1
|
Ji YW, Wen XY, Tang HP, Jin ZS, Su WT, Zhou L, Xia ZY, Xia ZY, Lei SQ. DJ-1: Potential target for treatment of myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 179:117383. [PMID: 39232383 DOI: 10.1016/j.biopha.2024.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Ischemic heart disease (IHD) is a significant global health concern, resulting in high rates of mortality and disability among patients. Although coronary blood flow reperfusion is a key treatment for IHD, it often leads to acute myocardial ischemia-reperfusion injury (IRI). Current intervention strategies have limitations in providing adequate protection for the ischemic myocardium. DJ-1, originally known as a Parkinson's disease related protein, is a highly conserved cytoprotective protein. It is involved in enhancing mitochondrial function, scavenging reactive oxygen species (ROS), regulating autophagy, inhibiting apoptosis, modulating anaerobic metabolism, and exerting anti-inflammatory effects. DJ-1 is also required for protective strategies, such as ischemic preconditioning, ischemic postconditioning, remote ischemic preconditioning and pharmacological conditioning. Therefore, DJ-1 emerges as a potential target for the treatment of myocardial IRI. Our comprehensive review delves into its protective mechanisms in myocardial IRI and the structural foundations underlying its functions.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Lin J, Zheng X, Xiong Z, Xiang Q, Zhao Y, Jiang S, Sun Z, Fan D, Sun C, Li W. DJ-1-mediated p62 degradation delays intervertebral disc degeneration by inhibiting apoptosis of nucleus pulposus cells. Apoptosis 2023; 28:1357-1371. [PMID: 37300741 DOI: 10.1007/s10495-023-01862-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Intervertebral disc degeneration (IDD) is the most important pathological basis of degenerative spinal diseases, for which effective interventions are still lacking. Oxidative stress is considered to be one of the leading pathological mechanisms contributing to IDD. However, the exact role of DJ-1 as an essential member of the antioxidant defense system in IDD is still unclear. Therefore, the aim of this study was to investigate the role played by DJ-1 in IDD and to reveal its potential molecular mechanisms. Western blot and immunohistochemical staining assays were performed to detect the expression of DJ-1 in degenerative nucleus pulposus cells (NPCs). After overexpression of DJ-1 in NPCs by lentiviral transfection, DCFH-DA and MitoSOX fluorescent probes were used to evaluate the levels of reactive oxygen species (ROS); while western blot, TUNEL staining, and Caspase-3 activity were used to assess apoptosis. Immunofluorescence staining was used to demonstrate the relationship between DJ-1 and p62. After inhibition of lysosomal degradation function with chloroquine, p62 degradation and apoptosis in DJ-1 overexpressing NPCs were further examined. In vivo, we assessed the therapeutic effect of upregulated DJ-1 on IDD by X-ray, MRI and Safranin O-Fast green staining. The protein expression of DJ-1 was significantly decreased in degenerated NPCs, accompanied by increased apoptosis. However, overexpression of DJ-1 significantly inhibited the elevated ROS levels and apoptosis in NPCs under oxidative stress. Mechanistically, our results showed that upregulation of DJ-1 promoted p62 degradation via the autophagic lysosomal pathway and that the protective effect of DJ-1 on NPCs under oxidative stress was partially mediated by promoting lysosomal pathway degradation of p62. Moreover, intradiscal injection of adeno-associated virus for overexpression of DJ-1 mitigated the progression of IDD in rats. This study reveals that DJ-1 maintains the homeostasis of NPCs by promoting the degradation of p62 through the autophagic lysosomal pathway, suggesting that DJ-1 is a promising new target for IDD intervention.
Collapse
Affiliation(s)
- Jialiang Lin
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Xuanqi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Zhencheng Xiong
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qian Xiang
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Yongzhao Zhao
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Shuai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Zhuoran Sun
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Dongwei Fan
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Chuiguo Sun
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China.
| |
Collapse
|
3
|
Hu H, Su M, Ba H, Chen G, Luo J, Liu F, Liao X, Cao Z, Zeng J, Lu H, Xiong G, Chen J. ZIF-8 nanoparticles induce neurobehavioral disorders through the regulation of ROS-mediated oxidative stress in zebrafish embryos. CHEMOSPHERE 2022; 305:135453. [PMID: 35752317 DOI: 10.1016/j.chemosphere.2022.135453] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Zeolite imidazolate framework-8 (ZIF-8) is a nanomaterial of metal-organic frameworks (MOFs), which have various applications in drug delivery and water pollution remediation. However, little is known about its developmental neurotoxicity in aquatic organisms, especially on the low-level exposure. In the present study, we investigated the toxic effects of ZIF-8 NPs on the neuron development, behavioral traits, oxidative stress and gene expression in zebrafish embryos. Firstly, our results showed that ZIF-8 induced significantly embryonic malformations and abnormal development of nervous system in zebrafish embryos with a concentration-dependent manner. Meanwhile, the locomotor behavior was obviously inhibited while the anxiety behavior was greatly increased after ZIF-8 exposure. Secondly, the levels of ROS and antioxidant enzyme activities (CAT, SOD and MDA) together with AChE and ATPase were substantially increased in the ZIF-8 exposed groups. At the molecular level, ZIF-8 NPs could down-regulate the expression profiles of neural development-related genes (gap43, synapsin 2a and neurogenin 1) and PD-like related genes (dj-1, dynactin and parkin), but up-regulate the expression levels of neuro-inflammatory genes (nox-1, glip1a and glip1b) in larval zebrafish. In addition, we further explored the molecular mechanism of neurotoxicity induced by ZIF-8 with pharmacological experiments. The results showed that specific inhibition of ROS-mediated oxidative stress by the astaxanthin could reverse the expression patterns of ATPase, AChE and neurodevelopmental genes. Moreover, astaxanthin can partially rescue the ZIF-8-modulated locomotor behavior. Taken together, our results demonstrated that ZIF-8 had the potential to cause neurotoxicity in zebrafish embryos. These informations presented in this study will help to elucidate the molecular mechanisms of ZIF-8 nanoparticles exposure in zebrafish, which providing a scientific evaluation of its safety to aquatic ecosystems.
Collapse
Affiliation(s)
- Hongmei Hu
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Meile Su
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Huixia Ba
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Guilan Chen
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Jiaqi Luo
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Fasheng Liu
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Zigang Cao
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Junquan Zeng
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Huiqiang Lu
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China
| | - Guanghua Xiong
- Center of Clinical Medicine Research, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China.
| | - Jianjun Chen
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| |
Collapse
|
4
|
Guatteo E, Berretta N, Monda V, Ledonne A, Mercuri NB. Pathophysiological Features of Nigral Dopaminergic Neurons in Animal Models of Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23094508. [PMID: 35562898 PMCID: PMC9102081 DOI: 10.3390/ijms23094508] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 12/21/2022] Open
Abstract
The degeneration of nigral dopaminergic neurons is considered the hallmark of Parkinson’s disease (PD), and it is triggered by different factors, including mitochondrial dysfunction, Lewy body accumulation, neuroinflammation, excitotoxicity and metal accumulation. Despite the extensive literature devoted to unravelling the signalling pathways involved in neuronal degeneration, little is known about the functional impairments occurring in these cells during illness progression. Of course, it is not possible to obtain direct information on the properties of the dopaminergic cells in patients. However, several data are available in the literature reporting changes in the function of these cells in PD animal models. In the present manuscript, we focus on dopaminergic neuron functional properties and summarize shared or peculiar features of neuronal dysfunction in different PD animal models at different stages of the disease in an attempt to design a picture of the functional modifications occurring in nigral dopaminergic neurons during disease progression preceding their eventual death.
