1
|
Yang P, Shuai W, Wang X, Hu X, Zhao M, Wang A, Wu Y, Ouyang L, Wang G. Mitophagy in Neurodegenerative Diseases: Mechanisms of Action and the Advances of Drug Discovery. J Med Chem 2025; 68:3970-3994. [PMID: 39908485 DOI: 10.1021/acs.jmedchem.4c01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Neurodegenerative diseases (NDDs), such as Parkinson's disease (PD) and Alzheimer's disease (AD), are devastating brain diseases and are incurable at the moment. Increasing evidence indicates that NDDs are associated with mitochondrial dysfunction. Mitophagy removes defective or redundant mitochondria to maintain cell homeostasis, whereas deficient mitophagy accelerates the accumulation of damaged mitochondria to mediate the pathologies of NDDs. Therefore, targeting mitophagy has become a valuable therapeutic pathway for the treatment of NDDs. Several mitophagy modulators have been shown to ameliorate neurodegeneration in PD and AD. However, it remains to be further investigated for other NDDs. Here, we describe the mechanism and key signaling pathway of mitophagy and summarize the roles of defective mitophagy on the pathogenesis of NDDs. Further, we underline the development advances of mitophagy modulators for PD and AD therapy, discuss the therapeutic challenges and limitations of the existing modulators, and provide guidelines for mitophagy mechanism exploration and drug design.
Collapse
Affiliation(s)
- Panpan Yang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xiuying Hu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| |
Collapse
|
2
|
Singh G, Mittra N, Singh C. Impaired Mitochondrial Function and Ubiquitin Proteasome System Activate α-Synuclein Aggregation in Zinc-Induced Neurotoxicity: Effect of Antioxidants. J Mol Neurosci 2025; 75:16. [PMID: 39907853 DOI: 10.1007/s12031-024-02293-5] [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: 10/01/2024] [Accepted: 11/30/2024] [Indexed: 02/06/2025]
Abstract
Impairment in mitochondrial function and ubiquitin-proteasome system (UPS) and alpha-synuclein (α-Syn) aggregation are implicated in Zn-induced neurotoxicity. A link among these events leading to Zn-induced neurotoxicity is not yet properly deciphered. Therefore, the study intended to check the existence of a crosstalk between the mitochondria and UPS and its further link to α-Syn aggregation. The study also aimed to investigate the efficacy of tempol, a SOD mimetic and silymarin, a natural antioxidant, against Zn-induced alterations in animals and differentiated cells. Zn reduced the locomotor activity, dopamine content and tyrosine hydroxylase (TH) expression in the exposed animals. Zn augmented the levels of mitochondrial reactive oxygen species, α-Syn and protein-ubiquitin conjugates. Mitochondrial membrane potential, adenosine triphosphate (ATP) production, UPS-associated enzymatic activities and levels of UPS subunits (SUG-1 and β-5) were attenuated in Zn-exposed animals. While Zn augmented the expression of heat shock protein 110 (HSP110), peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) and Parkin translocation, the mitochondrial PTEN-induced kinase-1 (PINK-1) level was attenuated. In addition to tempol and silymarin, a mitochondrial permeability transition pore inhibitor, cyclosporine A, also alleviated the Zn-induced changes in animals. Similar trends in a few parameters were also observed in the differentiated human neuroblastoma SH-SY-5Y cells. Besides, UPS inhibitor, MG132, enhanced Zn-induced UPS impairment, protein aggregation and mitochondrial dysfunction in differentiated cells. These results suggest that mitochondrial dysfunction triggers UPS impairment or vice versa that elevates α-Syn aggregation and consequent neuronal death. Furthermore, tempol and silymarin ameliorate the mitochondrial and UPS impairments and α-Syn aggregation thereby providing protection from Zn-induced neurotoxicity.
Collapse
Affiliation(s)
- Garima Singh
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Post Box No. 80, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Namrata Mittra
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Post Box No. 80, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Chetna Singh
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Post Box No. 80, Lucknow, 226001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
3
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
4
|
Liu M, Liu S, Lin Z, Chen X, Jiao Q, Du X, Jiang H. Targeting the Interplay Between Autophagy and the Nrf2 Pathway in Parkinson's Disease with Potential Therapeutic Implications. Biomolecules 2025; 15:149. [PMID: 39858542 PMCID: PMC11764135 DOI: 10.3390/biom15010149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder marked by the progressive degeneration of midbrain dopaminergic neurons and resultant locomotor dysfunction. Despite over two centuries of recognition as a chronic disease, the exact pathogenesis of PD remains elusive. The onset and progression of PD involve multiple complex pathological processes, with dysfunctional autophagy and elevated oxidative stress serving as critical contributors. Notably, emerging research has underscored the interplay between autophagy and oxidative stress in PD pathogenesis. Given the limited efficacy of therapies targeting either autophagy dysfunction or oxidative stress, it is crucial to elucidate the intricate mechanisms governing their interplay in PD to develop more effective therapeutics. This review overviews the role of autophagy and nuclear factor erythroid 2-related factor 2 (Nrf2), a pivotal transcriptional regulator orchestrating cellular defense mechanisms against oxidative stress, and the complex interplay between these processes. By elucidating the intricate interplay between these key pathological processes in PD, this review will deepen our comprehensive understanding of the multifaceted pathological processes underlying PD and may uncover potential strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Mengru Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Siqi Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Zihan Lin
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (M.L.); (S.L.)
| | - Hong Jiang
- Qingdao Key Laboratory of Neurorehabilitation, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| |
Collapse
|
5
|
Singh S, Chauhan K. Pharmacological approach using doxycycline and tocopherol in rotenone induced oxidative stress, neuroinflammation and Parkinson's like symptoms. Int J Neurosci 2024; 134:866-881. [PMID: 36453937 DOI: 10.1080/00207454.2022.2154670] [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: 02/24/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a second most common neurodegenerative disorder characterized by the selective and progressive degeneration of dopaminergic neurons in substantia nigra pars compacta. Rotenone is a neurotoxin which selectively degenerate dopaminergic neurons in striatum, leading to cause PD like symptoms. METHOD Rotenone was administered at a dose of 1.5 mg/kg, i.p. from day 1 to day 40. Treatment with doxycycline (50 and 100 mg/kg, p.o), tocopherol (5 mg and 10 mg/kg, p.o) alone, doxycycline (50 mg/kg, p.o) in combination with tocopherol (10 mg/kg, p.o), and ropinirole (0.5 mg/kg, i.p.) was given for 40 days 1 h prior to administration of rotenone. All behavioral parameters were analyzed on weekly basis. On day 41, animals were sacrificed and the striatum region was isolated for neurotransmitters estimation (dopamine, serotonin, norepinephrine, GABA and glutamate), biochemical analysis (GSH, nitrite, LPO, mitochondrial complexes I and IV), inflammatory markers estimation (IL-6, IL-1β and TNF-α) and activity of MAO-A, MAO-B. RESULT Doxycycline and tocopherol in combination significantly attenuated behavioral, neurotransmitters and biochemical alterations induced by rotenone in experimental rats as compared to alone treatment with DOX and TOCO. Similarly, DOX and TOCO combination significantly reduced the level of inflammatory markers, prevented the biochemical changes, decreased MAO-A and MAO-B and improved complex-I, complex-IV, cAMP levels significantly. CONCLUSION The current study revealed that a combination of doxycycline with tocopherol contributed to the prevention of PD like symptoms in rats by antioxidant, anti-inflammatory, MAO inhibitory and neuromodulatory mechanisms.
Collapse
Affiliation(s)
- Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Kanupriya Chauhan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
6
|
Liang H, Ma Z, Zhong W, Liu J, Sugimoto K, Chen H. Regulation of mitophagy and mitochondrial function: Natural compounds as potential therapeutic strategies for Parkinson's disease. Phytother Res 2024; 38:1838-1862. [PMID: 38356178 DOI: 10.1002/ptr.8156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Mitochondrial damage is associated with the development of Parkinson's disease (PD), indicating that mitochondrial-targeted treatments could hold promise as disease-modifying approaches for PD. Notably, natural compounds have demonstrated the ability to modulate mitochondrial-related processes. In this review article, we discussed the possible neuroprotective mechanisms of natural compounds against PD in modulating mitophagy and mitochondrial function. A comprehensive literature search on natural compounds related to the treatment of PD by regulating mitophagy and mitochondrial function was conducted from PubMed, Web of Science and Chinese National Knowledge Infrastructure databases from their inception until April 2023. We summarize recent advancements in mitophagy's molecular mechanisms, including upstream and downstream processes, and its relationship with PD-related genes or proteins. Importantly, we highlight how natural compounds can therapeutically regulate various mitochondrial processes through multiple targets and pathways to alleviate oxidative stress, neuroinflammation, Lewy's body aggregation and apoptosis, which are key contributors to PD pathogenesis. Unlike the single-target strategy of modern medicine, natural compounds provide neuroprotection against PD by modulating various mitochondrial-related processes, including ameliorating mitophagy by targeting the PINK1/parkin pathway, the NIX/BNIP3 pathway, and autophagosome formation (i.e., LC3 and p62). Given the prevalence of mitochondrial damage in various neurodegenerative diseases, exploring the exact mechanism of natural compounds on mitophagy and mitochondrial dysfunction could shed light on the development of highly effective disease-modifying or adjuvant therapies targeting PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Hao Liang
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Zhenwang Ma
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Wei Zhong
- Department of Rheumatology and Immunology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, China
| | - Jia Liu
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Kazuo Sugimoto
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Chen
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
- Department of TCM Geriatric, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Bartman S, Coppotelli G, Ross JM. Mitochondrial Dysfunction: A Key Player in Brain Aging and Diseases. Curr Issues Mol Biol 2024; 46:1987-2026. [PMID: 38534746 DOI: 10.3390/cimb46030130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Mitochondria are thought to have become incorporated within the eukaryotic cell approximately 2 billion years ago and play a role in a variety of cellular processes, such as energy production, calcium buffering and homeostasis, steroid synthesis, cell growth, and apoptosis, as well as inflammation and ROS production. Considering that mitochondria are involved in a multitude of cellular processes, mitochondrial dysfunction has been shown to play a role within several age-related diseases, including cancers, diabetes (type 2), and neurodegenerative diseases, although the underlying mechanisms are not entirely understood. The significant increase in lifespan and increased incidence of age-related diseases over recent decades has confirmed the necessity to understand the mechanisms by which mitochondrial dysfunction impacts the process of aging and age-related diseases. In this review, we will offer a brief overview of mitochondria, along with structure and function of this important organelle. We will then discuss the cause and consequence of mitochondrial dysfunction in the aging process, with a particular focus on its role in inflammation, cognitive decline, and neurodegenerative diseases, such as Huntington's disease, Parkinson's disease, and Alzheimer's disease. We will offer insight into therapies and interventions currently used to preserve or restore mitochondrial functioning during aging and neurodegeneration.
