1
|
Jain S, Vohora D. Primary components of MCT ketogenic diet are detrimental to bone loss associated with accelerated aging and age-related neurotoxicity in mice. Bone 2025; 192:117383. [PMID: 39732448 DOI: 10.1016/j.bone.2024.117383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Medium chained triglycerides (MCT) ketogenic diet is being extensively investigated for its neuroprotective effects against adverse effects associated with aging and neurodegenerative disorders. Aging is a common risk factor for the development of both osteoporosis and neurological disorders. Hence, suppression of aging and age-related neurodegeneration might contribute to delaying skeletal aging. The present study was designed to investigate the effects of the primary components of the MCT diet, against bone resorption associated with D-gal-induced accelerated aging and D-gal /AlCl3-induced age-related toxicity. We report bone loss in accelerated aged mice and age-related neurotoxic mice through declined Sirtuin1 (SIRT1) expression, depleted bone turnover markers, (P1NP and β-CTX-1), low bone mineral density (BMD), and deterioration of trabecular bone microarchitecture in both the distal femur and proximal tibia bones. Administration of MCT dietary components decanoic acid and octanoic acid, led to a decrease in body weight and only octanoic acid increased serum levels of ketone body, β-hydroxybutyrate (β-HB), but both of them failed to reverse the diminishing effects on bone health associated with aging and age-related neurotoxicity. Surprisingly, decanoic acid, octanoic acid, and their combination also exhibited negative effects on trabecular bone microarchitecture and BMD in the distal femur and proximal tibia bones of healthy mice. The findings from this study provide supporting evidence on the deterioration of bone health associated with aging and age-related neurotoxicity, and the bone resorption potential of MCT dietary supplements that are being prescribed in healthy older populations and elderly persons diagnosed with neurological disorders.
Collapse
Affiliation(s)
- Shreshta Jain
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
2
|
Ali I, Adil M, Imran M, Qureshi SA, Qureshi S, Hasan N, Ahmad FJ. Nanotechnology in Parkinson's Disease: overcoming drug delivery challenges and enhancing therapeutic outcomes. Drug Deliv Transl Res 2025:10.1007/s13346-025-01799-8. [PMID: 39878857 DOI: 10.1007/s13346-025-01799-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
The global prevalence of Parkinson's Disease (PD) is on the rise, driven by an ageing population and ongoing environmental conditions. To gain a better understanding of PD pathogenesis, it is essential to consider its relationship with the ageing process, as ageing stands out as the most significant risk factor for this neurodegenerative condition. PD risk factors encompass genetic predisposition, exposure to environmental toxins, and lifestyle influences, collectively increasing the chance of PD development. Moreover, early and precise PD diagnosis remains elusive, relying on clinical assessments, neuroimaging techniques, and emerging biomarkers. Conventional management of PD involves dopaminergic medications and surgical interventions, but these treatments often become less effective over time and do not address disease treatment. Challenges persist due to the blood-brain barrier's (BBB) impermeability, hindering drug delivery. Recent advancements in nanotechnology offer promising novel approaches for PD management. Various drug delivery systems (DDS), including nanosized polymers, lipid-based carriers, and nanoparticles (such as metal/metal oxide, protein, and carbonaceous particles), aim to enhance drug and gene delivery. These modifications seek to improve BBB permeability, ultimately benefiting PD patients. This review underscores the critical role of ageing in PD development and explores how age-related neuronal decline contributes to substantia nigra loss and PD manifestation in susceptible individuals. The review also highlights the advancements and ongoing challenges in nanotechnology-based therapies for PD.
Collapse
Affiliation(s)
- Irfan Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Adil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Imran
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, 4102, Australia
| | - Saba Asif Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saima Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
3
|
Gunawardena S. Alpha-synuclein in mitochondrial dysfunction: opportunities or obstacles. Neural Regen Res 2025; 20:197-198. [PMID: 39657088 DOI: 10.4103/nrr.nrr-d-23-01966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/05/2024] [Indexed: 12/17/2024] Open
Affiliation(s)
- Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
4
|
Hu Y, Yang H, Song C, Tian L, Wang P, Li T, Cheng C, AlNusaif M, Li S, Liang Z, Le W. LRRK2 G2019S Gene Mutation Causes Skeletal Muscle Impairment in Animal Model of Parkinson's Disease. J Cachexia Sarcopenia Muscle 2024; 15:2595-2607. [PMID: 39310961 PMCID: PMC11634472 DOI: 10.1002/jcsm.13604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 08/12/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND While the gradually aggravated motor and non-motor disorders of Parkinson's disease (PD) lead to progressive disability and frequent falling, skeletal muscle impairment may contribute to this condition. The leucine-rich repeat kinase2 (LRRK2) is a common disease-causing gene in PD. Little is known about its role in skeletal muscle impairment and its underlying mechanisms. METHODS To investigate whether the mutation in LRRK2 causes skeletal muscle impairment, we used 3-month-old (3mo) and 14-month-old (14mo) LRRK2G2019S transgenic (TG) mice as a model of PD, compared with the age-matched littermate wild-type (WT) controls. We measured the muscle mass and strength, ultrastructure, inflammatory infiltration, mitochondrial morphology and dynamics dysfunction through behavioural analysis, electromyography (EMG), immunostaining, transmission electron microscopy (TEM) and other molecular biology techniques. RESULTS The 3mo-TG mice display mild skeletal muscle impairment with spontaneous potentials in EMG (increased by 130%, p < 0.05), myofibre necrosis (p < 0.05) and myosin heavy chain-II changes (reduced by 19%, p < 0.01). The inflammatory cells and macrophage infiltration are significantly increased (CD8a+ and CD68+ cells up 1060% and 579%, respectively, both p < 0.0001) compared with the WT mice. All of the above pathogenic processes are aggravated by aging. The 14mo-TG mice EMG examinations show a reduced duration (by 31%, p < 0.01) and increased polyphasic waves of motor unit action potentials (by 28%, p < 0.05). The 14mo-TG mice present motor behavioural deficits (p < 0.05), muscle strength and mass reduction by 37% and 8% (p < 0.05 and p < 0.01, respectively). A remarkable increase in inflammatory infiltration is accompanied by pro-inflammatory cytokines in the skeletal muscles. TEM analysis shows muscle fibre regeneration with the reduced length of sarcomeres (by 6%;p < 0.05). The muscle regeneration is activated as Pax7+ cells increased by 106% (p < 0.0001), andmyoblast determination protein elevated by 71% (p < 0.01). We also document the morphological changes and dynamics dysfunction of mitochondria with the increase of mitofusin1 by 43% (p < 0.05) and voltage-dependent anion channel 1 by 115% (p < 0.001) in the skeletal muscles of 14mo-TG mice. CONCLUSIONS Taken together, these findings may provide new insights into the clinical and pathogenic involvement of LRRK2G2019 mutation in muscles, suggesting that the diseases may affect not only midbrain dopaminergic neurons, but also other tissues, and it may help overall clinical management of this devastating disease.
Collapse
Affiliation(s)
- Yiying Hu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of NeurologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Huijia Yang
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Chunli Song
- Department of NeurologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lulu Tian
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Panpan Wang
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Tianbai Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Cheng Cheng
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Murad AlNusaif
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Song Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhanhua Liang
- Department of NeurologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Weidong Le
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological DiseasesThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
5
|
Hattori N, Sato S. Mitochondrial dysfunction in Parkinson's disease. J Neural Transm (Vienna) 2024; 131:1415-1428. [PMID: 39585446 DOI: 10.1007/s00702-024-02863-2] [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: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
The exact cause of nigral cell death in Parkinson's disease (PD) is still unknown. However, research on MPTP-induced experimental parkinsonism has significantly advanced our understanding. In this model, it is widely accepted that mitochondrial respiratory failure is the primary mechanism of cell death. Studies have shown that a toxic metabolite of MPTP inhibits Complex I and alpha-ketoglutarate dehydrogenase activities in mitochondria. Since then, many research groups have focused on mitochondrial dysfunction in PD, identifying deficiencies in Complex I or III in PD patients' brains, skeletal muscle, and platelets. There is some debate about the decline in mitochondrial function in peripheral organs. However, since α-synuclein, the main component protein of Lewy bodies, accumulates in peripheral organs, it is reasonable to consider PD a systemic disease. Additionally, mutant mitochondrial DNA with a 4,977 base pair deletion has been found in the brains of PD patients, suggesting that age-related accumulation of deleted mtDNA is accelerated in the striatum and may contribute to the pathophysiology of PD. While the cause of PD remains unknown, mitochondrial dysfunction is undoubtedly a factor in cell death in PD. In addition, the causative gene for familial PD, parkin (now PRKN), and PTEN-induced putative kinase 1 (PINK1), both gene products are also involved in mitochondrial quality control. Moreover, we have successfully isolated and identified CHCHD2, which is involved in the mitochondrial electron transfer system. There is no doubt that mitochondrial dysfunction contributes to cell death in PD.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1-Hirosawa, Wako-Shi, Saitama, 351-0198, Japan.