Collapse
Affiliation(s)
- Ezia Guatteo
- Department of Motor Science and Wellness, University of Naples Parthenope, 80133 Naples, Italy; (E.G.); (V.M.)
- Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
| | - Nicola Berretta
- Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
| | - Vincenzo Monda
- Department of Motor Science and Wellness, University of Naples Parthenope, 80133 Naples, Italy; (E.G.); (V.M.)
| | - Ada Ledonne
- Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
- Correspondence: (A.L.); (N.B.M.)
| | - Nicola Biagio Mercuri
- Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, 00143 Rome, Italy
- Correspondence: (A.L.); (N.B.M.)
| |
Collapse
|
5
|
Beckstead MJ, Howell RD. Progressive parkinsonism due to mitochondrial impairment: Lessons from the MitoPark mouse model. Exp Neurol 2021; 341:113707. [PMID: 33753138 PMCID: PMC8169575 DOI: 10.1016/j.expneurol.2021.113707] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/21/2022]
Abstract
The cardinal pathophysiological finding of Parkinson's disease (PD) is a chronic, progressive degeneration of dopamine (DA) neurons in the substantia nigra, which is responsible for the motor and some of the non-motor symptomatology. While the primary causes of nigrostriatal degeneration are hotly debated, considerable evidence supports a central role for impaired mitochondrial function. Postmortem analysis of PD patients reveals impaired respiratory chains and increased mutations of mitochondrial DNA (mtDNA), in addition to increased markers of oxidative stress indicative of mitochondrial impairment. Most animal models of PD, both genetic and toxin-based, target some component of mitochondrial function to reproduce aspects of the human disease. One model that continues to gain attention is the MitoPark mouse, created through a cell type-specific knockout of mitochondrial transcription factor A specifically in midbrain DA neurons. This model effectively recapitulates the slowly developing, adult onset motor decline seen in PD due to mass loss of DA neurons. MitoPark mice therefore represent an effective tool for studying the sequence of events that occurs in the early stages of DA neuron degeneration following mitochondrial impairment, as well as for testing the efficacy of potential disease-modifying therapies in a progressive model of neurodegeneration. A targeted review of key findings from MitoPark mice has not been published since the early years following the initial report of the model in 2007. The current review synthesizes findings from several groups that are exploring MitoPark mice and discusses implications for the future identification of disease-modifying treatments for PD.
Collapse
Affiliation(s)
- Michael J Beckstead
- Oklahoma Medical Research Foundation, Aging & Metabolism Research Program, USA.
| | - Rebecca D Howell
- Oklahoma Medical Research Foundation, Aging & Metabolism Research Program, USA
| |
Collapse
|
6
|
Zhao N, Wang T, Peng L, Li Y, Zhao Y, Yu S. Attenuation of Inflammation by DJ-1 May Be a Drug Target for Cerebral Ischemia-Reperfusion Injury. Neurochem Res 2021; 46:1470-1479. [PMID: 33683631 DOI: 10.1007/s11064-021-03288-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 12/28/2022]
Abstract
The pathophysiological process of cerebral apoplexy is complex, and there are currently no specific drugs for this condition. The study of effective drug targets has become a hot topic in neuroscience. Currently, adeno-associated viruses (AAVs) and polypeptides are commonly used in drug research. DJ-1 has been widely considered a neuroprotective target in recent times, but the mechanism of its neuroprotective effects is unclear. In this study, we simulated ischemic injury by establishing a middle cerebral artery occlusion reperfusion (MCAO/R) model to compare the protective effect of DJ-1 overexpression induced by DJ-1 AAV and ND-13 on cerebral ischemia-reperfusion (I/R) injury. We found that DJ-1 overexpression and ND-13 significantly reduced the neurological function scores and infarct volume and alleviated pathological damage to brain tissue. In addition, Western blotting, ELISA and immunofluorescence labeling revealed that DJ-1 overexpression and ND-13 increased the expression of the anti-inflammatory cytokines IL-10 and IL-4, and decreased the levels of the pro-inflammatory cytokines IL-1β and TNF-α. In summary, our study shows that DJ-1 overexpression and ND-13 can regulate the expression of inflammatory factors and alleviate cerebral I/R injury. Thus, DJ-1 is a possible drug target for cerebral I/R injury.
Collapse
Affiliation(s)
- Na Zhao
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tingting Wang
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Li Peng
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yumei Li
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yong Zhao
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shanshan Yu
- Department of Pathology, Basic Medical College, Chongqing Medical University, Yixueyuan Road 1, Chongqing, 400016, People's Republic of China.
- Molecular Medical Laboratory, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
7
|
De Lazzari F, Prag HA, Gruszczyk AV, Whitworth AJ, Bisaglia M. DJ-1: A promising therapeutic candidate for ischemia-reperfusion injury. Redox Biol 2021; 41:101884. [PMID: 33561740 PMCID: PMC7872972 DOI: 10.1016/j.redox.2021.101884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 12/31/2022] Open
Abstract
DJ-1 is a multifaceted protein with pleiotropic functions that has been implicated in multiple diseases, ranging from neurodegeneration to cancer and ischemia-reperfusion injury. Ischemia is a complex pathological state arising when tissues and organs do not receive adequate levels of oxygen and nutrients. When the blood flow is restored, significant damage occurs over and above that of ischemia alone and is termed ischemia-reperfusion injury. Despite great efforts in the scientific community to ameliorate this pathology, its complex nature has rendered it challenging to obtain satisfactory treatments that translate to the clinic. In this review, we will describe the recent findings on the participation of the protein DJ-1 in the pathophysiology of ischemia-reperfusion injury, firstly introducing the features and functions of DJ-1 and, successively highlighting the therapeutic potential of the protein. DJ-1 has been shown to confer protection in ischemia-reperfusion injury models. DJ-1 protection relies on the activation of antioxidant signaling pathways. DJ-1 regulates mitochondrial homeostasis during ischemia and reperfusion. DJ-1 seems to modulate ion homeostasis during ischemia and reperfusion. DJ-1 may represent a promising therapeutic target for ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Federica De Lazzari
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy
| | - Hiran A Prag
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Marco Bisaglia
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
8
|
Imbriani P, D'Angelo V, Platania P, Di Lazzaro G, Scalise S, Salimei C, El Atiallah I, Colona VL, Mercuri NB, Bonsi P, Pisani A, Schirinzi T, Martella G. Ischemic injury precipitates neuronal vulnerability in Parkinson's disease: Insights from PINK1 mouse model study and clinical retrospective data. Parkinsonism Relat Disord 2020; 74:57-63. [PMID: 32335490 DOI: 10.1016/j.parkreldis.2020.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Increasing evidence demonstrates the relevant association between Parkinson's disease (PD) and vascular diseases/risk factors, as well as a worse clinico-pathological progression in those patients with vascular comorbidity. The mechanisms underlying this relationship have not been clarified yet, although their comprehension is critical in a perspective of disease-modifying treatments development or prevention. METHODS We performed an experimental protocol of ischemic injury (glucose-oxygen deprivation, OGD) on PTEN-induced kinase 1 knockout (PINK1-/-) mice, a well-established PD model, looking at both electrophysiological and morphological changes in basal ganglia. In addition, 253 PD patients were retrospectively analysed, to estimate the prevalence of vascular risk factors. RESULTS In PINK1-/- mice, the OGD protocol induced electrophysiological (prolonged depolarization) and morphological alterations (picnotic cells, cellular loss and swelling, thickening of nuclear chromatin) in striatal medium spiny neurons and nigral dopaminergic neurons. Vascular comorbidity occurred in 75% of PD patients. CONCLUSIONS The ischemic injury precipitates neuronal vulnerability in basal ganglia of PINK1-/- mice, probably through an impairment of mitochondrial metabolism and higher oxidative stress. These experimental data may provide a potential mechanistic explanation for both the association between vascular diseases and PD and their reciprocal interactions in determining the clinico-pathological burden of PD patients.