Collapse
Affiliation(s)
- Sydney Bartman
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Giuseppe Coppotelli
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Jaime M Ross
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
8
|
Gu Y, Zhang J, Zhao X, Nie W, Xu X, Liu M, Zhang X. Olfactory dysfunction and its related molecular mechanisms in Parkinson's disease. Neural Regen Res 2024; 19:583-590. [PMID: 37721288 PMCID: PMC10581567 DOI: 10.4103/1673-5374.380875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 09/19/2023] Open
Abstract
Changes in olfactory function are considered to be early biomarkers of Parkinson's disease. Olfactory dysfunction is one of the earliest non-motor features of Parkinson's disease, appearing in about 90% of patients with early-stage Parkinson's disease, and can often predate the diagnosis by years. Therefore, olfactory dysfunction should be considered a reliable marker of the disease. However, the mechanisms responsible for olfactory dysfunction are currently unknown. In this article, we clearly explain the pathology and medical definition of olfactory function as a biomarker for early-stage Parkinson's disease. On the basis of the findings of clinical olfactory function tests and animal model experiments as well as neurotransmitter expression levels, we further characterize the relationship between olfactory dysfunction and neurodegenerative diseases as well as the molecular mechanisms underlying olfactory dysfunction in the pathology of early-stage Parkinson's disease. The findings highlighted in this review suggest that olfactory dysfunction is an important biomarker for preclinical-stage Parkinson's disease. Therefore, therapeutic drugs targeting non-motor symptoms such as olfactory dysfunction in the early stage of Parkinson's disease may prevent or delay dopaminergic neurodegeneration and reduce motor symptoms, highlighting the potential of identifying effective targets for treating Parkinson's disease by inhibiting the deterioration of olfactory dysfunction.
Collapse
Affiliation(s)
- Yingying Gu
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jiaying Zhang
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Xinru Zhao
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Wenyuan Nie
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Xiaole Xu
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Mingxuan Liu
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoling Zhang
- College of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
9
|
Yao MF, Dang T, Wang HJ, Zhu XZ, Qiao C. Mitochondrial homeostasis regulation: A promising therapeutic target for Parkinson's disease. Behav Brain Res 2024; 459:114811. [PMID: 38103871 DOI: 10.1016/j.bbr.2023.114811] [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: 10/20/2023] [Revised: 12/10/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of Lewy bodies (LBs) or Lewy neurites (LNs) which consist of α-synuclein (α-syn) and a complex mix of other biomolecules. Mitochondrial dysfunction is widely believed to play an essential role in the pathogenesis of PD and other related neurodegenerative diseases. But mitochondrial dysfunction is subject to complex genetic regulation. There is increasing evidence that PD-related genes directly or indirectly affect mitochondrial integrity. Therefore, targeted regulation of mitochondrial function has great clinical application prospects in the treatment of PD. However, lots of PD drugs targeting mitochondria have been developed but their clinical therapeutic effects are not ideal. This review aims to reveal the role of mitochondrial dysfunction in the pathogenesis of neurodegenerative diseases based on the mitochondrial structure and function, which may highlight potential interventions and therapeutic targets for the development of PD drugs to recover mitochondrial dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Meng-Fan Yao
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tao Dang
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hua-Jun Wang
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xiao-Zhong Zhu
- Department of Cardiothoracic Surgery, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Chen Qiao
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
10
|
Croucher KM, Fleming SM. ATP13A2 (PARK9) and basal ganglia function. Front Neurol 2024; 14:1252400. [PMID: 38249738 PMCID: PMC10796451 DOI: 10.3389/fneur.2023.1252400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
ATP13A2 is a lysosomal protein involved in polyamine transport with loss of function mutations associated with multiple neurodegenerative conditions. These include early onset Parkinson's disease, Kufor-Rakeb Syndrome, neuronal ceroid lipofuscinosis, hereditary spastic paraplegia, and amyotrophic lateral sclerosis. While ATP13A2 mutations may result in clinical heterogeneity, the basal ganglia appear to be impacted in the majority of cases. The basal ganglia is particularly vulnerable to environmental exposures such as heavy metals, pesticides, and industrial agents which are also established risk factors for many neurodegenerative conditions. Not surprisingly then, impaired function of ATP13A2 has been linked to heavy metal toxicity including manganese, iron, and zinc. This review discusses the role of ATP13A2 in basal ganglia function and dysfunction, potential common pathological mechanisms in ATP13A2-related disorders, and how gene x environment interactions may contribute to basal ganglia dysfunction.
Collapse
Affiliation(s)
- Kristina M. Croucher
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
- Biomedical Sciences Graduate Program, Kent State University, Kent, OH, United States
| | - Sheila M. Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
11
|
Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. The role of mitochondrial dynamics in disease. MedComm (Beijing) 2023; 4:e462. [PMID: 38156294 PMCID: PMC10753647 DOI: 10.1002/mco2.462] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Mitochondria are multifaceted and dynamic organelles regulating various important cellular processes from signal transduction to determining cell fate. As dynamic properties of mitochondria, fusion and fission accompanied with mitophagy, undergo constant changes in number and morphology to sustain mitochondrial homeostasis in response to cell context changes. Thus, the dysregulation of mitochondrial dynamics and mitophagy is unsurprisingly related with various diseases, but the unclear underlying mechanism hinders their clinical application. In this review, we summarize the recent developments in the molecular mechanism of mitochondrial dynamics and mitophagy, particularly the different roles of key components in mitochondrial dynamics in different context. We also summarize the roles of mitochondrial dynamics and target treatment in diseases related to the cardiovascular system, nervous system, respiratory system, and tumor cell metabolism demanding high-energy. In these diseases, it is common that excessive mitochondrial fission is dominant and accompanied by impaired fusion and mitophagy. But there have been many conflicting findings about them recently, which are specifically highlighted in this view. We look forward that these findings will help broaden our understanding of the roles of the mitochondrial dynamics in diseases and will be beneficial to the discovery of novel selective therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Xinyan Dai
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Hui Li
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huiling Jiang
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Junfu Zhou
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Shiying Zhang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jiacheng Guo
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Lidu Shen
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huantao Yang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jie Lin
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Hengxiu Yan
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| |
Collapse
|
12
|
Wang B, Fang T, Chen H. Zinc and Central Nervous System Disorders. Nutrients 2023; 15:2140. [PMID: 37432243 DOI: 10.3390/nu15092140] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023] Open
Abstract
Zinc (Zn2+) is the second most abundant necessary trace element in the human body, exerting a critical role in many physiological processes such as cellular proliferation, transcription, apoptosis, growth, immunity, and wound healing. It is an essential catalyst ion for many enzymes and transcription factors. The maintenance of Zn2+ homeostasis is essential for the central nervous system, in which Zn2+ is abundantly distributed and accumulates in presynaptic vesicles. Synaptic Zn2+ is necessary for neural transmission, playing a pivotal role in neurogenesis, cognition, memory, and learning. Emerging data suggest that disruption of Zn2+ homeostasis is associated with several central nervous system disorders including Alzheimer's disease, depression, Parkinson's disease, multiple sclerosis, schizophrenia, epilepsy, and traumatic brain injury. Here, we reviewed the correlation between Zn2+ and these central nervous system disorders. The potential mechanisms were also included. We hope that this review can provide new clues for the prevention and treatment of nervous system disorders.
Collapse
Affiliation(s)
- Bangqi Wang
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China
| | - Tianshu Fang
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China
| | - Hongping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
13
|
Dong-Chen X, Yong C, Yang X, Chen-Yu S, Li-Hua P. Signaling pathways in Parkinson's disease: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:73. [PMID: 36810524 PMCID: PMC9944326 DOI: 10.1038/s41392-023-01353-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 01/16/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, and its treatment remains a big challenge. The pathogenesis of PD may be related to environmental and genetic factors, and exposure to toxins and gene mutations may be the beginning of brain lesions. The identified mechanisms of PD include α-synuclein aggregation, oxidative stress, ferroptosis, mitochondrial dysfunction, neuroinflammation, and gut dysbiosis. The interactions among these molecular mechanisms complicate the pathogenesis of PD and pose great challenges to drug development. At the same time, the diagnosis and detection of PD are also one of obstacles to the treatment of PD due to its long latency and complex mechanism. Most conventional therapeutic interventions for PD possess limited effects and have serious side effects, heightening the need to develop novel treatments for this disease. In this review, we systematically summarized the pathogenesis, especially the molecular mechanisms of PD, the classical research models, clinical diagnostic criteria, and the reported drug therapy strategies, as well as the newly reported drug candidates in clinical trials. We also shed light on the components derived from medicinal plants that are newly identified for their effects in PD treatment, with the expectation to provide the summary and outlook for developing the next generation of drugs and preparations for PD therapy.
Collapse
Affiliation(s)
- Xu Dong-Chen
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Chen Yong
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Xu Yang
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - ShenTu Chen-Yu
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Peng Li-Hua
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China. .,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P. R. China.
| |
Collapse
|
14
|
García-Beltrán O, Urrutia PJ, Núñez MT. On the Chemical and Biological Characteristics of Multifunctional Compounds for the Treatment of Parkinson's Disease. Antioxidants (Basel) 2023; 12:214. [PMID: 36829773 PMCID: PMC9952574 DOI: 10.3390/antiox12020214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Protein aggregation, mitochondrial dysfunction, iron dyshomeostasis, increased oxidative damage and inflammation are pathognomonic features of Parkinson's disease (PD) and other neurodegenerative disorders characterized by abnormal iron accumulation. Moreover, the existence of positive feed-back loops between these pathological components, which accelerate, and sometimes make irreversible, the neurodegenerative process, is apparent. At present, the available treatments for PD aim to relieve the symptoms, thus improving quality of life, but no treatments to stop the progression of the disease are available. Recently, the use of multifunctional compounds with the capacity to attack several of the key components of neurodegenerative processes has been proposed as a strategy to slow down the progression of neurodegenerative processes. For the treatment of PD specifically, the necessary properties of new-generation drugs should include mitochondrial destination, the center of iron-reactive oxygen species interaction, iron chelation capacity to decrease iron-mediated oxidative damage, the capacity to quench free radicals to decrease the risk of ferroptotic neuronal death, the capacity to disrupt α-synuclein aggregates and the capacity to decrease inflammatory conditions. Desirable additional characteristics are dopaminergic neurons to lessen unwanted secondary effects during long-term treatment, and the inhibition of the MAO-B and COMPT activities to increase intraneuronal dopamine content. On the basis of the published evidence, in this work, we review the molecular basis underlying the pathological events associated with PD and the clinical trials that have used single-target drugs to stop the progress of the disease. We also review the current information on multifunctional compounds that may be used for the treatment of PD and discuss the chemical characteristics that underlie their functionality. As a projection, some of these compounds or modifications could be used to treat diseases that share common pathology features with PD, such as Friedreich's ataxia, Multiple sclerosis, Huntington disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Olimpo García-Beltrán
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730002, Colombia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, General Gana 1702, Santiago 8370854, Chile
| | - Pamela J. Urrutia
- Faculty of Medicine and Science, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Marco T. Núñez
- Faculty of Sciences, Universidad de Chile, Las Palmeras 3425, Santiago 7800024, Chile
| |
Collapse
|
15
|
Dhanushkodi NR, Abul Khair SB, Ardah MT, Haque ME. ATP13A2 Gene Silencing in Drosophila Affects Autophagic Degradation of A53T Mutant α-Synuclein. Int J Mol Sci 2023; 24:ijms24021775. [PMID: 36675288 PMCID: PMC9861907 DOI: 10.3390/ijms24021775] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Mutations in ATP13A2 (PARK9), an autophagy-related protein, cause Kufor-Rakeb syndrome, an autosomal recessive, juvenile-onset form of parkinsonism. α-Synuclein (α-syn) is a presynaptic neuronal protein that forms toxic aggregates in Parkinson's disease (PD). We studied α-syn aggregation and autophagic flux in ATP13A2-knockdown Drosophila expressing either wild-type (WT) or mutant α-syn. Dopaminergic (DA) neuron loss was studied by confocal microscopy. Sleep and circadian activity were evaluated in young and old flies using a Drosophila activity monitor. Thirty-day-old ATP13A2-RNAi A53T-α-syn flies had increased Triton-insoluble α-syn levels, compared to control A53T-α-syn flies without ATP13A2-RNAi. Whole-brain staining revealed significantly fewer dopaminergic (DA) neurons in the PPL2 cluster of 30-day-old ATP13A2-RNAi flies expressing WT-, A30P-, and A53T-α-syn than in that of controls. In ATP13A2-RNAi A53T-α-syn flies, autophagic flux was decreased, as indicated by increased accumulation of Ref(2)P, the Drosophila p62 homologue. ATP13A2 silencing decreased total locomotor activity in young, and enhanced sleep features, similar to PD (decreasing bout length), in old flies expressing A53T-α-syn. ATP13A2 silencing also altered the circadian locomotor activity of A30P- and A53T-α-syn flies. Thus, ATP13A2 may play a role in the autophagic degradation of A53T-α-syn.
