1
|
Lin Z, Ying C, Si X, Xue N, Liu Y, Zheng R, Chen Y, Pu J, Zhang B. NOX4 exacerbates Parkinson's disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen Res 2025; 20:2038-2052. [PMID: 38993139 PMCID: PMC11691449 DOI: 10.4103/nrr.nrr-d-23-01265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/18/2023] [Accepted: 03/18/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00026/figure1/v/2024-09-09T124005Z/r/image-tiff Parkinson's disease is primarily caused by the loss of dopaminergic neurons in the substantia nigra compacta. Ferroptosis, a novel form of regulated cell death characterized by iron accumulation and lipid peroxidation, plays a vital role in the death of dopaminergic neurons. However, the molecular mechanisms underlying ferroptosis in dopaminergic neurons have not yet been completely elucidated. NADPH oxidase 4 is related to oxidative stress, however, whether it regulates dopaminergic neuronal ferroptosis remains unknown. The aim of this study was to determine whether NADPH oxidase 4 is involved in dopaminergic neuronal ferroptosis, and if so, by what mechanism. We found that the transcriptional regulator activating transcription factor 3 increased NADPH oxidase 4 expression in dopaminergic neurons and astrocytes in an 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced Parkinson's disease model. NADPH oxidase 4 inhibition improved the behavioral impairments observed in the Parkinson's disease model animals and reduced the death of dopaminergic neurons. Moreover, NADPH oxidase 4 inhibition reduced lipid peroxidation and iron accumulation in the substantia nigra of the Parkinson's disease model animals. Mechanistically, we found that NADPH oxidase 4 interacted with activated protein kinase C α to prevent ferroptosis of dopaminergic neurons. Furthermore, by lowering the astrocytic lipocalin-2 expression, NADPH oxidase 4 inhibition reduced 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced neuroinflammation. These findings demonstrate that NADPH oxidase 4 promotes ferroptosis of dopaminergic neurons and neuroinflammation, which contribute to dopaminergic neuron death, suggesting that NADPH oxidase 4 is a possible therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhihao Lin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Changzhou Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaoli Si
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Naijia Xue
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yi Liu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ran Zheng
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Zhang J, Zhao M, Yan R, Liu J, Maddila S, Junn E, Mouradian MM. MicroRNA-7 Protects Against Neurodegeneration Induced by α-Synuclein Preformed Fibrils in the Mouse Brain. Neurotherapeutics 2021; 18:2529-2540. [PMID: 34697773 PMCID: PMC8804150 DOI: 10.1007/s13311-021-01130-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 01/01/2023] Open
Abstract
α-Synuclein is a key protein in the pathogenesis of Parkinson's disease as it accumulates in fibrillar form in affected brain regions. Misfolded α-synuclein seeds recruit monomeric α-synuclein to form aggregates, which can spread to anatomically connected brain regions, a phenomenon that correlates with clinical disease progression. Thus, downregulating α-synuclein levels could reduce seeding and inhibit aggregate formation and propagation. We previously reported that microRNA-7 (miR-7) protects neuronal cells by downregulating α-synuclein expression through its effect on the 3'-untranslated region of SNCA mRNA; however, whether miR-7 blocks α-synuclein seeding and propagation in vivo remains unknown. Here, we induced miR-7 overexpression in the mouse striatum unilaterally by infusing adeno-associated virus 1 (AAV-miR-7) followed by inoculation with recombinant α-synuclein preformed fibrils (PFF) a month later. Compared with control mice injected with non-targeting AAV-miR-NT followed by PFF, AAV-miR-7 pre-injected mice exhibited lower levels of monomeric and high-molecular-weight α-synuclein species in the striatum, and reduced amount of phosphorylated α-synuclein in the striatum and in nigral dopamine neurons. Accordingly, AAV-miR-7-injected mice had less pronounced degeneration of the nigrostriatal pathway and better behavioral performance. The neuroinflammatory reaction to α-synuclein PFF inoculation was also significantly attenuated. These data suggest that miR-7 inhibits the formation and propagation of pathological α-synuclein and protects against neurodegeneration induced by PFF. Collectively, these findings support the potential of miR-7 as a disease modifying biologic agent for Parkinson's disease and related α-synucleinopathies.
Collapse
Affiliation(s)
- Jie Zhang
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
| | - Mengyuan Zhao
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
| | - Run Yan
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
- Current address: Sanyou Biopharmaceuticals Co., Ltd., 3rd Floor, Building 6B-C, No. 188 Xinjunhuan Road, Minhang District, Shanghai, 201114, China
| | - Jun Liu
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
| | - Santhosh Maddila
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
| | - Eunsung Junn
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA
| | - M Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 180, Piscataway, NJ, 08854, USA.
| |
Collapse
|
3
|
Zhang M, Cui Y, Zhu W, Yu J, Cheng Y, Wu X, Zhang J, Xin W, Yu Y, Sun H. Attenuation of the mutual elevation of iron accumulation and oxidative stress may contribute to the neuroprotective and anti-seizure effects of xenon in neonatal hypoxia-induced seizures. Free Radic Biol Med 2020; 161:212-223. [PMID: 33075502 DOI: 10.1016/j.freeradbiomed.2020.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Previous studies have suggested that xenon inhalation has neuroprotective and antiepileptic effects; however, the underlying mechanisms involved remain unclear. This study aimed to investigate the possible xenon inhalation mechanisms involved in the neuroprotection and antiepileptic effects. A neonatal hypoxic C57BL/6J mouse model was used for the experiments. Immediately after hypoxia treatment, the treatment group inhaled a xenon mixture (70% xenon/21% oxygen/9% nitrogen) for 60 min, while the hypoxia group inhaled a non-xenon mixture (21% oxygen/79% nitrogen) for 60 min. Seizure activity was recorded at designated time points using electroencephalography. Oxidative stress levels, iron levels, neuronal injury, and learning and memory functions were also studied. The results showed that hypoxia increased the levels of iron, oxidative stress, mitophagy, and neurodegeneration, which were accompanied by seizures and learning and memory disorders. In addition, our results confirmed that xenon treatment significantly attenuated the hypoxia-induced seizures and cognitive defects in neonatal C57 mice. Moreover, the increased levels of iron, oxidative stress, mitophagy, and neuronal injury were reduced in xenon-treated mice. This study confirms the significant protective effects of a xenon mixture on hypoxia-induced damage in neonatal mice. Furthermore, our results suggest that reducing oxidative stress levels and iron accumulation may be the underlying mechanisms of xenon activity. Studying the protective mechanisms of xenon will advance its applications in potential therapeutic strategies.