Collapse
Affiliation(s)
- Paola Imbriani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Vincenza D'Angelo
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Paola Platania
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Simona Scalise
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Chiara Salimei
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Ilham El Atiallah
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Vito Luigi Colona
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Antonio Pisani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy.
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giuseppina Martella
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
9
|
Oliveira RB, Petiz LL, Lim R, Lipski J, Gravina FS, Brichta AM, Callister RJ, Leão RN, Helden DF. Crosstalk between mitochondria, calcium channels and actin cytoskeleton modulates noradrenergic activity of locus coeruleus neurons. J Neurochem 2019; 149:471-487. [DOI: 10.1111/jnc.14692] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Ramatis B. Oliveira
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
- Health and Medical Research Group School of Medicine University of the Taquari Valley ‐ Univates Lajeado RS Brazil
- Bairro Universitário Lajeado RS Brazil
| | - Lyvia L. Petiz
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
- Brain Institute Federal University of Rio Grande do Norte Natal Brazil
| | - Rebecca Lim
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
| | - Janusz Lipski
- Faculty of Medical and Health Sciences University of Auckland Auckland New Zealand
| | - Fernanda S. Gravina
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
| | - Alan M. Brichta
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
| | - Robert J. Callister
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
| | - Richardson N. Leão
- Brain Institute Federal University of Rio Grande do Norte Natal Brazil
- The Beijer Laboratory for Gene and Neurosciences Department of Neuroscience Uppsala University Uppsala Sweden
| | - Dirk F. Helden
- School of Biomedical Sciences and Pharmacy University of Newcastle and Hunter Medical Research Institute Callaghan NSW Australia
| |
Collapse
|
10
|
Lin AW, Gill KK, Castañeda MS, Matucci I, Eder N, Claxton S, Flynn H, Snijders AP, George R, Ultanir SK. Chemical genetic identification of GAK substrates reveals its role in regulating Na +/K +-ATPase. Life Sci Alliance 2018; 1:e201800118. [PMID: 30623173 PMCID: PMC6312924 DOI: 10.26508/lsa.201800118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/15/2022] Open
Abstract
Novel GAK phosphorylation targets are identified using chemical genetic methods. One of the substrates is the α subunit of the Na+/K+-ATPase, phosphorylation of which is necessary for its surface trafficking from endosomes. Conserved functions of NAK family kinases are described. Cyclin G–associated kinase (GAK) is a ubiquitous serine/threonine kinase that facilitates clathrin uncoating during vesicle trafficking. GAK phosphorylates a coat adaptor component, AP2M1, to help achieve this function. GAK is also implicated in Parkinson's disease through genome-wide association studies. However, GAK's role in mammalian neurons remains unclear, and insight may come from identification of further substrates. Employing a chemical genetics method, we show here that the sodium potassium pump (Na+/K+-ATPase) α-subunit Atp1a3 is a GAK target and that GAK regulates Na+/K+-ATPase trafficking to the plasma membrane. Whole-cell patch clamp recordings from CA1 pyramidal neurons in GAK conditional knockout mice show a larger change in resting membrane potential when exposed to the Na+/K+-ATPase blocker ouabain, indicating compromised Na+/K+-ATPase function in GAK knockouts. Our results suggest a modulatory role for GAK via phosphoregulation of substrates such as Atp1a3 during cargo trafficking.
Collapse
Affiliation(s)
- Amy W Lin
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Kalbinder K Gill
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | | | - Irene Matucci
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom.,Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | - Suzanne Claxton
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Helen Flynn
- Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Roger George
- Protein Purification Facility, The Francis Crick Institute, London, United Kingdom
| | - Sila K Ultanir
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
11
|
Himmelberg MM, West RJH, Elliott CJH, Wade AR. Abnormal visual gain control and excitotoxicity in early-onset Parkinson's disease Drosophila models. J Neurophysiol 2018; 119:957-970. [PMID: 29142100 PMCID: PMC5899316 DOI: 10.1152/jn.00681.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022] Open
Abstract
The excitotoxic theory of Parkinson's disease (PD) hypothesizes that a pathophysiological degeneration of dopaminergic neurons stems from neural hyperactivity at early stages of disease, leading to mitochondrial stress and cell death. Recent research has harnessed the visual system of Drosophila PD models to probe this hypothesis. Here, we investigate whether abnormal visual sensitivity and excitotoxicity occur in early-onset PD (EOPD) Drosophila models DJ-1αΔ72, DJ-1βΔ 93, and PINK15. We used an electroretinogram to record steady-state visually evoked potentials driven by temporal contrast stimuli. At 1 day of age, all EOPD mutants had a twofold increase in response amplitudes compared with w̄ controls. Furthermore, we found that excitotoxicity occurs in older EOPD models after increased neural activity is triggered by visual stimulation. In an additional analysis, we used a linear discriminant analysis to test whether there were subtle variations in neural gain control that could be used to classify Drosophila into their correct age and genotype. The discriminant analysis was highly accurate, classifying Drosophila into their correct genotypic class at all age groups at 50-70% accuracy (20% chance baseline). Differences in cellular processes link to subtle alterations in neural network operation in young flies, all of which lead to the same pathogenic outcome. Our data are the first to quantify abnormal gain control and excitotoxicity in EOPD Drosophila mutants. We conclude that EOPD mutations may be linked to more sensitive neuronal signaling in prodromal animals that may cause the expression of PD symptomologies later in life. NEW & NOTEWORTHY Steady-state visually evoked potential response amplitudes to multivariate temporal contrast stimuli were recorded in early-onset PD Drosophila models. Our data indicate that abnormal gain control and a subsequent visual loss occur in these PD mutants, supporting a broader excitotoxicity hypothesis in genetic PD. Furthermore, linear discriminant analysis could accurately classify Drosophila into their correct genotype at different ages throughout their lifespan. Our results suggest increased neural signaling in prodromal PD patients.
Collapse
Affiliation(s)
- Marc M Himmelberg
- Department of Psychology, The University of York , York , United Kingdom
| | - Ryan J H West
- Department of Biology, The University of York , York , United Kingdom
| | | | - Alex R Wade
- Department of Psychology, The University of York , York , United Kingdom
| |
Collapse
|
12
|
Abstract
Oxidative stress has been implicated as a core contributor to the initiation and progression of multiple neurological diseases. Genetic and environmental factors can produce oxidative stress through mitochondrial dysfunction leading to the degeneration of dopaminergic and other neurons underlying Parkinson disease (PD). Although clinical trials of antioxidants have thus far failed to demonstrate slowed progression of PD, oxidative stress remains a compelling target. Rather than prompting abandonment of antioxidant strategies, these failures have raised the bar for justifying drug and dosing selections and for improving study designs to test for disease modification by antioxidants. Urate, the main antioxidant found in plasma as well as the end product of purine metabolism in humans, has emerged as a promising potential neuroprotectant with advantages that distinguish it from previously tested antioxidant agents. Uniquely, higher urate levels in plasma or cerebrospinal fluid (CSF) have been linked to both a lower risk of developing PD and to a slower rate of its subsequent progression in numerous large prospective epidemiological and clinical cohorts. Laboratory evidence that urate confers neuroprotection in cellular and animal models of PD, possibly via the Nrf2 antioxidant response pathway, further strengthened its candidacy for rapid clinical translation. An early phase trial of the urate precursor inosine demonstrated its capacity to safely produce well tolerated, long-term elevation of plasma and CSF urate in early PD, supporting a phase 3 trial now underway to determine whether oral inosine dosed to elevate urate to concentrations predictive of favorable prognosis in PD slows clinical decline in people with recently diagnosed, dopamine transporter-deficient PD.
