51
|
Patergnani S, Fossati V, Bonora M, Giorgi C, Marchi S, Missiroli S, Rusielewicz T, Wieckowski MR, Pinton P. Mitochondria in Multiple Sclerosis: Molecular Mechanisms of Pathogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:49-103. [PMID: 28069137 DOI: 10.1016/bs.ircmb.2016.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria, the organelles that function as the powerhouse of the cell, have been increasingly linked to the pathogenesis of many neurological disorders, including multiple sclerosis (MS). MS is a chronic inflammatory demyelinating disease of the central nervous system (CNS) and a leading cause of neurological disability in young adults in the western world. Its etiology remains unknown, and while the inflammatory component of MS has been heavily investigated and targeted for therapeutic intervention, the failure of remyelination and the process of axonal degeneration are still poorly understood. Recent studies suggest a role of mitochondrial dysfunction in the neurodegenerative aspects of MS. This review is focused on mitochondrial functions under physiological conditions and the consequences of mitochondrial alterations in various CNS disorders. Moreover, we summarize recent findings linking mitochondrial dysfunction to MS and discuss novel therapeutic strategies targeting mitochondria-related pathways as well as emerging experimental approaches for modeling mitochondrial disease.
Collapse
Affiliation(s)
- S Patergnani
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - V Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - C Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Missiroli
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - T Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - P Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
52
|
Gazzin S, Vitek L, Watchko J, Shapiro SM, Tiribelli C. A Novel Perspective on the Biology of Bilirubin in Health and Disease. Trends Mol Med 2016; 22:758-768. [PMID: 27515064 DOI: 10.1016/j.molmed.2016.07.004] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/14/2016] [Indexed: 01/02/2023]
Abstract
Unconjugated bilirubin (UCB) is known to be one of the most potent endogenous antioxidant substances. While hyperbilirubinemia has long been recognized as an ominous sign of liver dysfunction, recent data strongly indicate that mildly elevated bilirubin (BLB) levels can be protective against an array of diseases associated with increased oxidative stress. These clinical observations are supported by new discoveries relating to the role of BLB in immunosuppression and inhibition of protein phosphorylation, resulting in the modulation of intracellular signaling pathways in vascular biology and cancer, among others. Collectively, the evidence suggests that targeting BLB metabolism could be considered a potential therapeutic approach to ameliorate a variety of conditions.
Collapse
Affiliation(s)
- Silvia Gazzin
- Liver Research Center, Italian Liver Foundation, SS14, Km 163.5, Trieste, Italy
| | - Libor Vitek
- 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - Jon Watchko
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven M Shapiro
- Division of Neurology, Department of Pediatrics, Children's Mercy Hospital & Clinics, Kansas City, MO, USA; University of Missouri-Kansas City, Kansas City, MO, USA; Department of Neurology, University of Kansas, Kansas City, KS, USA; Department of Pediatrics, University of Kansas, Kansas City, KS, USA
| | - Claudio Tiribelli
- Liver Research Center, Italian Liver Foundation, SS14, Km 163.5, Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
53
|
Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci 2016; 73:3221-47. [PMID: 27100828 PMCID: PMC4967105 DOI: 10.1007/s00018-016-2223-0] [Citation(s) in RCA: 1865] [Impact Index Per Article: 207.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022]
Abstract
The multifunctional regulator nuclear factor erythroid 2-related factor (Nrf2) is considered not only as a cytoprotective factor regulating the expression of genes coding for anti-oxidant, anti-inflammatory and detoxifying proteins, but it is also a powerful modulator of species longevity. The vertebrate Nrf2 belongs to Cap 'n' Collar (Cnc) bZIP family of transcription factors and shares a high homology with SKN-1 from Caenorhabditis elegans or CncC found in Drosophila melanogaster. The major characteristics of Nrf2 are to some extent mimicked by Nrf2-dependent genes and their proteins including heme oxygenase-1 (HO-1), which besides removing toxic heme, produces biliverdin, iron ions and carbon monoxide. HO-1 and their products exert beneficial effects through the protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. On the other hand, the disturbances in the proper HO-1 level are associated with the pathogenesis of some age-dependent disorders, including neurodegeneration, cancer or macular degeneration. This review summarizes our knowledge about Nrf2 and HO-1 across different phyla suggesting their conservative role as stress-protective and anti-aging factors.
Collapse
Affiliation(s)
- Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Milena Damulewicz
- Department of Cell Biology and Imaging, Faculty of Biology and Earth Sciences, Jagiellonian University, Krakow, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Faculty of Biology and Earth Sciences, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
54
|
Yama K, Sato K, Murao Y, Tatsunami R, Tampo Y. Epalrestat Upregulates Heme Oxygenase-1, Superoxide Dismutase, and Catalase in Cells of the Nervous System. Biol Pharm Bull 2016; 39:1523-30. [PMID: 27439473 DOI: 10.1248/bpb.b16-00332] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heme oxygenase (HO)-1 has potent antioxidant and anti-inflammatory functions. Recent studies have shown that the upregulation of HO-1 is beneficial to counteract neuroinflammation, making HO-1 a new therapeutic target for neurological diseases. We have reported that epalrestat (EPS), which is currently used for the treatment of diabetic neuropathy, increases HO-1 levels through the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) in bovine aortic endothelial cells. In this study, we tested the hypothesis that EPS upregulates HO-1 via Nrf2 activation in the component cells of the nervous system, by using rat Schwann cells and human SH-SY5Y cells. Treatment of Schwann cells with EPS at near-plasma concentration led to a dramatic increase in HO-1 levels. Nrf2 knockdown by small interfering RNA (siRNA) suppressed the EPS-induced HO-1 expression. EPS did not promote the intracellular accumulation of free ferrous ion and reactive oxygen species, by increasing ferritin via Nrf2 during HO-1 induction. Moreover, EPS stimulated the expression of superoxide dismutase 1 and catalase, which also are Nrf2 target gene products. It also markedly increased HO-1 levels in SH-SY5Y cells through the activation of Nrf2. We demonstrated for the first time that EPS upregulates HO-1, superoxide dismutase, and catalase by activating Nrf2. We suggest that EPS has the potential to prevent several neurological diseases.
Collapse
Affiliation(s)
- Kaori Yama
- Hokkaido Pharmaceutical University School of Pharmacy
| | | | | | | | | |
Collapse
|
55
|
Lin JG, Lee YC, Tseng CH, Chen DY, Shih CY, MacDonald I, Hung SY, Chen YH. Electroacupuncture inhibits pruritogen-induced spinal microglial activation in mice. Brain Res 2016; 1649:23-29. [PMID: 27396909 DOI: 10.1016/j.brainres.2016.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 06/20/2016] [Accepted: 07/07/2016] [Indexed: 12/18/2022]
Abstract
In this study, we examined whether electroacupuncture (EA) represses pruritogen-induced microglial activation. Immunohistochemical studies revealed that a subcutaneous (s.c.) injection of the pruritogen 5'-guanidinonaltrindole (GNTI; 0.3mg/kg) to the back of the neck in mice induced acute expression of the ionized calcium-binding adaptor molecule 1 (Iba1) in both gray and white matter of the spinal cord, with the highest expression in the dorsal horn area. EA application (2Hz) to LI4 and LI11 attenuated GNTI-induced scratching behavior and repressed GNTI-induced Iba1 expression and Iba1 (+) microglia in the dorsal horn. In contrast, EA at the ST36 acupoint had no such effects. Confocal image analysis revealed co-expression of phosphorylated p38 and Iba1 in microglia with EA at the ST36 acupoint, but not at the LI4 or LI11 acupoints. In Western blot analysis, s.c. injection of GNTI to the back of the neck increased Iba1 and phospho-p38 expression in the spinal cord as compared with injection of saline, while EA at LI4 and LI11 reduced GNTI-induced expression of Iba1 and phospho-p38. These findings indicate that EA at LI4 and LI11, but not at ST36, reduces GNTI-induced microglial activation in the mouse spinal cord.