| | - Shigeto Sato
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
- Center for Biomedical Research Resources, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
6
|
Sharma Y, Gupta JK, Babu MA, Singh S, Sindhu RK. Signaling Pathways Concerning Mitochondrial Dysfunction: Implications in Neurodegeneration and Possible Molecular Targets. J Mol Neurosci 2024; 74:101. [PMID: 39466510 DOI: 10.1007/s12031-024-02269-5] [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: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
Mitochondrion is an important organelle present in our cells responsible for meeting energy requirements. All higher organisms rely on efficient mitochondrial bioenergetic machinery to sustain life. No other respiratory process can produce as much power as generated by mitochondria in the form of ATPs. This review is written in order to get an insight into the magnificent working of mitochondrion and its implications in cellular homeostasis, bioenergetics, redox, calcium signaling, and cell death. However, if this machinery gets faulty, it may lead to several disease states. Mitochondrial dysfunctioning is of growing concern today as it is seen in the pathogenesis of several diseases which includes neurodegenerative disorders, cardiovascular disorders, diabetes mellitus, skeletal muscle defects, liver diseases, and so on. To cover all these aspects is beyond the scope of this article; hence, our study is restricted to neurodegenerative disorders only. Moreover, faulty functioning of this organelle can be one of the causes of early ageing in individuals. This review emphasizes mutations in the mitochondrial DNA, defects in oxidative phosphorylation, generation of ROS, and apoptosis. Researchers have looked into new approaches that might be able to control mitochondrial failure and show a lot of promise as treatments.
Collapse
Affiliation(s)
- Yati Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Jeetendra Kumar Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Sumitra Singh
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Rakesh K Sindhu
- School of Pharmacy, Sharda University, Gautam Buddha Nagar, Greater Noida, Uttar Paresdh, 201310, India.
| |
Collapse
|
7
|
Li Y, Zhang H, Yu C, Dong X, Yang F, Wang M, Wen Z, Su M, Li B, Yang L. New Insights into Mitochondria in Health and Diseases. Int J Mol Sci 2024; 25:9975. [PMID: 39337461 PMCID: PMC11432609 DOI: 10.3390/ijms25189975] [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: 08/28/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondria are a unique type of semi-autonomous organelle within the cell that carry out essential functions crucial for the cell's survival and well-being. They are the location where eukaryotic cells carry out energy metabolism. Aside from producing the majority of ATP through oxidative phosphorylation, which provides essential energy for cellular functions, mitochondria also participate in other metabolic processes within the cell, such as the electron transport chain, citric acid cycle, and β-oxidation of fatty acids. Furthermore, mitochondria regulate the production and elimination of ROS, the synthesis of nucleotides and amino acids, the balance of calcium ions, and the process of cell death. Therefore, it is widely accepted that mitochondrial dysfunction is a factor that causes or contributes to the development and advancement of various diseases. These include common systemic diseases, such as aging, diabetes, Parkinson's disease, and cancer, as well as rare metabolic disorders, like Kearns-Sayre syndrome, Leigh disease, and mitochondrial myopathy. This overview outlines the various mechanisms by which mitochondria are involved in numerous illnesses and cellular physiological activities. Additionally, it provides new discoveries regarding the involvement of mitochondria in both disorders and the maintenance of good health.
Collapse
Affiliation(s)
- Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Chunjuan Yu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Xiaolei Dong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Fanghao Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Mengjun Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Ziyuan Wen
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Mohan Su
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, China
| |
Collapse
|
8
|
Sagredo GT, Tanglay O, Shahdadpuri S, Fu Y, Halliday GM. ⍺-Synuclein levels in Parkinson's disease - Cell types and forms that contribute to pathogenesis. Exp Neurol 2024; 379:114887. [PMID: 39009177 DOI: 10.1016/j.expneurol.2024.114887] [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: 04/02/2024] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Parkinson's disease (PD) has two main pathological hallmarks, the loss of nigral dopamine neurons and the proteinaceous aggregations of ⍺-synuclein (⍺Syn) in neuronal Lewy pathology. These two co-existing features suggest a causative association between ⍺Syn aggregation and the underpinning mechanism of neuronal degeneration in PD. Both increased levels and post-translational modifications of ⍺Syn can contribute to the formation of pathological aggregations of ⍺Syn in neurons. Recent studies have shown that the protein is also expressed by multiple types of non-neuronal cells in the brain and peripheral tissues, suggesting additional roles of the protein and potential diversity in non-neuronal pathogenic triggers. It is important to determine (1) the threshold levels triggering ⍺Syn to convert from a biological to a pathologic form in different brain cells in PD; (2) the dominant form of pathologic ⍺Syn and the associated post-translational modification of the protein in each cell type involved in PD; and (3) the cell type associated biological processes impacted by pathologic ⍺Syn in PD. This review integrates these aspects and speculates on potential pathological mechanisms and their impact on neuronal and non-neuronal ⍺Syn in the brains of patients with PD.
Collapse
Affiliation(s)
- Giselle Tatiana Sagredo
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Onur Tanglay
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia
| | - Shrey Shahdadpuri
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia
| | - YuHong Fu
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Glenda M Halliday
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America.
| |
Collapse
|
9
|
Ying C, Zhang H, Wang T, Li Y, Mao W, Hu S, Zhao L, Cai Y. Plasma level of alpha-synuclein oligomers as a biomarker for isolated rapid eye movement sleep behavior disorder diagnosis and progression: a prospective cohort study. Front Neurol 2024; 15:1442173. [PMID: 39246606 PMCID: PMC11377258 DOI: 10.3389/fneur.2024.1442173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alpha-synuclein oligomers (o-α-syn) are pivotal in the pathogenesis of α-synucleinopathy. Isolated rapid eye movement (REM) sleep behavior disorder (iRBD) serves as an early indicator of the disease, offering insights into disease mechanisms and early intervention. Nevertheless, the diagnostic and predictive potential of o-α-syn in iRBD remains largely unexplored. This study aimed to evaluate the plasma levels of o-α-syn in patients and investigate their utility as biomarkers for diagnosis of and predicting phenoconversion in iRBD. Methods A total of 143 participants, including 77 polysomnography-confirmed iRBD patients and 66 normal controls (NC), were recruited for this longitudinal observational study. Baseline clinical assessments and plasma collection were conducted for all iRBD patients, with 72 of them undergoing regularly prospective follow-up assessments for parkinsonism or dementia. Plasma levels of o-α-syn were quantified using enzyme-linked immunosorbent assay, and were compared between groups using a general linear model adjusted for age and sex. The diagnostic performance of plasma o-α-syn in iRBD was evaluated by area under the receiver operating characteristic curve (AUC) with 95% CI. Cox regression analysis and Kaplan-Meier survival curves were employed to assess the predictive value of plasma o-α-syn for phenoconversion in iRBD. Results Plasma o-α-syn levels did not exhibit statistically significant differences among iRBD converter patients, iRBD nonconverter patients, and NC. The AUC for distinguishing NC from iRBD was 0.52 (95% CI: 0.42-0.62, p = 0.682). Spearman correlation analysis revealed a significant positive correlation between plasma o-α-syn levels and MOCA scores in the iRBD group (p < 0.001). Subgroup analyses indicated that iRBD patients with cognitive decline (p = 0.058) and depressive symptoms (p = 0.017) had notably lower o-α-syn levels compared to those without such symptoms. Over a median follow-up period of 5.83 years, 26 iRBD patients developed neurodegenerative synucleinopathies. Cox regression and Kaplan-Meier survival curve analyses indicated that plasma level of o-α-syn lacked a predictive value for disease conversion in iRBD patients. Conclusion Despite a potential role in the pathophysiology of iRBD, o-α-syn are not appropriate biomarkers for diagnosing or predicting disease progression. While this study offers insights into the pathogenesis of iRBD and neurodegenerative synucleinopathies, further large-scale longitudinal studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Chao Ying
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Hui Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ting Wang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yuan Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Mao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Songnian Hu
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Lifang Zhao
- Beijing Geriatric Medical Research Center, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Akyazı O, Korkmaz D, Cevher SC. Experimental Parkinson models and green chemistry approach. Behav Brain Res 2024; 471:115092. [PMID: 38844056 DOI: 10.1016/j.bbr.2024.115092] [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: 02/03/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Parkinson's is the most common neurodegenerative disease after Alzheimer's. Motor findings in Parkinson's occur as a result of the degeneration of dopaminergic neurons starting in the substantia nigra pars compacta and ending in the putamen and caudate nucleus. Loss of neurons and the formation of inclusions called Lewy bodies in existing neurons are characteristic histopathological findings of Parkinson's. The disease primarily impairs the functional capacity of the person with cardinal findings such as tremor, bradykinesia, etc., as a result of the loss of dopaminergic neurons in the substantia nigra. Experimental animal models of Parkinson's have been used extensively in recent years to investigate the pathology of this disease. These models are generally based on systemic or local(intracerebral) administration of neurotoxins, which can replicate many features of Parkinson's mammals. The development of transgenic models in recent years has allowed us to learn more about the modeling of Parkinson's. Applying animal modeling, which shows the most human-like effects in studies, is extremely important. It has been demonstrated that oxidative stress increases in many neurodegenerative diseases such as Parkinson's and various age-related degenerative diseases in humans and that neurons are sensitive to it. In cases where oxidative stress increases and antioxidant systems are inadequate, natural molecules such as flavonoids and polyphenols can be used as a new antioxidant treatment to reduce neuronal reactive oxygen species and improve the neurodegenerative process. Therefore, in this article, we examined experimental animal modeling in Parkinson's disease and the effect of green chemistry approaches on Parkinson's disease.