Collapse
Affiliation(s)
- Grace F Crotty
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Alberto Ascherio
- Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, MA, USA
| | | |
Collapse
|
13
|
Requejo-Aguilar R, Bolaños JP. Mitochondrial control of cell bioenergetics in Parkinson's disease. Free Radic Biol Med 2016; 100:123-137. [PMID: 27091692 PMCID: PMC5065935 DOI: 10.1016/j.freeradbiomed.2016.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra. The earliest biochemical signs of the disease involve failure in mitochondrial-endoplasmic reticulum cross talk and lysosomal function, mitochondrial electron chain impairment, mitochondrial dynamics alterations, and calcium and iron homeostasis abnormalities. These changes are associated with increased mitochondrial reactive oxygen species (mROS) and energy deficiency. Recently, it has been reported that, as an attempt to compensate for the mitochondrial dysfunction, neurons invoke glycolysis as a low-efficient mode of energy production in models of PD. Here, we review how mitochondria orchestrate the maintenance of cellular energetic status in PD, with special focus on the switch from oxidative phosphorylation to glycolysis, as well as the implication of endoplasmic reticulum and lysosomes in the control of bioenergetics.
Collapse
Affiliation(s)
- Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Institute Maimonides of Biomedical Investigation of Cordoba (IMIBIC), Cordoba, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC, Zacarias Gonzalez, 2, 37007 Salamanca, Spain.
| |
Collapse
|
14
|
Ruegsegger C, Maharjan N, Goswami A, Filézac de L'Etang A, Weis J, Troost D, Heller M, Gut H, Saxena S. Aberrant association of misfolded SOD1 with Na(+)/K(+)ATPase-α3 impairs its activity and contributes to motor neuron vulnerability in ALS. Acta Neuropathol 2016; 131:427-51. [PMID: 26619836 DOI: 10.1007/s00401-015-1510-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/03/2015] [Accepted: 11/14/2015] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult onset progressive motor neuron disease with no cure. Transgenic mice overexpressing familial ALS associated human mutant SOD1 are a commonly used model for examining disease mechanisms. Presently, it is well accepted that alterations in motor neuron excitability and spinal circuits are pathological hallmarks of ALS, but the underlying molecular mechanisms remain unresolved. Here, we sought to understand whether the expression of mutant SOD1 protein could contribute to altering processes governing motor neuron excitability. We used the conformation specific antibody B8H10 which recognizes a misfolded state of SOD1 (misfSOD1) to longitudinally identify its interactome during early disease stage in SOD1G93A mice. This strategy identified a direct isozyme-specific association of misfSOD1 with Na(+)/K(+)ATPase-α3 leading to the premature impairment of its ATPase activity. Pharmacological inhibition of Na(+)/K(+)ATPase-α3 altered glutamate receptor 2 expression, modified cholinergic inputs and accelerated disease pathology. After mapping the site of direct association of misfSOD1 with Na(+)/K(+)ATPase-α3 onto a 10 amino acid stretch that is unique to Na(+)/K(+)ATPase-α3 but not found in the closely related Na(+)/K(+)ATPase-α1 isozyme, we generated a misfSOD1 binding deficient, but fully functional Na(+)/K(+)ATPase-α3 pump. Adeno associated virus (AAV)-mediated expression of this chimeric Na(+)/K(+)ATPase-α3 restored Na(+)/K(+)ATPase-α3 activity in the spinal cord, delayed pathological alterations and prolonged survival of SOD1G93A mice. Additionally, altered Na(+)/K(+)ATPase-α3 expression was observed in the spinal cord of individuals with sporadic and familial ALS. A fraction of sporadic ALS cases also presented B8H10 positive misfSOD1 immunoreactivity, suggesting that similar mechanism might contribute to the pathology.
Collapse
Affiliation(s)
- Céline Ruegsegger
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Niran Maharjan
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Anand Goswami
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Audrey Filézac de L'Etang
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Neuroscience, Genentech, Inc., South San Francisco, California, USA
| | - Joachim Weis
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Dirk Troost
- Division of Neuropathology, Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Manfred Heller
- Department of Clinical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Heinz Gut
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Smita Saxena
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
15
|
van der Merwe C, Jalali Sefid Dashti Z, Christoffels A, Loos B, Bardien S. Evidence for a common biological pathway linking three Parkinson's disease-causing genes: parkin, PINK1 and DJ-1. Eur J Neurosci 2015; 41:1113-25. [PMID: 25761903 DOI: 10.1111/ejn.12872] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/29/2015] [Accepted: 02/10/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is characterised by the loss of dopaminergic neurons in the midbrain. Autosomal recessive, early-onset cases of PD are predominantly caused by mutations in the parkin, PINK1 and DJ-1 genes. Animal and cellular models have verified a direct link between parkin and PINK1, whereby PINK1 phosphorylates and activates parkin at the outer mitochondrial membrane, resulting in removal of dysfunctional mitochondria via mitophagy. Despite the overwhelming evidence for this interaction, few studies have been able to identify a link for DJ-1 with parkin or PINK1. The aim of this review is to summarise the functions of these three proteins, and to analyse the existing evidence for direct and indirect interactions between them. DJ-1 is able to rescue the phenotype of PINK1-knockout Drosophila models, but not of parkin-knockouts, suggesting that DJ-1 may act in a parallel pathway to that of the PINK1/parkin pathway. To further elucidate a commonality between these three proteins, bioinformatics analysis established that Miro (RHOT1) interacts with parkin and PINK1, and HSPA4 interacts with all three proteins. Furthermore, 30 transcription factors were found to be common amongst all three proteins, with many of them being involved in transcriptional regulation. Interestingly, expression of these proteins and their associated transcription factors are found to be significantly down-regulated in PD patients compared to healthy controls. In summary, this review provides insight into common pathways linking three PD-causing genes and highlights some key questions, the answers to which may provide critical insight into the disease process.