Collapse
Affiliation(s)
- Jaung-Geng Lin
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Yu-Chen Lee
- Department of Acupuncture, China Medical University Hospital, Taichung 40447, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| | - Chien-Hsin Tseng
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| | - Dai-Yin Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| | - Chieh-Yun Shih
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan
| | - Iona MacDonald
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| | - Shih-Ya Hung
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Division of Colorectal Surgery, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; Department of Photonics and Communication Engineering, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
56
|
Prevention of Hippocampal Neuronal Damage and Cognitive Function Deficits in Vascular Dementia by Dextromethorphan. Mol Neurobiol 2016; 53:3494-3502. [DOI: 10.1007/s12035-016-9786-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/31/2016] [Indexed: 11/25/2022]
|
57
|
Wang H, Wang Y, Zhao L, Cui Q, Wang Y, Du G. Pinocembrin attenuates MPP + -induced neurotoxicity by the induction of heme oxygenase-1 through ERK1/2 pathway. Neurosci Lett 2016; 612:104-109. [DOI: 10.1016/j.neulet.2015.11.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/27/2015] [Accepted: 11/27/2015] [Indexed: 10/22/2022]
|
58
|
Wu WYY, Kang KH, Chen SLS, Chiu SYH, Yen AMF, Fann JCY, Su CW, Liu HC, Lee CZ, Fu WM, Chen HH, Liou HH. Hepatitis C virus infection: a risk factor for Parkinson's disease. J Viral Hepat 2015; 22:784-91. [PMID: 25608223 DOI: 10.1111/jvh.12392] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/02/2014] [Indexed: 12/12/2022]
Abstract
Recent studies found that hepatitis C virus (HCV) may invade the central nervous system, and both HCV and Parkinson's disease (PD) have in common the overexpression of inflammatory biomarkers. We analysed data from a community-based integrated screening programme based on a total of 62,276 subjects. We used logistic regression models to investigate association between HCV infection and PD. The neurotoxicity of HCV was evaluated in the midbrain neuron-glia coculture system in rats. The cytokine/chemokine array was performed to measure the differences of amounts of cytokines released from midbrain in the presence and absence of HCV. The crude odds ratios (ORs) for having PD were 0.62 [95% confidence interval (CI), 0.48-0.81] and 1.91 (95% CI, 1.48-2.47) for hepatitis B virus (HBV) and HCV. After controlling for potential confounders, the association between HCV and PD remained statistically significant (adjusted OR = 1.39; 95% CI, 1.07-1.80), but not significantly different between HBV and PD. The HCV induced 60% dopaminergic neuron death in the midbrain neuron-glia coculture system in rats, similar to that of 1-methyl-4-phenylpyridinium (MPP(+) ) but not caused by HBV. This link was further supported by the finding that HCV infection may release the inflammatory cytokines, which may play a role in the pathogenesis of PD. In conclusion, our study demonstrated a significantly positive epidemiological association between HCV infection and PD and corroborated the dopaminergic toxicity of HCV similar to that of MPP(+) .
Collapse
Affiliation(s)
- W Y-Y Wu
- Graduate Institute of Epidemiology and Preventive medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - K-H Kang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - S L-S Chen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - S Y-H Chiu
- Department and Graduate Institute of Health Care Management, Chang Gung University, Tao-Yuan, Taiwan
| | - A M-F Yen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - J C-Y Fann
- Department of Health Industry Management, School of Health Care Management, Kainan University, Taoyuan, Taiwan
| | - C-W Su
- Graduate Institute of Epidemiology and Preventive medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - H-C Liu
- Graduate Institute of Epidemiology and Preventive medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - C-Z Lee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - W-M Fu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - H-H Chen
- Graduate Institute of Epidemiology and Preventive medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - H-H Liou
- Department of Neurology and Pharmacology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
59
|
Xu X, Song N, Wang R, Jiang H, Xie J. Preferential Heme Oxygenase-1 Activation in Striatal Astrocytes Antagonizes Dopaminergic Neuron Degeneration in MPTP-Intoxicated Mice. Mol Neurobiol 2015; 53:5056-65. [PMID: 26385576 DOI: 10.1007/s12035-015-9437-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/10/2015] [Indexed: 01/01/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) accompanied by increased oxidative damage. Astrocytes, which are the most abundant glial cell types in the brain, possess higher antioxidant potential partially due to preferentially activated nuclear factor E2-related factor 2 genes. Heme oxygenase isoform 1 (HO-1) is crucial for the response to oxidative stress via the catabolism of heme to carbon monoxide, bilirubin, and iron. In the present study, we aimed to investigate astroglial expression of HO-1 in the SNpc, especially in the striatum of a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mouse model of PD, and to investigate the neuroprotective effects of intraventricularly administrated HO-1 activator cobalt protoporphyrin IX (CoPPIX). The results showed that HO-1 was faintly distributed in neurons but not astrocytes in the normal SNpc and striatum. MPTP triggered a robust HO-1 response in the astrocytes of the striatum after 1-day treatment, but the HO-1 levels declined dramatically at day 3 and were completely undetectable at day 5. Intraventricular administration of CoPPIX for 8 days could preferentially activate HO-1 in astrocytes in the striatum but not SNpc. The content of striatal dopamine and its derivatives was restored in the subacute MPTP models. CoPPIX also increased the number of dopaminergic neurons and the tyrosine hydroxylase levels in the SNpc. These results suggest that inadequate HO-1 in striatal astrocytes might contribute to the limited antioxidant defense and dopaminergic neuron degeneration in PD, and preferential HO-1 activation in striatal astrocytes might be neuroprotective. The study thus sheds light on the targeting of HO-1 in striatal astrocytes for PD therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Ning Song
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Ranran Wang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Hong Jiang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Junxia Xie
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China. .,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China.
| |
Collapse
|
60
|
Aguiar AS, Duzzioni M, Remor AP, Tristão FSM, Matheus FC, Raisman-Vozari R, Latini A, Prediger RD. Moderate-Intensity Physical Exercise Protects Against Experimental 6-Hydroxydopamine-Induced Hemiparkinsonism Through Nrf2-Antioxidant Response Element Pathway. Neurochem Res 2015; 41:64-72. [PMID: 26323504 DOI: 10.1007/s11064-015-1709-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/12/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Exercise improves the motor symptoms of patients with Parkinson disease in a palliative manner. Existing evidence demonstrates that exercise induces neuroprotection based on the neurotrophic properties. We investigated the effect of exercise on mitochondrial physiology and oxidative stress in an animal model of hemiparkinsonism. METHODS C57BL/6 mice completed a 6-week exercise program on a treadmill. We injected 6-hydroxydopamine (6-OHDA; 4 μg/2 μl) into the midstriatum. The animals progressively developed bradykinesia and R(-)-apomorphine-induced rotations that were attenuated by exercise. Transcriptional activation of protective genes is mediated by the antioxidant response element (ARE). Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) binds to ARE. We investigated the Nrf2-ARE pathway in the striatum of animals. RESULTS Exercise protected 6-OHDA-induced loss of tyrosine hydroxylase immunolabeling and activated the Nrf2-ARE pathway in the nigrostriatal pathway. Exercise stimulated mitochondrial biogenesis in the striatum of animals that was more resistant to oxidant 6-OHDA and nitric oxide donor (±)-S-nitroso-N-acetylpenicillamine. CONCLUSIONS In mice, exercise activated Nrf2-ARE signaling in the nigrostriatal pathway that was protective against the development of hemiparkinsonism.
Collapse
Affiliation(s)
- Aderbal Silva Aguiar
- Laboratório Experimental de Doenças Neurodegenerativas (LEXDON), Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil. .,Laboratório de Bioenergética e Estresse Oxidativo (LABOX), Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil. .,Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
| | - Marcelo Duzzioni
- Laboratório Experimental de Doenças Neurodegenerativas (LEXDON), Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil
| | - Aline Pertile Remor
- Laboratório de Bioenergética e Estresse Oxidativo (LABOX), Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil
| | - Fabrine Sales Massafera Tristão
- INSERM UMR 975 (ex U679), CNRS UMR 7225, Hôpital de la Salpêtrière-Bâtiment, ICM (Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, CRICM), Thérapeutique Expérimentale de la Neurodégénérescence, Université Pierre et Marie Curie (UPMC), 47 Boulevard de l'Hôpital, 75651, Paris, France
| | - Filipe C Matheus
- Laboratório Experimental de Doenças Neurodegenerativas (LEXDON), Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil
| | - Rita Raisman-Vozari
- INSERM UMR 975 (ex U679), CNRS UMR 7225, Hôpital de la Salpêtrière-Bâtiment, ICM (Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, CRICM), Thérapeutique Expérimentale de la Neurodégénérescence, Université Pierre et Marie Curie (UPMC), 47 Boulevard de l'Hôpital, 75651, Paris, France
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo (LABOX), Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil
| | - Rui Daniel Prediger
- Laboratório Experimental de Doenças Neurodegenerativas (LEXDON), Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Brazil
| |
Collapse
|
61
|
Oliveira SR, Vieira HLA, Duarte CB. Effect of carbon monoxide on gene expression in cerebrocortical astrocytes: Validation of reference genes for quantitative real-time PCR. Nitric Oxide 2015. [PMID: 26196856 DOI: 10.1016/j.niox.2015.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) is a widely used technique to characterize changes in gene expression in complex cellular and tissue processes, such as cytoprotection or inflammation. The accurate assessment of changes in gene expression depends on the selection of adequate internal reference gene(s). Carbon monoxide (CO) affects several metabolic pathways and de novo protein synthesis is crucial in the cellular responses to this gasotransmitter. Herein a selection of commonly used reference genes was analyzed to identify the most suitable internal control genes to evaluate the effect of CO on gene expression in cultured cerebrocortical astrocytes. The cells were exposed to CO by treatment with CORM-A1 (CO releasing molecule A1) and four different algorithms (geNorm, NormFinder, Delta Ct and BestKeeper) were applied to evaluate the stability of eight putative reference genes. Our results indicate that Gapdh (glyceraldehyde-3-phosphate dehydrogenase) together with Ppia (peptidylpropyl isomerase A) is the most suitable gene pair for normalization of qRT-PCR results under the experimental conditions used. Pgk1 (phosphoglycerate kinase 1), Hprt1 (hypoxanthine guanine phosphoribosyl transferase I), Sdha (Succinate Dehydrogenase Complex, Subunit A), Tbp (TATA box binding protein), Actg1 (actin gamma 1) and Rn18s (18S rRNA) genes presented less stable expression profiles in cultured cortical astrocytes exposed to CORM-A1 for up to 60 min. For validation, we analyzed the effect of CO on the expression of Bdnf and bcl-2. Different results were obtained, depending on the reference genes used. A significant increase in the expression of both genes was found when the results were normalized with Gapdh and Ppia, in contrast with the results obtained when the other genes were used as reference. These findings highlight the need for a proper and accurate selection of the reference genes used in the quantification of qRT-PCR results in studies on the effect of CO in gene expression.