Collapse
Affiliation(s)
- Ozge Akyazı
- Gazi University, Institute of Science, Department of Biology, Ankara 06500, Turkey.
| | - Dılara Korkmaz
- Gazi University, Institute of Science, Department of Biology, Ankara 06500, Turkey
| | - Sule Coskun Cevher
- Gazi University, Faculty of Science, Department of Biology, Ankara 06500, Turkey
| |
Collapse
|
11
|
Xian M, Li J, Liu T, Hou K, Sun L, Wei J. β-Synuclein Intermediates α-Synuclein Neurotoxicity in Parkinson's Disease. ACS Chem Neurosci 2024; 15:2445-2453. [PMID: 38905183 DOI: 10.1021/acschemneuro.4c00263] [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] [Indexed: 06/23/2024] Open
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disease in the world, and synuclein is closely related to the onset and progression of PD. Synuclein is considered a therapeutic target for PD. Recent studies have found that abnormal aggregation of α-synuclein (α-Syn) in the brains of PD patients leads to mitochondrial dysfunction and neuroinflammation. Research in the field of neuroscience has confirmed that β-synuclein (β-Syn) also plays a role in Parkinson's disease. However, there has been little research on the role mechanisms and interactions between β-Syn and α-Syn in PD. Therefore, the purpose of this study is to clarify the relationship between α-Syn, β-Syn, and PD and to explore the roles and interactions of β-Syn and α-Syn in PD.
Collapse
Affiliation(s)
- Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jingwen Li
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Kaiying Hou
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| |
Collapse
|
12
|
Sanluca C, Spagnolo P, Mancinelli R, De Bartolo MI, Fava M, Maccarrone M, Carotti S, Gaudio E, Leuti A, Vivacqua G. Interaction between α-Synuclein and Bioactive Lipids: Neurodegeneration, Disease Biomarkers and Emerging Therapies. Metabolites 2024; 14:352. [PMID: 39057675 PMCID: PMC11278689 DOI: 10.3390/metabo14070352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
The present review provides a comprehensive examination of the intricate dynamics between α-synuclein, a protein crucially involved in the pathogenesis of several neurodegenerative diseases, including Parkinson's disease and multiple system atrophy, and endogenously-produced bioactive lipids, which play a pivotal role in neuroinflammation and neurodegeneration. The interaction of α-synuclein with bioactive lipids is emerging as a critical factor in the development and progression of neurodegenerative and neuroinflammatory diseases, offering new insights into disease mechanisms and novel perspectives in the identification of potential biomarkers and therapeutic targets. We delve into the molecular pathways through which α-synuclein interacts with biological membranes and bioactive lipids, influencing the aggregation of α-synuclein and triggering neuroinflammatory responses, highlighting the potential of bioactive lipids as biomarkers for early disease detection and progression monitoring. Moreover, we explore innovative therapeutic strategies aimed at modulating the interaction between α-synuclein and bioactive lipids, including the development of small molecules and nutritional interventions. Finally, the review addresses the significance of the gut-to-brain axis in mediating the effects of bioactive lipids on α-synuclein pathology and discusses the role of altered gut lipid metabolism and microbiota composition in neuroinflammation and neurodegeneration. The present review aims to underscore the potential of targeting α-synuclein-lipid interactions as a multifaceted approach for the detection and treatment of neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Chiara Sanluca
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Paolo Spagnolo
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Romina Mancinelli
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | | | - Marina Fava
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Simone Carotti
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| | - Eugenio Gaudio
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | - Alessandro Leuti
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Giorgio Vivacqua
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| |
Collapse
|
13
|
Bhatt V, Shukla H, Tiwari AK. Parkinson's Disease and Mitotherapy-Based Approaches towards α-Synucleinopathies. J Integr Neurosci 2024; 23:109. [PMID: 38940084 DOI: 10.31083/j.jin2306109] [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: 11/28/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain and the formation of intracellular protein aggregates known as Lewy bodies, of which a major component is the protein α-synuclein. Several studies have suggested that mitochondria play a central role in the pathogenesis of PD, encompassing both familial and sporadic forms of the disease. Mitochondrial dysfunction is attributed to bioenergetic impairment, increased oxidative stress, damage to mitochondrial DNA, and alteration in mitochondrial morphology. These alterations may contribute to improper functioning of the central nervous system and ultimately lead to neurodegeneration. The perturbation of mitochondrial function makes it a potential target, worthy of exploration for neuroprotective therapies and to improve mitochondrial health in PD. Thus, in the current review, we provide an update on mitochondria-based therapeutic approaches toward α-synucleinopathies in PD.
Collapse
Affiliation(s)
- Vidhi Bhatt
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Halak Shukla
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| |
Collapse
|
14
|
Hembach S, Schmidt S, Orschmann T, Burtscher I, Lickert H, Giesert F, Weisenhorn DV, Wurst W. Engrailed 1 deficiency induces changes in ciliogenesis during human neuronal differentiation. Neurobiol Dis 2024; 194:106474. [PMID: 38518837 DOI: 10.1016/j.nbd.2024.106474] [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: 01/23/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
A key pathological feature of Parkinson's Disease (PD) is the progressive degeneration of dopaminergic neurons (DAns) in the substantia nigra pars compacta. Considering the major role of EN1 in the development and maintenance of these DAns and the implications from En1 mouse models, it is highly interesting to study the molecular and protective effect of EN1 also in a human cellular model. Therefore, we generated EN1 knock-out (ko) human induced pluripotent stem cell (hiPSCs) lines and analyzed these during neuronal differentiation. Although the EN1 ko didn't interfere with neuronal differentiation and generation of tyrosine hydroxylase positive (TH+) neurons per se, the neurons exhibited shorter neurites. Furthermore, mitochondrial respiration, as well as mitochondrial complex I abundance was significantly reduced in fully differentiated neurons. To understand the implications of an EN1 ko during differentiation, we performed a transcriptome analysis of human neuronal precursor cells (hNPCs) which unveiled alterations in cilia-associated pathways. Further analysis of ciliary morphology revealed an elongation of primary cilia in EN1-deficient hNPCs. Besides, also Wnt signaling pathways were severely affected. Upon stimulating hNPCs with Wnt which drastically increased EN1 expression in WT lines, the phenotypes concerning mitochondrial function and cilia were exacerbated in EN1 ko hNPCs. They failed to enhance the expression of the complex I subunits NDUFS1 and 3, and now displayed a reduced mitochondrial respiration. Furthermore, Wnt stimulation decreased ciliogenesis in EN1 ko hNPCs but increased ciliary length even further. This further highlights the relevance of primary cilia next to mitochondria for the functionality and correct maintenance of human DAns and provides new possibilities to establish neuroprotective therapies for PD.
Collapse
Affiliation(s)
- Sina Hembach
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany; Munich School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Sebastian Schmidt
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany; Neurobiological Engineering, Munich Institute of Biomedical Engineering, TUM School of Natural Sciences, Garching, Germany; Deutsche Zentrum für Psychische Gesundheit (DZPG), Site Munich-Augsburg, Munich, Germany
| | - Tanja Orschmann
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technische Universität München, Munich, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany
| | | | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg, Germany; Deutsche Zentrum für Psychische Gesundheit (DZPG), Site Munich-Augsburg, Munich, Germany; Technische Universität München-Weihenstephan, Neuherberg, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| |
Collapse
|
15
|
Bej E, Cesare P, Volpe AR, d’Angelo M, Castelli V. Oxidative Stress and Neurodegeneration: Insights and Therapeutic Strategies for Parkinson's Disease. Neurol Int 2024; 16:502-517. [PMID: 38804477 PMCID: PMC11130796 DOI: 10.3390/neurolint16030037] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition marked by the gradual deterioration of dopaminergic neurons in the substantia nigra. Oxidative stress has been identified as a key player in the development of PD in recent studies. In the first part, we discuss the sources of oxidative stress in PD, including mitochondrial dysfunction, dopamine metabolism, and neuroinflammation. This paper delves into the possibility of mitigating oxidative stress as a potential treatment approach for PD. In addition, we examine the hurdles and potential of antioxidant therapy, including the challenge of delivering antioxidants to the brain and the requirement for biomarkers to track oxidative stress in PD patients. However, even if antioxidant therapy holds promise, further investigation is needed to determine its efficacy and safety in PD treatment.