Collapse
Affiliation(s)
- Celia van der Merwe
- Division of Molecular Biology & Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 19063, Cape Town, 7505, South Africa
| | | | | | | | | |
Collapse
|
16
|
Sampath U, Janardhanam VA. Asiaticoside, a trisaccaride triterpene induces biochemical and molecular variations in brain of mice with parkinsonism. Transl Neurodegener 2013; 2:23. [PMID: 24262283 PMCID: PMC4177538 DOI: 10.1186/2047-9158-2-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/19/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Parkinson's disease characterized by oxidative stress and mitochondrial damage in the pars compacta of substantia nigra remains a challenge to manage with an added disadvantage of side effects of L-levo dopa, the standard drug used for therapy. Thus, an alternative approach of utilizing natural components would be beneficial in the management of the disease. The present study was aimed to investigate the potential role of asiaticoside (As), a trisaccaride triterpene against1 - methyl 4 - phenyl 1,2,3,6 tetrahydropyridine (MPTP)-induced neurotoxicity in experimental mice. METHODS Mice were divided into 4 groups: Group I received vehicle saline, group II was treated with 20 mg/kg of body weight of MPTP (2 doses with 2 h intervals), group III received MPTP along with 50 mg/kg body weight of As for the 21 consecutive days starting from the day of MPTP intoxication. Group IV received 50 mg/kg body weight of asiaticoside for the same period serving as drug control. Animals were sacrificed at the end of experimental period and the striatum and midbrain samples were analyzed for enzyme assays, transmission electron microscopic (TEM) analysis. Immunofluorescent assay was performed to study the expression of GFAP to detect astrocyte, which are activated due to neuronal damage. Imunohistochemical studies were carried out to quantify the expression of Bax and Bcl2, the molecular signatures that would provide clues of the extent of neurodegeneration. RESULTS The activities of enzymes were increased on As administration when compared with those of group II animals. Expressions of Bax and Bcl2 along with GFAP did show significant variations (p < 0.05) on MPTP treatment when compared to control animals and the changes were found to be reversed significantly (p < 0.05) after treatment with asiaticoside. TEM analysis also showed attenuated degenerative architecture on As administration. The mice which received As alone (drug control IV) did not show significant variation from that of the control mice. CONCLUSION The observations suggest that asiaticoside may be efficacious in protecting neurons from the oxidative damage caused by the insult of MPTP.
Collapse
Affiliation(s)
- Uvarajan Sampath
- Department of Biochemistry, Indo-American College, Cheyyar, Tamilnadu, India
| | | |
Collapse
|
17
|
Evidence of oxidative stress in young and aged DJ-1-deficient mice. FEBS Lett 2013; 587:1562-70. [PMID: 23587484 DOI: 10.1016/j.febslet.2013.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/28/2013] [Accepted: 04/03/2013] [Indexed: 02/02/2023]
Abstract
Loss of DJ-1 function contributes to pathogenesis in Parkinson's disease. Here, we investigate the impact of aging and DJ-1 deficiency in transgenic mice. Ventral midbrain from young DJ-1-deficient mice revealed no change in 4-hydroxy-2-nonenal (4-HNE), but HSP60, HSP40 and striatal dopamine turnover were significantly elevated compared to wildtype. In aged mice, the chaperone response observed in wildtype animals was absent from DJ-1-deficient transgenics, and nigral 4-HNE immunoreactivity was enhanced. These changes were concomitant with increased striatal dopamine levels and uptake. Thus, increased oxidants and diminished protein quality control may contribute to nigral oxidative damage with aging in the model.
Collapse
|
18
|
Cookson MR. Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and oxidative stress and mitochondrial pathways. Cold Spring Harb Perspect Med 2012; 2:a009415. [PMID: 22951446 DOI: 10.1101/cshperspect.a009415] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three genes have been identified that cause, in humans, autosomally inherited parkinsonism. These are PARK2, encoding the E3 ubiquitin ligase parkin; PINK1, a mitochondrial kinase; and PARK7, which codes for the protein DJ-1. In several experimental systems, it has been shown that all three proteins impact mitochondrial function and/or oxidative stress responses. These are probably related because mitochondria produce oxidative stress in neurons. Moreover, it is clear that there are relationships between these genes, with a single pathway linking PINK1 and parkin and a parallel relationship with DJ-1. Work in progress in the field is aimed at understanding these relationships in more depth.
Collapse
Affiliation(s)
- Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Abstract
SIGNIFICANCE Studies of sporadic cases, toxin models, and genetic causes of Parkinson's disease suggest that mitochondrial dysfunction may be an early feature of pathogenesis. RECENT ADVANCES Compelling evidence of a causal relationship between mitochondrial function and disease was found with the identification of several genes for recessive parkinsonism, PINK1, DJ-1, and parkin. There is evidence that each of these regulates responses to cellular stresses, including oxidative stress and depolarization of the mitochondrial membrane. Specifically, PINK1 and parkin modulate mitochondrial dynamics by promoting autophagic removal of depolarized mitochondria. Mutations in all genes linked to Parkinson's disease lead to enhanced sensitivity to mitochondrial toxins and oxidative stress. CRITICAL ISSUES Both increased mitochondrial damage due to complex 1 inhibition, mishandling of calcium, oxidant stress, or impaired clearance of dysfunctional mitochondria would lead to the accumulation of nonfunctional organelles and could contribute to neuronal dysfunction. However, several unanswered questions remain about the underlying mechanism(s) involved. FUTURE DIRECTIONS PINK1 and parkin have been demonstrated to regulate mitochondrial dynamics, but the pathways linking PINK1 activity to parkin function are still unclear and warrant further investigation.
Collapse
Affiliation(s)
- Melissa K McCoy
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
20
|
Wyse ATS, Netto CA. Behavioral and neurochemical effects of proline. Metab Brain Dis 2011; 26:159-72. [PMID: 21643764 DOI: 10.1007/s11011-011-9246-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 05/12/2011] [Indexed: 12/11/2022]
Abstract
Proline is an amino acid with an essential role for primary metabolism and physiologic functions. Hyperprolinemia results from the deficiency of specific enzymes for proline catabolism, leading to tissue accumulation of this amino acid. Hyperprolinemic patients can present neurological symptoms and brain abnormalities, whose aetiopathogenesis is poorly understood. This review addresses some of the findings obtained, mainly from animal studies, indicating that high proline levels may be associated to neuropathophysiology of some disorders. In this context, it has been suggested that energy metabolism deficit, Na(+),K(+)-ATPase, kinase creatine, oxidative stress, excitotoxicity, lipid content, as well as purinergic and cholinergic systems are involved in the effect of proline on brain damage and spatial memory deficit. The discussion focuses on the relatively low antioxidant defenses of the brain and the vulnerability of neural tissue to reactive species. This offers new perspectives for potential therapeutic strategies for this condition, which may include the early use of appropriate antioxidants as a novel adjuvant therapy, besides the usual treatment based on special diets poor in proline.
Collapse
Affiliation(s)
- Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| | | |
Collapse
|
21
|
Nisticò R, Mehdawy B, Piccirilli S, Mercuri N. Paraquat- and rotenone-induced models of Parkinson's disease. Int J Immunopathol Pharmacol 2011; 24:313-22. [PMID: 21658306 DOI: 10.1177/039463201102400205] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder mainly characterized by a loss of dopaminergic (DA) neurons in the substantia nigra pars compacta. In recent years, several new genes and environmental factors have been implicated in PD, and their impact on DA neuronal cell death is slowly emerging. However, PD etiology remains unknown, whereas its pathogenesis begins to be clarified as a multifactorial cascade of deleterious factors. Recent epidemiological studies have linked exposure to environmental agents, including pesticides, with an increased risk of developing the disease. As a result, over the last two decades the "environmental hypothesis" of PD has gained considerable interest. This speculates that agricultural chemicals in the environment, by producing selective dopaminergic cell death, can contribute to the development of the disease. However, a causal role for pesticides in the etiology of PD has yet to be definitively established. Importantly, most insights into PD pathogenesis came from investigations performed in experimental models of PD, especially those produced by neurotoxins. This review presents data obtained in our laboratories along with current views on the neurotoxic actions induced by the two most popular parkinsonian pesticide neurotoxins, namely paraquat and rotenone. Although confined to these two chemicals, mechanistic studies underlying dopaminergic cell death are of the utmost importance to identify new drug targets for the treatment of PD.