Collapse
Affiliation(s)
- Sara R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
62
|
Youn JK, Kim DW, Kim ST, Park SY, Yeo EJ, Choi YJ, Lee HR, Kim DS, Cho SW, Han KH, Park J, Eum WS, Hwang HS, Choi SY. PEP-1-HO-1 prevents MPTP-induced degeneration of dopaminergic neurons in a Parkinson's disease mouse model. BMB Rep 2015; 47:569-74. [PMID: 24499676 PMCID: PMC4261515 DOI: 10.5483/bmbrep.2014.47.10.286] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Indexed: 11/20/2022] Open
Abstract
Heme oxygenase-1 (HO-1) degrades heme to carbon dioxide, biliverdin, and Fe2+, which play important roles in various biochemical processes. In this study, we examined the protective function of HO-1 against oxidative stress in SH-SY5Y cells and in a Parkinson's disease mouse model. Western blot and fluorescence microscopy analysis demonstrated that PEP-1-HO-1, fused with a PEP-1 peptide can cross the cellular membranes of human neuroblastoma SH-SY5Y cells. In addition, the transduced PEP-1-HO-1 inhibited generation of reactive oxygen species (ROS) and cell death caused by 1-methyl-4-phenylpyridinium ion (MPP+). In contrast, HO-1, which has no ability to transduce into SH-SY5Y cells, failed to reduce MPP+-induced cellular toxicity and ROS production. Furthermore, intraperitoneal injected PEP-1-HO-1 crossed the blood-brain barrier in mouse brains. In a PD mouse model, PEP-1-HO-1 significantly protected against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced toxicity and dopaminergic neuronal death. Therefore, PEP-1-HO-1 could be a useful agent in treating oxidative stress induced ailments including PD.
Collapse
Affiliation(s)
- Jong Kyu Youn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Gangneung 210-702, Korea
| | - Seung Tae Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Sung Yeon Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Hae-Ran Lee
- Department of Pediatrics, Hallym University Medical Center, Pyungchon 431-796, Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 330-090, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Hyun Sook Hwang
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 200-702, Korea
| |
Collapse
|
63
|
Aguirre-Vidal Y, Montes S, Tristan-López L, Anaya-Ramos L, Teiber J, Ríos C, Baron-Flores V, Monroy-Noyola A. The neuroprotective effect of lovastatin on MPP(+)-induced neurotoxicity is not mediated by PON2. Neurotoxicology 2015; 48:166-70. [PMID: 25842176 DOI: 10.1016/j.neuro.2015.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/19/2015] [Accepted: 03/24/2015] [Indexed: 01/09/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of the pigmented dopaminergic neurons in the substantia nigra pars compacta with subsequent striatal dopamine (DA) deficiency and increased lipid peroxidation. The etiology of the disease is still unclear and it is thought that PD may be caused by a combination of genetic and environmental factors. In the search of new pharmacological options, statins have been recognized for their potential application to treat PD, due to their antioxidant effect. The aim of this work is to contribute in the characterization of the neuroprotective effect of lovastatin in a model of PD induced by 1-methyl-4-phenylpyridinium (MPP(+)). Male Wistar rats (200-250 g) were randomly allocated into 4 groups and administered for 7 days with different pharmacological treatments. Lovastatin administration (5 mg/kg) diminished 40% of the apomorphine-induced circling behavior, prevented the striatal DA depletion and lipid peroxides formation by MPP(+) intrastriatal injection, as compared to the group of animals treated only with MPP(+). Lovastatin produced no change in paraoxonase-2 (PON2) activity. It is evident that lovastatin conferred neuroprotection against MPP(+)-induced protection but this effect was not associated with the induction of PON2 in the rat striatum.
Collapse
Affiliation(s)
- Yoshajandith Aguirre-Vidal
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, M.V.S., D.F., Mexico
| | - Luis Tristan-López
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, M.V.S., D.F., Mexico
| | - Laura Anaya-Ramos
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - John Teiber
- Division of Epidemiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, M.V.S., D.F., Mexico; Laboratorio de Neurofarmacologia Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, D.F., Mexico
| | - Verónica Baron-Flores
- Laboratorio de Neurofarmacologia Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, D.F., Mexico
| | - Antonio Monroy-Noyola
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
64
|
Kumar V, Singh BK, Chauhan AK, Singh D, Patel DK, Singh C. Minocycline Rescues from Zinc-Induced Nigrostriatal Dopaminergic Neurodegeneration: Biochemical and Molecular Interventions. Mol Neurobiol 2015; 53:2761-2777. [PMID: 25764516 DOI: 10.1007/s12035-015-9137-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/22/2015] [Indexed: 12/29/2022]
Abstract
Accumulation of zinc (Zn) in dopaminergic neurons is implicated in Parkinson's disease (PD), and microglial activation plays a critical role in toxin-induced Parkinsonism. Oxidative stress is accused in Zn-induced dopaminergic neurodegeneration; however, its connection with microglial activation is still not known. This study was undertaken to elucidate the role and underlying mechanism of microglial activation in Zn-induced nigrostriatal dopaminergic neurodegeneration. Male Wistar rats were treated intraperitoneally with/without zinc sulphate (20 mg/kg) in the presence/absence of minocycline (30 mg/kg), a microglial activation inhibitor, for 2-12 weeks. While neurobehavioral and biochemical indexes of PD and number of dopaminergic neurons were reduced, the number of microglial cells was increased in the substantia nigra of the Zn-exposed animals. Similarly, Zn elevated lipid peroxidation (LPO) and activities of superoxide dismutase (SOD) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase; however, catalase activity was reduced. Besides, Zn increased an association of NADPH oxidase subunit p67(phox) with membrane, cytochrome c release from the mitochondria and cleavage of pro-caspase 3. Zn attenuated the expression of tyrosine hydroxylase (TH) and vesicular monoamine transporter-2 (VMAT-2) while augmented the expression of dopamine transporter (DAT) and heme oxygenase-1 (HO-1). Minocycline alleviated Zn-induced behavioural impairments, loss of TH-positive neurons, activated microglial cells and biochemical indexes and modulated the expression of studied genes/proteins towards normalcy. The results demonstrate that minocycline reduces the number of activated microglial cells and oxidative stress, which rescue from Zn-induced changes in the expression of monoamine transporter and nigrostriatal dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Vinod Kumar
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, New Delhi, 110 025, India
| | - Brajesh Kumar Singh
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Amit Kumar Chauhan
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, New Delhi, 110 025, India
| | - Deepali Singh
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, New Delhi, 110 025, India
| | - Devendra Kumar Patel
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Chetna Singh
- CSIR-Indian Institute of Toxicology Research, Post Box No. 80, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research, New Delhi, 110 025, India.
| |
Collapse
|
65
|
Moccia M, Picillo M, Erro R, Longo K, Amboni M, Santangelo G, Palladino R, Allocca R, Caporale O, Triassi M, Pellecchia MT, Barone P, Vitale C. Increased bilirubin levels in de novo Parkinson's disease. Eur J Neurol 2015; 22:954-9. [PMID: 25740378 DOI: 10.1111/ene.12688] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 12/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Oxidative stress is a central pathogenic mechanism of Parkinson's disease (PD), and the heme oxygenase (HO) bilirubin pathway is one of the main mammalian antioxidative defences. Indeed, there is growing evidence of HO-bilirubin upregulation from early phases of PD. Our aim was to investigate bilirubin as a possible biomarker of PD diagnosis and progression. METHODS A cross-sectional case-control study was performed to evaluate differences in bilirubin levels between newly diagnosed, drug-naïve PD subjects and controls. Afterwards, PD subjects were included in a 2-year longitudinal study to evaluate disease progression in relation to baseline bilirubin levels. RESULTS Seventy-five de novo PD subjects were selected and matched with 75 controls by propensity score. Analysis of variance showed higher bilirubin levels in PD patients compared with controls (P < 0.001). Linear regression analysis failed to show a relationship between bilirubin and Unified Parkinson's Disease Rating Scale (UPDRS) part III (P = 0.283) at baseline evaluation. At 2-year follow-up, indirect relationships between bilirubin levels and UPDRS part III (P = 0.028) and between bilirubin levels and levodopa-equivalent daily dosage (P = 0.012) were found. CONCLUSIONS Parkinson's disease subjects showed higher levels of bilirubin compared with controls. Bilirubin increase might be due to HO overexpression as a compensatory response to oxidative stress occurring from early stages of PD.
Collapse
Affiliation(s)
- M Moccia
- Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Queiroga CSF, Vercelli A, Vieira HLA. Carbon monoxide and the CNS: challenges and achievements. Br J Pharmacol 2015; 172:1533-45. [PMID: 24758548 PMCID: PMC4369262 DOI: 10.1111/bph.12729] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/20/2014] [Accepted: 03/26/2014] [Indexed: 01/12/2023] Open
Abstract
Haem oxygenase (HO) and its product carbon monoxide (CO) are associated with cytoprotection and maintenance of homeostasis in several different organs and tissues. This review focuses upon the role of exogenous and endogenous CO (via HO activity and expression) in various CNS pathologies, based upon data from experimental models, as well as from some clinical data on human patients. The pathophysiological conditions reviewed are cerebral ischaemia, chronic neurodegenerative diseases (Alzheimer's and Parkinson's diseases), multiple sclerosis and pain. Among these pathophysiological conditions, a variety of cellular mechanisms and processes are considered, namely cytoprotection, cell death, inflammation, cell metabolism, cellular redox responses and vasomodulation, as well as the different targeted neural cells. Finally, novel potential methods and strategies for delivering exogenous CO as a drug are discussed, particularly approaches based upon CO-releasing molecules, their limitations and challenges. The diagnostic and prognostic value of HO expression in clinical use for brain pathologies is also addressed.