Collapse
Affiliation(s)
| | | | | | | | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (A.R.V.); (M.d.)
| |
Collapse
|
16
|
Zhang Z, Wang R, Zhou H, Wu D, Cao Y, Zhang C, Sun H, Mu C, Hao Z, Ren H, Wang N, Yu S, Zhang J, Tao M, Wang C, Liu Y, Liu L, Liu Y, Zang J, Wang G. Inhibition of EHMT1/2 rescues synaptic damage and motor impairment in a PD mouse model. Cell Mol Life Sci 2024; 81:128. [PMID: 38472451 DOI: 10.1007/s00018-024-05176-5] [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: 10/17/2023] [Revised: 01/16/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
Epigenetic dysregulation that leads to alterations in gene expression and is suggested to be one of the key pathophysiological factors of Parkinson's disease (PD). Here, we found that α-synuclein preformed fibrils (PFFs) induced histone H3 dimethylation at lysine 9 (H3K9me2) and increased the euchromatic histone methyltransferases EHMT1 and EHMT2, which were accompanied by neuronal synaptic damage, including loss of synapses and diminished expression levels of synaptic-related proteins. Furthermore, the levels of H3K9me2 at promoters in genes that encode the synaptic-related proteins SNAP25, PSD95, Synapsin 1 and vGLUT1 were increased in primary neurons after PFF treatment, which suggests a linkage between H3K9 dimethylation and synaptic dysfunction. Inhibition of EHMT1/2 with the specific inhibitor A-366 or shRNA suppressed histone methylation and alleviated synaptic damage in primary neurons that were treated with PFFs. In addition, the synaptic damage and motor impairment in mice that were injected with PFFs were repressed by treatment with the EHMT1/2 inhibitor A-366. Thus, our findings reveal the role of histone H3 modification by EHMT1/2 in synaptic damage and motor impairment in a PFF animal model, suggesting the involvement of epigenetic dysregulation in PD pathogenesis.
Collapse
Affiliation(s)
- Zhixiong Zhang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Rui Wang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hui Zhou
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Dan Wu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yifan Cao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chuang Zhang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hongyang Sun
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chenchen Mu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zongbing Hao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
- Jiangsu Provincial Medical Innovation Center of Trauma Medicine, Institute of Trauma Medicine, Soochow University, Suzhou, 215123, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Nana Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Mengdan Tao
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Can Wang
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Liu
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Liu Liu
- Department of Pharmacy, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Changsha, 410005, China
| | - Yanli Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Jianye Zang
- Hefei National Laboratory for Physical Sciences at Microscale CAS Center for Excellence in Biomacromolecules, Collaborative Innovation Center of Chemistry for Life Sciences, and School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, 215123, Jiangsu, China.
- Center of Translational Medicine, First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, 215400, China.
| |
Collapse
|
17
|
Brash-Arias D, García LI, Pérez-Estudillo CA, Rojas-Durán F, Aranda-Abreu GE, Herrera-Covarrubias D, Chi-Castañeda D. The Role of Astrocytes and Alpha-Synuclein in Parkinson's Disease: A Review. NEUROSCI 2024; 5:71-86. [PMID: 39483813 PMCID: PMC11523690 DOI: 10.3390/neurosci5010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 11/03/2024] Open
Abstract
The search for new therapies to reduce symptoms and find a cure for Parkinson's disease has focused attention on two key points: the accumulation of alpha-synuclein aggregates and astrocytes. The former is a hallmark of the disease, while the latter corresponds to a type of glial cell with an important role in both the prevention and development of this neurodegenerative disorder. Traditionally, research has focused on therapies targeting dopaminergic neurons. Currently, as more is known about the genetic and molecular factors and the neuroglial interaction in the disease, great emphasis has been placed on the neuroprotective role of astrocytes in the early stages of the disease and on the astrocytic capture of alpha-synuclein under both physiological and pathological conditions. This review aims to analyze the contribution of alpha-synuclein and astrocytes to the development and progression of Parkinson's disease, as well as to evaluate recent therapeutic proposals specifically focused on synucleopathies and astroglial cells as potential therapies for the disease.
Collapse
Affiliation(s)
- David Brash-Arias
- Doctorado en Investigaciones Cerebrales, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico;
| | - Luis I. García
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| | | | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| | | | | | - Donaji Chi-Castañeda
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| |
Collapse
|
18
|
Amrein Almira A, Chen MW, El Demerdash N, Javdan C, Park D, Lee JK, Martin LJ. Proteasome localization and activity in pig brain and in vivo small molecule screening for activators. Front Cell Neurosci 2024; 18:1353542. [PMID: 38469354 PMCID: PMC10925635 DOI: 10.3389/fncel.2024.1353542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Loss of proteasome function, proteinopathy, and proteotoxicity may cause neurodegeneration across the human lifespan in several forms of brain injury and disease. Drugs that activate brain proteasomes in vivo could thus have a broad therapeutic impact in neurology. Methods Using pigs, a clinically relevant large animal with a functionally compartmental gyrencephalic cerebral cortex, we evaluated the localization and biochemical activity of brain proteasomes and tested the ability of small molecules to activate brain proteasomes. Results By Western blotting, proteasome protein subunit PSMB5 and PSMA3 levels were similar in different pig brain regions. Immunohistochemistry for PSMB5 showed localization in the cytoplasm (diffuse and particulate) and nucleus (cytoplasm < nucleus). Some PSMB5 immunoreactivity was colocalized with mitochondrial (voltage-gated anion channel and cyclophilin D) and cell death (Aven) proteins in the neuronal soma and neuropil in the neocortex of pig and human brains. In the nucleus, PSMB5 immunoreactivity was diffuse, particulate, and clustered, including perinucleolar decorations. By fluorogenic assay, proteasome chymotrypsin-like activities (CTL) in crude tissue soluble fractions were generally similar within eight different pig brain regions. Proteasome CTL activity in the hippocampus was correlated with activity in nasal mucosa biopsies. In pilot analyses of subcellular fractions of pig cerebral cortex, proteasome CTL activity was highest in the cytosol and then ~50% lower in nuclear fractions; ~15-20% of total CTL activity was in pure mitochondrial fractions. With in-gel activity assay, 26S-singly and -doubly capped proteasomes were the dominant forms in the pig cerebral cortex. With a novel in situ histochemical activity assay, MG132-inhibitable proteasome CTL activity was localized to the neuropil, as a mosaic, and to cell bodies, nuclei, and centrosome-like perinuclear satellites. In piglets treated intravenously with pyrazolone derivative and chlorpromazine over 24 h, brain proteasome CTL activity was modestly increased. Discussion This study shows that the proteasome in the pig brain has relative regional uniformity, prominent nuclear and perinuclear presence with catalytic activity, a mitochondrial association with activity, 26S-single cap dominance, and indications from small molecule systemic administration of pyrazolone derivative and chlorpromazine that brain proteasome function appears safely activable.
Collapse
Affiliation(s)
- Adriana Amrein Almira
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - May W. Chen
- Departments of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nagat El Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cameron Javdan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dongseok Park
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jennifer K. Lee
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lee J. Martin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Keomanivong C, Schamp J, Tabakovic E, Thangavel R, Aldridge G, Pieper AA, Narayanan NS. Mice Expressing A53T/A30P Mutant Alpha-Synuclein in Dopamine Neurons Do Not Display Behavioral Deficits. eNeuro 2024; 11:ENEURO.0170-23.2023. [PMID: 38351057 PMCID: PMC10866330 DOI: 10.1523/eneuro.0170-23.2023] [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: 05/22/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024] Open
Abstract
Alpha-synuclein has been implicated in neurodegenerative diseases such as Parkinson's disease and dementia with Lewy bodies, with A53T and A30P mutations shown to be disease causing. It has been reported that hemizygous transgenic mice with tyrosine hydroxylase promotor-driven expression of A53T/A30P mutant alpha-synuclein in dopamine neurons provide a useful preclinical model of these conditions by virtue of developing behavioral deficits. Here, we report a lack of replication of this finding. Despite detecting robust overexpression of A53T/A30P mutant alpha-synuclein in dopamine neurons, we did not observe decreased tyrosine hydroxylase immunofluorescence or behavioral deficits in these mice. Our results demonstrate that preclinical models of synucleinopathy need careful validation in the field.
Collapse
Affiliation(s)
| | | | - Ervina Tabakovic
- Carver College of Medicine, University of Iowa, Iowa City 52245, Iowa
| | | | | | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland 44106, Ohio; Department of Psychiatry, Case Western Reserve University, Cleveland 44106, Ohio; Geriatric Research Education and Clinical Center (GRECC), Louis Stokes Cleveland VA Medical Center, Cleveland 44106, Ohio; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, Ohio; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland 44106, Ohio; Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland 44106, Ohio; Translational Therapeutics Core, Cleveland Alzheimer's Disease Research Center, Cleveland, Ohio
| | | |
Collapse
|
20
|
Sturchio A, Rocha EM, Kauffman MA, Marsili L, Mahajan A, Saraf AA, Vizcarra JA, Guo Z, Espay AJ. Recalibrating the Why and Whom of Animal Models in Parkinson Disease: A Clinician's Perspective. Brain Sci 2024; 14:151. [PMID: 38391726 PMCID: PMC10887152 DOI: 10.3390/brainsci14020151] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 02/24/2024] Open
Abstract
Animal models have been used to gain pathophysiologic insights into Parkinson's disease (PD) and aid in the translational efforts of interventions with therapeutic potential in human clinical trials. However, no disease-modifying therapy for PD has successfully emerged from model predictions. These translational disappointments warrant a reappraisal of the types of preclinical questions asked of animal models. Besides the limitations of experimental designs, the one-size convergence and oversimplification yielded by a model cannot recapitulate the molecular diversity within and between PD patients. Here, we compare the strengths and pitfalls of different models, review the discrepancies between animal and human data on similar pathologic and molecular mechanisms, assess the potential of organoids as novel modeling tools, and evaluate the types of questions for which models can guide and misguide. We propose that animal models may be of greatest utility in the evaluation of molecular mechanisms, neural pathways, drug toxicity, and safety but can be unreliable or misleading when used to generate pathophysiologic hypotheses or predict therapeutic efficacy for compounds with potential neuroprotective effects in humans. To enhance the translational disease-modification potential, the modeling must reflect the biology not of a diseased population but of subtypes of diseased humans to distinguish What data are relevant and to Whom.
