of the RV-G pseudotype in mice. In addition, injection of the FuG-B-pseudotyped vector into monkey striatum (caudate and putamen) allowed for highly efficient gene delivery into the nigrostriatal dopamine system, which is a major target for gene therapy of Parkinson's disease. Our strategy provides a powerful tool for the treatment of certain neurological and neurodegenerative diseases by promoting retrograde gene delivery via a lentiviral vector.
Collapse
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Gonzalez AM, Berlanga O, Leadbeater WE, Cooper-Charles L, Sims K, Logan A, Eliceiri B, Berry M, Baird A. The deployment of adenovirus-containing gene activated matrices onto severed axons after central nervous system injury leads to transgene expression in target neuronal cell bodies. J Gene Med 2009; 11:679-88. [PMID: 19507166 DOI: 10.1002/jgm.1354] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In previous studies, we showed that gene activated matrices (GAMs) containing nonviral vectors successfully deliver genes to neurons after optic nerve and spinal cord injury. In the present study, we evaluated whether adenoviral vectors delivered within a GAM increase the efficiency of local gene delivery to injured CNS neurons. Lyophilized GAMs containing collagen and adenoviral vectors were assessed in vitro and in vivo. METHODS We evaluated viral vector stability, release kinetics and efficiency of transduction for this GAM formulation in vitro using the quantitative polymerase chain reaction (qPCR), flow cytometry and fluorescence microscopy. Using PCR, reverse transcriptase-PCR and confocal microscopy, we assessed viral DNA retrograde axonal transport, green fluorescent protein (GFP) expression in retinal ganglion cells (RGCs) after GAM implantation into the wound of the rat transected optic nerve. RESULTS qPCR analyses demonstrated that 100% of viral particles were retained within the collagen after lyophilization. In vitro studies demonstrated that 60% of the particles within the GAM were infective and not released from the collagen matrix when placed in water. By 24 h, GFP expression was detected within cells that have invaded the GAM. In vivo studies demonstrated that adenoviral particles were retrogradely transported in axons from the GAM implanted at the lesion site to the RGC in the retina where the corresponding mRNA was expressed. Analysis of the efficiency of cell transduction indicated that 69% of RGC express GFP. CONCLUSIONS These studies demonstrate that lyophilized GAMs containing adenoviral particles within collagen are stable, retain a significant proportion of their infectivity and successfully and efficiently deliver genes to neurons after central nervous system injury.
Collapse
Affiliation(s)
- Ana Maria Gonzalez
- Molecular Neuroscience Group, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yon JM, Baek IJ, Lee SR, Kim MR, Lee BJ, Yun YW, Nam SY. Immunohistochemical identification and quantitative analysis of cytoplasmic Cu/Zn superoxide dismutase in mouse organogenesis. J Vet Sci 2008; 9:233-40. [PMID: 18716442 PMCID: PMC2811834 DOI: 10.4142/jvs.2008.9.3.233] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cytoplasmic Cu/Zn superoxide dismutase (SOD1) is an antioxidant enzyme that converts superoxide to hydrogen peroxide in cells. Its spatial distribution matches that of superoxide production, allowing it to protect cells from oxidative stress. SOD1 deficiencies result in embryonic lethality and a wide range of pathologies in mice, but little is known about normal SOD1 protein expression in developing embryos. In this study, the expression pattern of SOD1 was investigated in post-implantation mouse embryos and extraembryonic tissues, including placenta, using Western blotting and immunohistochemical analyses. SOD1 was detected in embryos and extraembryonic tissues from embryonic day (ED) 8.5 to 18.5. The signal in embryos was observed at the lowest level on ED 9.5-11.5, and the highest level on ED 17.5-18.5, while levels remained constant in the surrounding extraembryonic tissues during all developmental stages examined. Immunohistochemical analysis of SOD1 expression on ED 13.5-18.5 revealed its ubiquitous distribution throughout developing organs. In particular, high levels of SOD1 expression were observed in the ependymal epithelium of the choroid plexus, ganglia, sensory cells of the olfactory and vestibulocochlear epithelia, blood cells and vessels, hepatocytes and hematopoietic cells of the liver, lymph nodes, osteogenic tissues, and skin. Thus, SOD1 is highly expressed at late stages of embryonic development in a cell- and tissue-specific manner, and can function as an important antioxidant enzyme during organogenesis in mouse embryos.