Collapse
Affiliation(s)
- Cláudia S F Queiroga
- Chronic Diseases Research Center (CEDOC), Faculdade de Ciências Médicas, Universidade Nova de LisboaLisbon, Portugal
| | - Alessandro Vercelli
- Department of Neuroscience, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of TurinTurin, Italy
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC), Faculdade de Ciências Médicas, Universidade Nova de LisboaLisbon, Portugal
- Instituto de Biologia Experimental e Tecnológica (IBET)Oeiras, Portugal
| |
Collapse
|
67
|
Hung SY, Huang WP, Liou HC, Fu WM. LC3 overexpression reduces Aβ neurotoxicity through increasing α7nAchR expression and autophagic activity in neurons and mice. Neuropharmacology 2015; 93:243-51. [PMID: 25686800 DOI: 10.1016/j.neuropharm.2015.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 01/26/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Autophagy is an intracellular degradation pathway with dynamic interactions for eliminating damaged organelles and protein aggregates by lysosomal digestion. The EGFP-conjugated microtubule-associated protein 1 light chain 3 (EGFP-LC3) serves to monitor autophagic process. Extracellular β-amyloid peptide accumulation is reported as a major cause in Alzheimer's disease (AD) pathogenesis; large numbers of autophagic vacuoles accumulate in patients' brains. We previously demonstrated that extracellular Aβ (eAβ) induces strong autophagic response and α7nAChR acts as a carrier to bind with eAβ; which further inhibits Aβ-induced neurotoxicity via autophagic degradation. In the present study, we overexpressed LC3 in both neuroblastoma cells (SH-SY5Y/pEGFP-LC3) and mice (TgEGFP-LC3) to assess the effect of LC3 overexpression on Aβ neurotoxicity. SH-SY5Y/pEGFP-LC3 cells and primary cortical neuron cultures derived from E17 (embryonic day 17) TgEGFP-LC3 mice showed not only better resistance against Aβ neurotoxicity but also higher α7nAChR expression and autophagic activity than control. Administration of α-bungarotoxin (α-BTX) to block α7nAChR antagonized the neuroprotective action of SH-SY5Y/pECGF-LC3 cells, suggesting that eAβ binding with α7nAChR is an important step in Aβ detoxification. LC3 overexpression thus exerts neuroprotection through increasing α7nAChR expression for eAβ binding and further enhancing autophagic activity for Aβ clearance in vitro and in vivo.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Wei-Pang Huang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Houng-Chi Liou
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
68
|
Ai Q, Sun G, Luo Y, Dong X, Hu R, Meng X, Sun X. Ginsenoside Rb1 prevents hypoxia-reoxygenation-induced apoptosis in H9c2 cardiomyocytes via an estrogen receptor-dependent crosstalk among the Akt, JNK, and ERK 1/2 pathways using a label-free quantitative proteomics analysis. RSC Adv 2015. [DOI: 10.1039/c5ra02432c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rb1 prevents H/R-induced apoptosis of H9c2 cells via an estrogen receptor-dependent crosstalk among the Akt, JNK, and ERK 1/2 pathways.
Collapse
Affiliation(s)
- Qidi Ai
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| | - Guibo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| | - Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| | - Xi Dong
- Wenzhou Medical University
- Wenzhou
- P. R. China
| | - Ruifeng Hu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| | - Xiangbao Meng
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine
- Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
| |
Collapse
|
69
|
Khan A, Jamwal S, Bijjem KRV, Prakash A, Kumar P. Neuroprotective effect of hemeoxygenase-1/glycogen synthase kinase-3β modulators in 3-nitropropionic acid-induced neurotoxicity in rats. Neuroscience 2014; 287:66-77. [PMID: 25536048 DOI: 10.1016/j.neuroscience.2014.12.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 01/27/2023]
Abstract
The present study has been designed to explore the possible interaction between hemeoxygenase-1 (HO-1) and glycogen synthase kinase-3β (GSK-3β) pathway in 3-nitropropionic acid (3-NP)-induced neurotoxicity in rats. 3-NP produces neurotoxicity by inhibition of the mitochondrial complex II (enzyme succinate dehydrogenase) and by sensitizing the N-methyl-D-aspartate receptor. Recent studies have reported the therapeutic potential of HO-1/GSK-3β modulators in different neurodegenerative disorders. However, their exact role is yet to be explored. The present study is an attempt to investigate the effect of pharmacological modulation of HO-1/GSK-3β pathway against 3-NP-induced behavioral, biochemical and molecular alterations in rat. Behavioral observation, oxidative stress, pro-inflammatory [tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β)], HO-1 and GSK-3β activity were evaluated post 3-NP treatment. Findings of the present study demonstrate a significant alteration in the locomotor activity, motor coordination, oxidative burden (increased lipid peroxidation, nitrite concentration and decreased endogenous antioxidants), pro-inflammatory mediators [TNF-α, IL-1β], HO-1 and GSK-3β activity in 3-NP-treated animals. Further, administration of hemin (10- and 30-mg/kg; i.p.) and lithium chloride (LiCl) (25- and 50-mg/kg; i.p.) prevented the alteration in body weight, motor impairments, oxidative stress and cellular markers. In addition, combined administration of hemin (10-mg/kg) and LiCl (25-mg/kg) showed synergistic effect on 3-NP-treated rats. Pretreatment with Tin (IV) protoporphyrin (40 μM/kg), HO-1 inhibitor reversed the beneficial effect of LiCl and hemin. Outcomes of the present study suggest that HO-1 and GSK-3β enzymes are involved in the pathophysiology of HD. The modulators of both the pathways might be used as adjuvants or prophylactic therapy for the treatment of HD-like symptoms.
Collapse
Affiliation(s)
- A Khan
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - S Jamwal
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India; Research Scholar, Punjab Technical University, Jalandhar, India
| | - K R V Bijjem
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - A Prakash
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - P Kumar
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India.
| |
Collapse
|
70
|
Tsou YH, Shih CT, Ching CH, Huang JY, Jen CJ, Yu L, Kuo YM, Wu FS, Chuang JI. Treadmill exercise activates Nrf2 antioxidant system to protect the nigrostriatal dopaminergic neurons from MPP+ toxicity. Exp Neurol 2014; 263:50-62. [PMID: 25286336 DOI: 10.1016/j.expneurol.2014.09.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/22/2014] [Accepted: 09/27/2014] [Indexed: 10/24/2022]
Abstract
Exercise induces oxidative stress, which may activate adaptive antioxidant responses. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays an important role in the defense of oxidative stress by regulating the expression of antioxidant enzymes, gamma-glutamylcysteine ligase (γGCL) and heme oxygenase-1 (HO-1). We investigated whether treadmill exercise protects dopaminergic neurons by regulating the Nrf2 antioxidant system in a 1-methyl-4-phenylpyridine (MPP(+))-induced parkinsonian rat model. We found that MPP(+) induced early decreases in total glutathione level and Nrf2/γGCLC (catalytic subunit of γGCL) expression, but late upregulation of HO-1 expression in association with loss of nigral dopaminergic neurons and downregulation of tyrosine hydroxylase and dopamine transporter expression in the striatum. Treadmill exercise for 4weeks induced upregulation of Nrf2 and γGCLC expression, and also prevented the MPP(+)-induced downregulation of Nrf2/γGCLC/glutathione, HO-1 upregulation, and nigrostriatal dopaminergic neurodegeneration. Moreover, the protective effect of exercise was blocked by the knockdown of Nrf2 using a lentivirus-carried shNrf2 delivery system. These results demonstrate an essential role of Nrf2 in the exercise-mediated protective effect that exercise enhances the nigrostriatal Nrf2 antioxidant defense capacity to protect dopaminergic neurons against the MPP(+)-induced toxicity.
Collapse
Affiliation(s)
- Yi-Hsien Tsou
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Ching-Ting Shih
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Cheng-Hsin Ching
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jui-Yen Huang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Chauying J Jen
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Lung Yu
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Fong-Sen Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jih-Ing Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
71
|
Cheng B, Guo Y, Li C, Ji B, Pan Y, Chen J, Bai B. Edaravone protected PC12 cells against MPP(+)-cytoxicity via inhibiting oxidative stress and up-regulating heme oxygenase-1 expression. J Neurol Sci 2014; 343:115-9. [PMID: 24930399 DOI: 10.1016/j.jns.2014.05.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/20/2014] [Accepted: 05/22/2014] [Indexed: 01/30/2023]
Abstract
Oxidative stress is involved in the pathogenesis of Parkinson's disease (PD). Edaravone has been shown to have a neuroprotective effect. In the present work, we investigated the effect of edaravone on 1-methyl-4-phenylpyridinium (MPP(+))-treated PC12 cells. Edaravone inhibited the decrease of cell viability and apoptosis induced by MPP(+) in PC12 cells. In addition, edaravone alleviated intracellular reactive oxygen species (ROS) production. MPP(+) induced heme oxygenase-1 (HO-1) expression, which was further enhanced by edaravone. The inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of edaravone. So edaravone protected PC12 cells against MPP(+)-cytoxicity via inhibiting oxidative stress and up-regulating HO-1 expression. The data showed that edaravone was neuroprotective and could be potentially therapeutics for PD in future.