Collapse
Affiliation(s)
- Mengdi Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yaru Cui
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Wei Zhu
- Institute of Radiation Medicine, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, 250062, China
| | - Jie Yu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiangdong Wu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Jinjin Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Wenyu Xin
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yan Yu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
4
|
Heimfarth L, Dos Anjos KS, de Carvalho YMBG, Dos Santos BL, Serafini MR, de Carvalho Neto AG, Nunes PS, Beserra Filho JIA, da Silva SP, Ribeiro AM, Bezerra DP, Marreto RN, de Souza Siqueira Quintans J, de Souza Araújo AA, Melo Coutinho HD, Scotti MT, Scotti L, Quintans-Júnior LJ. Characterization of β-cyclodextrin/myrtenol complex and its protective effect against nociceptive behavior and cognitive impairment in a chronic musculoskeletal pain model. Carbohydr Polym 2020; 244:116448. [PMID: 32536383 DOI: 10.1016/j.carbpol.2020.116448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022]
Abstract
Myrtenol has gained wide interest because of its pharmacological profiles, mainly for treatment of chronic diseases. To improve the solubility of myrtenol, the formation of inclusion complexes with β-cyclodextrin was performed by physical mixture, kneading process or slurry complexation (SC) methods and characterized using thermal analysis, XRD, SEM and NMR. From these results, myrtenol complexed by SC was successfully complexed into β-cyclodextrin cavity. The interaction between myrtenol and β-cyclodextrin was confirmed by molecular docking. Hence, the SC β-cyclodextrin-myrtenol complex was evaluate for its anti-hyperalgesic, anxiolytic and antioxidant activity in a fibromyalgia model. Results show that myrtenol and β-cyclodextrin form a stable complex and have anti-hyperalgesic effect, improve the cognitive impairment caused and have an anxiolytic-like effect. Furthermore, the β-cyclodextrin/myrtenol complex decrease lipoperoxidation, increased catalase activity and a reduce SOD/CAT ratio. Therefore, β-cyclodextrin/myrtenol complex reduce painful behavior, improves motor skills and emotional behavior and decreases oxidative stress in a fibromyalgia model.
Collapse
Affiliation(s)
- Luana Heimfarth
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | | | | | | | | | - Paula Santos Nunes
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Sara Pereira da Silva
- Department of Biosciences, Federal University of São Paulo/UNIFESP, Santos, SP, Brazil
| | | | - Daniel Pereira Bezerra
- Oswaldo Cruz Foundation, Laboratory of Tissue Engineering and Immunopharmacology, Salvador, BA, Brazil
| | | | | | | | - Henrique Douglas Melo Coutinho
- Department of Biological Chemistry, Regional University of Cariri, URCA, Av Cel. Antonio Luiz, 1161, Pimenta, Crato, CE, 63105-000, Brazil.
| | - Marcus T Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | | |
Collapse
|
5
|
Autophagy as a Cellular Stress Response Mechanism in the Nervous System. J Mol Biol 2020; 432:2560-2588. [PMID: 31962122 DOI: 10.1016/j.jmb.2020.01.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Cells of an organism face with various types of insults during their lifetime. Exposure to toxins, metabolic problems, ischaemia/reperfusion, physical trauma, genetic diseases, neurodegenerative diseases are among the conditions that trigger cellular stress responses. In this context, autophagy is one of the mechanisms that supports cell survival under stressful conditions. Autophagic vesicle engulfs the cargo and transports it to lysosome for degradation and turnover. As such, autophagy eliminates abnormal proteins, clears damaged organelles, limits oxidative stress and helps to improve metabolic balance. Nervous system cells and particularly postmitotic neurons are highly sensitive to a spectrum of insults, and autophagy emerges as one of the key stress response mechanism, ensuring health and survival of these vulnerable cell types. In this review, we will overview mechanisms through which cells cope with stress, and how these stress responses regulate autophagy, with a special focus on the nervous system.
Collapse
|
6
|
Zhang J, Park ES, Park HJ, Yan R, Grudniewska M, Zhang X, Oh S, Yang X, Baum J, Mouradian MM. Apoptosis signal regulating kinase 1 deletion mitigates α-synuclein pre-formed fibril propagation in mice. Neurobiol Aging 2020; 85:49-57. [PMID: 31734439 PMCID: PMC7064162 DOI: 10.1016/j.neurobiolaging.2019.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
α-Synuclein (α-Syn) is a key pathogenic protein in α-synucleinopathies including Parkinson disease and dementia with Lewy bodies. Accumulating evidence has shown that misfolded fibrillar α-Syn is transmitted from cell-to-cell, a phenomenon that correlates with clinical progression of the disease. We previously showed that deleting the MAP3 kinase apoptosis signal-regulating kinase 1 (ASK1), which is a central player linking oxidative stress with neuroinflammation, mitigates the phenotype of α-Syn transgenic mice. However, whether ASK1 impacts pathology and disease progression induced by recombinant α-Syn pre-formed fibrils (PFF) remains unknown. Here, we compared the neuropathological and behavioral phenotype of ASK1 knock-out mice with that of wild-type mice following intrastriatal injections of α-Syn PFF. At 6 months post-injections, ASK1 null mice exhibited reduced amount of phosphorylated α-Syn aggregates in the striatum and cortex, and less pronounced degeneration of the nigrostriatal pathway. Additionally, the neuroinflammatory reaction to α-Syn PFF injection and propagation seen in wild-type mice was attenuated in ASK1 knock-out animals. These neuropathological markers were associated with better behavioral performance. These data suggest that ASK1 plays an important role in pathological α-Syn fibril transmission and, consequently, may impact disease progression. These findings collectively support inhibiting ASK1 as a disease modifying therapeutic strategy for Parkinson disease and related α-synucleinopathies.
Collapse
Affiliation(s)
- Jie Zhang
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Eun S Park
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Hye-Jin Park
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Run Yan
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Magda Grudniewska
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Xiaopei Zhang
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Stephanie Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Xue Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, USA
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, USA
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA.
| |
Collapse
|
7
|
Mohite GM, Navalkar A, Kumar R, Mehra S, Das S, Gadhe LG, Ghosh D, Alias B, Chandrawanshi V, Ramakrishnan A, Mehra S, Maji SK. The Familial α-Synuclein A53E Mutation Enhances Cell Death in Response to Environmental Toxins Due to a Larger Population of Oligomers. Biochemistry 2018; 57:5014-5028. [DOI: 10.1021/acs.biochem.8b00321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ganesh M. Mohite
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Subhadeep Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
- IITB-Monash Research Academy, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Laxmikant G. Gadhe
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Dhiman Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Basil Alias
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Vikas Chandrawanshi
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Aishwarya Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Sarika Mehra
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| |
Collapse
|
8
|
Brown J, Horrocks MH. A sticky situation: Aberrant protein-protein interactions in Parkinson's disease. Semin Cell Dev Biol 2018; 99:65-77. [PMID: 29738882 DOI: 10.1016/j.semcdb.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
Abstract
The aberrant aggregation of normally soluble proteins into amyloid fibrils is the pathological hallmark of several neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Understanding this process will be key to developing both diagnostic and therapeutic approaches for neurodegenerative diseases. Recent advances in biophysical techniques, coupled with kinetic analyses have enabled a thorough description of the key molecular steps involved in protein aggregation. In this review, we discuss these advances and how they have been applied to study the ability of one such protein, α-Synuclein, to form neurotoxic oligomers.