Collapse
|
16
|
Oh J, Min C, Park D, Han MS. Oligonucleotide-Chemosensor Conjugate as a Dual Responsive Detection Platform and Its Application for Simultaneous Detection of ATP and Zn 2. ACS Sens 2022; 7:3933-3939. [PMID: 36503238 DOI: 10.1021/acssensors.2c02006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Simultaneous detection, which helps understand complex physiological processes and accurately diagnose diseases, has been achieved using dual responsive probes. The dual responsive probe can ideally distinguish four cases, which are a combination of the absence and presence of two analytes, with characteristic fluorescence emissions. Owing to the demanding conditions of its development, most previous studies have focused on the simple linkage between small-molecule chemosensors that have an individual target and spectral range. In this study, a new dual responsive detection platform, oligonucleotide-chemosensor conjugate, was developed using a linkage between versatile oligonucleotide probes and small-molecule chemosensors to expand the applicable scaffold and detectable target for simultaneous detection. As a proof of concept, the ATP aptamer probe and Zn2+ chemosensor were conjugated as the levels of ATP and Zn2+ are intimately correlated in several signaling pathways and diseases. Each probe could detect an analyte independently within a conjugate probe, and simultaneous detection was also demonstrated without spectral crosstalk or interference between the receptors. In addition, the introduced cholesterol modification allowed the developed probe to detect changes in analytes on the plasma membrane of live cells through flow cytometry and confocal microscopy.
Collapse
Affiliation(s)
- Jinyoung Oh
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Chanhyuk Min
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Daeho Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Min Su Han
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
17
|
Silva DF, Empadinhas N, Cardoso SM, Esteves AR. Neurodegenerative Microbially-Shaped Diseases: Oxidative Stress Meets Neuroinflammation. Antioxidants (Basel) 2022; 11:2141. [PMID: 36358513 PMCID: PMC9686748 DOI: 10.3390/antiox11112141] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 04/18/2025] Open
Abstract
Inflammation and oxidative stress characterize a number of chronic conditions including neurodegenerative diseases and aging. Inflammation is a key component of the innate immune response in Alzheimer's disease and Parkinson's disease of which oxidative stress is an important hallmark. Immune dysregulation and mitochondrial dysfunction with concomitant reactive oxygen species accumulation have also been implicated in both diseases, both systemically and within the Central Nervous System. Mitochondria are a centrally positioned signalling hub for inflammatory responses and inflammatory cells can release reactive species at the site of inflammation often leading to exaggerated oxidative stress. A growing body of evidence suggests that disruption of normal gut microbiota composition may induce increased permeability of the gut barrier leading to chronic systemic inflammation, which may, in turn, impair the blood-brain barrier function and promote neuroinflammation and neurodegeneration. The gastrointestinal tract is constantly exposed to myriad exogenous substances and microbial pathogens, which are abundant sources of reactive oxygen species, oxidative damage and pro-inflammatory events. Several studies have demonstrated that microbial infections may also affect the balance in gut microbiota composition (involving oxidant and inflammatory processes by the host and indigenous microbiota) and influence downstream Alzheimer's disease and Parkinson's disease pathogenesis, in which blood-brain barrier damage ultimately occurs. Therefore, the oxidant/inflammatory insults triggered by a disrupted gut microbiota and chronic dysbiosis often lead to compromised gut barrier function, allowing inflammation to "escape" as well as uncontrolled immune responses that may ultimately disrupt mitochondrial function upwards the brain. Future therapeutic strategies should be designed to "restrain" gut inflammation, a goal that could ideally be attained by microbiota modulation strategies, in alternative to classic anti-inflammatory agents with unpredictable effects on the microbiota architecture itself.
Collapse
Affiliation(s)
- Diana Filipa Silva
- CNC—Center for Neuroscience and Cell Biology and CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC—Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Nuno Empadinhas
- CNC—Center for Neuroscience and Cell Biology and CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC—Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC—Center for Neuroscience and Cell Biology and CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Raquel Esteves
- CNC—Center for Neuroscience and Cell Biology and CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC—Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
18
|
Lang M, Pramstaller PP, Pichler I. Crosstalk of organelles in Parkinson's disease - MiT family transcription factors as central players in signaling pathways connecting mitochondria and lysosomes. Mol Neurodegener 2022; 17:50. [PMID: 35842725 PMCID: PMC9288732 DOI: 10.1186/s13024-022-00555-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Living organisms constantly need to adapt to their surrounding environment and have evolved sophisticated mechanisms to deal with stress. Mitochondria and lysosomes are central organelles in the response to energy and nutrient availability within a cell and act through interconnected mechanisms. However, when such processes become overwhelmed, it can lead to pathologies. Parkinson's disease (PD) is a common neurodegenerative disorder (NDD) characterized by proteinaceous intracellular inclusions and progressive loss of dopaminergic neurons, which causes motor and non-motor symptoms. Genetic and environmental factors may contribute to the disease etiology. Mitochondrial dysfunction has long been recognized as a hallmark of PD pathogenesis, and several aspects of mitochondrial biology are impaired in PD patients and models. In addition, defects of the autophagy-lysosomal pathway have extensively been observed in cell and animal models as well as PD patients' brains, where constitutive autophagy is indispensable for adaptation to stress and energy deficiency. Genetic and molecular studies have shown that the functions of mitochondria and lysosomal compartments are tightly linked and influence each other. Connections between these organelles are constituted among others by mitophagy, organellar dynamics and cellular signaling cascades, such as calcium (Ca2+) and mTOR (mammalian target of rapamycin) signaling and the activation of transcription factors. Members of the Microphthalmia-associated transcription factor family (MiT), including MITF, TFE3 and TFEB, play a central role in regulating cellular homeostasis in response to metabolic pressure and are considered master regulators of lysosomal biogenesis. As such, they are part of the interconnection between mitochondria and lysosome functions and therefore represent attractive targets for therapeutic approaches against NDD, including PD. The activation of MiT transcription factors through genetic and pharmacological approaches have shown encouraging results at ameliorating PD-related phenotypes in in vitro and in vivo models. In this review, we summarize the relationship between mitochondrial and autophagy-lysosomal functions in the context of PD etiology and focus on the role of the MiT pathway and its potential as pharmacological target against PD.
Collapse
Affiliation(s)
- Martin Lang
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.,Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
19
|
Zhang F, Wu Z, Long F, Tan J, Gong N, Li X, Lin C. The Roles of ATP13A2 Gene Mutations Leading to Abnormal Aggregation of α-Synuclein in Parkinson’s Disease. Front Cell Neurosci 2022; 16:927682. [PMID: 35875356 PMCID: PMC9296842 DOI: 10.3389/fncel.2022.927682] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PARK9 (also known as ATP13A2) is recognized as one of the key genes that cause PD, and a mutation in this gene was first discovered in a rare case of PD in an adolescent. Lewy bodies (LBs) formed by abnormal aggregation of α-synuclein, which is encoded by the SNCA gene, are one of the pathological diagnostic criteria for PD. LBs are also recognized as one of the most important features of PD pathogenesis. In this article, we first summarize the types of mutations in the ATP13A2 gene and their effects on ATP13A2 mRNA and protein structure; then, we discuss lysosomal autophagy inhibition and the molecular mechanism of abnormal α-synuclein accumulation caused by decreased levels and dysfunction of the ATP13A2 protein in lysosomes. Finally, this article provides a new direction for future research on the pathogenesis and therapeutic targets for ATP13A2 gene-related PD from the perspective of ATP13A2 gene mutations and abnormal aggregation of α-synuclein.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhiwei Wu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fei Long
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jieqiong Tan
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Key Laboratory of Molecular Precision Medicine of Hunan Province, Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Ni Gong
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaorong Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- *Correspondence: Changwei Lin, orcid.org/0000-0003-1676-0912
| |
Collapse
|
20
|
Kowalczyk A, Gbadamosi O, Kolor K, Sosa J, Andrzejczuk L, Gibson G, Croix C, Chikina M, Aizenman E, Clark N, Kiselyov K. Evolutionary rate covariation identifies SLC30A9 (ZnT9) as a mitochondrial zinc transporter. Biochem J 2021; 478:3205-3220. [PMID: 34397090 PMCID: PMC10491466 DOI: 10.1042/bcj20210342] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022]
Abstract
Recent advances in genome sequencing have led to the identification of new ion and metabolite transporters, many of which have not been characterized. Due to the variety of subcellular localizations, cargo and transport mechanisms, such characterization is a daunting task, and predictive approaches focused on the functional context of transporters are very much needed. Here we present a case for identifying a transporter localization using evolutionary rate covariation (ERC), a computational approach based on pairwise correlations of amino acid sequence evolutionary rates across the mammalian phylogeny. As a case study, we find that poorly characterized transporter SLC30A9 (ZnT9) coevolves with several components of the mitochondrial oxidative phosphorylation chain, suggesting mitochondrial localization. We confirmed this computational finding experimentally using recombinant human SLC30A9. SLC30A9 loss caused zinc mishandling in the mitochondria, suggesting that under normal conditions it acts as a zinc exporter. We therefore propose that ERC can be used to predict the functional context of novel transporters and other poorly characterized proteins.