Collapse
Affiliation(s)
- Baohua Cheng
- Neurobiology Institute, Jining Medical University, Jining 272067, PR China
| | - Yunliang Guo
- Shandong University School of Medicine, Jinan 250012, PR China
| | - Chuangang Li
- The Second Hospital of Shandong University, Jinan 250033, PR China
| | - Bingyuan Ji
- Neurobiology Institute, Jining Medical University, Jining 272067, PR China
| | - Yanyou Pan
- Neurobiology Institute, Jining Medical University, Jining 272067, PR China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining 272067, PR China.
| | - Bo Bai
- Neurobiology Institute, Jining Medical University, Jining 272067, PR China.
| |
Collapse
|
72
|
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: 98] [Impact Index Per Article: 8.9] [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
|
73
|
Jung UJ, Jeon MT, Choi MS, Kim SR. Silibinin attenuates MPP⁺-induced neurotoxicity in the substantia nigra in vivo. J Med Food 2014; 17:599-605. [PMID: 24660866 DOI: 10.1089/jmf.2013.2926] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is characterized by degeneration of the nigrostriatal dopaminergic (DA) pathway. The cause of neuronal death in PD is largely unknown, but it is becoming clear that inflammation plays a significant role in the pathophysiology of PD. Silibinin is a major flavonoid in milk thistle which has an anti-inflammatory activity. We investigated whether silibinin could have neuroprotective effects on DA neurons in the 1-methyl-4-phenylpyridinium ion (MPP(+))-treated animal model of PD in vivo. To address this question, animals received intraperitoneal (i.p.) injections 10, 50, or 100 mg/kg of silibinin, starting 1 day before MPP(+) injection and continued daily until 6 days post-lesion for tyrosine hydroxylase (TH) staining, or until 1 hour prior to the MPP(+) injection to examine the expression levels of inflammatory proteins. Finally, their brains were harvested at the indicated time points for the analyses. Silibinin treatment with 10 mg/kg had no significantly neuroprotective effects in the substantia nigra (SN). However, 50 and 100 mg/kg of silibinin ameliorated the MPP(+)-induced neurotoxicity in the SN in a dose-dependent manner, and the increased levels of inflammatory molecules such as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β) and inducible nitric oxide synthase (iNOS) by MPP(+) treatment were attenuated by treatment with 100 mg/kg of silibinin. These results indicate that silibinin could be a useful and beneficial natural product offering promise for the prevention of DA neuronal degeneration involved in PD.
Collapse
Affiliation(s)
- Un Ju Jung
- 1 Department of Food Science and Nutrition, Kyungpook National University , Daegu, Korea
| | | | | | | |
Collapse
|
74
|
Yang SM, Ka SM, Wu HL, Yeh YC, Kuo CH, Hua KF, Shi GY, Hung YJ, Hsiao FC, Yang SS, Shieh YS, Lin SH, Wei CW, Lee JS, Yang CY, Chen A. Thrombomodulin domain 1 ameliorates diabetic nephropathy in mice via anti-NF-κB/NLRP3 inflammasome-mediated inflammation, enhancement of NRF2 antioxidant activity and inhibition of apoptosis. Diabetologia 2014; 57:424-34. [PMID: 24317792 DOI: 10.1007/s00125-013-3115-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/29/2013] [Indexed: 01/30/2023]
Abstract
AIMS/HYPOTHESIS Chronic inflammatory processes have been increasingly shown to be involved in the pathogenesis of diabetes and diabetic nephropathy. Recently, we demonstrated that a lectin-like domain of thrombomodulin (THBD), which is known as THBD domain 1 (THBDD1) and which acts independently of protein C activation, neutralised an inflammatory response in a mouse model of sepsis. Here, therapeutic effects of gene therapy with adeno-associated virus (AAV)-carried THBDD1 (AAV-THBDD1) were tested in a mouse model of type 2 diabetic nephropathy. METHODS To assess the therapeutic potential of THBDD1 and the mechanisms involved, we delivered AAV-THBDD1 (10(11) genome copies) into db/db mice and tested the effects of recombinant THBDD1 on conditionally immortalised podocytes. RESULTS A single dose of AAV-THBDD1 improved albuminuria, renal interstitial inflammation and glomerular sclerosis, as well as renal function in db/db mice. These effects were closely associated with: (1) inhibited activation of the nuclear factor κB (NF-κB) pathway and the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome; (2) promotion of nuclear factor (erythroid-derived 2)-like 2 (NRF2) nuclear translocation; and (3) suppression of mitochondria-derived apoptosis in the kidney of treated mice. CONCLUSIONS/INTERPRETATION AAV-THBDD1 gene therapy resulted in improvements in a model of diabetic nephropathy by suppressing the NF-κB-NLRP3 inflammasome-mediated inflammatory process, enhancing the NRF2 antioxidant pathway and inhibiting apoptosis in the kidney.
Collapse
Affiliation(s)
- Shun-Min Yang
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gung Road, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Resveratrol partially prevents rotenone-induced neurotoxicity in dopaminergic SH-SY5Y cells through induction of heme oxygenase-1 dependent autophagy. Int J Mol Sci 2014; 15:1625-46. [PMID: 24451142 PMCID: PMC3907890 DOI: 10.3390/ijms15011625] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/08/2014] [Accepted: 01/14/2014] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder characterized by a progressive loss of dopaminergic neurons. Mitochondrial dysfunction, oxidative stress or protein misfolding and aggregation may underlie this process. Autophagy is an intracellular catabolic mechanism responsible for protein degradation and recycling of damaged proteins and cytoplasmic organelles. Autophagic dysfunction may hasten the progression of neuronal degeneration. In this study, resveratrol promoted autophagic flux and protected dopaminergic neurons against rotenone-induced apoptosis. In an in vivo PD model, rotenone induced loss of dopaminergic neurons, increased oxidation of mitochondrial proteins and promoted autophagic vesicle development in brain tissue. The natural phytoalexin resveratrol prevented rotenone-induced neuronal apoptosis in vitro, and this pro-survival effect was abolished by an autophagic inhibitor. Although both rotenone and resveratrol promoted LC3-II accumulation, autophagic flux was inhibited by rotenone and augmented by resveratrol. Further, rotenone reduced heme oxygenase-1 (HO-1) expression, whereas resveratrol increased HO-1 expression. Pharmacological inhibition of HO-1 abolished resveratrol-mediated autophagy and neuroprotection. Notably, the effects of a pharmacological inducer of HO-1 were similar to those of resveratrol, and protected against rotenone-induced cell death in an autophagy-dependent manner, validating the hypothesis of HO-1 dependent autophagy in preventing neuronal death in the in vitro PD model. Collectively, our findings suggest that resveratrol induces HO-1 expression and prevents dopaminergic cell death by regulating autophagic flux; thus protecting against rotenone-induced neuronal apoptosis.
Collapse
|
76
|
Phillipson OT. Management of the aging risk factor for Parkinson's disease. Neurobiol Aging 2013; 35:847-57. [PMID: 24246717 DOI: 10.1016/j.neurobiolaging.2013.10.073] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 01/12/2023]
Abstract
The aging risk factor for Parkinson's disease is described in terms of specific disease markers including mitochondrial and gene dysfunctions relevant to energy metabolism. This review details evidence for the ability of nutritional agents to manage these aging risk factors. The combination of alpha lipoic acid, acetyl-l-carnitine, coenzyme Q10, and melatonin supports energy metabolism via carbohydrate and fatty acid utilization, assists electron transport and adenosine triphosphate synthesis, counters oxidative and nitrosative stress, and raises defenses against protein misfolding, inflammatory stimuli, iron, and other endogenous or xenobiotic toxins. These effects are supported by gene expression via the antioxidant response element (ARE; Keap/Nrf2 pathway), and by peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1 alpha), a transcription coactivator, which regulates gene expression for energy metabolism and mitochondrial biogenesis, and maintains the structural integrity of mitochondria. The effectiveness and synergies of the combination against disease risks are discussed in relation to gene action, dopamine cell loss, and the accumulation and spread of pathology via misfolded alpha-synuclein. In addition there are potential synergies to support a neurorestorative role via glial derived neurotrophic factor expression.