Collapse
Affiliation(s)
- Jung-Min Yon
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Leak RK, Zigmond MJ, Liou AKF. Adaptation to chronic MG132 reduces oxidative toxicity by a CuZnSOD-dependent mechanism. J Neurochem 2008; 106:860-74. [PMID: 18466318 DOI: 10.1111/j.1471-4159.2008.05459.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To study whether and how cells adapt to chronic cellular stress, we exposed PC12 cells to the proteasome inhibitor MG132 (0.1 microM) for 2 weeks and longer. This treatment reduced chymotrypsin-like proteasome activity by 47% and was associated with protection against both 6-hydroxydopamine (6-OHDA; 100 microM) and higher dose MG132 (40 microM). Protection developed slowly over the course of the first 2 weeks of exposure and was chronic thereafter. There was no change in total GSH levels after MG132. Buthionine sulfoximine (100 microM) reduced GSH levels by 60%, but exacerbated 6-OHDA toxicity to the same extent in both MG132-treated and control cells and failed to reduce MG132-induced protection. Chronic MG132 resulted in elevated antioxidant proteins CuZn superoxide dismutase (SOD; +55%), MnSOD (+21%), and catalase (+15%), as well as chaperone heat-shock protein 70 (+42%). Examination of SOD enzyme activity revealed higher levels of CuZnSOD (+40%), with no change in MnSOD. We further assessed the mechanism of protection by reducing CuZnSOD levels with two independent siRNA sequences, both of which successfully attenuated protection against 6-OHDA. Previous reports suggested that artificial over-expression of CuZnSOD in dopaminergic cells is protective. Our data complement such observations, revealing that dopaminergic cells are also able to use endogenous CuZnSOD in self-defensive adaptations to chronic stress, and that they can even do so in the face of extensive GSH loss.
Collapse
Affiliation(s)
- Rehana K Leak
- Department of Neurology, Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | |
Collapse
|
20
|
Boulos S, Meloni BP, Arthur PG, Bojarski C, Knuckey NW. Peroxiredoxin 2 overexpression protects cortical neuronal cultures from ischemic and oxidative injury but not glutamate excitotoxicity, whereas Cu/Zn superoxide dismutase 1 overexpression protects only against oxidative injury. J Neurosci Res 2008; 85:3089-97. [PMID: 17663478 DOI: 10.1002/jnr.21429] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously reported that peroxiredoxin 2 (PRDX2) and Cu/Zn superoxide dismutase 1 (SOD1) proteins are up-regulated in rat primary neuronal cultures following erythropoietin (EPO) preconditioning. In the present study, we have demonstrated that adenovirally mediated overexpression of PRDX2 in cortical neuronal cultures can protect neurons from in vitro ischemia (oxygen-glucose deprivation) and an oxidative insult (cumene hydroperoxide) but not glutamate excitotoxicity. We have also demonstrated that adenovirally mediated overexpression of SOD1 in cortical neuronal cultures protected neurons only against the oxidative insult. Interestingly, we did not detect up-regulation of PRDX2 or SOD1 protein in the rat hippocampus following exposure to either 3 min or 8 min of global cerebral ischemia. Further characterization of PRDX2's neuroprotective mechanisms may aid in the development of a neuroprotective therapy.
Collapse
Affiliation(s)
- Sherif Boulos
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Australian Neuromuscular Research Institute, Nedlands, Western Australia, Australia.
| | | | | | | | | |
Collapse
|
21
|
Kato S, Inoue KI, Kobayashi K, Yasoshima Y, Miyachi S, Inoue S, Hanawa H, Shimada T, Takada M, Kobayashi K. Efficient Gene Transfer via Retrograde Transport in Rodent and Primate Brains Using a Human Immunodeficiency Virus Type 1-Based Vector Pseudotyped with Rabies Virus Glycoprotein. Hum Gene Ther 2007; 18:1141-51. [DOI: 10.1089/hum.2007.082] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 9601295, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Cooperation, Kawaguchi 332-0012, Japan
| | - Ken-Ichi Inoue
- Core Research for Evolutional Science and Technology, Japan Science and Technology Cooperation, Kawaguchi 332-0012, Japan
- Department of System Neuroscience, Tokyo Metropolitan Institute for Neuroscience, Tokyo 183-8526, Japan
| | - Kenta Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 9601295, Japan
| | - Yasunobu Yasoshima
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 9601295, Japan
| | - Shigehiro Miyachi
- Core Research for Evolutional Science and Technology, Japan Science and Technology Cooperation, Kawaguchi 332-0012, Japan
- Department of Behavioral and Brain Sciences, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan
| | - Satoshi Inoue
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hideki Hanawa
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo 183-8526, Japan
| | - Takashi Shimada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo 183-8526, Japan
| | - Masahiko Takada