Collapse
Affiliation(s)
- Amanda Kowalczyk
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, PA 15213, U.S.A
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, U.S.A
| | - Omotola Gbadamosi
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Kathryn Kolor
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Jahree Sosa
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Livia Andrzejczuk
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Gregory Gibson
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Claudette Croix
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Maria Chikina
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, U.S.A
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Nathan Clark
- Department of Human Genetics, University of Utah, Utah 84112, U.S.A
| | - Kirill Kiselyov
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| |
Collapse
|
21
|
Oxidative Stress, Mitochondrial Dysfunction, and Neuroprotection of Polyphenols with Respect to Resveratrol in Parkinson's Disease. Biomedicines 2021; 9:biomedicines9080918. [PMID: 34440122 PMCID: PMC8389563 DOI: 10.3390/biomedicines9080918] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease and is characterized by dopaminergic neuronal loss. The exact pathogenesis of PD is complex and not yet completely understood, but research has established the critical role mitochondrial dysfunction plays in the development of PD. As the main producer of cytosolic reactive oxygen species (ROS), mitochondria are particularly susceptible to oxidative stress once an imbalance between ROS generation and the organelle’s antioxidative system occurs. An overabundance of ROS in the mitochondria can lead to mitochondrial dysfunction and further vicious cycles. Once enough damage accumulates, the cell may undergo mitochondria-dependent apoptosis or necrosis, resulting in the neuronal loss of PD. Polyphenols are a group of natural compounds that have been shown to offer protection against various diseases, including PD. Among these, the plant-derived polyphenol, resveratrol, exhibits neuroprotective effects through its antioxidative capabilities and provides mitochondria protection. Resveratrol also modulates crucial genes involved in antioxidative enzymes regulation, mitochondrial dynamics, and cellular survival. Additionally, resveratrol offers neuroprotective effects by upregulating mitophagy through multiple pathways, including SIRT-1 and AMPK/ERK pathways. This compound may provide potential neuroprotective effects, and more clinical research is needed to establish the efficacy of resveratrol in clinical settings.
Collapse
|
22
|
Li W, Fu Y, Halliday GM, Sue CM. PARK Genes Link Mitochondrial Dysfunction and Alpha-Synuclein Pathology in Sporadic Parkinson's Disease. Front Cell Dev Biol 2021; 9:612476. [PMID: 34295884 PMCID: PMC8291125 DOI: 10.3389/fcell.2021.612476] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 06/10/2021] [Indexed: 11/28/2022] Open
Abstract
Parkinson’s disease (PD) is an age-related neurodegenerative disorder affecting millions of people worldwide. The disease is characterized by the progressive loss of dopaminergic neurons and spread of Lewy pathology (α-synuclein aggregates) in the brain but the pathogenesis remains elusive. PD presents substantial clinical and genetic variability. Although its complex etiology and pathogenesis has hampered the breakthrough in targeting disease modification, recent genetic tools advanced our approaches. As such, mitochondrial dysfunction has been identified as a major pathogenic hub for both familial and sporadic PD. In this review, we summarize the effect of mutations in 11 PARK genes (SNCA, PRKN, PINK1, DJ-1, LRRK2, ATP13A2, PLA2G6, FBXO7, VPS35, CHCHD2, and VPS13C) on mitochondrial function as well as their relevance in the formation of Lewy pathology. Overall, these genes play key roles in mitochondrial homeostatic control (biogenesis and mitophagy) and functions (e.g., energy production and oxidative stress), which may crosstalk with the autophagy pathway, induce proinflammatory immune responses, and increase oxidative stress that facilitate the aggregation of α-synuclein. Thus, rectifying mitochondrial dysregulation represents a promising therapeutic approach for neuroprotection in PD.
Collapse
Affiliation(s)
- Wen Li
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - YuHong Fu
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Carolyn M Sue
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
23
|
Abstract
Since the discovery of manifest Zn deficiency in 1961, the increasing number of studies demonstrated the association between altered Zn status and multiple diseases. In this chapter, we provide a review of the most recent advances on the role of Zn in health and disease (2010-20), with a special focus on the role of Zn in neurodegenerative and neurodevelopmental disorders, diabetes and obesity, male and female reproduction, as well as COVID-19. In parallel with the revealed tight association between ASD risk and severity and Zn status, the particular mechanisms linking Zn2+ and ASD pathogenesis like modulation of synaptic plasticity through ProSAP/Shank scaffold, neurotransmitter metabolism, and gut microbiota, have been elucidated. The increasing body of data indicate the potential involvement of Zn2+ metabolism in neurodegeneration. Systemic Zn levels in Alzheimer's and Parkinson's disease were found to be reduced, whereas its sequestration in brain may result in modulation of amyloid β and α-synuclein processing with subsequent toxic effects. Zn2+ was shown to possess adipotropic effects through the role of zinc transporters, zinc finger proteins, and Zn-α2-glycoprotein in adipose tissue physiology, underlying its particular role in pathogenesis of obesity and diabetes mellitus type 2. Recent findings also contribute to further understanding of the role of Zn2+ in spermatogenesis and sperm functioning, as well as oocyte development and fertilization. Finally, Zn2+ was shown to be the potential adjuvant therapy in management of novel coronavirus infection (COVID-19), underlining the perspectives of zinc in management of old and new threats.
Collapse
Affiliation(s)
- Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia.
| |
Collapse
|
24
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
25
|
Basak I, Wicky HE, McDonald KO, Xu JB, Palmer JE, Best HL, Lefrancois S, Lee SY, Schoderboeck L, Hughes SM. A lysosomal enigma CLN5 and its significance in understanding neuronal ceroid lipofuscinosis. Cell Mol Life Sci 2021; 78:4735-4763. [PMID: 33792748 PMCID: PMC8195759 DOI: 10.1007/s00018-021-03813-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/09/2023]
Abstract
Neuronal Ceroid Lipofuscinosis (NCL), also known as Batten disease, is an incurable childhood brain disease. The thirteen forms of NCL are caused by mutations in thirteen CLN genes. Mutations in one CLN gene, CLN5, cause variant late-infantile NCL, with an age of onset between 4 and 7 years. The CLN5 protein is ubiquitously expressed in the majority of tissues studied and in the brain, CLN5 shows both neuronal and glial cell expression. Mutations in CLN5 are associated with the accumulation of autofluorescent storage material in lysosomes, the recycling units of the cell, in the brain and peripheral tissues. CLN5 resides in the lysosome and its function is still elusive. Initial studies suggested CLN5 was a transmembrane protein, which was later revealed to be processed into a soluble form. Multiple glycosylation sites have been reported, which may dictate its localisation and function. CLN5 interacts with several CLN proteins, and other lysosomal proteins, making it an important candidate to understand lysosomal biology. The existing knowledge on CLN5 biology stems from studies using several model organisms, including mice, sheep, cattle, dogs, social amoeba and cell cultures. Each model organism has its advantages and limitations, making it crucial to adopt a combinatorial approach, using both human cells and model organisms, to understand CLN5 pathologies and design drug therapies. In this comprehensive review, we have summarised and critiqued existing literature on CLN5 and have discussed the missing pieces of the puzzle that need to be addressed to develop an efficient therapy for CLN5 Batten disease.
Collapse
Affiliation(s)
- I Basak
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - H E Wicky
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - K O McDonald
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - J B Xu
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - J E Palmer
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - H L Best
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
- School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Wales, CF10 3AX, United Kingdom
| | - S Lefrancois
- Centre INRS-Institut Armand-Frappier, INRS, Laval, H7V 1B7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, H3A 2B2, Canada
| | - S Y Lee
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - L Schoderboeck
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand
| | - S M Hughes
- Neurodegenerative and Lysosomal Disease Laboratory, Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, 710 Cumberland Street, Dunedin, 9016, New Zealand.
| |
Collapse
|
26
|
Synaptic Zinc: An Emerging Player in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094724. [PMID: 33946908 PMCID: PMC8125092 DOI: 10.3390/ijms22094724] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/03/2023] Open
Abstract
Alterations of zinc homeostasis have long been implicated in Parkinson's disease (PD). Zinc plays a complex role as both deficiency and excess of intracellular zinc levels have been incriminated in the pathophysiology of the disease. Besides its role in multiple cellular functions, Zn2+ also acts as a synaptic transmitter in the brain. In the forebrain, subset of glutamatergic neurons, namely cortical neurons projecting to the striatum, use Zn2+ as a messenger alongside glutamate. Overactivation of the cortico-striatal glutamatergic system is a key feature contributing to the development of PD symptoms and dopaminergic neurotoxicity. Here, we will cover recent evidence implicating synaptic Zn2+ in the pathophysiology of PD and discuss its potential mechanisms of actions. Emphasis will be placed on the functional interaction between Zn2+ and glutamatergic NMDA receptors, the most extensively studied synaptic target of Zn2+.
Collapse
|
27
|
The Multifaceted Roles of Zinc in Neuronal Mitochondrial Dysfunction. Biomedicines 2021; 9:biomedicines9050489. [PMID: 33946782 PMCID: PMC8145363 DOI: 10.3390/biomedicines9050489] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
Zinc is a highly abundant cation in the brain, essential for cellular functions, including transcription, enzymatic activity, and cell signaling. However, zinc can also trigger injurious cascades in neurons, contributing to the pathology of neurodegenerative diseases. Mitochondria, critical for meeting the high energy demands of the central nervous system (CNS), are a principal target of the deleterious actions of zinc. An increasing body of work suggests that intracellular zinc can, under certain circumstances, contribute to neuronal damage by inhibiting mitochondrial energy processes, including dissipation of the mitochondrial membrane potential (MMP), leading to ATP depletion. Additional consequences of zinc-mediated mitochondrial damage include reactive oxygen species (ROS) generation, mitochondrial permeability transition, and excitotoxic calcium deregulation. Zinc can also induce mitochondrial fission, resulting in mitochondrial fragmentation, as well as inhibition of mitochondrial motility. Here, we review the known mechanisms responsible for the deleterious actions of zinc on the organelle, within the context of neuronal injury associated with neurodegenerative processes. Elucidating the critical contributions of zinc-induced mitochondrial defects to neurotoxicity and neurodegeneration may provide insight into novel therapeutic targets in the clinical setting.