Collapse
Affiliation(s)
- Oliver T Phillipson
- School of Medical Sciences, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
77
|
Dowell JA, Johnson JA. Mechanisms of Nrf2 protection in astrocytes as identified by quantitative proteomics and siRNA screening. PLoS One 2013; 8:e70163. [PMID: 23922950 PMCID: PMC3726381 DOI: 10.1371/journal.pone.0070163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/16/2013] [Indexed: 12/30/2022] Open
Abstract
The Nrf2 (NF-E2 related factor 2)-ARE (antioxidant response element) pathway controls a powerful array of endogenous cellular antioxidant systems and is an important pathway in the detoxification of reactive oxygen species (ROS) in the brain. Using a combination of quantitative proteomics and siRNA screening, we have identified novel protective mechanisms of the Nrf2-ARE pathway against oxidative stress in astrocytes. Studies from our lab and others have shown Nrf2 overexpression protects astrocytes from oxidative stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In this study, we show that induction of Nrf2 reduces levels of reactive oxygen species (ROS) produced by various oxidative stressors and results in robust cytoprotection. To identify the enzymes responsible for these effects, we used stable isotope labeling by amino acids in cell culture (SILAC) and quantitative shotgun proteomics to identify 72 Nrf2-regulated proteins in astrocytes. We hypothesized a subset of these proteins might play a critical role in Nrf2 protection. In order to identify these critical proteins, we used bioinformatics to narrow our target list of proteins and then systematically screened each candidate with siRNA to assess the role of each in Nrf2 protection. We screened each target against H2O2, tert-butyl hydroperoxide, and 4-hydroxynonenal and subsequently identified three enzymes-catalase, prostaglandin reductase-1, and peroxiredoxin-6-that are critical for Nrf2-mediated protection in astrocytes.
Collapse
Affiliation(s)
- James A. Dowell
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jeffrey A. Johnson
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
78
|
Teng YC, Tai YI, Lee YH, Lin AMY. Role of HO-1 in the Arsenite-Induced Neurotoxicity in Primary Cultured Cortical Neurons. Mol Neurobiol 2013; 48:281-7. [DOI: 10.1007/s12035-013-8492-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 06/16/2013] [Indexed: 11/29/2022]
|
79
|
Kang KH, Liou HH, Hour MJ, Liou HC, Fu WM. Protection of dopaminergic neurons by 5-lipoxygenase inhibitor. Neuropharmacology 2013; 73:380-7. [PMID: 23800665 DOI: 10.1016/j.neuropharm.2013.06.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 05/29/2013] [Accepted: 06/10/2013] [Indexed: 02/06/2023]
Abstract
Neuroinflammation and oxidative stress are important factors that induce neurodegeneration in age-related neurological disorders. 5-Lipoxygenase (5-LOX) is the enzyme responsible for catalysing the synthesis of leukotriene or 5-HETE from arachidonic acid. 5-LOX is expressed in the central nervous system and may cause neurodegenerative disease. In this study, we investigated the effect of the pharmacological inhibition of 5-lipoxygenase on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/MPP(+)-induced dopaminergic neuronal death in midbrain neuron-glia co-cultures and in mice. It was found that 5-LOX was over-expressed in astrocytes after the injection of MPTP into C57BL6 mice. MK-886, a specific inhibitor of 5-LOX activating protein (FLAP), significantly increased [(3)H]-dopamine uptake, a functional indicator of the integrity of dopaminergic neurons, in midbrain cultures or the SH-SY5Y human dopaminergic cell line following MPP(+) treatment. In addition, LTB₄, one of 5-LOX's downstream products, was increased in the striatum and substantia nigra following MPTP injection in mice. LTB₄ but not LTD₄ and 5-HETE enhanced MPP(+)-induced neurotoxicity in primary midbrain cultures. MK-886 administration increased the number of tyrosine hydroxylase-positive neurons in the substantia nigra and the dopamine content in the striatum in MPTP-induced parkinsonian mice. Furthermore, the MPTP-induced upregulation of LTB₄ in the striatum and substantia nigra was antagonised by MK-886. These results suggest that 5-LOX inhibitors may be developed as novel neuroprotective agents and LTB₄ may play an important pathological role in Parkinson's disease.
Collapse
Affiliation(s)
- Kai-Hsiang Kang
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
80
|
Decursin Isolated from Angelica gigas Nakai Rescues PC12 Cells from Amyloid β-Protein-Induced Neurotoxicity through Nrf2-Mediated Upregulation of Heme Oxygenase-1: Potential Roles of MAPK. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:467245. [PMID: 23762139 PMCID: PMC3665219 DOI: 10.1155/2013/467245] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/27/2013] [Accepted: 04/07/2013] [Indexed: 01/15/2023]
Abstract
Decursin (D), purified from Angelica gigas Nakai, has been proven to exert neuroprotective property. Previous study revealed that D reduced A β 25 ‒ 35-induced cytotoxicity in PC12 cells. Our study explored the underlying mechanisms by which D mediates its therapeutic effects in vitro. Pretreatment of cells with D diminished intracellular generation of ROS in response to A β 25 ‒ 35. Western blot revealed that D significantly increased the expression and activity of HO-1, which was correlated with its protection against A β 25 ‒ 35-induced injury. Addition of ZnPP, an HO-1 competitive inhibitor, significantly attenuated its protective effect in A β 25 ‒ 35-treated cells, indicating the vital role of HO-1 resistance to oxidative injury. Moreover, D induced Nrf2 nuclear translocation, the upstream of HO-1 expression. While investigating the signaling pathways responsible for HO-1 induction, D activated ERK and dephosphorylated p38 in PC12 cells. Addition of U0126, a selective inhibitor of ERK, blocked D-induced Nrf2 activation and HO-1 induction and meanwhile reversed the protection of D against A β 25 ‒ 35-induced cell death. These findings suggest D augments cellular antioxidant defense capacity through both intrinsic free radical scavenging activity and activation of MAPK signal pathways that leads to Nrf2 activation, and subsequently HO-1 induction, thereby protecting the PC12 cells from A β 25 ‒ 35-induced oxidative cytotoxicity.
Collapse
|
81
|
Chien WL, Lee TR, Hung SY, Kang KH, Wu RM, Lee MJ, Fu WM. Increase of oxidative stress by a novel PINK1 mutation, P209A. Free Radic Biol Med 2013; 58:160-9. [PMID: 23261939 DOI: 10.1016/j.freeradbiomed.2012.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/30/2012] [Accepted: 12/09/2012] [Indexed: 12/30/2022]
Abstract
Mutation in the human PTEN-induced protein kinase 1 (PINK1) gene is responsible for the second most common form of recessive Parkinson disease (PD). We have identified a single heterozygous PINK1 mutation, P209A, from a cohort of 68 patients with early onset PD. From age 31, this patient developed an asymmetric bradykinesia with rigidity that was L-DOPA responsive. An [(18)F]-fluorodopa PET scan showed reduced DOPA uptake in the bilateral basal ganglia. The H2O2-induced cell death, ROS production, and caspase-3 activation in SH-SY5Y cells were enhanced by the transfection of the PINK1 P209A mutant. The heme oxygenase-1 (HO-1) induction in response to H2O2 and MPP(+) treatment was impaired by the overexpression of the PINK1 P209A mutant. In addition, SOD2 induction after TNFα treatment was also inhibited by the PINK1 P209A mutation. Akt and ERK are involved in HO-1 induction after oxidative stress. The phosphorylation of Akt and ERK after exposure to H2O2 or MPP(+) was also inhibited in PINK1 P209A mutant cells compared with empty-vector-transfected cells. These results indicate a novel pathway by which the P209A defect in the PINK1 kinase domain inhibits oxidative stress-induced HO-1 and SOD2 induction, which may accelerate the neurodegeneration in PD with PINK1 defect.