Collapse
Affiliation(s)
- James Brown
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney, NSW, 2032, Australia.
| | - Mathew H Horrocks
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
9
|
Li T, Feng Y, Yang R, Wu L, Li R, Huang L, Yang Q, Chen J. Salidroside Promotes the Pathological α-Synuclein Clearance Through Ubiquitin-Proteasome System in SH-SY5Y Cells. Front Pharmacol 2018; 9:377. [PMID: 29725300 PMCID: PMC5917065 DOI: 10.3389/fphar.2018.00377] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/03/2018] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of Lewy bodies (LBs) in the surviving SNc neurons. LBs formation is caused by the accumulation of α-synuclein (α-syn) or phosphorylated α-syn at serine-129 (pSer129-α-syn), which is implicated in the pathological progression of PD. Salidroside (Sal), the main active ingredient of the root of Rhodiola rosea L., has been reported to have potent neuroprotective properties in our previous investigations. Here, we investigated the effects of Sal on 6-OHDA and overexpresssion of WT/A30P-α-syn-induced pathological α-syn increase and the mechanism behind it in SH-SY5Y cells. We found Sal displays neuroprotective effects against 6-hydroxydopamine (6-OHDA)-induced cytotoxicity. Sal decreased the pSer129-α-syn level mainly by maintaining the normal function of ubiquitin-proteasome system (UPS). Furthermore, Sal promoted the clearance of α-syn and protected the cell viability mainly through recovered the 20S proteasome activity in WT/A30P-α-syn-transfected cells. These data provide new mechanistic insights into the neuroprotective effects of Sal and Sal may be a promising therapy to slow neurodegeneration in PD. Highlights: Sal protects cells and decreases the pSer129-α-syn protein level in 6-OHDA-induced impairmental and dysfunctional SH-SY5Y cells. Sal promotes the clearance of α-syn and protects the cell viability mainly through recovering the 20S proteasome activity in WT/A30P-α-syn plasmids transfected cells. Maintaining the normal function of the UPS may be one of the important mechanisms of Sal in neuroprotective effects.
Collapse
Affiliation(s)
- Tao Li
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Yang Feng
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Ruixin Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Leitao Wu
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Ruru Li
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Lu Huang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Qian Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Jianzong Chen
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| |
Collapse
|
10
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 PMCID: PMC5635514 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
11
|
Han J, Plummer J, Liu L, Byrd A, Aschner M, Erikson KM. The impact of obesity on brain iron levels and α-synuclein expression is regionally dependent. Nutr Neurosci 2017; 22:335-343. [PMID: 29034829 DOI: 10.1080/1028415x.2017.1387720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The importance of iron homeostasis is particularly apparent in the brain, where iron deficiency results in impaired cognition and iron accumulation is associated with neurodegenerative diseases. Obesity is linked to iron deficiency systemically, but the effects of obesity on brain iron and its associated consequences, including neurodegenerative processes remain unexplored. This preliminary study examined the effect of dietary-induced obesity on brain regional iron, α-synuclein expression, and F2-isoprostane (oxidative stress marker) concentrations in selected brain regions. OBJECTIVE The objective of the study was to elucidate the vulnerability of selected brain regions (e.g. midbrain, hippocampus) to the possible process of neurodegeneration due to the altered iron content associated with obesity. METHODS Twenty-one-day-old male C57BL/6J mice were fed with a high-fat diet (60% kcal from fat) or a control-fat diet (10% kcal from fat) for 20 weeks. Brain samples were collected and dissected into hippocampus, midbrain, striatum, and thalamus regions. Iron content, ferritin H (FtH) and α-synuclein protein and mRNA expressions, and F2-isoprostane were measured in selected regions. RESULTS The results indicated that obesity caused significant differences in iron levels in the midbrain and thalamus, but not in the hippocampus or striatum, compared to control mice. Furthermore, markers of neurodegeneration (α-synuclein mRNA expression and F2-isoprostanes) were increased in the midbrain. DISCUSSION These results support previous findings that brain iron metabolism responds to environmental stress in a regionally distinct manner and suggests that alterations in brain iron metabolism due to obesity may be relevant in neurodegeneration.
Collapse
Affiliation(s)
- Jian Han
- a Department of Biology , North Carolina Agricultural and Technical State University , Greensboro , NC 27411 , USA
| | - Justin Plummer
- b Department of Nutrition , The University of North Carolina at Greensboro , Greensboro , NC 27412 , USA
| | - Lumei Liu
- a Department of Biology , North Carolina Agricultural and Technical State University , Greensboro , NC 27411 , USA
| | - Aria Byrd
- c Department of Toxicology and Cancer Biology , University of Kentucky , Lexington , KY 40536 , USA
| | - Michael Aschner
- d Department of Molecular Pharmacology , Albert Einstein School of Medicine , Bronx , NY 10461 , USA
| | - Keith M Erikson
- b Department of Nutrition , The University of North Carolina at Greensboro , Greensboro , NC 27412 , USA
| |
Collapse
|
12
|
Superoxide drives progression of Parkin/PINK1-dependent mitophagy following translocation of Parkin to mitochondria. Cell Death Dis 2017; 8:e3097. [PMID: 29022898 PMCID: PMC5680585 DOI: 10.1038/cddis.2017.463] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/05/2017] [Accepted: 07/24/2017] [Indexed: 11/13/2022]
Abstract
Reactive oxygen species (ROS) and mitophagy are profoundly implicated in the pathogenesis of neurodegenerative diseases, such as Parkinson’s disease (PD). Several studies have suggested that ROS are not involved in mitochondrial translocation of Parkin which primes mitochondria for autophagic elimination. However, whether ROS play a role in the execution of mitophagy is unknown. In the present study, we show that carbonyl cyanide m-chlorophenylhydrazone (CCCP) treatment induced both mitochondrial depolarization and generation of ROS that were needed for the mitophagy process. Cells failed to proceed to complete mitophagy if CCCP treatment was discontinued even after recruitment of Parkin and autophagy machinery to mitochondria. Notably, treatment of pro-oxidant was able to replace CCCP treatment to take mitophagy forward, while it alone was insufficient to induce translocation of Parkin to mitochondria or autophagic clearance of mitochondria. In addition, an SOD mimetic that attenuated the superoxide level suppressed mitophagy, while an SOD inhibitor accumulated cellular superoxide and promoted mitophagy. Furthermore, blockage of the p38 signaling pathway inhibited mitophagy induced by ROS, suggesting that it may contribute to the activation of ROS-mediated mitophagy. Together, our study sheds light on the link between ROS and mitophagy at a molecular level, and suggests the therapeutic potential of regulating mitophagy through the superoxide–p38–mitophagy axis.
Collapse
|
13
|
Trehalose Inhibits A53T Mutant α-Synuclein Overexpression and Neurotoxicity in Transduced PC12 Cells. Molecules 2017; 22:molecules22081293. [PMID: 28786917 PMCID: PMC6152154 DOI: 10.3390/molecules22081293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/01/2017] [Indexed: 12/03/2022] Open
Abstract
Fibrillar accumulation of A53T mutant α-synuclein (A53T-AS) in Lewy bodies is a symptom of Parkinsonism. Inhibitions of the overexpression and fibrillar aggregation of α-synuclein (AS) in vivo could be a promising strategy for treating Parkinson’s disease (PD). In this study, at concentrations lower than 1 mM, trehalose decreased the A53T-AS expression level in transduced PC12 cells. Although H2O2 and aluminum ions increased the expression level and neurotoxicity of A53T-AS in cells, proper trehalose concentrations inhibited the event. These studies adequately prove that trehalose at an appropriate dose would be potentially useful for PD treatment.