Collapse
|
28
|
Luo Y, Fu Y, Huang Z, Li M. Transition metals and metal complexes in autophagy and diseases. J Cell Physiol 2021; 236:7144-7158. [PMID: 33694161 DOI: 10.1002/jcp.30359] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 12/19/2022]
Abstract
Transition metals refer to the elements in the d and ds blocks of the periodic table. Since the success of cisplatin and auranofin, transition metal-based compounds have become a prospective source for drug development, particularly in cancer treatment. In recent years, extensive studies have shown that numerous transition metal-based compounds could modulate autophagy, promising a new therapeutic strategy for metal-related diseases and the design of metal-based agents. Copper, zinc, and manganese, which are common components in physiological pathways, play important roles in the progression of cancer, neurodegenerative diseases, and cardiovascular diseases. Furthermore, enrichment of copper, zinc, or manganese can regulate autophagy. Thus, we summarized the current advances in elucidating the mechanisms of some metals/metal-based compounds and their functions in autophagy regulation, which is conducive to explore the intricate roles of autophagy and exploit novel therapeutic drugs for human diseases.
Collapse
Affiliation(s)
- Yuping Luo
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Fu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiying Huang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Min Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
ATP13A2 Regulates Cellular α-Synuclein Multimerization, Membrane Association, and Externalization. Int J Mol Sci 2021; 22:ijms22052689. [PMID: 33799982 PMCID: PMC7962109 DOI: 10.3390/ijms22052689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/02/2022] Open
Abstract
ATP13A2, a late endo-/lysosomal polyamine transporter, is implicated in a variety of neurodegenerative diseases, including Parkinson’s disease and Kufor–Rakeb syndrome, an early-onset atypical form of parkinsonism. Loss-of-function mutations in ATP13A2 result in lysosomal deficiency as a consequence of impaired lysosomal export of the polyamines spermine/spermidine. Furthermore, accumulating evidence suggests the involvement of ATP13A2 in regulating the fate of α-synuclein, such as cytoplasmic accumulation and external release. However, no consensus has yet been reached on the mechanisms underlying these effects. Here, we aimed to gain more insight into how ATP13A2 is linked to α-synuclein biology in cell models with modified ATP13A2 activity. We found that loss of ATP13A2 impairs lysosomal membrane integrity and induces α-synuclein multimerization at the membrane, which is enhanced in conditions of oxidative stress or exposure to spermine. In contrast, overexpression of ATP13A2 wildtype (WT) had a protective effect on α-synuclein multimerization, which corresponded with reduced αsyn membrane association and stimulation of the ubiquitin-proteasome system. We also found that ATP13A2 promoted the secretion of α-synuclein through nanovesicles. Interestingly, the catalytically inactive ATP13A2 D508N mutant also affected polyubiquitination and externalization of α-synuclein multimers, suggesting a regulatory function independent of the ATPase and transport activity. In conclusion, our study demonstrates the impact of ATP13A2 on α-synuclein multimerization via polyamine transport dependent and independent functions.
Collapse
|
30
|
Prasuhn J, Davis RL, Kumar KR. Targeting Mitochondrial Impairment in Parkinson's Disease: Challenges and Opportunities. Front Cell Dev Biol 2021; 8:615461. [PMID: 33469539 PMCID: PMC7813753 DOI: 10.3389/fcell.2020.615461] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The underlying pathophysiology of Parkinson's disease is complex, but mitochondrial dysfunction has an established and prominent role. This is supported by an already large and rapidly growing body of evidence showing that the role of mitochondrial (dys)function is central and multifaceted. However, there are clear gaps in knowledge, including the dilemma of explaining why inherited mitochondriopathies do not usually present with parkinsonian symptoms. Many aspects of mitochondrial function are potential therapeutic targets, including reactive oxygen species production, mitophagy, mitochondrial biogenesis, mitochondrial dynamics and trafficking, mitochondrial metal ion homeostasis, sirtuins, and endoplasmic reticulum links with mitochondria. Potential therapeutic strategies may also incorporate exercise, microRNAs, mitochondrial transplantation, stem cell therapies, and photobiomodulation. Despite multiple studies adopting numerous treatment strategies, clinical trials to date have generally failed to show benefit. To overcome this hurdle, more accurate biomarkers of mitochondrial dysfunction are required to detect subtle beneficial effects. Furthermore, selecting study participants early in the disease course, studying them for suitable durations, and stratifying them according to genetic and neuroimaging findings may increase the likelihood of successful clinical trials. Moreover, treatments involving combined approaches will likely better address the complexity of mitochondrial dysfunction in Parkinson's disease. Therefore, selecting the right patients, at the right time, and using targeted combination treatments, may offer the best chance for development of an effective novel therapy targeting mitochondrial dysfunction in Parkinson's disease.
Collapse
Affiliation(s)
- Jannik Prasuhn
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Department of Neurology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Center for Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Ryan L Davis
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, Sydney, NSW, Australia.,Department of Neurogenetics, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Kishore R Kumar
- Molecular Medicine Laboratory and Department of Neurology, Concord Repatriation General Hospital, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
31
|
Clark EH, Vázquez de la Torre A, Hoshikawa T, Briston T. Targeting mitophagy in Parkinson's disease. J Biol Chem 2021; 296:100209. [PMID: 33372898 PMCID: PMC7948953 DOI: 10.1074/jbc.rev120.014294] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
The genetics and pathophysiology of Parkinson's disease (PD) strongly implicate mitochondria in disease aetiology. Elegant studies over the last two decades have elucidated complex molecular signaling governing the identification and removal of dysfunctional mitochondria from the cell, a process of mitochondrial quality control known as mitophagy. Mitochondrial deficits and specifically reduced mitophagy are evident in both sporadic and familial PD. Mendelian genetics attributes loss-of-function mutations in key mitophagy regulators PINK1 and Parkin to early-onset PD. Pharmacologically enhancing mitophagy and accelerating the removal of damaged mitochondria are of interest for developing a disease-modifying PD therapeutic. However, despite significant understanding of both PINK1-Parkin-dependent and -independent mitochondrial quality control pathways, the therapeutic potential of targeting mitophagy remains to be fully explored. Here, we provide a summary of the genetic evidence supporting the role for mitophagy failure as a pathogenic mechanism in PD. We assess the tractability of mitophagy pathways and prospects for drug discovery and consider intervention points for mitophagy enhancement. We explore the numerous hit molecules beginning to emerge from high-content/high-throughput screening as well as the biochemical and phenotypic assays that enabled these screens. The chemical and biological properties of these reference compounds suggest many could be used to interrogate and perturb mitochondrial biology to validate promising drug targets. Finally, we address key considerations and challenges in achieving preclinical proof-of-concept, including in vivo mitophagy reporter methodologies and disease models, as well as patient stratification and biomarker development for mitochondrial forms of the disease.
Collapse
Affiliation(s)
- Emily H Clark
- Hatfield Research Laboratories, Neurology Innovation Centre, Eisai Ltd, Hatfield, United Kingdom
| | | | - Tamaki Hoshikawa
- Hatfield Research Laboratories, Neurology Innovation Centre, Eisai Ltd, Hatfield, United Kingdom
| | - Thomas Briston
- Hatfield Research Laboratories, Neurology Innovation Centre, Eisai Ltd, Hatfield, United Kingdom.
| |
Collapse
|
32
|
Gopurappilly R. Pluripotent Stem Cell Derived Neurons as In Vitro Models for Studying Autosomal Recessive Parkinson's Disease (ARPD): PLA2G6 and Other Gene Loci. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:115-133. [PMID: 33990932 PMCID: PMC7612166 DOI: 10.1007/5584_2021_643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative motor disorder which is largely sporadic; however, some familial forms have been identified. Genetic PD can be inherited by autosomal, dominant or recessive mutations. While the dominant mutations mirror the prototype of PD with adult-onset and L-dopa-responsive cases, autosomal recessive PD (ARPD) exhibit atypical phenotypes with additional clinical manifestations. Young-onset PD is also very common with mutations in recessive gene loci. The main genes associated with ARPD are Parkin, PINK1, DJ-1, ATP13A2, FBXO7 and PLA2G6. Calcium dyshomeostasis is a mainstay in all types of PD, be it genetic or sporadic. Intriguingly, calcium imbalances manifesting as altered Store-Operated Calcium Entry (SOCE) is suggested in PLA2G6-linked PARK 14 PD. The common pathways underlying ARPD pathology, including mitochondrial abnormalities and autophagic dysfunction, can be investigated ex vivo using induced pluripotent stem cell (iPSC) technology and are discussed here. PD pathophysiology is not faithfully replicated by animal models, and, therefore, nigral dopaminergic neurons generated from iPSC serve as improved human cellular models. With no cure to date and treatments aiming at symptomatic relief, these in vitro models derived through midbrain floor-plate induction provide a platform to understand the molecular and biochemical pathways underlying PD etiology in a patient-specific manner.
Collapse
Affiliation(s)
- Renjitha Gopurappilly
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India.
| |
Collapse
|
33
|
ATP13A2-mediated endo-lysosomal polyamine export counters mitochondrial oxidative stress. Proc Natl Acad Sci U S A 2020; 117:31198-31207. [PMID: 33229544 PMCID: PMC7733819 DOI: 10.1073/pnas.1922342117] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mutations in ATP13A2 cause a spectrum of related neurodegenerative disorders. ATP13A2 is a lysosomal exporter of polyamines that contributes to lysosomal health and controls cellular polyamine content. Conversely, loss of ATP13A2 leads to lysosomal dysfunction, a hallmark of neurodegeneration. Here, we show that polyamines transported by ATP13A2 provide cellular protection by lowering reactive oxygen species (ROS), which may relate to the antioxidant properties of polyamines. Consequently, dysfunctional ATP13A2 sensitizes cells to oxidative stress, which impairs mitochondria, and induces toxicity and cell death. ATP13A2-mediated polyamine transport represents a conserved pathway that protects against mitochondrial oxidative stress. The combined protective impact of ATP13A2 on lysosomal health and mitochondrial oxidative stress may explain why ATP13A2 exerts potent neuroprotective effects. Recessive loss-of-function mutations in ATP13A2 (PARK9) are associated with a spectrum of neurodegenerative disorders, including Parkinson’s disease (PD). We recently revealed that the late endo-lysosomal transporter ATP13A2 pumps polyamines like spermine into the cytosol, whereas ATP13A2 dysfunction causes lysosomal polyamine accumulation and rupture. Here, we investigate how ATP13A2 provides protection against mitochondrial toxins such as rotenone, an environmental PD risk factor. Rotenone promoted mitochondrial-generated superoxide (MitoROS), which was exacerbated by ATP13A2 deficiency in SH-SY5Y cells and patient-derived fibroblasts, disturbing mitochondrial functionality and inducing toxicity and cell death. Moreover, ATP13A2 knockdown induced an ATF4-CHOP-dependent stress response following rotenone exposure. MitoROS and ATF4-CHOP were blocked by MitoTEMPO, a mitochondrial antioxidant, suggesting that the impact of ATP13A2 on MitoROS may relate to the antioxidant properties of spermine. Pharmacological inhibition of intracellular polyamine synthesis with α-difluoromethylornithine (DFMO) also increased MitoROS and ATF4 when ATP13A2 was deficient. The polyamine transport activity of ATP13A2 was required for lowering rotenone/DFMO-induced MitoROS, whereas exogenous spermine quenched rotenone-induced MitoROS via ATP13A2. Interestingly, fluorescently labeled spermine uptake in the mitochondria dropped as a consequence of ATP13A2 transport deficiency. Our cellular observations were recapitulated in vivo, in a Caenorhabditis elegans strain deficient in the ATP13A2 ortholog catp-6. These animals exhibited a basal elevated MitoROS level, mitochondrial dysfunction, and enhanced stress response regulated by atfs-1, the C. elegans ortholog of ATF4, causing hypersensitivity to rotenone, which was reversible with MitoTEMPO. Together, our study reveals a conserved cell protective pathway that counters mitochondrial oxidative stress via ATP13A2-mediated lysosomal spermine export.