Collapse
Affiliation(s)
- Wei-Lin Chien
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
82
|
Pan LL, Liu XH, Jia YL, Wu D, Xiong QH, Gong QH, Wang Y, Zhu YZ. A novel compound derived from danshensu inhibits apoptosis via upregulation of heme oxygenase-1 expression in SH-SY5Y cells. Biochim Biophys Acta Gen Subj 2013; 1830:2861-71. [DOI: 10.1016/j.bbagen.2013.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/16/2012] [Accepted: 01/08/2013] [Indexed: 01/06/2023]
|
83
|
Akt/Nrf2 Activated Upregulation of Heme Oxygenase-1 Involves in the Role of Rg1 Against Ferrous Iron-Induced Neurotoxicity in SK-N-SH Cells. Neurotox Res 2012. [DOI: 10.1007/s12640-012-9362-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
84
|
Kurauchi Y, Hisatsune A, Isohama Y, Mishima S, Katsuki H. Caffeic acid phenethyl ester protects nigral dopaminergic neurons via dual mechanisms involving haem oxygenase-1 and brain-derived neurotrophic factor. Br J Pharmacol 2012; 166:1151-68. [PMID: 22224485 DOI: 10.1111/j.1476-5381.2012.01833.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Caffeic acid phenethyl ester (CAPE) is a component of honey bee propolis that can induce expression of haem oxygenase-1 (HO-1). Because HO-1 induction has been suggested to protect dopaminergic neurons in the substantia nigra, we examined the effect of CAPE in experimental models of dopaminergic neurodegeneration. EXPERIMENTAL APPROACH Neuroprotective effect of CAPE was investigated in rat organotypic midbrain slice cultures and in vivo, using a mouse model of dopaminergic neurodegeneration induced by intranigral injection of LPS and intrastriatal injection of 6-hydroxydopamine. KEY RESULTS CAPE protected dopaminergic neurons in slice cultures from IFN-γ/LPS-induced injury. The effect of CAPE was inhibited by zinc protoporphyrin IX, an HO-1 inhibitor, and by neutralizing antibody against brain-derived neurotrophic factor (BDNF). A p38 MAPK inhibitor SB203580 prevented activation of NF-E2-related factor 2, attenuated increased expression of HO-1 and BDNF, and blocked the neuroprotective actions of CAPE. In the LPS-injected mouse model, daily intraperitoneal administration of CAPE protected dopaminergic neurons, up-regulated HO-1 and BDNF, and reduced the increase of activated microglia/macrophages. Neuroprotective effects of CAPE against LPS-induced injury was prevented by zinc protoporphyrin IX or anti-BDNF antibody. CAPE protected dopaminergic neurons and alleviated methamphetamine-induced rotational behaviour also in 6-hydroxydopamine hemiparkinsonian mice. CONCLUSION AND IMPLICATIONS CAPE is a novel type of neuroprotective agent whose actions are mediated by both HO-1 and BDNF. These findings may provide novel clues to develop neuroprotective agents for treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Y Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamto, Japan
| | | | | | | | | |
Collapse
|
85
|
Shin JH, Kim SW, Jin Y, Kim ID, Lee JK. Ethyl pyruvate-mediated Nrf2 activation and hemeoxygenase 1 induction in astrocytes confer protective effects via autocrine and paracrine mechanisms. Neurochem Int 2012; 61:89-99. [DOI: 10.1016/j.neuint.2012.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 03/22/2012] [Accepted: 04/05/2012] [Indexed: 12/30/2022]
|
86
|
Resveratrol protects rats from Aβ-induced neurotoxicity by the reduction of iNOS expression and lipid peroxidation. PLoS One 2011; 6:e29102. [PMID: 22220203 PMCID: PMC3248406 DOI: 10.1371/journal.pone.0029102] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 11/21/2011] [Indexed: 01/14/2023] Open
Abstract
Alzheimer disease (AD) is an age-dependent neurodegenerative disease characterized by the formation of β–amyloid (Aβ)-containing senile plaque. The disease could be induced by the administration of Aβ peptide, which was also known to upregulate inducible nitric oxide synthase (iNOS) and stimulate neuronal apoptosis. The present study is aimed to elucidate the cellular effect of resveratrol, a natural phytoestrogen with neuroprotective activities, on Aβ-induced hippocampal neuron loss and memory impairment. On adult Sprague-Dawley rats, we found the injection of Aβ could result in a significant impairment in spatial memory, a marked increase in the cellular level of iNOS and lipid peroxidation, and an apparent decrease in the expression of heme oxygenase-1 (HO-1). By combining the treatment with Aβ, resveratrol was able to confer a significant improvement in spatial memory, and protect animals from Aβ-induced neurotoxicity. These neurological protection effects of resveratrol were associated with a reduction in the cellular levels of iNOS and lipid peroxidation and an increase in the production of HO-1. Moreover, the similar neurological and cellular response were also observed when Aβ treatment was combined with the administration of a NOS inhibitor, N(G)-nitro-L-arginine methyl ester hydrochloride (L-NAME). These findings strongly implicate that iNOS is involved in the Aβ-induced lipid peroxidation and HO-1 downregulation, and resveratrol protects animals from Aβ-induced neurotoxicity by suppressing iNOS production.
Collapse
|
87
|
Wang HQ, Xu YX, Zhu CQ. Upregulation of heme oxygenase-1 by acteoside through ERK and PI3 K/Akt pathway confer neuroprotection against beta-amyloid-induced neurotoxicity. Neurotox Res 2011; 21:368-78. [PMID: 22147269 DOI: 10.1007/s12640-011-9292-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 11/05/2011] [Accepted: 11/23/2011] [Indexed: 12/30/2022]
Abstract
Our previous study has shown that acteoside, an antioxidative phenylethanoid glycoside, protect against beta-amyloid (Aβ)-induced cytotoxicity in vitro. However, the precise protective mechanisms remains unclear. Heme oxygenase-1 (HO-1) is a crucial factor in the response to oxidative injury, protecting neurons against Aβ-induced injury. In the present study we examined to determine whether acteoside upregulates HO-1 expression, and thereby protects PC12 cells against Aβ-induced cell death. It was revealed that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We showed that acteoside increased HO-1 expression in vitro and in vivo. Acteoside treatment resulted in nuclear translocation of the transcription factor NF-E2-related factor 2 (Nrf2). Acteoside activated both ERK and PI3 K/Akt, and treatments with the specific ERK inhibitor PD98059, the PI3 K inhibitor LY294002, and the specific Nrf2 siRNA suppressed the acteoside-induced HO-1 expression. The HO-1 inhibitor ZnPP, PD98059, and LY294002 markedly abolished the neuroprotective effect of acteoside against Aβ-induced neurotoxicity. Taken together, these results demonstrate that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We also showed that acteoside increased HO-1 expression through activation of ERK and PI3 K/Akt signal pathways in vitro. Upregulation of HO-1 by acteoside may involve in the neuroprotection against Aβ-induced neurotoxicity.
Collapse
Affiliation(s)
- Hong-Quan Wang
- Department of Neurology, The Affiliated Hospital of Chifeng University, Chifeng 024000, Inner Mongolia, People's Republic of China
| | | | | |
Collapse
|
88
|
A polymorphism located at an ATG transcription start site of the heme oxygenase-2 gene is associated with classical Parkinson's disease. Pharmacogenet Genomics 2011; 21:565-71. [PMID: 21709601 DOI: 10.1097/fpc.0b013e328348f729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM Oxidative stress and iron deposition is related to Parkinson's disease (PD). Heme oxygenase 2 (HMOX2) catalyzes the cleavage of the heme ring to form biliverdin with release of iron and carbon monoxide. This study aims to analyze variations in the HMOX2 gene in patients with PD. MATERIALS AND METHODS We mapped four single nucleotide polymorphisms (SNPs) and copy number variations of the HMOX2 gene in 691 patients with PD and 747 healthy individuals. RESULTS We identified a highly homogeneous association of the HMOX2 SNP rs2270363 homozygous G/G genotype with patients with classical PD phenotype compared with healthy individuals. We identified three patients with PD and two control individuals with a single copy of the HMOX2 gene. No individuals with zero or more than two gene copies were identified. CONCLUSION We describe for the first time, copy number variations in the HMOX2 gene and an association of the SNP rs2270363 with PD risk.
Collapse
|
89
|
Feng LR, Maguire-Zeiss KA. Dopamine and paraquat enhance α-synuclein-induced alterations in membrane conductance. Neurotox Res 2011; 20:387-401. [PMID: 21735318 PMCID: PMC3175296 DOI: 10.1007/s12640-011-9255-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 06/03/2011] [Accepted: 06/22/2011] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated that α-synuclein overexpression increases the membrane conductance of dopaminergic-like cells. Although α-synuclein is thought to play a central role in the pathogenesis of several neurodegenerative diseases including Parkinson's disease, multiple system atrophy, and diffuse Lewy body disease, the mechanism of action is not completely understood. In this study, we sought to determine whether multiple factors act together with α-synuclein to engender cell vulnerability through an augmentation of membrane conductance. In this article, we employed a cell model that mimics dopaminergic neurons coupled with α-synuclein overexpression and oxidative stressors. We demonstrate an enhancement of α-synuclein-induced toxicity in the presence of combined treatment with dopamine and paraquat, two molecules known to incite oxidative stress. In addition, we show that combined dopamine and paraquat treatment increases the expression of heme oxygenase-1, an antioxidant response protein. Finally, we demonstrate for the first time that combined treatment of dopaminergic cells with paraquat and dopamine enhances α-synuclein-induced leak channel properties resulting in increased membrane conductance. Importantly, these increases are most robust when both paraquat and dopamine are present suggesting the need for multiple oxidative insults to augment α-synuclein-induced disruption of membrane integrity.
Collapse
Affiliation(s)
- Li Rebekah Feng
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC 20057, USA
| | | |
Collapse
|
90
|
Dodd CA, Filipov NM. Manganese potentiates LPS-induced heme-oxygenase 1 in microglia but not dopaminergic cells: role in controlling microglial hydrogen peroxide and inflammatory cytokine output. Neurotoxicology 2011; 32:683-92. [PMID: 21963524 DOI: 10.1016/j.neuro.2011.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 08/03/2011] [Accepted: 09/02/2011] [Indexed: 01/21/2023]
Abstract
Excessive manganese (Mn) exposure increases output of glial-derived inflammatory products, which may indirectly contribute to the neurotoxic effects of this essential metal. In microglia, Mn increases hydrogen peroxide (H(2)O(2)) release and potentiates lipopolysaccharide (LPS)-induced cytokines (TNF-α, IL-6) and nitric oxide (NO). Inducible heme-oxygenase (HO-1) plays a role in the regulation of inflammation and its expression is upregulated in response to oxidative stressors, including metals and LPS. Because Mn can oxidatively affect neurons both directly and indirectly, we investigated the effect of Mn exposure on the induction of HO-1 in resting and LPS-activated microglia (N9) and dopaminergic neurons (N27). In microglia, 24h exposure to Mn (up to 250 μM) had minimal effects on its own, but it markedly potentiated LPS (100 ng/ml)-induced HO-1 protein and mRNA. Inhibition of microglial HO-1 activity with two different inhibitors indicated that HO-1 is a positive regulator of the Mn-potentiated cytokine output and a negative regulator of the Mn-induced H(2)O(2) output. Mn enhancement of LPS-induced HO-1 does not appear to be dependent on H(2)O(2) or NO, as Mn+LPS-induced H(2)O(2) release was not greater than the increase induced by Mn alone and inhibition of iNOS did not change Mn potentiation of HO-1. However, because Mn exposure potentiated the LPS-induced nuclear expression of small Maf proteins, this may be one mechanism Mn uses to affect the expression of HO-1 in activated microglia. Finally, the potentiating effects of Mn on HO-1 appear to be glia-specific for Mn, LPS, or Mn+LPS did not induce HO-1 in N27 neuronal cells.