Collapse
|
14
|
Mingori MR, Heimfarth L, Ferreira CF, Gomes HM, Moresco KS, Delgado J, Roncato S, Zeidán-Chuliá F, Gelain DP, Moreira JCF. Effect of Paullinia cupana Mart. Commercial Extract During the Aging of Middle Age Wistar Rats: Differential Effects on the Hippocampus and Striatum. Neurochem Res 2017; 42:2257-2273. [DOI: 10.1007/s11064-017-2238-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/19/2023]
|
15
|
Menges S, Minakaki G, Schaefer PM, Meixner H, Prots I, Schlötzer-Schrehardt U, Friedland K, Winner B, Outeiro TF, Winklhofer KF, von Arnim CAF, Xiang W, Winkler J, Klucken J. Alpha-synuclein prevents the formation of spherical mitochondria and apoptosis under oxidative stress. Sci Rep 2017; 7:42942. [PMID: 28224980 PMCID: PMC5320486 DOI: 10.1038/srep42942] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/17/2017] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress (OS), mitochondrial dysfunction, and dysregulation of alpha-synuclein (aSyn) homeostasis are key pathogenic factors in Parkinson's disease. Nevertheless, the role of aSyn in mitochondrial physiology remains elusive. Thus, we addressed the impact of aSyn specifically on mitochondrial response to OS in neural cells. We characterize a distinct type of mitochondrial fragmentation, following H2O2 or 6-OHDA-induced OS, defined by spherically-shaped and hyperpolarized mitochondria, termed "mitospheres". Mitosphere formation mechanistically depended on the fission factor Drp1, and was paralleled by reduced mitochondrial fusion. Furthermore, mitospheres were linked to a decrease in mitochondrial activity, and preceded Caspase3 activation. Even though fragmentation of dysfunctional mitochondria is considered to be a prerequisite for mitochondrial degradation, mitospheres were not degraded via Parkin-mediated mitophagy. Importantly, we provide compelling evidence that aSyn prevents mitosphere formation and reduces apoptosis under OS. In contrast, aSyn did not protect against Rotenone, which led to a different, previously described donut-shaped mitochondrial morphology. Our findings reveal a dichotomic role of aSyn in mitochondrial biology, which is linked to distinct types of stress-induced mitochondrial fragmentation. Specifically, aSyn may be part of a cellular defense mechanism preserving neural mitochondrial homeostasis in the presence of increased OS levels, while not protecting against stressors directly affecting mitochondrial function.
Collapse
Affiliation(s)
- Stefanie Menges
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Georgia Minakaki
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Holger Meixner
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Iryna Prots
- IZKF Junior Research Group III and BMBF Research Group Neuroscience, IZKF, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.,Department of Stem Cell Biology, Institute of Human Genetics, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Kristina Friedland
- Molecular and Clinical Pharmacy, Department of Chemistry and Pharmacy, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Beate Winner
- IZKF Junior Research Group III and BMBF Research Group Neuroscience, IZKF, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.,Department of Stem Cell Biology, Institute of Human Genetics, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tiago F Outeiro
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, 37073 Göttingen, Germany.,Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Konstanze F Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr-University Bochum, 44801 Bochum, Germany
| | | | - Wei Xiang
- Institute of Biochemistry, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jochen Klucken
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
16
|
Lee KW, Woo JM, Im JY, Park ES, He L, Ichijo H, Junn E, Mouradian MM. Apoptosis signal-regulating kinase 1 modulates the phenotype of α-synuclein transgenic mice. Neurobiol Aging 2014; 36:519-26. [PMID: 25219466 DOI: 10.1016/j.neurobiolaging.2014.07.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/12/2014] [Accepted: 07/26/2014] [Indexed: 10/25/2022]
Abstract
α-Synuclein is a key pathogenic protein in α-synucleinopathies including Parkinson's disease, and its overexpression and aggregation in model systems are associated with a neuroinflammatory response and increased oxidative stress. Apoptosis signal-regulating kinase 1 (ASK1) is activated upon stress signaling events such as oxidative stress and is a central player linking oxidative stress with neuroinflammation. Here, we demonstrate that overexpression of human α-synuclein activates ASK1 in both PC12 cells and in the brains of α-synuclein transgenic mice. Deleting ASK1 in mice mitigates the neuronal damage and neuroinflammation induced by α-synuclein and improves performance of the animals on the rotarod. ASK1 deletion does not impact the aggregation profile or phosphorylation state of α-synuclein in the mouse brain. These results collectively implicate ASK1 in the cascade of events triggered by α-synuclein overexpression, likely because of the inflammatory response and oxidative stress that lead to ASK1 activation. These conclusions raise the possibility that potent antioxidants and anti-inflammatory agents may ameliorate the phenotype of α-synucleinopathies.
Collapse
Affiliation(s)
- Kang-Woo Lee
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jong-Min Woo
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Joo-Young Im
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Eun S Park
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Liqiang He
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Eunsung Junn
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - M Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| |
Collapse
|
17
|
Awan MUF, Deng Y. Role of autophagy and its significance in cellular homeostasis. Appl Microbiol Biotechnol 2014; 98:5319-28. [PMID: 24743981 DOI: 10.1007/s00253-014-5721-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/21/2014] [Accepted: 03/22/2014] [Indexed: 12/18/2022]
Abstract
Autophagy is a catabolic pathway that regulates homeostasis in cells. It is an exceptional pathway of membrane trafficking. Autophagy is characterized by the formation of double-membrane vesicles; autophagosomes that are responsible for delivering damaged organelle and extra proteins to lysosome for recycling. A series of actions including environmental and genetic factors are responsible for induction of autophagy. In the past few decades, the research on autophagy has been immensely expanded because it is a vital process in maintaining cellular balance as well as deeply connected with pathogenesis of a number of diseases. The aim of this review is to present an overview of modern work on autophagy and highlight some essential genetic role in the induction of autophagy. There is an emerging need to identify, quantify, and manipulate the pathway of autophagy, due to its close relationship with a variety of developmental pathways and functions especially in cancer, diabetes, neurodegenerative disorders, and infectious diseases.
Collapse
Affiliation(s)
- M Umer Farooq Awan
- School of Life Sciences, Beijing Institute of Technology, No. 5 Zhongguancunn South Street, Beijing, 100081, People's Republic of China
| | | |
Collapse
|
18
|
Ma L, Wei L, Wu F, Hu Z, Liu Z, Yuan W. Advances with microRNAs in Parkinson's disease research. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:1103-13. [PMID: 24109179 PMCID: PMC3792848 DOI: 10.2147/dddt.s48500] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second-most common age-dependent neurodegenerative disorder and is caused by severe degeneration of dopaminergic neurons in the substantia nigra pars compacta. Unfortunately, current treatment only targets symptoms and involves dopamine replacement therapy, which does not counteract progressive degeneration. MicroRNAs (miRNAs) are a class of small RNA molecules implicated in post-transcriptional regulation of gene expression during development. Recent studies show that miRNAs are playing an important role in the pathophysiology of PD. miRNA-based therapy is a powerful tool with which to study gene function, investigate the mechanism of the disease, and validate drug targets. In this review, we focus on the recent advances of the use of miRNAs in the pathogenesis of PD.
Collapse
Affiliation(s)
- Liuqing Ma
- Department of Neurology, Xinhua Hospital Affiliated with Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China ; School of Pharmacy, Shanghai JiaoTong University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Increased α-synuclein levels and mutations in mitochondria-associated proteins both cause familial Parkinson's disease (PD), and synuclein and mitochondria also play central, but poorly understood, roles in the pathogenesis of idiopathic PD. A fraction of synuclein interacts with mitochondria, and synuclein can produce mitochondrial fragmentation and impair mitochondrial complex I activity. However, the consequences of these mitochondrial changes for bioenergetic and other mitochondrial functions remain poorly defined, as does the role of synuclein-mitochondria interactions in the normal and pathologic effects of synuclein. Understanding the functional consequences of synuclein's interactions with mitochondria is likely to provide important insights into disease pathophysiology, and may also reveal therapeutic strategies.