Collapse
|
22
|
Crema L, Schlabitz M, Tagliari B, Cunha A, Simão F, Krolow R, Pettenuzzo L, Salbego C, Vendite D, Wyse ATS, Dalmaz C. Na+, K+ ATPase activity is reduced in amygdala of rats with chronic stress-induced anxiety-like behavior. Neurochem Res 2010; 35:1787-95. [PMID: 20717721 DOI: 10.1007/s11064-010-0245-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2010] [Indexed: 01/24/2023]
Abstract
In this study, we examined the effects of two chronic stress regimens upon anxiety-like behavior, Na(+), K(+)-ATPase activity and immunocontent, and oxidative stress parameters (antioxidant enzymes and reactive oxygen species production) in the amygdala. Male rats were subjected to chronic unpredictable and to chronic restraint stress for 40 days. Subsequently, anxiety-like behavior was examined. Both stressed groups presented increased anxiety-like behavior. Reduced amygdalal Na(+), K(+)-ATPase activity in the synaptic plasma membranes was also observed, without alterations in the amygdala immunocontent. In addition, when analyzing oxidative stress parameters, only superoxide dismutase activity was decreased in the amygdala of animals subjected to unpredictable stress. We conclude that both models of chronic stress lead to anxiety-like behavior and decreased amygdalal Na(+), K(+)-ATPase activity, which appears not to be related to oxidative imbalance. The relationship between this decreased activity and anxiety-like behavior remains to be studied.
Collapse
Affiliation(s)
- Leonardo Crema
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Cookson MR. DJ-1, PINK1, and their effects on mitochondrial pathways. Mov Disord 2010; 25 Suppl 1:S44-8. [PMID: 20187230 DOI: 10.1002/mds.22713] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genetic forms of parkinsonism are interesting for two particular reasons. First, finding a gene identifies a cause for a disease that would otherwise be unexplained. Second, finding several genes for the same disorder allows us to reconstruct molecular pathways that, in the example of Parkinson's disease, are be associated with the survival of dopamine neurons in the substantia nigra. Two rare causes of parkinsonism, DJ-1 and PINK1, are associated with mitochondria. This organelle has long been linked with Parkinson's disease, and recent results are starting to show how mutations impact mitochondrial function. In this short review, I will discuss how we can use some of this information to understand why it is that neurons become dysfunctional in PD.
Collapse
Affiliation(s)
- Mark R Cookson
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, Maryland 20982-3707, USA.
| |
Collapse
|
24
|
Foti R, Zucchelli S, Biagioli M, Roncaglia P, Vilotti S, Calligaris R, Krmac H, Girardini JE, Del Sal G, Gustincich S. Parkinson disease-associated DJ-1 is required for the expression of the glial cell line-derived neurotrophic factor receptor RET in human neuroblastoma cells. J Biol Chem 2010; 285:18565-74. [PMID: 20395301 DOI: 10.1074/jbc.m109.088294] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations in PARK7/DJ-1 are associated with autosomal recessive, early onset Parkinson disease (PD). DJ-1 is an atypical peroxiredoxin-like peroxidase that may act as a redox-dependent chaperone and a regulator of transcription. Here we show that DJ-1 plays an essential role in the expression of rearranged during transfection (RET), a receptor for the glial cell line-derived neurotrophic factor, a neuroprotective molecule for dopaminergic neurons, the main target of degeneration in PD. The inducible loss of DJ-1 triggers the establishment of hypoxia and the production of reactive oxygen species that stabilize the hypoxia-inducible factor-1alpha (HIF-1a). HIF-1a expression is required for RET down-regulation. This study establishes for the first time a molecular link between the lack of functional DJ-1 and the glial cell line-derived neurotrophic factor signaling pathway that may explain the adult-onset loss of dopaminergic neurons. Furthermore, it suggests that hypoxia may play an important role in PD.
Collapse
Affiliation(s)
- Rossana Foti
- Sector of Neurobiology, International School for Advanced Studies, Giovanni Armenise-Harvard Foundation Laboratory, AREA Science Park, SS 14, Km 163.5, Basovizza, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
DJ-1 protects the nigrostriatal axis from the neurotoxin MPTP by modulation of the AKT pathway. Proc Natl Acad Sci U S A 2010; 107:3186-91. [PMID: 20133695 DOI: 10.1073/pnas.0914876107] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Loss-of-function DJ-1 (PARK7) mutations have been linked with a familial form of early onset Parkinson disease. Numerous studies have supported the role of DJ-1 in neuronal survival and function. Our initial studies using DJ-1-deficient neurons indicated that DJ-1 specifically protects the neurons against the damage induced by oxidative injury in multiple neuronal types and degenerative experimental paradigms, both in vitro and in vivo. However, the manner by which oxidative stress-induced death is ameliorated by DJ-1 is not completely clear. We now present data that show the involvement of DJ-1 in modulation of AKT, a major neuronal prosurvival pathway induced upon oxidative stress. We provide evidence that DJ-1 promotes AKT phosphorylation in response to oxidative stress induced by H(2)O(2) in vitro and in vivo following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. Moreover, we show that DJ-1 is necessary for normal AKT-mediated protective effects, which can be bypassed by expression of a constitutively active form of AKT. Taken together, these data suggest that DJ-1 is crucial for full activation of AKT upon oxidative injury, which serves as one explanation for the protective effects of DJ-1.
Collapse
|
26
|
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
|
27
|
Chan SL, Wei Z, Chigurupati S, Tu W. Compromised respiratory adaptation and thermoregulation in aging and age-related diseases. Ageing Res Rev 2010; 9:20-40. [PMID: 19800420 DOI: 10.1016/j.arr.2009.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 09/22/2009] [Accepted: 09/23/2009] [Indexed: 02/04/2023]
Abstract
Mitochondrial dysfunction and reactive oxygen species (ROS) production are at the heart of the aging process and are thought to underpin age-related diseases. Mitochondria are not only the primary energy-generating system but also the dominant cellular source of metabolically derived ROS. Recent studies unravel the existence of mechanisms that serve to modulate the balance between energy metabolism and ROS production. Among these is the regulation of proton conductance across the inner mitochondrial membrane that affects the efficiency of respiration and heat production. The field of mitochondrial respiration research has provided important insight into the role of altered energy balance in obesity and diabetes. The notion that respiration and oxidative capacity are mechanistically linked is making significant headway into the field of aging and age-related diseases. Here we review the regulation of cellular energy and ROS balance in biological systems and survey some of the recent relevant studies that suggest that respiratory adaptation and thermodynamics are important in aging and age-related diseases.
Collapse
|
28
|
Snyder CM, Chandel NS. Mitochondrial regulation of cell survival and death during low-oxygen conditions. Antioxid Redox Signal 2009; 11:2673-83. [PMID: 19580395 PMCID: PMC2821141 DOI: 10.1089/ars.2009.2730] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mitochondria can initiate cell death or activate genes that promote cell survival in response to low oxygen. The BCL-2 family of proteins regulate cell death in response to anoxia (0-0.5% O2). By contrast, under hypoxia (0.5-3% O2), mitochondrial oxidative stress activates hypoxia-inducible factors (HIFs) to promote cell survival. In this review, we discuss how mitochondria, BCL-2 proteins, and HIFs are crucial for cellular responses to low oxygen.