Collapse
Affiliation(s)
- Celia A Dodd
- Department of Physiology and Pharmacology, 501 D.W. Brooks Drive, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | | |
Collapse
|
91
|
Tung WH, Hsieh HL, Lee IT, Yang CM. Enterovirus 71 induces integrin β1/EGFR-Rac1-dependent oxidative stress in SK-N-SH cells: Role of HO-1/CO in viral replication. J Cell Physiol 2011; 226:3316-29. [DOI: 10.1002/jcp.22677] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
92
|
Singh BK, Kumar A, Ahmad I, Kumar V, Patel DK, Jain SK, Singh C. Oxidative stress in zinc-induced dopaminergic neurodegeneration: Implications of superoxide dismutase and heme oxygenase-1. Free Radic Res 2011; 45:1207-22. [DOI: 10.3109/10715762.2011.607164] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
93
|
Kim S, Shim S, Choi DS, Kim HB, Kim JH, Lee JC, Cho ES, Kwon J. Up-regulation of heme oxygenase-1 as a resistance mechanism against MPP+ induced cytotoxicity in Raw 264.7 macrophages. Mol Cell Toxicol 2011. [DOI: 10.1007/s13273-011-0016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
94
|
Numakawa T, Matsumoto T, Numakawa Y, Richards M, Yamawaki S, Kunugi H. Protective Action of Neurotrophic Factors and Estrogen against Oxidative Stress-Mediated Neurodegeneration. J Toxicol 2011; 2011:405194. [PMID: 21776259 PMCID: PMC3135156 DOI: 10.1155/2011/405194] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/28/2011] [Accepted: 03/29/2011] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress is involved in the pathogenesis of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Low levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important for maintenance of neuronal function, though elevated levels lead to neuronal cell death. A complex series of events including excitotoxicity, Ca(2+) overload, and mitochondrial dysfunction contributes to oxidative stress-mediated neurodegeneration. As expected, many antioxidants like phytochemicals and vitamins are known to reduce oxidative toxicity. Additionally, growing evidence indicates that neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and estrogens significantly prevent neuronal damage caused by oxidative stress. Here, we review and discuss recent studies addressing the protective mechanisms of neurotrophic factors and estrogen within this system.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| | - Tomoya Matsumoto
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yumiko Numakawa
- Peptide-prima Co., Ltd., 1-25-81, Nuyamazu, Kumamoto 861-2102, Japan
| | - Misty Richards
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- The Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | - Shigeto Yamawaki
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
95
|
Chien WL, Lee TR, Hung SY, Kang KH, Lee MJ, Fu WM. Impairment of oxidative stress-induced heme oxygenase-1 expression by the defect of Parkinson-related gene of PINK1. J Neurochem 2011; 117:643-53. [PMID: 21366594 DOI: 10.1111/j.1471-4159.2011.07229.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Mutation in the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1) gene causes an autosomal recessive form of PD. However, the etiology related to PINK1 is still not clear. Here, we examined the effect of PINK1 on heme oxygenase (HO)-1 induction in SH-SY5Y neuronal cells following H(2)O(2) or 1-methyl-4-phenylpyridinium (MPP(+)) treatment. The HO-1 induction in response to H(2)O(2) and MPP(+) treatment was impaired by the expression of recombinant PINK1 G309D mutant. PINK1 G309D mutation increased the apoptosis of SH-SY5Y cells following H(2)O(2) treatment and cell survival was rescued by the over-expression of HO-1 using adenovirus (Ad) infection. In addition, knockdown of tumor necrosis factor receptor-associated protein-1 (TRAP1), which is the substrate of PINK1 kinase, in SH-SY5Y cells also inhibited the expression of HO-1 in response to oxidative stress. The up-regulation of TRAP1 expression following H(2)O(2) treatment was inhibited by the expression of recombinant PINK1 G309D mutant. The H(2)O(2)-induced HO-1 induction was Akt- and ERK-dependent. The phosphorylation of ERK and Akt but not p38 was inhibited in cells expressing the PINK1 G309D mutant and knockdown of TRAP1. These results indicate a novel pathway by which the defect of PINK1 inhibits the oxidative stress-induced HO-1 production. Impairment of HO-1 production following oxidative stress may accelerate the dopaminergic neurodegeneration in Parkinson patients with PINK1 defect.
Collapse
Affiliation(s)
- Wei-Lin Chien
- Department of Pharmacology, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
96
|
van Horssen J, Witte ME, Schreibelt G, de Vries HE. Radical changes in multiple sclerosis pathogenesis. Biochim Biophys Acta Mol Basis Dis 2011; 1812:141-50. [DOI: 10.1016/j.bbadis.2010.06.011] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 06/08/2010] [Accepted: 06/16/2010] [Indexed: 12/20/2022]
|
97
|
Quesada A, Ogi J, Schultz J, Handforth A. C-terminal mechano-growth factor induces heme oxygenase-1-mediated neuroprotection of SH-SY5Y cells via the protein kinase Cϵ/Nrf2 pathway. J Neurosci Res 2011; 89:394-405. [DOI: 10.1002/jnr.22543] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/28/2010] [Accepted: 10/08/2010] [Indexed: 11/10/2022]
|
98
|
Guan L, Zhang YL, Wen T, Wang XF, Zhu MX, Zhao JY. Dynamic changes of heme oxygenase-1 in the hippocampus of rats after acute carbon monoxide poisoning. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2011; 60:165-172. [PMID: 20422170 DOI: 10.1007/s00244-010-9524-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 04/05/2010] [Indexed: 05/29/2023]
Abstract
Heme oxygenase-1 (HO-1), an inducible enzyme, degrades heme to carbon monoxide (CO), iron, and bilirubin. We have investigated the relationship among HO-1 protein expression, HO activity, and CO concentrations in the hippocampus of CO-exposed rats. Western blotting and immunohistochemistry revealed that the enzyme is predominantly localized in hippocampal CA1 and CA3 pyramidal cells and in granule cells of the dentate gyrus. HO enzyme activity was reduced immediately following CO exposure, while expression of HO-1 protein was consistently upregulated in a time-dependent manner. Local CO concentrations in hippocampus rose immediately following exposure, but the elevation was maintained for ~20 h despite the decline in blood carboxyhemoglobin levels toward baseline. We suggest that CO initially inhibits HO enzyme activity, whereas at later time points the inhibition is released and local CO generation is maintained by the activity of the endogenous HO enzyme. And the noninducible form of heme oxygenase, HO-2, was not altered following CO administration. Together these results indicate that the HO/CO pathway in the rat hippocampus is induced by acute CO exposure; local CO production may play a regulatory role in brain injury following CO poisoning.
Collapse
Affiliation(s)
- Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
99
|
Maternal separation affects the number, proliferation and apoptosis of glia cells in the substantia nigra and ventral tegmental area of juvenile rats. Neuroscience 2011; 173:1-18. [DOI: 10.1016/j.neuroscience.2010.11.037] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 11/08/2010] [Accepted: 11/17/2010] [Indexed: 12/15/2022]
|
100
|
Huang JY, Chuang JI. Fibroblast growth factor 9 upregulates heme oxygenase-1 and gamma-glutamylcysteine synthetase expression to protect neurons from 1-methyl-4-phenylpyridinium toxicity. Free Radic Biol Med 2010; 49:1099-108. [PMID: 20615462 DOI: 10.1016/j.freeradbiomed.2010.06.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 05/14/2010] [Accepted: 06/22/2010] [Indexed: 10/19/2022]
Abstract
Oxidative stress and lower levels of trophic factors involved in nigrostriatal dopaminergic neurodegeneration are a hallmark of Parkinson disease. Our previous studies found that fibroblast growth factor 9 (FGF9) prevented 1-methyl-4-phenylpyridinium (MPP(+))-induced nigral dopaminergic neuron death and was involved in the neuroprotection of the antioxidant melatonin. However, the protective mechanisms mediated by FGF9 remain unclear. Herein, we explored whether FGF9 regulated the cellular antioxidant defense protecting dopaminergic neurons against MPP(+) intoxication. We found that FGF9 treatment alone induced a decrease in hydrogen peroxide (H(2)O(2)) level, an increase in glutathione content, and an upregulation of gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase 1 (HO-1) expression in primary cortical neurons but not in astrocytes. Simultaneous treatment with FGF9 and MPP(+) prevented MPP(+)-induced neuron death and H(2)O(2) overproduction but did not affect the FGF9-increased gamma-GCS and HO-1 protein expression. Inhibition of gamma-GCS or HO-1 prevented the inhibitory effect of FGF9 on MPP(+)-induced H(2)O(2) production and death in mesencephalic dopaminergic and cortical neurons. However, in the absence of MPP(+), the FGF9-induced H(2)O(2) reduction was blocked by HO-1 inhibitors, but not by gamma-GCS inhibitors. These results indicate that FGF9 upregulates gamma-GCS and HO-1 expression to protect cortical and dopaminergic neurons from MPP(+)-induced oxidative insult.
Collapse
Affiliation(s)
- Jui-Yen Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | | |
Collapse
|