Collapse
Affiliation(s)
- Ken Nakamura
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158, USA.
| |
Collapse
|
20
|
Liddell JR, Obando D, Liu J, Ganio G, Volitakis I, Mok SS, Crouch PJ, White AR, Codd R. Lipophilic adamantyl- or deferasirox-based conjugates of desferrioxamine B have enhanced neuroprotective capacity: implications for Parkinson disease. Free Radic Biol Med 2013; 60:147-56. [PMID: 23391576 DOI: 10.1016/j.freeradbiomed.2013.01.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 01/22/2013] [Accepted: 01/29/2013] [Indexed: 02/03/2023]
Abstract
Parkinson disease (PD) is a neurodegenerative disease characterized by death of dopaminergic neurons in the substantia nigra region of the brain. Iron content is also elevated in this region in PD and is implicated in the pathobiology of the disease. Desferrioxamine B (DFOB) is a high-affinity iron chelator and has shown efficacy in animal models of Parkinson disease. The high water solubility of DFOB, however, attenuates its ability to enter the brain. In this study, we have conjugated DFOB to derivatives of adamantane or the clinical iron chelator deferasirox to produce lipophilic compounds designed to increase the bioavailability of DFOB to brain cells. We found that the novel compounds are highly effective in preventing iron-mediated paraquat and hydrogen peroxide toxicity in neuronal-like BE2-M17 dopaminergic cells, primary neurons, and iron-loaded or glutathione-depleted primary astrocytes. The compounds also alleviated paraquat toxicity in BE2-M17 cells that express the PD-causing A30P mutation of α-synuclein. This protection was ∼66-fold more potent than DFOB alone and also more effective than other cell-permeative metal chelators, clioquinol and phenanthroline. These results demonstrate that increasing the bioavailability of DFOB through the conjugation of lipophilic fragments greatly enhances its protective capacity. These novel compounds have potential as therapeutics for the treatment of PD and other conditions of Fe dyshomeostasis.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Pathology, University of Melbourne, and Mental Health Research Institute, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Jiang T, Yu WB, Yao T, Zhi XL, Pan LF, Wang J, Zhou P. Trehalose inhibits wild-type α-synuclein fibrillation and overexpression and protects against the protein neurotoxicity in transduced PC12 cells. RSC Adv 2013. [DOI: 10.1039/c3ra40600h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
22
|
Kansara S, Trivedi A, Chen S, Jankovic J, Le W. Early diagnosis and therapy of Parkinson’s disease: can disease progression be curbed? J Neural Transm (Vienna) 2012; 120:197-210. [DOI: 10.1007/s00702-012-0840-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 05/26/2012] [Indexed: 12/15/2022]
|
23
|
MicroRNAs in Parkinson's disease. Neurobiol Dis 2012; 46:279-84. [PMID: 22245218 DOI: 10.1016/j.nbd.2011.12.046] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 12/15/2011] [Accepted: 12/26/2011] [Indexed: 02/03/2023] Open
Abstract
Besides the classic mutations in coding regions of genes, the critical role of gene expression regulators in disease states is increasingly recognized. The network of small non-coding microRNAs is crucial for the normal development and survival of distinct neuronal populations that are vulnerable in various neurodegenerative disorders. In midbrain dopaminergic neurons, which degenerate in Parkinson's disease (PD) causing motor signs and symptoms, disruption of this network results in their progressive loss associated with impaired motor activity in Drosophila and mouse models. Studies of families with dominantly inherited PD linked to multiplication of the α-synuclein gene locus indicate that the amount of this key pathogenic protein in neurons is an important determinant of its tendency to aggregate pathologically and increase neuronal susceptibility. Recent reports demonstrate that the α-synuclein mRNA is under negative control by at least two microRNAs, miR-7 and miR-153. In addition to studying the regulation of candidate genes by specific microRNA species, different profiling approaches are uncovering variations in the abundance of certain microRNAs that may prove to be relevant to the disease. For example, miR-133b is deficient in the PD midbrain as well as in mouse models, and miR-34b/34c are decreased in several affected brain regions in PD and incidental Lewy body disease. Polymorphisms in the 3'-untranslated region of microRNA target mRNAs, including in the gene encoding α-synuclein found in Genome Wide Association studies, are another potential reason for variations in the rate of protein production and thus disease risk. And finally, the impact of a disease associated gene product, and in particular LRRK2, on the microRNA network compounds the complexity of the interplay between the microRNA system and pathogenic proteins. The wealth of knowledge accumulating from these studies in a few short years holds considerable promise to harness its potential and translate it into therapeutic strategies for PD.
Collapse
|
24
|
Abstract
Aggregated a-synuclein is the major component of inclusions in Parkinson's disease and other synucleinopathy brains indicating that a-syn aggregation is associated with the pathogenesis of neurodegenerative disorders. Although the mechanisms underlying a-syn aggregation and toxicity are not fully elucidated, it is clear that a-syn undergoes post-translational modifications and interacts with numerous proteins and other macromolecules, metals, hormones, neurotransmitters, drugs and poisons that can all modulate its aggregation propensity. The current and most recent findings regarding the factors modulating a-syn aggregation process are discussed in detail.
Collapse
|
25
|
Li J, Yang J, Zhao P, Li S, Zhang R, Zhang X, Liu D, Zhang B. Neuromelanin enhances the toxicity of α-synuclein in SK-N-SH cells. J Neural Transm (Vienna) 2011; 119:685-91. [PMID: 22200858 DOI: 10.1007/s00702-011-0753-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/13/2011] [Indexed: 10/14/2022]
Abstract
The key pathological feature of Parkinson's disease (PD) is selective degeneration of the neuromelanin (NM)-pigmented dopaminergic neurons in the substantia nigra (SN). NM, like other risk factors, such as oxidative stress (OS) and α-synuclein (α-syn), is involved in the pathogenesis of PD. But whether or not NM synergizes with α-syn or OS in the pathogenesis of PD remains unexplored. In the present study, we examined the effects of NM on cellular viability, apoptosis and free radical production in α-syn over-expressing human neuroblastoma cell line (SK-N-SH) in the presence or absence of the oxidizer Fenton's Reagent (FR). We showed that NM synergized with FR in suppressing cell viability, and in inducing apoptosis and hydroxyl radical production in all SK-N-SH cell lines. α-Syn over-expressing cells exhibited more pronounced effect, especially the A53T mutation. Our findings suggest that NM synergizes with both OS and α-syn in conferring dopaminergic vulnerability, adding to our understanding of the pathogenesis of PD.