Collapse
Affiliation(s)
- Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Emily M. Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Marcelo A. Kauffman
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología José María Ramos Mejía, Buenos Aires C1221ADC, Argentina;
| | - Luca Marsili
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Abhimanyu Mahajan
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Ameya A. Saraf
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Joaquin A. Vizcarra
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 15213, USA;
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA;
| | - Alberto J. Espay
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| |
Collapse
|
21
|
Yamanaka T, Matsui H. Modeling familial and sporadic Parkinson's disease in small fishes. Dev Growth Differ 2024; 66:4-20. [PMID: 37991125 DOI: 10.1111/dgd.12904] [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: 09/01/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
The establishment of animal models for Parkinson's disease (PD) has been challenging. Nevertheless, once established, they will serve as valuable tools for elucidating the causes and pathogenesis of PD, as well as for developing new strategies for its treatment. Following the recent discovery of a series of PD causative genes in familial cases, teleost fishes, including zebrafish and medaka, have often been used to establish genetic PD models because of their ease of breeding and gene manipulation, as well as the high conservation of gene orthologs. Some of the fish lines can recapitulate PD phenotypes, which are often more pronounced than those in rodent genetic models. In addition, a new experimental teleost fish, turquoise killifish, can be used as a sporadic PD model, because it spontaneously manifests age-dependent PD phenotypes. Several PD fish models have already made significant contributions to the discovery of novel PD pathological features, such as cytosolic leakage of mitochondrial DNA and pathogenic phosphorylation in α-synuclein. Therefore, utilizing various PD fish models with distinct degenerative phenotypes will be an effective strategy for identifying emerging facets of PD pathogenesis and therapeutic modalities.
Collapse
Affiliation(s)
- Tomoyuki Yamanaka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
22
|
Choong CJ, Mochizuki H. Involvement of Mitochondria in Parkinson's Disease. Int J Mol Sci 2023; 24:17027. [PMID: 38069350 PMCID: PMC10707101 DOI: 10.3390/ijms242317027] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Mitochondrial dysregulation, such as mitochondrial complex I deficiency, increased oxidative stress, perturbation of mitochondrial dynamics and mitophagy, has long been implicated in the pathogenesis of PD. Initiating from the observation that mitochondrial toxins cause PD-like symptoms and mitochondrial DNA mutations are associated with increased risk of PD, many mutated genes linked to familial forms of PD, including PRKN, PINK1, DJ-1 and SNCA, have also been found to affect the mitochondrial features. Recent research has uncovered a much more complex involvement of mitochondria in PD. Disruption of mitochondrial quality control coupled with abnormal secretion of mitochondrial contents to dispose damaged organelles may play a role in the pathogenesis of PD. Furthermore, due to its bacterial ancestry, circulating mitochondrial DNAs can function as damage-associated molecular patterns eliciting inflammatory response. In this review, we summarize and discuss the connection between mitochondrial dysfunction and PD, highlighting the molecular triggers of the disease process, the intra- and extracellular roles of mitochondria in PD as well as the therapeutic potential of mitochondrial transplantation.
Collapse
Affiliation(s)
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan;
| |
Collapse
|
23
|
Li HY, Liu DS, Zhang YB, Rong H, Zhang XJ. The interaction between alpha-synuclein and mitochondrial dysfunction in Parkinson's disease. Biophys Chem 2023; 303:107122. [PMID: 37839353 DOI: 10.1016/j.bpc.2023.107122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/18/2023] [Accepted: 10/07/2023] [Indexed: 10/17/2023]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder with the hallmark of abnormal aggregates of alpha-synuclein (α-syn) in Lewy bodies (LBs) and Lewy neurites (LNs). Currently, pathogenic α-syn and mitochondrial dysfunction have been considered as prominent roles that give impetus to the PD onset. This review describes the α-syn pathology and mitochondrial alterations in PD, and focuses on how α-syn interacts with multiple aspects of mitochondrial homeostasis in the pathogenesis of PD.
Collapse
Affiliation(s)
- Hong-Yan Li
- Department of Basic Medical College, Heilongjiang University of Chinese Medicine, Haerbin 150000, PR China
| | - De-Shui Liu
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Ying-Bo Zhang
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Hua Rong
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Xiao-Jie Zhang
- Department of Basic Medical College, Heilongjiang University of Chinese Medicine, Haerbin 150000, PR China; Heilongjiang Nursing College, Haerbin 150000, PR China.
| |
Collapse
|
24
|
Khan MR, Yin X, Kang SU, Mitra J, Wang H, Ryu T, Brahmachari S, Karuppagounder SS, Kimura Y, Jhaldiyal A, Kim HH, Gu H, Chen R, Redding-Ochoa J, Troncoso J, Na CH, Ha T, Dawson VL, Dawson TM. Enhanced mTORC1 signaling and protein synthesis in pathologic α-synuclein cellular and animal models of Parkinson's disease. Sci Transl Med 2023; 15:eadd0499. [PMID: 38019930 DOI: 10.1126/scitranslmed.add0499] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/10/2023] [Indexed: 12/01/2023]
Abstract
Pathologic α-synuclein plays an important role in the pathogenesis of α-synucleinopathies such as Parkinson's disease (PD). Disruption of proteostasis is thought to be central to pathologic α-synuclein toxicity; however, the molecular mechanism of this deregulation is poorly understood. Complementary proteomic approaches in cellular and animal models of PD were used to identify and characterize the pathologic α-synuclein interactome. We report that the highest biological processes that interacted with pathologic α-synuclein in mice included RNA processing and translation initiation. Regulation of catabolic processes that include autophagy were also identified. Pathologic α-synuclein was found to bind with the tuberous sclerosis protein 2 (TSC2) and to trigger the activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), which augmented mRNA translation and protein synthesis, leading to neurodegeneration. Genetic and pharmacologic inhibition of mTOR and protein synthesis rescued the dopamine neuron loss, behavioral deficits, and aberrant biochemical signaling in the α-synuclein preformed fibril mouse model and Drosophila transgenic models of pathologic α-synuclein-induced degeneration. Pathologic α-synuclein furthermore led to a destabilization of the TSC1-TSC2 complex, which plays an important role in mTORC1 activity. Constitutive overexpression of TSC2 rescued motor deficits and neuropathology in α-synuclein flies. Biochemical examination of PD postmortem brain tissues also suggested deregulated mTORC1 signaling. These findings establish a connection between mRNA translation deregulation and mTORC1 pathway activation that is induced by pathologic α-synuclein in cellular and animal models of PD.
Collapse
Affiliation(s)
- Mohammed Repon Khan
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Xiling Yin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Jaba Mitra
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hu Wang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Taekyung Ryu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Saurav Brahmachari
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Yasuyoshi Kimura
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aanishaa Jhaldiyal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyun Hee Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hao Gu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rong Chen
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Javier Redding-Ochoa
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juan Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Howard Hughes Medical Institute, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
Diaw SH, Borsche M, Streubel-Gallasch L, Dulovic-Mahlow M, Hermes J, Lenz I, Seibler P, Klein C, Brüggemann N, Vos M, Lohmann K. Characterization of the pathogenic α-Synuclein Variant V15A in Parkinson´s disease. NPJ Parkinsons Dis 2023; 9:148. [PMID: 37903765 PMCID: PMC10616187 DOI: 10.1038/s41531-023-00584-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/02/2023] [Indexed: 11/01/2023] Open
Abstract
Despite being a major component of Lewy bodies and Lewy neurites, pathogenic variants in the gene encoding alpha-Synuclein (α-Syn) are rare. To date, only four missense variants in the SNCA gene, encoding α-Syn have unequivocally been shown to be disease-causing. We here describe a Parkinson´s disease patient with early cognitive decline carrying an as yet not fully characterized variant in SNCA (NM_001146055: c.44T > C, p.V15A). We used different cellular models, including stably transfected neuroblastoma (SH-SY5Y) cell cultures, induced pluripotent stem cell (iPSC)-derived neuronal cultures, and generated a Drosophila model to elucidate the impact of the p.V15A variant on α-Syn function and aggregation properties compared to other known pathogenic variants. We demonstrate that p.V15A increased the aggregation potential of α-Syn and the levels of apoptotic markers, and impaired the mitochondrial network. Moreover, p.V15A affects the flying ability and survival of mutant flies. Thus, we provide supporting evidence for the pathogenicity of the p.V15A variant, suggesting its inclusion in genetic testing approaches.