- Core Research for Evolutional Science and Technology, Japan Science and Technology Cooperation, Kawaguchi 332-0012, Japan
- Department of System Neuroscience, Tokyo Metropolitan Institute for Neuroscience, Tokyo 183-8526, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 9601295, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Cooperation, Kawaguchi 332-0012, Japan
| |
Collapse
|
22
|
Zhu Y, Hoell P, Ahlemeyer B, Sure U, Bertalanffy H, Krieglstein J. Implication of PTEN in production of reactive oxygen species and neuronal death in in vitro models of stroke and Parkinson's disease. Neurochem Int 2006; 50:507-16. [PMID: 17169462 DOI: 10.1016/j.neuint.2006.10.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 10/17/2006] [Accepted: 10/25/2006] [Indexed: 11/28/2022]
Abstract
Oxidative stress plays crucial role in the pathogenesis of neurodegenerative diseases. However, the precise mechanism for an increased production of reactive oxygen species (ROS) under pathological conditions is not yet fully understood. We have recently demonstrated an implication of phosphatase and tensin homologue deleted on chromosome 10 (PTEN), a tumor suppressor, in ROS generation and neuronal apoptosis induced by staurosporine. These findings raised further interest whether PTEN functions as a common mediator of oxidative stress in neurodegenerative processes. To address this issue, neural cells were exposed to oxygen-glucose deprivation (OGD) and to the neurotoxin 1-methyl-4-phenylpyridinium iodide (MPP(+)), which mimic cerebral ischemia and Parkinson's disease, respectively. OGD for 4 h followed by 16 h of reoxygenation or incubation with MPP(+) (250 microM) for 48 h induced 33% and 45% neuronal death in rat hippocampal and in human dopaminergic SH-SY5Y neurons, respectively, accompanied by a gradual increase in the intracellular level of ROS. The increase in ROS by OGD and by MPP(+) did not cause oxidative inactivation of PTEN and thus, PTEN remains constitutively active. In support, the protein level of PTEN was not reduced in both cell cultures after challenging with OGD or MPP(+). Importantly, the elevated intracellular ROS levels and the neuronal death caused by OGD or by MPP(+) toxicity were significantly inhibited when PTEN was downregulated by a specific antisense oligonucleotide or by siRNA. Because SOD2 protein level is not altered either by knockdown of PTEN nor by an inhibition of the PI3K/Akt signalling, we suggest that SOD2 do not contribute to the pathomechanism of oxidative stress induced by PTEN or by inhibiting the related Akt signalling. The present study highlights PTEN as a crucial and common mediator of ROS generation and neuronal death and suggests that PTEN could become a potential therapeutic target for interfering with neurodegeneration.
Collapse
Affiliation(s)
- Yuan Zhu
- Institut für Pharmakologie und Toxikologie, Philipps-Universität Marburg, Baldingerstrasse, D-35032 Marburg, Germany.
| | | | | | | | | | | |
Collapse
|
23
|
Angelova P, Müller W. Oxidative modulation of the transient potassium current IAby intracellular arachidonic acid in rat CA1 pyramidal neurons. Eur J Neurosci 2006; 23:2375-84. [PMID: 16706845 DOI: 10.1111/j.1460-9568.2006.04767.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Oxidative stress affects cellular membrane lipids and proteins. Using whole-cell patch-clamp recording we demonstrate differential oxidative inhibition of voltage-gated transient (IA) and delayed rectifier [IK(V)] K+ currents by arachidonic acid (AA) and H2O2 in CA1 neurons in hippocampal slice. We show that intracellular application of 1 pm AA or its non-metabolizable analog eicosatetraynoic acid (100 pm) reduced IA by approximately 42% but did not affect IK(V). AA shifted the voltage dependence of steady-state inactivation of IA by 12 mV to more negative potentials whereas the rate of inactivation was unchanged. Surprisingly, intracellular glutathione (GSH, 20 mm) enhanced the effect of AA on maximal IA (-62%) and with AA slowed inactivation of IA. The combination of GSH and extracellular ascorbate (0.4 mm) prevented reduction of IA by AA. Intracellular Trolox (a vitamin E analog, 10 microm) reduced IA by 61%and IK(V) by 39%. Like AA, intracellular Trolox caused a 10-mV left shift of IA steady-state inactivation but Trolox and AA did not cause a shift when coapplied. Extracellular Trolox (100 microm) had no effects on IA. H2O2 (80 microm) reduced both IA and IK(V) in a GSH- and ascorbate-sensitive manner and slowed the rate of inactivation of IA by a factor of 2. Coapplication of H2O2 with GSH and extracellular ascorbate caused approximately 22 mV negative shifts of both steady-state inactivation and activation. We conclude that AA is extremely potent in affecting IA by oxidative modifications. Antioxidants can augment these effects, probably by catalysis of the underlying reactions between oxidants and IA channel proteins.
Collapse
Affiliation(s)
- Plamena Angelova
- Neuroscience Research Institute, Charité, Humboldt University, Berlin, Germany
| | | |
Collapse
|