Collapse
|
34
|
Quinn PMJ, Moreira PI, Ambrósio AF, Alves CH. PINK1/PARKIN signalling in neurodegeneration and neuroinflammation. Acta Neuropathol Commun 2020; 8:189. [PMID: 33168089 PMCID: PMC7654589 DOI: 10.1186/s40478-020-01062-w] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations in the PTEN-induced kinase 1 (PINK1) and Parkin RBR E3 ubiquitin-protein ligase (PARKIN) genes are associated with familial forms of Parkinson’s disease (PD). PINK1, a protein kinase, and PARKIN, an E3 ubiquitin ligase, control the specific elimination of dysfunctional or superfluous mitochondria, thus fine-tuning mitochondrial network and preserving energy metabolism. PINK1 regulates PARKIN translocation in impaired mitochondria and drives their removal via selective autophagy, a process known as mitophagy. As knowledge obtained using different PINK1 and PARKIN transgenic animal models is being gathered, growing evidence supports the contribution of mitophagy impairment to several human pathologies, including PD and Alzheimer’s diseases (AD). Therefore, therapeutic interventions aiming to modulate PINK1/PARKIN signalling might have the potential to treat these diseases. In this review, we will start by discussing how the interplay of PINK1 and PARKIN signalling helps mediate mitochondrial physiology. We will continue by debating the role of mitochondrial dysfunction in disorders such as amyotrophic lateral sclerosis, Alzheimer’s, Huntington’s and Parkinson’s diseases, as well as eye diseases such as age-related macular degeneration and glaucoma, and the causative factors leading to PINK1/PARKIN-mediated neurodegeneration and neuroinflammation. Finally, we will discuss PINK1/PARKIN gene augmentation possibilities with a particular focus on AD, PD and glaucoma.
Collapse
|
35
|
Hinarejos I, Machuca C, Sancho P, Espinós C. Mitochondrial Dysfunction, Oxidative Stress and Neuroinflammation in Neurodegeneration with Brain Iron Accumulation (NBIA). Antioxidants (Basel) 2020; 9:antiox9101020. [PMID: 33092153 PMCID: PMC7589120 DOI: 10.3390/antiox9101020] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
The syndromes of neurodegeneration with brain iron accumulation (NBIA) encompass a group of invalidating and progressive rare diseases that share the abnormal accumulation of iron in the basal ganglia. The onset of NBIA disorders ranges from infancy to adulthood. Main clinical signs are related to extrapyramidal features (dystonia, parkinsonism and choreoathetosis), and neuropsychiatric abnormalities. Ten NBIA forms are widely accepted to be caused by mutations in the genes PANK2, PLA2G6, WDR45, C19ORF12, FA2H, ATP13A2, COASY, FTL1, CP, and DCAF17. Nonetheless, many patients remain without a conclusive genetic diagnosis, which shows that there must be additional as yet undiscovered NBIA genes. In line with this, isolated cases of known monogenic disorders, and also, new genetic diseases, which present with abnormal brain iron phenotypes compatible with NBIA, have been described. Several pathways are involved in NBIA syndromes: iron and lipid metabolism, mitochondrial dynamics, and autophagy. However, many neurodegenerative conditions share features such as mitochondrial dysfunction and oxidative stress, given the bioenergetics requirements of neurons. This review aims to describe the existing link between the classical ten NBIA forms by examining their connection with mitochondrial impairment as well as oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Isabel Hinarejos
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (I.H.); (C.M.); (P.S.)
- Rare Diseases Joint Units, CIPF-IIS La Fe & INCLIVA, 46012 Valencia, Spain
| | - Candela Machuca
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (I.H.); (C.M.); (P.S.)
- Rare Diseases Joint Units, CIPF-IIS La Fe & INCLIVA, 46012 Valencia, Spain
- Unit of Stem Cells Therapies in Neurodegenerative Diseases, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain
| | - Paula Sancho
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (I.H.); (C.M.); (P.S.)
- Rare Diseases Joint Units, CIPF-IIS La Fe & INCLIVA, 46012 Valencia, Spain
| | - Carmen Espinós
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain; (I.H.); (C.M.); (P.S.)
- Rare Diseases Joint Units, CIPF-IIS La Fe & INCLIVA, 46012 Valencia, Spain
- Department of Genetics, University of Valencia, 46100 Valencia, Spain
- Correspondence: ; Tel.: +34-963-289-680
| |
Collapse
|
36
|
Soto-Avellaneda A, Morrison BE. Signaling and other functions of lipids in autophagy: a review. Lipids Health Dis 2020; 19:214. [PMID: 32998777 PMCID: PMC7525950 DOI: 10.1186/s12944-020-01389-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
The process of autophagy is integral to cellular function. In this process, proteins, organelles, and metabolites are engulfed in a lipid vesicle and trafficked to a lysosome for degradation. Its central role in protein and organelle homeostasis has piqued interest for autophagy dysfunction as a driver of pathology for a number of diseases including cancer, muscular disorders, neurological disorders, and non-alcoholic fatty liver disease. For much of its history, the study of autophagy has centered around proteins, however, due to advances in mass spectrometry and refined methodologies, the role of lipids in this essential cellular process has become more apparent. This review discusses the diverse endogenous lipid compounds shown to mediate autophagy. Downstream lipid signaling pathways are also reviewed in the context of autophagy regulation. Specific focus is placed upon the Mammalian Target of Rapamycin (mTOR) and Peroxisome Proliferator-Activated Receptor (PPAR) signaling pathways as integration hubs for lipid regulation of autophagy.
Collapse
Affiliation(s)
| | - Brad E Morrison
- Biomolecular Sciences Graduate programs, Boise State University, Boise, ID, 83725, USA.
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA.
| |
Collapse
|
37
|
Trinh D, Israwi AR, Arathoon LR, Gleave JA, Nash JE. The multi-faceted role of mitochondria in the pathology of Parkinson's disease. J Neurochem 2020; 156:715-752. [PMID: 33616931 DOI: 10.1111/jnc.15154] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria are essential for neuronal function. They produce ATP to meet energy demands, regulate homeostasis of ion levels such as calcium and regulate reactive oxygen species that cause oxidative cellular stress. Mitochondria have also been shown to regulate protein synthesis within themselves, as well as within the nucleus, and also influence synaptic plasticity. These roles are especially important for neurons, which have higher energy demands and greater susceptibility to stress. Dysfunction of mitochondria has been associated with several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, Huntington's disease, Glaucoma and Amyotrophic Lateral Sclerosis. The focus of this review is on how and why mitochondrial function is linked to the pathology of Parkinson's disease (PD). Many of the PD-linked genetic mutations which have been identified result in dysfunctional mitochondria, through a wide-spread number of mechanisms. In this review, we describe how susceptible neurons are predisposed to be vulnerable to the toxic events that occur during the neurodegenerative process of PD, and how mitochondria are central to these pathways. We also discuss ways in which proteins linked with familial PD control mitochondrial function, both physiologically and pathologically, along with their implications in genome-wide association studies and risk assessment. Finally, we review potential strategies for disease modification through mitochondrial enhancement. Ultimately, agents capable of both improving and/or restoring mitochondrial function, either alone, or in conjunction with other disease-modifying agents may halt or slow the progression of neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Dennison Trinh
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Ahmad R Israwi
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Lindsay R Arathoon
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Jacqueline A Gleave
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| | - Joanne E Nash
- Department of Biological Sciences, University of Toronto Scarborough, Centre for Neurobiology of Stress, Toronto, ON, Canada
| |
Collapse
|
38
|
Butz ES, Chandrachud U, Mole SE, Cotman SL. Moving towards a new era of genomics in the neuronal ceroid lipofuscinoses. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165571. [DOI: 10.1016/j.bbadis.2019.165571] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
|
39
|
Watanabe H, Hattori T, Kume A, Misu K, Ito T, Koike Y, Johnson TA, Kamitsuji S, Kamatani N, Sobue G. Improved Parkinsons disease motor score in a single-arm open-label trial of febuxostat and inosine. Medicine (Baltimore) 2020; 99:e21576. [PMID: 32871874 PMCID: PMC7458241 DOI: 10.1097/md.0000000000021576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cellular energetics play an important role in Parkinsons disease etiology, but no treatments directly address this deficiency. Our past research showed that treatment with febuxostat and inosine increased blood hypoxanthine and ATP in healthy adults, and a preliminary trial in 3 Parkinson's disease patients suggested some symptomatic improvements with no adverse effects. METHODS To examine the efficacy on symptoms and safety in a larger group of Parkinsons disease patients, we conducted a single-arm, open-label trial at 5 Japanese neurology clinics and enrolled thirty patients (nmales = 11; nfemales = 19); 26 patients completed the study (nmales = 10; nfemales = 16). Each patient was administered febuxostat 20 mg and inosine 500 mg twice-per-day (after breakfast and dinner) for 8 weeks. The primary endpoint was the difference of MDS-UPDRS Part III score immediately before and after 57 days of treatment. RESULTS Serum hypoxanthine concentrations were raised significantly after treatment (Pre = 11.4 μM; Post = 38.1 μM; P < .0001). MDS-UPDRS Part III score was significantly lower after treatment (Pre = 28.1 ± 9.3; Post = 24.7 ± 10.8; mean ± SD; P = .0146). Sixteen adverse events occurred in 13/29 (44.8%) patients, including 1 serious adverse event (fracture of the second lumbar vertebra) that was considered not related to the treatment. CONCLUSIONS The results of this study suggest that co-administration of febuxostat and inosine is relatively safe and effective for improving symptoms of Parkinsons disease patients. Further controlled trials need to be performed to confirm the symptomatic improvement and to examine the disease-modifying effect in long-term trials.