Collapse
Affiliation(s)
- Jie Li
- Department of Psychiatry, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Distinct roles in vivo for the ubiquitin-proteasome system and the autophagy-lysosomal pathway in the degradation of α-synuclein. J Neurosci 2011; 31:14508-20. [PMID: 21994367 DOI: 10.1523/jneurosci.1560-11.2011] [Citation(s) in RCA: 300] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increased intracellular levels of α-synuclein are implicated in Parkinson's disease and related disorders and may be caused by alterations in the ubiquitin-proteasome system (UPS) or the autophagy-lysosomal pathway (ALP). A critical question remains how α-synuclein is degraded by neurons in vivo. To address this, our study uses α-synuclein transgenic mice, expressing human α-synuclein or α-synuclein-eGFP under the (h)PDGF-β promoter, in combination with in vivo pharmacologic and multiphoton imaging strategies to systematically test degradation pathways in the living mouse brain. We demonstrate that the UPS is the main degradation pathway for α-synuclein under normal conditions in vivo while with increased α-synuclein burden the ALP is recruited. Moreover, we report alterations of the UPS in α-synuclein transgenic mice and age dependence to the role of the UPS in α-synuclein degradation. In addition, we provide evidence that the UPS and ALP might be functionally connected such that impairment of one can upregulate the other. These results provide a novel link between the UPS, the ALP, and α-synuclein pathology and may have important implications for future therapeutics targeting degradation pathways.
Collapse
|
27
|
Liu Z, Yu Y, Li X, Ross CA, Smith WW. Curcumin protects against A53T alpha-synuclein-induced toxicity in a PC12 inducible cell model for Parkinsonism. Pharmacol Res 2011; 63:439-44. [DOI: 10.1016/j.phrs.2011.01.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/04/2011] [Accepted: 01/05/2011] [Indexed: 11/26/2022]
|
28
|
ER stress response plays an important role in aggregation of α-synuclein. Mol Neurodegener 2010; 5:56. [PMID: 21144044 PMCID: PMC3016345 DOI: 10.1186/1750-1326-5-56] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 12/13/2010] [Indexed: 01/04/2023] Open
Abstract
Background Accumulation of filamentous α-synuclein as Lewy bodies is a hallmark of Parkinson's disease. To identify the mechanisms involved in α-synuclein assembly and determine whether the assemblies are cytotoxic, we developed a cell model (3D5) that inducibly expresses wild-type human α-synuclein and forms inclusions that reproduce many morphological and biochemical characteristics of Lewy bodies. In the present study, we evaluated the effects of several histone deacetylase inhibitors on α-synuclein aggregation in 3D5 cells and primary neuronal cultures. These drugs have been demonstrated to protect cells transiently overexpressing α-synuclein from its toxicity. Results Contrary to transient transfectants, the drug treatment did not benefit 3D5 cells and primary cultures. The treated were less viable and contained more α-synuclein oligomers, active caspases 3 and 9, as well as ER stress markers than non-treated counterparts. The drug-treated, induced-3D5 cells, or primary cultures from transgenic mice overexpressing (<2 fold) α-synuclein, displayed more α-synuclein oligomers and ER stress markers than non-induced or non-transgenic counterparts. Similar effects were demonstrated in cultures treated with tunicamycin, an ER stressor. These effects were blocked by co-treatment with salubrinal, an ER stress inhibitor. In comparison, co-treatment with a pan caspase inhibitor protected cells from demise but did not reduce α-synuclein oligomer accumulation. Conclusions Our results indicate that an increase of wild-type α-synuclein can elicit ER stress response and sensitize cells to further insults. Most importantly, an increase of ER stress response can promote the aggregation of wild type α-synuclein.
Collapse
|
29
|
Wang P, Jiang S, Cui Y, Yue Z, Su C, Sun J, Sheng S, Tian J. The n-terminal 5-MER peptide analogue P165 of amyloid precursor protein exerts protective effects on SH-SY5Y cells and rat hippocampus neuronal synapses. Neuroscience 2010; 173:169-78. [PMID: 21055450 DOI: 10.1016/j.neuroscience.2010.10.069] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/08/2010] [Accepted: 10/26/2010] [Indexed: 11/28/2022]
Abstract
The disturbance of the insulin-signaling pathway plays an important role in Alzheimer's disease. Resistance to insulin signaling renders neurons energy-deficient and vulnerable to oxidization or other metabolic insults and impairs synaptic plasticity. In search of neuroprotective drugs, we synthesized a peptide analogue, P165, an active domain of the soluble amyloid precursor protein, which is resistant to degradation and is suitable for oral administration in a clinical setting. Initially, we confirmed that P165 can protect cells from streptozotocin-caused damage and stimulate cell outgrowth using cultured SH-SY5Y cell lines treated with streptozotocin. P165 significantly reduced lactate dehydrogenase leakage from damaged cells, thereby rescuing cell energy production. Insulin signaling such as insulin receptor substrate-1 (IRS-1) and phosphoinositide 3-kinase (PI3K) proteins were upregulated to stimulate cell survival and growth. We proceeded to investigate the effect of P165 on streptozotocin-treated Alzheimer's disease (AD) rats. The data showed that P165 protected synaptic loss and dysfunction by increasing synaptophysin and PSD-95 (post synaptic density-95), while simultaneously decreasing α-synuclein expression. Moreover, animal behavior testing clearly showed that P165 increased rats' learning and memory activity. Overall, these results constitute evidence that peptide analogue 165 may protect synapse and improve learning and memory ability in AD.
Collapse
Affiliation(s)
- P Wang
- Key Laboratory of Chinese Internal Medicine (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Qian L, Flood PM, Hong JS. Neuroinflammation is a key player in Parkinson's disease and a prime target for therapy. J Neural Transm (Vienna) 2010; 117:971-9. [PMID: 20571837 PMCID: PMC3392895 DOI: 10.1007/s00702-010-0428-1] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 05/22/2010] [Indexed: 11/27/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative movement disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra and depletion of dopamine in the striatum, which lead to pathological and clinical abnormalities. Increasing evidence has demonstrated that inflammation is the fundamental process contributing to neuron death in PD. Neuroinflammation, which is characterized by activated microglia and infiltrating T cells at sites of neuronal injury, is a prominent contributor to the pathogenesis of progressive PD. Microglia play a critical role in forming a self-propelling cycle leading to sustained chronic neuroinflammation and driving the progressive neurodegeneration in PD. This activation depends heavily on the respiratory burst within the microglia, which in turn regulates a number of downstream pro-inflammatory activities. On the other hand, the adaptive immune responses, most notably T cells, are now emerging as important components of the inflammatory response that contribute to the pathogenesis of PD. This review paper focus on the understanding of the inflammatory etiology of PD, as well as the molecular signaling involved in this inflammatory response, with the aim to provide more effective treatments to slow down or halt the progression of chronic inflammation-induced CNS disorders, such as PD.
Collapse
Affiliation(s)
- Li Qian
- Comprehensive Center for Inflammatory Disorders, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
31
|
Abstract
Autophagy is a term used to describe the process by which lysosomes degrade intracellular components. Known originally as an adaptive response to nutrient deprivation, autophagy has now been recognized to play important roles in several human disorders including neurodegenerative diseases. Experimental results from genetic, cellular, and toxicological studies indicate that many of the etiological factors associated with Parkinson disease (PD) can perturb the autophagic process in various model systems. Thus, the emerging data support the view that dysregulation of autophagy may play a critical role in the pathogenic process of PD.