Collapse
Affiliation(s)
| | - Max Borsche
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | | | | | - Julia Hermes
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Insa Lenz
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | - Melissa Vos
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany.
| |
Collapse
|
26
|
Patikas N, Ansari R, Metzakopian E. Single-cell transcriptomics identifies perturbed molecular pathways in midbrain organoids using α-synuclein triplication Parkinson's disease patient-derived iPSCs. Neurosci Res 2023; 195:13-28. [PMID: 37271312 DOI: 10.1016/j.neures.2023.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/26/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Three-dimensional (3D) brain organoids provide a platform to study brain development, cellular coordination, and disease using human tissue. Here, we generate midbrain dopaminergic (mDA) organoids from induced pluripotent stem cells (iPSC) from healthy and Parkinson's Disease (PD) donors and assess them as a human PD model using single-cell RNAseq. We characterize cell types in our organoid cultures and analyze our model's Dopamine (DA) neurons using cytotoxic and genetic stressors. Our study provides the first in-depth, single-cell analysis of SNCA triplication and shows evidence for molecular dysfunction in oxidative phosphorylation, translation, and ER protein-folding in DA neurons. We perform an in-silico identification of rotenone-sensitive DA neurons and characterization of corresponding transcriptomic profiles associated with synaptic signalling and cholesterol biosynthesis. Finally, we show a novel chimera organoid model from healthy and PD iPSCs allowing the study of DA neurons from different individuals within the same tissue.
Collapse
Affiliation(s)
- Nikolaos Patikas
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0AH, UK.
| | - Rizwan Ansari
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0AH, UK
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0AH, UK.
| |
Collapse
|
27
|
Ramezani M, Wagenknecht-Wiesner A, Wang T, Holowka DA, Eliezer D, Baird BA. Alpha synuclein modulates mitochondrial Ca 2+ uptake from ER during cell stimulation and under stress conditions. NPJ Parkinsons Dis 2023; 9:137. [PMID: 37741841 PMCID: PMC10518018 DOI: 10.1038/s41531-023-00578-x] [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: 04/18/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023] Open
Abstract
Alpha synuclein (a-syn) is an intrinsically disordered protein prevalent in neurons, and aggregated forms are associated with synucleinopathies including Parkinson's disease (PD). Despite the biomedical importance and extensive studies, the physiological role of a-syn and its participation in etiology of PD remain uncertain. We showed previously in model RBL cells that a-syn colocalizes with mitochondrial membranes, depending on formation of N-terminal helices and increasing with mitochondrial stress1. We have now characterized this colocalization and functional correlates in RBL, HEK293, and N2a cells. We find that expression of a-syn enhances stimulated mitochondrial uptake of Ca2+ from the ER, depending on formation of its N-terminal helices but not on its disordered C-terminal tail. Our results are consistent with a-syn acting as a tether between mitochondria and ER, and we show increased contacts between these two organelles using structured illumination microscopy. We tested mitochondrial stress caused by toxins related to PD, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP/MPP+) and carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and found that a-syn prevents recovery of stimulated mitochondrial Ca2+ uptake. The C-terminal tail, and not N-terminal helices, is involved in this inhibitory activity, which is abrogated when phosphorylation site serine-129 is mutated (S129A). Correspondingly, we find that MPTP/MPP+ and CCCP stress is accompanied by both phosphorylation (pS129) and aggregation of a-syn. Overall, our results indicate that a-syn can participate as a tethering protein to modulate Ca2+ flux between ER and mitochondria, with potential physiological significance. A-syn can also prevent cellular recovery from toxin-induced mitochondrial dysfunction, which may represent a pathological role of a-syn in the etiology of PD.
Collapse
Affiliation(s)
- Meraj Ramezani
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | | | - Tong Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - David A Holowka
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - David Eliezer
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Barbara A Baird
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
28
|
Hou X, Chen TH, Koga S, Bredenberg JM, Faroqi AH, Delenclos M, Bu G, Wszolek ZK, Carr JA, Ross OA, McLean PJ, Murray ME, Dickson DW, Fiesel FC, Springer W. Alpha-synuclein-associated changes in PINK1-PRKN-mediated mitophagy are disease context dependent. Brain Pathol 2023; 33:e13175. [PMID: 37259617 PMCID: PMC10467041 DOI: 10.1111/bpa.13175] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Alpha-synuclein (αsyn) aggregates are pathological features of several neurodegenerative conditions including Parkinson disease (PD), dementia with Lewy bodies, and multiple system atrophy (MSA). Accumulating evidence suggests that mitochondrial dysfunction and impairments of the autophagic-lysosomal system can contribute to the deposition of αsyn, which in turn may interfere with health and function of these organelles in a potentially vicious cycle. Here we investigated a potential convergence of αsyn with the PINK1-PRKN-mediated mitochondrial autophagy pathway in cell models, αsyn transgenic mice, and human autopsy brain. PINK1 and PRKN identify and selectively label damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) to mark them for degradation (mitophagy). We found that disease-causing multiplications of αsyn resulted in accumulation of the ubiquitin ligase PRKN in cells. This effect could be normalized by starvation-induced autophagy activation and by CRISPR/Cas9-mediated αsyn knockout. Upon acute mitochondrial damage, the increased levels of PRKN protein contributed to an enhanced pS65-Ub response. We further confirmed increased pS65-Ub-immunopositive signals in mouse brain with αsyn overexpression and in postmortem human disease brain. Of note, increased pS65-Ub was associated with neuronal Lewy body-type αsyn pathology, but not glial cytoplasmic inclusions of αsyn as seen in MSA. While our results add another layer of complexity to the crosstalk between αsyn and the PINK1-PRKN pathway, distinct mechanisms may underlie in cells and brain tissue despite similar outcomes. Notwithstanding, our finding suggests that pS65-Ub may be useful as a biomarker to discriminate different synucleinopathies and may serve as a potential therapeutic target for Lewy body disease.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Ayman H. Faroqi
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Jonathan A. Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Pamela J. McLean
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Melissa E. Murray
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
29
|
Chen X, Tsika E, Levine N, Moore DJ. VPS35 and α-Synuclein fail to interact to modulate neurodegeneration in rodent models of Parkinson's disease. Mol Neurodegener 2023; 18:51. [PMID: 37542299 PMCID: PMC10403858 DOI: 10.1186/s13024-023-00641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/11/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Mutations in the vacuolar protein sorting 35 ortholog (VPS35) gene cause late-onset, autosomal dominant Parkinson's disease (PD), with a single missense mutation (Asp620Asn, D620N) known to segregate with disease in families with PD. The VPS35 gene encodes a core component of the retromer complex, involved in the endosomal sorting and recycling of transmembrane cargo proteins. VPS35-linked PD is clinically indistinguishable from sporadic PD, although it is not yet known whether VPS35-PD brains exhibit α-synuclein-positive brainstem Lewy pathology that is characteristic of sporadic cases. Prior studies have suggested a functional interaction between VPS35 and the PD-linked gene product α-synuclein in lower organisms, where VPS35 deletion enhances α-synuclein-induced toxicity. In mice, VPS35 overexpression is reported to rescue hippocampal neuronal loss in human α-synuclein transgenic mice, potentially suggesting a retromer deficiency in these mice. METHODS Here, we employ multiple well-established genetic rodent models to explore a functional or pathological interaction between VPS35 and α-synuclein in vivo. RESULTS We find that endogenous α-synuclein is dispensable for nigrostriatal pathway dopaminergic neurodegeneration induced by the viral-mediated delivery of human D620N VPS35 in mice, suggesting that α-synuclein does not operate downstream of VPS35. We next evaluated retromer levels in affected brain regions from human A53T-α-synuclein transgenic mice, but find normal levels of the core subunits VPS35, VPS26 or VPS29. We further find that heterozygous VPS35 deletion fails to alter the lethal neurodegenerative phenotype of these A53T-α-synuclein transgenic mice, suggesting the absence of retromer deficiency in this PD model. Finally, we explored the neuroprotective capacity of increasing VPS35 expression in a viral-based human wild-type α-synuclein rat model of PD. However, we find that the overexpression of wild-type VPS35 is not sufficient for protection against α-synuclein-induced nigral dopaminergic neurodegeneration, α-synuclein pathology and reactive gliosis. CONCLUSION Collectively, our data suggest a limited interaction of VPS35 and α-synuclein in neurodegenerative models of PD, and do not provide support for their interaction within a common pathophysiological pathway.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Elpida Tsika
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Vaud, 1015, Switzerland
- AC Immune SA, EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Nathan Levine
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
30
|
Dovonou A, Bolduc C, Soto Linan V, Gora C, Peralta Iii MR, Lévesque M. Animal models of Parkinson's disease: bridging the gap between disease hallmarks and research questions. Transl Neurodegener 2023; 12:36. [PMID: 37468944 PMCID: PMC10354932 DOI: 10.1186/s40035-023-00368-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still provide valuable information that contributes to our understanding of the disease and the limitations of our treatment options. This review comprehensively summarizes the different animal models available for Parkinson's research, with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to realize that the strengths and weaknesses of each model and the induction technique at our disposal are determined by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete discernment of classical and novel animal models in PD research.