Collapse
Affiliation(s)
- Colleen M Snyder
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Medical School , Chicago, Illinois, USA
| | | |
Collapse
|
29
|
|
30
|
Ashley AK, Hanneman WH, Katoh T, Moreno JA, Pollack A, Tjalkens RB, Legare ME. Analysis of targeted mutation in DJ-1 on cellular function in primary astrocytes. Toxicol Lett 2009; 184:186-91. [PMID: 19063952 PMCID: PMC4632527 DOI: 10.1016/j.toxlet.2008.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 11/13/2008] [Accepted: 11/13/2008] [Indexed: 12/21/2022]
Abstract
DJ-1 mutation induces early-onset Parkinson's disease, and conversely over-expression of DJ-1 is associated with cancer in numerous tissues. A gene-trap screening library conducted in embryonic stem cells was utilized for generation of a DJ-1 mutant mouse. Real-time PCR and immunoblotting were utilized to confirm functional mutation of the DJ-1 gene. Normal DJ-1 protein expression in adult mouse tissue was characterized and demonstrates high expression in brain tissue with wide systemic distribution. Primary astrocytes isolated from DJ-1(-/-) mice reveal a decreased nuclear localization of DJ-1 protein in response to rotenone or LPS, with a concomitant increase in mitochondrial localization of DJ-1 found only in the rotenone exposure. Resting mitochondrial membrane potential was significantly lower in DJ-1(-/-) astrocytes, as compared to controls. Our DJ-1 knockout mouse provides an exciting tool for exploring the molecular and physiological roles of DJ-1 to further explicate its functions in neurodegeneration.
Collapse
Affiliation(s)
- Amanda K. Ashley
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80523, United States
| | - William H. Hanneman
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80523, United States
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Takeshi Katoh
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Julie A. Moreno
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80523, United States
| | - Ashley Pollack
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Ronald B. Tjalkens
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80523, United States
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Marie E. Legare
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80523, United States
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
31
|
DJ-1/PARK7 is an important mediator of hypoxia-induced cellular responses. Proc Natl Acad Sci U S A 2009; 106:1111-6. [PMID: 19144925 DOI: 10.1073/pnas.0812745106] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In cancer, DJ-1/PARK7 acts as an oncogene that drives Akt-mediated cell survival. Although amplification of DJ-1 has been described in several types of tumors, the mechanistic basis of DJ-1's oncogenic effect remains incompletely understood. A tumor's ability to adapt to hypoxia is absolutely critical for its survival and progression, and this adaptation is largely mediated by the transcription factor HIF1. The stabilization of HIF1 subunits during hypoxia is at least partly dependent on the PI3K/Akt/mTOR pathway. We hypothesized that DJ-1, a positive regulator of Akt when over-expressed, might be involved in regulating HIF1 transcriptional activity under hypoxic conditions. Our results show that loss of DJ-1 in human cell lines and transformed mouse fibroblasts decreases the transcription of a variety of HIF1-responsive genes during hypoxia. Moreover, DJ-1 expression is critical for the Akt and mTOR activities that sustain HIF1 stability. Surprisingly, DJ-1 also regulates the activity of the metabolic sensor AMPK, especially during hypoxia. Finally, DJ-1 appears to protect cells against hypoxia-induced cell death and is required for their adaptation to severe hypoxic stress. Our work positions DJ-1 as an upstream activator of HIF1 function in cancer cells and establishes that DJ-1's oncogenic activity stems from its ability to increase a cell's resistance to hypoxic stress through DJ-1's regulatory effects on mTOR and AMPK. The discovery of these functions of DJ-1 strengthens the case for the development of therapeutics that target DJ-1 activity in cancer cells.
Collapse
|
32
|
Bagh MB, Maiti AK, Jana S, Banerjee K, Roy A, Chakrabarti S. Quinone and oxyradical scavenging properties of N-acetylcysteine prevent dopamine mediated inhibition of Na+, K+-ATPase and mitochondrial electron transport chain activity in rat brain: implications in the neuroprotective therapy of Parkinson's disease. Free Radic Res 2008; 42:574-81. [PMID: 18569015 DOI: 10.1080/10715760802158430] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dopamine oxidation products such as H2O2 and reactive quinones have been held responsible for various toxic actions of dopamine, which have implications in the aetiopathogenesis of Parkinson's disease. This study has shown that N-acetylcysteine (0.25-1 mm) is a potent scavenger of both H2O2 and toxic quinones derived from dopamine and it further prevents dopamine mediated inhibition of Na+,K+-ATPase activity and mitochondrial respiratory chain function. The quinone scavenging ability of N-acetylcysteine is presumably related to its protective effect against dopamine mediated inhibition of mitochondrial respiratory chain activity. However, both H2O2 scavenging and quinone scavenging properties of N-acetylcysteine probably account for its protective effect against Na+,K+-ATPase inhibition induced by dopamine. The results have important implications in the neuroprotective therapy of sporadic Parkinson's disease since inactivation of mitochondrial respiratory activity and Na+,K+-ATPase may trigger intracellular damage pathways leading to the death of nigral dopaminergic neurons.
Collapse
Affiliation(s)
- Maria B Bagh
- Department of Biochemistry, Institute of Post Graduate Medical Education & Research, Dr B. C. Roy Post Graduate Institute of Basic Medical Sciences, India
| | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Chandran JS, Lin X, Zapata A, Höke A, Shimoji M, Moore SO, Galloway MP, Laird FM, Wong PC, Price DL, Bailey KR, Crawley JN, Shippenberg T, Cai H. Progressive behavioral deficits in DJ-1-deficient mice are associated with normal nigrostriatal function. Neurobiol Dis 2007; 29:505-14. [PMID: 18187333 DOI: 10.1016/j.nbd.2007.11.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 11/14/2007] [Accepted: 11/17/2007] [Indexed: 12/13/2022] Open
Abstract
Loss-of-function mutations in the DJ-1 gene account for an autosomal recessive form of Parkinson's disease (PD). To investigate the physiological functions of DJ-1 in vivo, we generated DJ-1 knockout (DJ-1(-/-)) mice. Younger (<1 year) DJ-1(-/-) mice were hypoactive and had mild gait abnormalities. Older DJ-1(-/-), however, showed decreased body weight and grip strength and more severe gait irregularities compared to wild-type littermates. The basal level of extracellular dopamine, evoked dopamine release and dopamine receptor D2 sensitivity appeared normal in the striatum of DJ-1(-/-) mice, which was consistent with similar results between DJ-1(-/-) and controls in behavioral paradigms specific for the dopaminergic system. An examination of spinal cord, nerve and muscle tissues failed to identify any pathological changes that were consistent with the noted motor deficits. Taken together, our findings suggest that loss of DJ-1 leads to progressive behavioral changes without significant alterations in nigrostriatal dopaminergic and spinal motor systems.