Collapse
Affiliation(s)
- Qian Yang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322-3090, USA
| | | |
Collapse
|
32
|
Suárez I, Bodega G, Fernández B. Upregulation of alpha-synuclein expression in the rat cerebellum in experimental hepatic encephalopathy. Neuropathol Appl Neurobiol 2010; 36:422-35. [PMID: 20345648 DOI: 10.1111/j.1365-2990.2010.01083.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS The overexpression of alpha-synuclein has been associated with neurodegenerative diseases, especially when the protein aggregates to form insoluble structures. The present study examined the effect of chronic hyperammonaemia on alpha-synuclein expression in the rat cerebellum following portacaval anastomosis (PCA). METHODS Immunohistochemical and western blot determinations were performed 1 month and 6 months after the PCA procedure. RESULTS A time-dependent increase in alpha-synuclein expression was seen in the cerebellar grey matter compared with the controls. At 1 month post PCA, alpha-synuclein-immunopositive material was observed in the molecular layer, while the Purkinje cells showed weak alpha-synuclein expression, and alpha-synuclein aggregates were observed throughout the granular layer. At 6 months post PCA, alpha-synuclein expression was significantly increased compared with the controls. alpha-synuclein-immunostained astroglial cells were also found; the Bergmann glial cells showed alpha-synuclein-positive processes in the molecular layer of PCA-exposed rats, and in the granular layer, perivascular astrocytes showed intense alpha-synuclein immunoreactivity, as indicated by colocalization of alpha-synuclein with glial fibrillary acidic protein (GFAP). In addition, ubiquitin-immunoreactive inclusions were present in PCA-exposed rats, although they did not colocalize with alpha-synuclein. Western blotting performed at 6 months post PCA showed a reduction in the level of soluble alpha-synuclein compared with 1 month post PCA and the controls; this reduction was concomitant with an increase in the insoluble form of alpha-synuclein. CONCLUSIONS Although the precise mechanism by which alpha-synuclein aggregates in PCA-treated rats remains unknown, the present data suggest an important role for this protein in the onset and progression of hepatic encephalopathy, probably via its expression in astroglial cells.
Collapse
Affiliation(s)
- I Suárez
- Departamento de Biología Celular y Genética, Universidad de Alcalá, 28871 Madrid, Spain.
| | | | | |
Collapse
|
33
|
Ramsey CP, Tsika E, Ischiropoulos H, Giasson BI. DJ-1 deficient mice demonstrate similar vulnerability to pathogenic Ala53Thr human alpha-syn toxicity. Hum Mol Genet 2010; 19:1425-37. [PMID: 20089532 DOI: 10.1093/hmg/ddq017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder. A pathological hallmark of PD is the presence of intraneuronal inclusions composed of fibrillized alpha-synuclein (alpha-syn) in affected brain regions. Mutations in the gene, PARK7, which encodes DJ-1, can cause autosomal recessive early-onset PD. Although DJ-1 has been shown to be involved in diverse biological processes, several in vitro studies suggest that it can inhibit the formation and protect against the effects of alpha-syn aggregation. We previously established and characterized transgenic mice expressing pathogenic Ala53Thr human alpha-syn (M83 mice) that develop extensive alpha-syn pathologies in the neuroaxis resulting in severe motor impairments and eventual fatality. In the current study, we have crossbred M83 mice on a DJ-1 null background (M83-DJnull mice) in efforts to determine the effects of the lack of DJ-1 in these mice. Animals were assessed and compared for survival rate, distribution of alpha-syn inclusions, biochemical properties of alpha-syn protein, demise and function of nigral dopaminergic neurons, and extent of gliosis in the neuroaxis. M83 and M83-DJnull mice displayed a similar onset of disease and pathological changes, and none of the analyses to assess for changes in pathogenesis revealed any significant differences between M83 and M83-DJnull mice. These findings suggest that DJ-1 may not function to directly modulate alpha-syn nor does DJ-1 appear to play a role in protecting against the deleterious effects of expressing pathogenic Ala53Thr alpha-syn in vivo. It is possible that alpha-syn and DJ-1 mutations may lead to PD via independent mechanisms.
Collapse
Affiliation(s)
- Chenere P Ramsey
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6084, USA
| | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Neurodegenerative brain diseases, including Alzheimer's disease, frontotemporal degeneration and Lewy body disease, are the most frequent pathologies underlying cognitive disorders in old age. This review outlines recent advances in the understanding of key molecular mechanisms involved in these neurodegenerations, particularly with regard to the abnormal processing of proteins. The consequences of these novel insights for therapeutic interventions are also explained. RECENT FINDINGS Aberrant processing, misfolding, and subsequent deposition of amyloid beta protein, TAU, alpha-synuclein, and TDP-43 are key events in the pathological cascades of neurodegenerations leading to cognitive impairment and dementia. The nonpolymerized, oligomeric forms of these proteins have neurotoxic properties including the disruption of synaptic function and the induction of oxidative stress. The aggregation and deposition of these proteins may represent a neuronal repair mechanism which ultimately worsens the deleterious effects of the preaggregated forms. Novel disease-modifying treatment strategies aim at down-regulating protein production, inhibiting polymerization, or removing preaggregated forms of the proteins from the brain. SUMMARY Recent research has elucidated important molecular events in neurodegenerative diseases upstream of the aggregation and deposition of proteins which forms their histopathological hallmarks. These insights translate into novel therapeutic strategies which are currently evaluated in clinical trials.
Collapse
|
35
|
Repression of alpha-synuclein expression and toxicity by microRNA-7. Proc Natl Acad Sci U S A 2009; 106:13052-7. [PMID: 19628698 DOI: 10.1073/pnas.0906277106] [Citation(s) in RCA: 513] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
alpha-Synuclein is a key protein in Parkinson's disease (PD) because it accumulates as fibrillar aggregates in pathologic hallmark features in affected brain regions, most notably in nigral dopaminergic neurons. Intraneuronal levels of this protein appear critical in mediating its toxicity, because multiplication of its gene locus leads to autosomal dominant PD, and transgenic animal models overexpressing human alpha-synuclein manifest impaired function or decreased survival of dopaminergic neurons. Here, we show that microRNA-7 (miR-7), which is expressed mainly in neurons, represses alpha-synuclein protein levels through the 3'-untranslated region (UTR) of alpha-synuclein mRNA. Importantly, miR-7-induced down-regulation of alpha-synuclein protects cells against oxidative stress. Further, in the MPTP-induced neurotoxin model of PD in cultured cells and in mice, miR-7 expression decreases, possibly contributing to increased alpha-synuclein expression. These findings provide a mechanism by which alpha-synuclein levels are regulated in neurons, have implications for the pathogenesis of PD, and suggest miR-7 as a therapeutic target for PD and other alpha-synucleinopathies.
Collapse
|
36
|
Krenz A, Falkenburger BH, Gerhardt E, Drinkut A, Schulz JB. Aggregate formation and toxicity by wild-type and R621C synphilin-1 in the nigrostriatal system of mice using adenoviral vectors. J Neurochem 2009; 108:139-46. [PMID: 19094062 DOI: 10.1111/j.1471-4159.2008.05755.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synphilin-1 was described as a protein interacting with alpha-synuclein and is commonly found in Lewy bodies, the pathological hallmark of Parkinson's disease (PD). Our group has previously described and characterized in vitro a mutation in the synphilin-1 gene (R621C) in PD patients. Providing the first characterization of synphilin-1 expression in an animal model, we here used adenoviral gene transfer to study the effects of wild-type (WT) and R621C synphilin-1 in dopaminergic neurons in mouse brain. As synphilin-1 is commonly used to trigger aggregation of alpha-synuclein in cell culture, we investigated not only non-transgenic C57Bl/6 mice but also A30P-alpha-synuclein transgenic animals. Both WT synphilin-1 and R621C synphilin-1 led to the formation of Thioflavine-S positive inclusions in C57Bl/6 mice and degeneration of dopaminergic neurons in the substantia nigra. R621C synphilin-1 induced more aggregate formation than WT synphilin-1 in A30P-alpha-synuclein transgenic mice, consistent with the role of the R621C mutation as a susceptibility factor for PD. Synphilin-1 expression may be used to improve current mouse models of PD, as it induced both the formation of aggregates and degeneration of dopaminergic neurons, two core characteristics of PD that have not been well reproduced with expression of alpha-synuclein.