Collapse
Affiliation(s)
- Hirohisa Watanabe
- Nagoya University Graduate School of Medicine, Brain and Mind Research Center, Nagoya
- Fujita Health University School of Medicine, Department of Neurology, Toyoake
| | | | | | | | | | | | | | | | | | - Gen Sobue
- Nagoya University Graduate School of Medicine, Brain and Mind Research Center, Nagoya
| |
Collapse
|
40
|
Wali G, Kumar KR, Liyanage E, Davis RL, Mackay-Sim A, Sue CM. Mitochondrial Function in Hereditary Spastic Paraplegia: Deficits in SPG7 but Not SPAST Patient-Derived Stem Cells. Front Neurosci 2020; 14:820. [PMID: 32973427 PMCID: PMC7469654 DOI: 10.3389/fnins.2020.00820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022] Open
Abstract
Mutations in SPG7 and SPAST are common causes of hereditary spastic paraplegia (HSP). While some SPG7 mutations cause paraplegin deficiency, other SPG7 mutations cause increased paraplegin expression. Mitochondrial function has been studied in models that are paraplegin-deficient (human, mouse, and Drosophila models with large exonic deletions, null mutations, or knockout models) but not in models of mutations that express paraplegin. Here, we evaluated mitochondrial function in olfactory neurosphere-derived cells, derived from patients with a variety of SPG7 mutations that express paraplegin and compared them to cells derived from healthy controls and HSP patients with SPAST mutations, as a disease control. We quantified paraplegin expression and an extensive range of mitochondrial morphology measures (fragmentation, interconnectivity, and mass), mitochondrial function measures (membrane potential, oxidative phosphorylation, and oxidative stress), and cell proliferation. Compared to control cells, SPG7 patient cells had increased paraplegin expression, fragmented mitochondria with low interconnectivity, reduced mitochondrial mass, decreased mitochondrial membrane potential, reduced oxidative phosphorylation, reduced ATP content, increased mitochondrial oxidative stress, and reduced cellular proliferation. Mitochondrial dysfunction was specific to SPG7 patient cells and not present in SPAST patient cells, which displayed mitochondrial functions similar to control cells. The mitochondrial dysfunction observed here in SPG7 patient cells that express paraplegin was similar to the dysfunction reported in cell models without paraplegin expression. The p.A510V mutation was common to all patients and was the likely species associated with increased expression, albeit seemingly non-functional. The lack of a mitochondrial phenotype in SPAST patient cells indicates genotype-specific mechanisms of disease in these HSP patients.
Collapse
Affiliation(s)
- Gautam Wali
- Department of Neurogenetics, Royal North Shore Hospital, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Kishore Raj Kumar
- Department of Neurogenetics, Royal North Shore Hospital, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Department of Neurology, Concord Hospital, Sydney, NSW, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Department of Neurology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Erandhi Liyanage
- Department of Neurogenetics, Royal North Shore Hospital, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Ryan L Davis
- Department of Neurogenetics, Royal North Shore Hospital, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Alan Mackay-Sim
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | - Carolyn M Sue
- Department of Neurogenetics, Royal North Shore Hospital, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Neurology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| |
Collapse
|
41
|
Chang KH, Chen CM. The Role of Oxidative Stress in Parkinson's Disease. Antioxidants (Basel) 2020; 9:antiox9070597. [PMID: 32650609 PMCID: PMC7402083 DOI: 10.3390/antiox9070597] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Parkinson’s disease (PD) is caused by progressive neurodegeneration of dopaminergic (DAergic) neurons with abnormal accumulation of α-synuclein in substantia nigra (SN). Studies have suggested the potential involvement of dopamine, iron, calcium, mitochondria and neuroinflammation in contributing to overwhelmed oxidative stress and neurodegeneration in PD. Function studies on PD-causative mutations of SNCA, PRKN, PINK1, DJ-1, LRRK2, FBXO7 and ATP13A2 further indicate the role of oxidative stress in the pathogenesis of PD. Therefore, it is reasonable that molecules involved in oxidative stress, such as DJ-1, coenzyme Q10, uric acid, 8-hydroxy-2’-deoxyguanosin, homocysteine, retinoic acid/carotenes, vitamin E, glutathione peroxidase, superoxide dismutase, xanthine oxidase and products of lipid peroxidation, could be candidate biomarkers for PD. Applications of antioxidants to modulate oxidative stress could be a strategy in treating PD. Although a number of antioxidants, such as creatine, vitamin E, coenzyme Q10, pioglitazone, melatonin and desferrioxamine, have been tested in clinical trials, none of them have demonstrated conclusive evidence to ameliorate the neurodegeneration in PD patients. Difficulties in clinical studies may be caused by the long-standing progression of neurodegeneration, lack of biomarkers for premotor stage of PD and inadequate drug delivery across blood–brain barrier. Solutions for these challenges will be warranted for future studies with novel antioxidative treatment in PD patients.
Collapse
Affiliation(s)
| | - Chiung-Mei Chen
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8347); Fax: +886-3-3288849
| |
Collapse
|
42
|
Martín-Jiménez R, Lurette O, Hebert-Chatelain E. Damage in Mitochondrial DNA Associated with Parkinson's Disease. DNA Cell Biol 2020; 39:1421-1430. [PMID: 32397749 DOI: 10.1089/dna.2020.5398] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the only organelles that contain their own genetic material (mtDNA). Mitochondria are involved in several key physiological functions, including ATP production, Ca2+ homeostasis, and metabolism of neurotransmitters. Since these organelles perform crucial processes to maintain neuronal homeostasis, mitochondrial dysfunctions can lead to various neurodegenerative diseases. Several mitochondrial proteins involved in ATP production are encoded by mtDNA. Thus, any mtDNA alteration can ultimately lead to mitochondrial dysfunction and cell death. Accumulation of mutations, deletions, and rearrangements in mtDNA has been observed in animal models and patients suffering from Parkinson's disease (PD). Also, specific inherited variations associated with mtDNA genetic groups (known as mtDNA haplogroups) are associated with lower or higher risk of developing PD. Consequently, mtDNA alterations should now be considered important hallmarks of this neurodegenerative disease. This review provides an update about the role of mtDNA alterations in the physiopathology of PD.
Collapse
Affiliation(s)
- Rebeca Martín-Jiménez
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Olivier Lurette
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Etienne Hebert-Chatelain
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| |
Collapse
|
43
|
Smolders S, Van Broeckhoven C. Genetic perspective on the synergistic connection between vesicular transport, lysosomal and mitochondrial pathways associated with Parkinson's disease pathogenesis. Acta Neuropathol Commun 2020; 8:63. [PMID: 32375870 PMCID: PMC7201634 DOI: 10.1186/s40478-020-00935-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) and atypical parkinsonian syndromes (APS) are symptomatically characterized by parkinsonism, with the latter presenting additionally a distinctive range of atypical features. Although the majority of patients with PD and APS appear to be sporadic, genetic causes of several rare monogenic disease variants were identified. The knowledge acquired from these genetic factors indicated that defects in vesicular transport pathways, endo-lysosomal dysfunction, impaired autophagy-lysosomal protein and organelle degradation pathways, α-synuclein aggregation and mitochondrial dysfunction play key roles in PD pathogenesis. Moreover, membrane dynamics are increasingly recognized as a key player in the disease pathogenesis due lipid homeostasis alterations, associated with lysosomal dysfunction, caused by mutations in several PD and APS genes. The importance of lysosomal dysfunction and lipid homeostasis is strengthened by both genetic discoveries and clinical epidemiology of the association between parkinsonism and lysosomal storage disorders (LSDs), caused by the disruption of lysosomal biogenesis or function. A synergistic coordination between vesicular trafficking, lysosomal and mitochondria defects exist whereby mutations in PD and APS genes encoding proteins primarily involved one PD pathway are frequently associated with defects in other PD pathways as a secondary effect. Moreover, accumulating clinical and genetic observations suggest more complex inheritance patters of familial PD exist, including oligogenic and polygenic inheritance of genes in the same or interconnected PD pathways, further strengthening their synergistic connection.Here, we provide a comprehensive overview of PD and APS genes with functions in vesicular transport, lysosomal and mitochondrial pathways, and highlight functional and genetic evidence of the synergistic connection between these PD associated pathways.
Collapse
Affiliation(s)
- Stefanie Smolders
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, University of Antwerp - CDE, Universiteitsplein 1, 2610, Antwerpen, Belgium
- Biomedical Sciences, University of Antwerp, Antwerpen, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, University of Antwerp - CDE, Universiteitsplein 1, 2610, Antwerpen, Belgium.
- Biomedical Sciences, University of Antwerp, Antwerpen, Belgium.
| |
Collapse
|
44
|
Bourdenx M, Nioche A, Dovero S, Arotcarena ML, Camus S, Porras G, Thiolat ML, Rougier NP, Prigent A, Aubert P, Bohic S, Sandt C, Laferrière F, Doudnikoff E, Kruse N, Mollenhauer B, Novello S, Morari M, Leste-Lasserre T, Trigo-Damas I, Goillandeau M, Perier C, Estrada C, Garcia-Carrillo N, Recasens A, Vaikath NN, El-Agnaf OMA, Herrero MT, Derkinderen P, Vila M, Obeso JA, Dehay B, Bezard E. Identification of distinct pathological signatures induced by patient-derived α-synuclein structures in nonhuman primates. SCIENCE ADVANCES 2020; 6:eaaz9165. [PMID: 32426502 PMCID: PMC7220339 DOI: 10.1126/sciadv.aaz9165] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/06/2020] [Indexed: 06/11/2023]
Abstract
Dopaminergic neuronal cell death, associated with intracellular α-synuclein (α-syn)-rich protein aggregates [termed "Lewy bodies" (LBs)], is a well-established characteristic of Parkinson's disease (PD). Much evidence, accumulated from multiple experimental models, has suggested that α-syn plays a role in PD pathogenesis, not only as a trigger of pathology but also as a mediator of disease progression through pathological spreading. Here, we have used a machine learning-based approach to identify unique signatures of neurodegeneration in monkeys induced by distinct α-syn pathogenic structures derived from patients with PD. Unexpectedly, our results show that, in nonhuman primates, a small amount of singular α-syn aggregates is as toxic as larger amyloid fibrils present in the LBs, thus reinforcing the need for preclinical research in this species. Furthermore, our results provide evidence supporting the true multifactorial nature of PD, as multiple causes can induce a similar outcome regarding dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- M. Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - A. Nioche
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- Institut Jean Nicod, Département d’études cognitives, ENS, EHESS, PSL Research University, 75005 Paris, France
- Institut Jean Nicod, Département d’études cognitives, CNRS, UMR 8129, Paris, France
| | - S. Dovero
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - M.-L. Arotcarena
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - S. Camus
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - G. Porras
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - M.-L. Thiolat
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - N. P. Rougier
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- INRIA Bordeaux Sud-Ouest, 33405 Talence, France
| | - A. Prigent
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - P. Aubert
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - S. Bohic
- EA-7442 Rayonnement Synchrotron et Recherche Medicale, RSRM, University of Grenoble Alpes, 38000 Grenoble, France
| | - C. Sandt
- SMIS beamline, Synchrotron SOLEIL, l’orme des merisiers, 91192 Gif sur Yvette, France
| | - F. Laferrière
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - E. Doudnikoff
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - N. Kruse
- Paracelsus-Elena-Klinik, Kassel, Germany
- University Medical Center Goettingen, Institute of Neuropathology, Goettingen, Germany
| | - B. Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany
- University Medical Center Goettingen, Institute of Neuropathology, Goettingen, Germany
| | - S. Novello
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
- Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - M. Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
- Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - T. Leste-Lasserre
- INSERM, Neurocentre Magendie, U1215, Physiopathologie de la Plasticité Neuronale, F-33000 Bordeaux, France
| | - I. Trigo-Damas
- HM CINAC, HM Puerta del Sur and CEU–San Pablo University Madrid, E-28938 Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - M. Goillandeau
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - C. Perier
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - C. Estrada
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - N. Garcia-Carrillo
- Centro Experimental en Investigaciones Biomédica (CEIB), Universidad de Murcia, Murcia, Spain
| | - A. Recasens
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - N. N. Vaikath
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar
| | - O. M. A. El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar
| | - M. T. Herrero
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - P. Derkinderen
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - M. Vila
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - J. A. Obeso
- HM CINAC, HM Puerta del Sur and CEU–San Pablo University Madrid, E-28938 Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - B. Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - E. Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
45
|
Ge P, Dawson VL, Dawson TM. PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson's disease. Mol Neurodegener 2020; 15:20. [PMID: 32169097 PMCID: PMC7071653 DOI: 10.1186/s13024-020-00367-7] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
That certain cell types in the central nervous system are more likely to undergo neurodegeneration in Parkinson's disease is a widely appreciated but poorly understood phenomenon. Many vulnerable subpopulations, including dopamine neurons in the substantia nigra pars compacta, have a shared phenotype of large, widely distributed axonal networks, dense synaptic connections, and high basal levels of neural activity. These features come at substantial bioenergetic cost, suggesting that these neurons experience a high degree of mitochondrial stress. In such a context, mechanisms of mitochondrial quality control play an especially important role in maintaining neuronal survival. In this review, we focus on understanding the unique challenges faced by the mitochondria in neurons vulnerable to neurodegeneration in Parkinson's and summarize evidence that mitochondrial dysfunction contributes to disease pathogenesis and to cell death in these subpopulations. We then review mechanisms of mitochondrial quality control mediated by activation of PINK1 and Parkin, two genes that carry mutations associated with autosomal recessive Parkinson's disease. We conclude by pinpointing critical gaps in our knowledge of PINK1 and Parkin function, and propose that understanding the connection between the mechanisms of sporadic Parkinson's and defects in mitochondrial quality control will lead us to greater insights into the question of selective vulnerability.