Collapse
Affiliation(s)
- Jayanth S Chandran
- Unit of Transgenesis, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, MSC 3707, 35 Convent Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kumaran R, Kingsbury A, Coulter I, Lashley T, Williams D, de Silva R, Mann D, Revesz T, Lees A, Bandopadhyay R. DJ-1 (PARK7) is associated with 3R and 4R tau neuronal and glial inclusions in neurodegenerative disorders. Neurobiol Dis 2007; 28:122-32. [PMID: 17719794 DOI: 10.1016/j.nbd.2007.07.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 06/25/2007] [Accepted: 07/01/2007] [Indexed: 12/13/2022] Open
Abstract
Mutations in the DJ-1 gene are associated with autosomal recessive Parkinson's disease (PD), but its role in disease pathogenesis is unknown. This study examines DJ-1 immunoreactivity (DJ-1 IR) in a variety of neurodegenerative disorders, Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD) with Pick bodies, FTLD with MAPT mutations, progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), in which hyperphosphorylated tau inclusions are the major pathological signature. DJ-1 IR was seen in a subset of neurofibrillary tangles (NFTs), neuropil threads (NTs), and neurites in extracellular plaques in AD; tau inclusions in AD contained both 3R and 4R tau. A subset of Pick bodies in FTLD showed DJ-1 IR. In PSP, DJ-1 IR was present in a few NFTs, NTs and glial cell inclusions. In CBD, DJ-1 IR was seen only in astrocytic plaques. In cases of FTLD with MAPT mutations that were 4R tau positive (i.e. N279K and exon 10+16 mutations), DJ-1 IR was present mostly in oligodendroglial coiled bodies. However, in MAPT R406W mutation cases, DJ-1 IR was associated mainly with NFTs and NTs and these were both 3R and 4R tau positive. No DJ-1 IR was present in FTLD with ubiquitin inclusions (FTLD-U). In AD and FTLD with Pick bodies, DJ-1 protein was enriched in the sarkosyl-insoluble fractions of frozen brain tissue containing insoluble hyperphosphorylated tau, thus strengthening the association of DJ-1 with tau pathology. Additionally using two-dimensional gel electrophoresis, we demonstrated accumulation of acidic pI isoforms of DJ-1 in AD brain, which may compromise its normal function. Our observations confirm previous findings that DJ-1 is present in a subpopulation of glial and neuronal tau inclusions in tau diseases and associated with both 3R and 4R tau isoforms.
Collapse
Affiliation(s)
- Ravindran Kumaran
- Reta Lila Weston Institute of Neurological Studies, Institute of Neurology, 1, Wakefield Street, WC1N 1PJ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Stefanello FM, Scherer EBS, Kurek AG, Mattos CB, Wyse ATS. Effect of hypermethioninemia on some parameters of oxidative stress and on Na(+),K (+)-ATPase activity in hippocampus of rats. Metab Brain Dis 2007; 22:172-82. [PMID: 17473966 DOI: 10.1007/s11011-007-9052-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Accepted: 12/12/2006] [Indexed: 01/24/2023]
Abstract
In the present study we investigated the effect of chronic administration of methionine, a metabolite accumulated in many inherited pathological conditions such as methionine adenosyltransferase deficiency and homocystinuria, on some parameters of oxidative stress, namely thiobarbituric acid reactive substances (TBARS), catalase activity and total thiol content, as well as on Na(+),K(+)-ATPase activity in rat hippocampus. For chronic treatment, rats received subcutaneous injections of methionine (1.34-2.68 mumol/g of body weight), twice a day, from the 6th to the 28th day of age and controls received saline. Animals were killed 12 h after the last injection. Results showed that chronic hypermethioninemia significantly increased TBARS, decreased Na(+),K(+)-ATPase activity but did not alter catalase and total thiol content. Since chronic hypermethioninemia altered TBARS and Na(+),K(+)-ATPase activity at 12 h after methionine administration, we also investigated the effect of acute administration of this amino acid on the same parameters studied after chronic methionine administration. For acute treatment,29-day-old rats received one single injection of methionine (2.68 mumol/g of body weight) or saline and were killed 1, 3 or 12 h later. Results showed that rats subjected to acute hypermethioninemia presented a reduction of Na(+),K(+)-ATPase activity and an increase in TBARS when the animals were killed at 3 and 12 h, but not at 1 h, after methionine administration. These data indicate that hypermethioninemia increases lipid peroxidation which may, at least partially, explain the effect of methionine on the reduction in Na(+),K(+)-ATPase activity. If confirmed in human beings, our findings could suggest that the induction of oxidative stress and the inhibition of Na(+),K(+)-ATPase activity caused by methionine might contribute to the neurophysiopathology observed in patients with severe hypermethioninemia.
Collapse
Affiliation(s)
- Francieli M Stefanello
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
37
|
Wu YN, Johnson SW. Rotenone potentiates NMDA currents in substantia nigra dopamine neurons. Neurosci Lett 2007; 421:96-100. [PMID: 17560718 DOI: 10.1016/j.neulet.2007.05.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 05/17/2007] [Accepted: 05/21/2007] [Indexed: 12/21/2022]
Abstract
Rotenone is a pesticide that produces a rodent model of Parkinson's disease. Although much evidence suggests that oxidative stress mediates the toxicity of rotenone on dopamine neurons, rotenone can also potentiate glutamate excitotoxicity. We used whole-cell patch pipettes to investigate actions of rotenone on currents evoked by N-methyl-d-aspartate (NMDA) in dopamine neurons in slices of rat midbrain. After superfusing the slice for 20-30 min, rotenone (100 nM) caused a 162% increase in the average amplitude of inward current evoked by 30 microM NMDA. This effect of rotenone was mimicked by the sodium pump inhibitor strophanthidin (10 microM) and was abolished when pipettes contained an ATP regeneration solution. Although strophanthidin also significantly increased the amplitude of inward currents evoked by (+/-)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA; 10 microM), rotenone failed to potentiate AMPA currents. Because rotenone potentiated NMDA- but not AMPA-dependent currents, this suggests that rotenone acts selectively to augment NMDA receptor function. Furthermore, the failure of rotenone to mimic strophanthidin suggests that rotenone does not inhibit sodium pump activity. Our results suggest that an excitotoxic mechanism might contribute to rotenone neurotoxicity.
Collapse
Affiliation(s)
- Yan-Na Wu
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|
38
|
Manning-Boğ AB, Caudle WM, Perez XA, Reaney SH, Paletzki R, Isla MZ, Chou VP, McCormack AL, Miller GW, Langston JW, Gerfen CR, Dimonte DA. Increased vulnerability of nigrostriatal terminals in DJ-1-deficient mice is mediated by the dopamine transporter. Neurobiol Dis 2007; 27:141-50. [PMID: 17560790 DOI: 10.1016/j.nbd.2007.03.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 03/26/2007] [Accepted: 03/29/2007] [Indexed: 11/29/2022] Open
Abstract
Mutations in the gene for DJ-1 have been associated with early-onset autosomal recessive parkinsonism. Previous studies of null DJ-1 mice have shown alterations in striatal dopamine (DA) transmission with no DAergic cell loss. Here we characterize a new line of DJ-1-deficient mice. A subtle locomotor deficit was present in the absence of a change in striatal DA levels. However, increased [(3)H]-DA synaptosomal uptake and [(125)I]-RTI-121 binding were measured in null DJ-1 vs. wild-type mice. Western analyses of synaptosomes revealed significantly higher dopamine transporter (DAT) levels in pre-synaptic membrane fractions. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) exposure exacerbated striatal DA depletion in null DJ-1 mice with no difference in DAergic nigral cell loss. Furthermore, increased 1-methyl-4-phenylpyridinium (MPP(+)) synaptosomal uptake and enhanced MPP(+) accumulation were measured in DJ-1-deficient vs. control striatum. Thus, under null DJ-1 conditions, DAT changes likely contribute to altered DA neurotransmission and enhanced sensitivity to toxins that utilize DAT for nigrostriatal entry.
Collapse
Affiliation(s)
- Amy B Manning-Boğ
- Department of Basic Research, The Parkinson's Institute, 1170 Morse Avenue, Sunnyvale, CA 94089, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|