Collapse
Affiliation(s)
- Antje Krenz
- Department of Neurodegeneration and Restorative Research, DFG Research Center for Molecular Physiology of Brain and Center for Neurological Medicine, University of Göttingen, Göttingen, Germany
| | | | | | | | | |
Collapse
|
37
|
Abstract
PD is the second most common neurodegenerative disease, and affects 5% of the population by the age of 85. PD is a multi-factorial disease with a complex etiology including genetic risk factors, environmental exposure and aging. The pathogenesis is not fully understood. Here we review research on the genetic and environmental causes of PD and the current research models. None of the single models replicate all the features of PD. Genetic models (possibly including more than one mutation) in combination with toxins or other environmental manipulation may provide better models of PD pathogenesis.
Collapse
|
38
|
Cookson MR. alpha-Synuclein and neuronal cell death. Mol Neurodegener 2009; 4:9. [PMID: 19193223 PMCID: PMC2646729 DOI: 10.1186/1750-1326-4-9] [Citation(s) in RCA: 262] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 02/04/2009] [Indexed: 11/16/2022] Open
Abstract
α-Synuclein is a small protein that has special relevance for understanding Parkinson disease and related disorders. Not only is α-synuclein found in Lewy bodies characteristic of Parkinson disease, but also mutations in the gene for α-synuclein can cause an inherited form of Parkinson disease and expression of normal α-synuclein can increase the risk of developing Parkinson disease in sporadic, or non-familial, cases. Both sporadic and familial Parkinson disease are characterized by substantial loss of several groups of neurons, including the dopaminergic cells of the substantia nigra that are the target of most current symptomatic therapies. Therefore, it is predicted that α-synuclein, especially in its mutant forms or under conditions where its expression levels are increased, is a toxic protein in the sense that it is associated with an increased rate of neuronal cell death. This review will discuss the experimental contexts in which α-synuclein has been demonstrated to be toxic. I will also outline what is known about the mechanisms by which α-synuclein triggers neuronal damage, and identify some of the current gaps in our knowledge about this subject. Finally, the therapeutic implications of toxicity of α-synuclein will be discussed.
Collapse
Affiliation(s)
- Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Building 35, Room 1A116, MSC 3707, 35 Convent Drive, Bethesda, MD 20982-3707, USA.
| |
Collapse
|
39
|
Cui M, Huang Y, Zhao Y, Zheng J. New insights for FOXO and cell-fate decision in HIV infection and HIV associated neurocognitive disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 665:143-59. [PMID: 20429422 DOI: 10.1007/978-1-4419-1599-3_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human immunodeficiency virus Type 1 (HIV-1) infection and associated diseases continue to represent major health problem worldwide. FOXO transcriptional factors play an important role in the regulation of cell apoptosis, cell cycle arrest, stress resistance, metabolism and differentiation. This chapter will discuss the diverse functions of FOXO in different cell types including T-cells, macrophages, neurons and astrocytes within the context of HIV-1 infection. Given the overwhelming evidence that FOXO proteins influence the cell fate of immune cells and involve in the homeostasis of the central nervous system (CNS), we will also discuss the potential role of FOXO factors in HIV-1-associated neurological disorders.
Collapse
Affiliation(s)
- Min Cui
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|
40
|
Houghton EA, Nicholas KM. In vitro reactive oxygen species production by histatins and copper(I,II). J Biol Inorg Chem 2008; 14:243-51. [DOI: 10.1007/s00775-008-0444-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 10/09/2008] [Indexed: 12/15/2022]
|
41
|
Cole NB, Dieuliis D, Leo P, Mitchell DC, Nussbaum RL. Mitochondrial translocation of alpha-synuclein is promoted by intracellular acidification. Exp Cell Res 2008; 314:2076-89. [PMID: 18440504 DOI: 10.1016/j.yexcr.2008.03.012] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 03/11/2008] [Accepted: 03/12/2008] [Indexed: 10/22/2022]
Abstract
Mitochondrial dysfunction plays a central role in the selective vulnerability of dopaminergic neurons in Parkinson's disease (PD) and is influenced by both environmental and genetic factors. Expression of the PD protein alpha-synuclein or its familial mutants often sensitizes neurons to oxidative stress and to damage by mitochondrial toxins. This effect is thought to be indirect, since little evidence physically linking alpha-synuclein to mitochondria has been reported. Here, we show that the distribution of alpha-synuclein within neuronal and non-neuronal cells is dependent on intracellular pH. Cytosolic acidification induces translocation of alpha-synuclein from the cytosol onto the surface of mitochondria. Translocation occurs rapidly under artificially-induced low pH conditions and as a result of pH changes during oxidative or metabolic stress. Binding is likely facilitated by low pH-induced exposure of the mitochondria-specific lipid cardiolipin. These results imply a direct role for alpha-synuclein in mitochondrial physiology, especially under pathological conditions, and in principle, link alpha-synuclein to other PD genes in regulating mitochondrial homeostasis.
Collapse
Affiliation(s)
- Nelson B Cole
- Genetic Diseases Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
42
|
Petrucelli L, Dickson DW. Neuropathology of Parkinson's Disease. PARKINSONS DISEASE 2008. [DOI: 10.1016/b978-0-12-374028-1.00003-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
43
|
Cookson MR, van der Brug M. Cell systems and the toxic mechanism(s) of alpha-synuclein. Exp Neurol 2007; 209:5-11. [PMID: 17603039 PMCID: PMC2231843 DOI: 10.1016/j.expneurol.2007.05.022] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 05/02/2007] [Accepted: 05/28/2007] [Indexed: 10/25/2022]
Abstract
Mutations in the SNCA gene are causal for familial Parkinson disease/Lewy body disease. alpha-Synuclein is a small acidic protein that binds loosely to the surface of vesicles and may play a role in synaptic dynamics, although its normal function remains somewhat unclear. What is clear is that point mutations or increased expression of wild type alpha-synuclein causes disease. A great deal of literature supports the overall hypothesis that alpha-synuclein is damaging to neurons because it is inherently prone to aggregation; mutations or increased concentration of the protein both increase this tendency. An unproven, but popular, contention is that the toxic species are small oligomers that are relatively soluble, which may react with membranes to damage key processes within the cell. The details of this process, especially in determining the order of events and the requirement of particular processes in cell death, are unclear. Derangements in vesicle processing, including synaptic function, protein turnover, mitochondrial function and oxidative stress, have all been suggested to occur. Whether there is a sequence of events or whether these are interacting effects is unclear, but the outcome is to trigger cell death, by both apoptotic and non-apoptotic mechanisms depending on the system studied. In this article, we develop a framework for thinking about alpha-synuclein in terms of initiating events and secondary processes that are required to trigger neuronal dysfunction and cell death.
Collapse
Affiliation(s)
- Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20982-3707, USA.
| | | |
Collapse
|