Collapse
Affiliation(s)
- Preston Ge
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
- Present address: Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Present address: Picower Institute for Learning and Memory, Cambridge, MA 02139 USA
- Present address: Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, MA 02115 USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| |
Collapse
|
46
|
Sironi L, Restelli LM, Tolnay M, Neutzner A, Frank S. Dysregulated Interorganellar Crosstalk of Mitochondria in the Pathogenesis of Parkinson's Disease. Cells 2020; 9:cells9010233. [PMID: 31963435 PMCID: PMC7016713 DOI: 10.3390/cells9010233] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of Parkinson’s disease (PD), the second most common neurodegenerative disorder, is complex and involves the impairment of crucial intracellular physiological processes. Importantly, in addition to abnormal α-synuclein aggregation, the dysfunction of various mitochondria-dependent processes has been prominently implicated in PD pathogenesis. Besides the long-known loss of the organelles’ bioenergetics function resulting in diminished ATP synthesis, more recent studies in the field have increasingly focused on compromised mitochondrial quality control as well as impaired biochemical processes specifically localized to ER–mitochondria interfaces (such as lipid biosynthesis and calcium homeostasis). In this review, we will discuss how dysregulated mitochondrial crosstalk with other organelles contributes to PD pathogenesis.
Collapse
Affiliation(s)
- Lara Sironi
- Division of Neuropathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.M.R.); (M.T.)
- Correspondence: (L.S.); (S.F.); Tel.: +41-61-265-2776 (L.S. & S.F.)
| | - Lisa Michelle Restelli
- Division of Neuropathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.M.R.); (M.T.)
| | - Markus Tolnay
- Division of Neuropathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.M.R.); (M.T.)
| | - Albert Neutzner
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland;
- Department of Ophthalmology University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Stephan Frank
- Division of Neuropathology, Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.M.R.); (M.T.)
- Correspondence: (L.S.); (S.F.); Tel.: +41-61-265-2776 (L.S. & S.F.)
| |
Collapse
|
47
|
GBA mutation promotes early mitochondrial dysfunction in 3D neurosphere models. Aging (Albany NY) 2019; 11:10338-10355. [PMID: 31751314 PMCID: PMC6914435 DOI: 10.18632/aging.102460] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/08/2019] [Indexed: 12/31/2022]
Abstract
Glucocerebrosidase (GBA) mutations are the most important genetic risk factor for the development of Parkinson disease (PD). GBA encodes the lysosomal enzyme glucocerebrosidase (GCase). Loss-of-GCase activity in cellular models has implicated lysosomal and mitochondrial dysfunction in PD disease pathogenesis, although the exact mechanisms remain unclear. We hypothesize that GBA mutations impair mitochondria quality control in a neurosphere model. We have characterized mitochondrial content, mitochondrial function and macroautophagy flux in 3D-neurosphere-model derived from neural crest stem cells containing heterozygous and homozygous N370SGBA mutations, under carbonyl cyanide-m-chlorophenyl-hydrazine (CCCP)- induced mitophagy. Our findings on mitochondrial markers and ATP levels indicate that mitochondrial accumulation occurs in mutant N370SGBA neurospheres under basal conditions, and clearance of depolarised mitochondria is impaired following CCCP-treatment. A significant increase in TFEB-mRNA levels, the master regulator of lysosomal and autophagy genes, may explain an unchanged macroautophagy flux in N370SGBA neurospheres. PGC1α-mRNA levels were also significantly increased following CCCP-treatment in heterozygote, but not homozygote neurospheres, and might contribute to the increased mitochondrial content seen in cells with this genotype, probably as a compensatory mechanism that is absent in homozygous lines. Mitochondrial impairment occurs early in the development of GCase-deficient neurons. Furthermore, impaired turnover of depolarised mitochondria is associated with early mitochondrial dysfunction. In summary, the presence of GBA mutation may be associated with higher levels of mitochondrial content in homozygous lines and lower clearance of damaged mitochondria in our neurosphere model.
Collapse
|
48
|
Heins-Marroquin U, Jung PP, Cordero-Maldonado ML, Crawford AD, Linster CL. Phenotypic assays in yeast and zebrafish reveal drugs that rescue ATP13A2 deficiency. Brain Commun 2019; 1:fcz019. [PMID: 32954262 PMCID: PMC7425419 DOI: 10.1093/braincomms/fcz019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/27/2019] [Accepted: 08/16/2019] [Indexed: 12/21/2022] Open
Abstract
Mutations in ATP13A2 (PARK9) are causally linked to the rare neurodegenerative disorders Kufor-Rakeb syndrome, hereditary spastic paraplegia and neuronal ceroid lipofuscinosis. This suggests that ATP13A2, a lysosomal cation-transporting ATPase, plays a crucial role in neuronal cells. The heterogeneity of the clinical spectrum of ATP13A2-associated disorders is not yet well understood and currently, these diseases remain without effective treatment. Interestingly, ATP13A2 is widely conserved among eukaryotes, and the yeast model for ATP13A2 deficiency was the first to indicate a role in heavy metal homeostasis, which was later confirmed in human cells. In this study, we show that the deletion of YPK9 (the yeast orthologue of ATP13A2) in Saccharomyces cerevisiae leads to growth impairment in the presence of Zn2+, Mn2+, Co2+ and Ni2+, with the strongest phenotype being observed in the presence of zinc. Using the ypk9Δ mutant, we developed a high-throughput growth rescue screen based on the Zn2+ sensitivity phenotype. Screening of two libraries of Food and Drug Administration-approved drugs identified 11 compounds that rescued growth. Subsequently, we generated a zebrafish model for ATP13A2 deficiency and found that both partial and complete loss of atp13a2 function led to increased sensitivity to Mn2+. Based on this phenotype, we confirmed two of the drugs found in the yeast screen to also exert a rescue effect in zebrafish-N-acetylcysteine, a potent antioxidant, and furaltadone, a nitrofuran antibiotic. This study further supports that combining the high-throughput screening capacity of yeast with rapid in vivo drug testing in zebrafish can represent an efficient drug repurposing strategy in the context of rare inherited disorders involving conserved genes. This work also deepens the understanding of the role of ATP13A2 in heavy metal detoxification and provides a new in vivo model for investigating ATP13A2 deficiency.
Collapse
Affiliation(s)
- Ursula Heins-Marroquin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Paul P Jung
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | | | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
- Institute for Orphan Drug Discovery, Bremer Innovations- und Technologiezentrum, 28359 Bremen, Germany
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
49
|
Baesler J, Kopp JF, Pohl G, Aschner M, Haase H, Schwerdtle T, Bornhorst J. Zn homeostasis in genetic models of Parkinson's disease in Caenorhabditis elegans. J Trace Elem Med Biol 2019; 55:44-49. [PMID: 31345364 PMCID: PMC6676891 DOI: 10.1016/j.jtemb.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 01/13/2023]
Abstract
While the underlying mechanisms of Parkinson's disease (PD) are still insufficiently studied, a complex interaction between genetic and environmental factors is emphasized. Nevertheless, the role of the essential trace element zinc (Zn) in this regard remains controversial. In this study we altered Zn balance within PD models of the versatile model organism Caenorhabditis elegans (C. elegans) in order to examine whether a genetic predisposition in selected genes with relevance for PD affects Zn homeostasis. Protein-bound and labile Zn species act in various areas, such as enzymatic catalysis, protein stabilization pathways and cell signaling. Therefore, total Zn and labile Zn were quantitatively determined in living nematodes as individual biomarkers of Zn uptake and bioavailability with inductively coupled plasma tandem mass spectrometry (ICP-MS/MS) or a multi-well method using the fluorescent probe ZinPyr-1. Young and middle-aged deletion mutants of catp-6 and pdr-1, which are orthologues of mammalian ATP13A2 (PARK9) and parkin (PARK2), showed altered Zn homeostasis following Zn exposure compared to wildtype worms. Furthermore, age-specific differences in Zn uptake were observed in wildtype worms for total as well as labile Zn species. These data emphasize the importance of differentiation between Zn species as meaningful biomarkers of Zn uptake as well as the need for further studies investigating the role of dysregulated Zn homeostasis in the etiology of PD.
Collapse
Affiliation(s)
- Jessica Baesler
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany
| | - Johannes F Kopp
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany
| | - Michael Aschner
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461 Bronx, NY, USA
| | - Hajo Haase
- TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany; Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany
| | - Julia Bornhorst
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit FOR 2558, Berlin, Potsdam, Jena, Germany; Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany.
| |
Collapse
|
50
|
Grünewald A, Kumar KR, Sue CM. New insights into the complex role of mitochondria in Parkinson’s disease. Prog Neurobiol 2019; 177:73-93. [DOI: 10.1016/j.pneurobio.2018.09.003] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/09/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
|