Collapse
Affiliation(s)
- Axelle Dovonou
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Cyril Bolduc
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Victoria Soto Linan
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Charles Gora
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Modesto R Peralta Iii
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
31
|
Maor G, Dubreuil RR, Feany MB. α-synuclein promotes neuronal dysfunction and death by disrupting the binding of ankyrin to ß-spectrin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543481. [PMID: 37333277 PMCID: PMC10274672 DOI: 10.1101/2023.06.02.543481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
α-synuclein plays a key role in the pathogenesis of Parkinson's disease and related disorders, but critical interacting partners and molecular mechanisms mediating neurotoxicity are incompletely understood. We show that α-synuclein binds directly to ß-spectrin. Using males and females in a Drosophila model of α-synuclein-related disorders we demonstrate that ß-spectrin is critical for α-synuclein neurotoxicity. Further, the ankyrin binding domain of ß-spectrin is required for α-synuclein binding and neurotoxicity. A key plasma membrane target of ankyrin, Na+/K+ ATPase, is mislocalized when human α-synuclein is expressed in Drosophila. Accordingly, membrane potential is depolarized in α-synuclein transgenic fly brains. We examine the same pathway in human neurons and find that Parkinson's disease patient-derived neurons with a triplication of the α-synuclein locus show disruption of the spectrin cytoskeleton, mislocalization of ankyrin and Na+/K+ ATPase, and membrane potential depolarization. Our findings define a specific molecular mechanism by which elevated levels of α-synuclein in Parkinson's disease and related α-synucleinopathies leads to neuronal dysfunction and death.
Collapse
Affiliation(s)
- Gali Maor
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ronald R. Dubreuil
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
32
|
Khan E, Hasan I, Haque ME. Parkinson's Disease: Exploring Different Animal Model Systems. Int J Mol Sci 2023; 24:ijms24109088. [PMID: 37240432 DOI: 10.3390/ijms24109088] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Disease modeling in non-human subjects is an essential part of any clinical research. To gain proper understanding of the etiology and pathophysiology of any disease, experimental models are required to replicate the disease process. Due to the huge diversity in pathophysiology and prognosis in different diseases, animal modeling is customized and specific accordingly. As in other neurodegenerative diseases, Parkinson's disease is a progressive disorder coupled with varying forms of physical and mental disabilities. The pathological hallmarks of Parkinson's disease are associated with the accumulation of misfolded protein called α-synuclein as Lewy body, and degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc) area affecting the patient's motor activity. Extensive research has already been conducted regarding animal modeling of Parkinson's diseases. These include animal systems with induction of Parkinson's, either pharmacologically or via genetic manipulation. In this review, we will be summarizing and discussing some of the commonly employed Parkinson's disease animal model systems and their applications and limitations.
Collapse
Affiliation(s)
- Engila Khan
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ikramul Hasan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - M Emdadul Haque
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
33
|
Ramezani M, Wagenknecht-Wiesner A, Wang T, Holowka DA, Eliezer D, Baird BA. Alpha Synuclein Modulates Mitochondrial Ca 2+ Uptake from ER During Cell Stimulation and Under Stress Conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.23.537965. [PMID: 37163091 PMCID: PMC10168219 DOI: 10.1101/2023.04.23.537965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Alpha synuclein (a-syn) is an intrinsically disordered protein prevalent in neurons, and aggregated forms are associated with synucleinopathies including Parkinson' disease (PD). Despite the biomedical importance and extensive studies, the physiological role of a-syn and its participation in etiology of PD remain uncertain. We showed previously in model RBL cells that a-syn colocalizes with mitochondrial membranes, depending on formation of N-terminal helices and increasing with mitochondrial stress. 1 We have now characterized this colocalization and functional correlates in RBL, HEK293, and N2a cells. We find that expression of a-syn enhances stimulated mitochondrial uptake of Ca 2+ from the ER, depending on formation of its N-terminal helices but not on its disordered C-terminal tail. Our results are consistent with a-syn acting as a tether between mitochondria and ER, and we show increased contacts between these two organelles using structured illumination microscopy. We tested mitochondrial stress caused by toxins related to PD, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP/MPP+) and carbonyl cyanide m-chlorophenyl hydrazone (CCCP), and found that a-syn prevents recovery of stimulated mitochondrial Ca 2+ uptake. The C-terminal tail, and not N-terminal helices, is involved in this inhibitory activity, which is abrogated when phosphorylation site serine-129 is mutated (S129A). Correspondingly, we find that MPTP/MPP+ and CCCP stress is accompanied by both phosphorylation (pS129) and aggregation of a-syn. Overall, our results indicate that a-syn can participate as a tethering protein to modulate Ca 2+ flux between ER and mitochondria, with potential physiological significance. A-syn can also prevent cellular recovery from toxin-induced mitochondrial dysfunction, which may represent a pathological role of a-syn in the etiology of PD.
Collapse
Affiliation(s)
- Meraj Ramezani
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | | | - Tong Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | - David A. Holowka
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | - David Eliezer
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065
| | - Barbara A. Baird
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| |
Collapse
|
34
|
Tassone A, Meringolo M, Ponterio G, Bonsi P, Schirinzi T, Martella G. Mitochondrial Bioenergy in Neurodegenerative Disease: Huntington and Parkinson. Int J Mol Sci 2023; 24:ijms24087221. [PMID: 37108382 PMCID: PMC10138549 DOI: 10.3390/ijms24087221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Strong evidence suggests a correlation between degeneration and mitochondrial deficiency. Typical cases of degeneration can be observed in physiological phenomena (i.e., ageing) as well as in neurological neurodegenerative diseases and cancer. All these pathologies have the dyshomeostasis of mitochondrial bioenergy as a common denominator. Neurodegenerative diseases show bioenergetic imbalances in their pathogenesis or progression. Huntington's chorea and Parkinson's disease are both neurodegenerative diseases, but while Huntington's disease is genetic and progressive with early manifestation and severe penetrance, Parkinson's disease is a pathology with multifactorial aspects. Indeed, there are different types of Parkinson/Parkinsonism. Many forms are early-onset diseases linked to gene mutations, while others could be idiopathic, appear in young adults, or be post-injury senescence conditions. Although Huntington's is defined as a hyperkinetic disorder, Parkinson's is a hypokinetic disorder. However, they both share a lot of similarities, such as neuronal excitability, the loss of striatal function, psychiatric comorbidity, etc. In this review, we will describe the start and development of both diseases in relation to mitochondrial dysfunction. These dysfunctions act on energy metabolism and reduce the vitality of neurons in many different brain areas.
Collapse
Affiliation(s)
- Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
35
|
Banarase TA, Sammeta SS, Wankhede NL, Mangrulkar SV, Rahangdale SR, Aglawe MM, Taksande BG, Upaganlawar AB, Umekar MJ, Kale MB. Mitophagy regulation in aging and neurodegenerative disease. Biophys Rev 2023; 15:239-255. [PMID: 37124925 PMCID: PMC10133433 DOI: 10.1007/s12551-023-01057-6] [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: 09/10/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are the primary cellular energy generators, supplying the majority of adenosine triphosphate through oxidative phosphorylation, which is necessary for neuron function and survival. Mitophagy is the metabolic process of eliminating dysfunctional or redundant mitochondria. It is a type of autophagy and it is crucial for maintaining mitochondrial and neuronal health. Impaired mitophagy leads to an accumulation of damaged mitochondria and proteins leading to the dysregulation of mitochondrial quality control processes. Recent research shows the vital role of mitophagy in neurons and the pathogenesis of major neurodegenerative diseases. Mitophagy also plays a major role in the process of aging. This review describes the alterations that are being caused in the mitophagy process at the molecular level in aging and in neurodegenerative diseases, particularly Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis, also looks at how mitophagy can be exploited as a therapeutic target for these diseases.
Collapse
Affiliation(s)
- Trupti A. Banarase
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shivkumar S. Sammeta
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Nitu L. Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shubhada V. Mangrulkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Sandip R. Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Manish M. Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Brijesh G. Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Aman B. Upaganlawar
- SNJB’s Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra India 423101
| | - Milind J. Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Mayur B. Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| |
Collapse
|
36
|
Moradi Vastegani S, Nasrolahi A, Ghaderi S, Belali R, Rashno M, Farzaneh M, Khoshnam SE. Mitochondrial Dysfunction and Parkinson's Disease: Pathogenesis and Therapeutic Strategies. Neurochem Res 2023:10.1007/s11064-023-03904-0. [PMID: 36943668 DOI: 10.1007/s11064-023-03904-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023]
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder whose pathogenesis is not completely understood. Mitochondrial dysfunction and increased oxidative stress have been considered as major causes and central events responsible for the progressive degeneration of dopaminergic (DA) neurons in PD. Therefore, investigating mitochondrial disorders plays a role in understanding the pathogenesis of PD and can be an important therapeutic target for this disease. This study discusses the effect of environmental, genetic and biological factors on mitochondrial dysfunction and also focuses on the mitochondrial molecular mechanisms underlying neurodegeneration, and its possible therapeutic targets in PD, including reactive oxygen species generation, calcium overload, inflammasome activation, apoptosis, mitophagy, mitochondrial biogenesis, and mitochondrial dynamics. Other potential therapeutic strategies such as mitochondrial transfer/transplantation, targeting microRNAs, using stem cells, photobiomodulation, diet, and exercise were also discussed in this review, which may provide valuable insights into clinical aspects. A better understanding of the roles of mitochondria in the pathophysiology of PD may provide a rationale for designing novel therapeutic interventions in our fight against PD.
Collapse
Affiliation(s)
- Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Imam Khomeini Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rafie Belali
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran
- Student Research Committee, Asadabad School of Medical Sciences, Asadabad, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
37
|
Chen Y, Lu H, Ding Y, Liu S, Ding Y, Lu B, Xiao J, Zhou X. Dietary Protective Potential of Fucoxanthin as an Active Food Component on Neurological Disorders. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3599-3619. [PMID: 36802555 DOI: 10.1021/acs.jafc.2c08249] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The prevalence of neurodegenerative, cerebrovascular, and psychiatric diseases and other neurological disorders has increased dramatically worldwide. Fucoxanthin is an algal pigment with many biological functions, and there is rising evidence that fucoxanthin plays a preventive and therapeutic role in neurological disorders. This review focuses on the metabolism, bioavailability, and blood-brain barrier penetration of fucoxanthin. Furthermore, the neuroprotective potential of fucoxanthin in neurodegenerative diseases, cerebrovascular diseases, and psychiatric diseases as well as other neurological disorders such as epilepsy, neuropathic pain, and brain tumors by acting on multiple targets will be summarized. The multiple targets include regulating apoptosis, reducing oxidative stress, activating the autophagy pathway, inhibiting Aβ aggregation, improving dopamine secretion, reducing α-synuclein aggregation, attenuating neuroinflammation, modulating gut microbiota, and activating brain-derived neurotrophic factor, etc. Additionally, we look forward to brain-targeted oral transport systems due to the low bioavailability and blood-brain barrier permeability of fucoxanthin. We also propose exploring the systemic mechanisms of fucoxanthin metabolism and transport through the gut-brain process and envision new therapeutic targets for fucoxanthin to act on the central nervous system. Finally, we propose dietary fucoxanthin delivery interventions to achieve preventive effects on neurological disorders. This review provides a reference for the application of fucoxanthin in the neural field.
Collapse
Affiliation(s)
- Yufeng Chen
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Hao Lu
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yicheng Ding
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Shulai Liu
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yuting Ding
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo-Ourense Campus, E-32004 Ourense, Spain
| | - Xuxia Zhou
- College of Food Science and Technology, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, National R&D Branch Center for Pelagic Aquatic Products Processing, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
38
|
Maor G, Dubreuil RR, Feany MB. α-Synuclein Promotes Neuronal Dysfunction and Death by Disrupting the Binding of Ankyrin to β-Spectrin. J Neurosci 2023; 43:1614-1626. [PMID: 36653193 PMCID: PMC10008058 DOI: 10.1523/jneurosci.1922-22.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 01/20/2023] Open
Abstract
α-Synuclein plays a key role in the pathogenesis of Parkinson's disease and related disorders, but critical interacting partners and molecular mechanisms mediating neurotoxicity are incompletely understood. We show that α-synuclein binds directly to β-spectrin. Using males and females in a Drosophila model of α-synuclein-related disorders, we demonstrate that β-spectrin is critical for α-synuclein neurotoxicity. Further, the ankyrin binding domain of β-spectrin is required for α-synuclein binding and neurotoxicity. A key plasma membrane target of ankyrin, Na+/K+ ATPase, is mislocalized when human α-synuclein is expressed in Drosophila Accordingly, membrane potential is depolarized in α-synuclein transgenic fly brains. We examine the same pathway in human neurons and find that Parkinson's disease patient-derived neurons with a triplication of the α-synuclein locus show disruption of the spectrin cytoskeleton, mislocalization of ankyrin and Na+/K+ ATPase, and membrane potential depolarization. Our findings define a specific molecular mechanism by which elevated levels of α-synuclein in Parkinson's disease and related α-synucleinopathies lead to neuronal dysfunction and death.SIGNIFICANCE STATEMENT The small synaptic vesicle associate protein α-synuclein plays a critical role in the pathogenesis of Parkinson's disease and related disorders, but the disease-relevant binding partners of α-synuclein and proximate pathways critical for neurotoxicity require further definition. We show that α-synuclein binds directly to β-spectrin, a key cytoskeletal protein required for localization of plasma membrane proteins and maintenance of neuronal viability. Binding of α-synuclein to β-spectrin alters the organization of the spectrin-ankyrin complex, which is critical for localization and function of integral membrane proteins, including Na+/K+ ATPase. These finding outline a previously undescribed mechanism of α-synuclein neurotoxicity and thus suggest potential new therapeutic approaches in Parkinson's disease and related disorders.
Collapse
Affiliation(s)
- Gali Maor
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ronald R Dubreuil
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
39
|
Yang Q, Wang Y, Zhao C, Pang S, Lu J, Chan P. α-Synuclein aggregation causes muscle atrophy through neuromuscular junction degeneration. J Cachexia Sarcopenia Muscle 2023; 14:226-242. [PMID: 36416282 PMCID: PMC9891985 DOI: 10.1002/jcsm.13123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 09/15/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Sarcopenia is common in patients with Parkinson's disease (PD), showing mitochondrial oxidative stress in skeletal muscle. The aggregation of α-synuclein (α-Syn) to induce oxidative stress is a key pathogenic process of PD; nevertheless, we know little about its potential role in regulating peripheral nerves and the function of the muscles they innervate. METHODS To investigate the role of α-Syn aggregation on neuromuscular system, we used the Thy1 promoter to overexpress human α-Syn transgenic mice (mThy1-hSNCA). hα-Syn expression was evaluated by western blot, and its localization was determined by confocal microscopy. The impact of α-Syn aggregation on the structure and function of skeletal muscle mitochondria and neuromuscular junctions (NMJs), as well as muscle mass and function were characterized by flow cytometry, transmission electron microscopy, Seahorse XF24 metabolic assay, and AAV9 in vivo injection. We assessed the regenerative effect of mitochondrial-targeted superoxide dismutase (Mito-TEMPO) after skeletal muscle injury in mThy1-hSNCA mice. RESULTS Overexpressed hα-Syn protein localized in motor neuron axons and NMJs in muscle and formed aggregates. α-Syn aggregation increased the number of abnormal mitochondrial in the intramuscular axons and NMJs by over 60% (P < 0.01), which inhibited the release of acetylcholine (ACh) from presynaptic vesicles in NMJs (P < 0.05). The expression of genes associated with NMJ activity, neurotransmission and regulation of reactive oxygen species (ROS) metabolic process were significantly decreased in mThy1-hSNCA mice, resulting in ROS production elevated by ~220% (P < 0.05), thereby exacerbating oxidative stress. Such process altered mitochondrial spatial relationships to sarcomeric structures, decreased Z-line spacing by 36% (P < 0.05) and increased myofibre apoptosis by ~10% (P < 0.05). Overexpression of α-Syn altered the metabolic profile of muscle satellite cells (MuSCs), including basal respiratory capacity (~170% reduction) and glycolytic capacity (~150% reduction) (P < 0.05) and decreased cell migration and fusion during muscle regeneration (~60% and ~40%, respectively) (P < 0.05). We demonstrated that Mito-TEMPO treatment could restore the oxidative stress status (the complex I/V protein and enzyme activities increased ~200% and ~150%, respectively), which caused by α-Syn aggregation, and improve the ability of muscle regeneration after injury. In addition, the NMJ receptor fragmentation and ACh secretion were also improved. CONCLUSIONS These results reveal that the α-synuclein aggregation plays an important role in regulating acetylcholine release from neuromuscular junctions and induces intramuscular mitochondrial oxidative stress, which can provide new insights into the aetiology of muscle atrophy in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Qiumei Yang
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yanyan Wang
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chunsong Zhao
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shimin Pang
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Lu
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Piu Chan
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.,Clinical Center for Parkinson's Disease, Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.,Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Gao V, Briano JA, Komer LE, Burré J. Functional and Pathological Effects of α-Synuclein on Synaptic SNARE Complexes. J Mol Biol 2023; 435:167714. [PMID: 35787839 PMCID: PMC10472340 DOI: 10.1016/j.jmb.2022.167714] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
α-Synuclein is an abundant protein at the neuronal synapse that has been implicated in Parkinson's disease for over 25 years and characterizes the hallmark pathology of a group of neurodegenerative diseases now known as the synucleinopathies. Physiologically, α-synuclein exists in an equilibrium between a synaptic vesicle membrane-bound α-helical multimer and a cytosolic largely unstructured monomer. Through its membrane-bound state, α-synuclein functions in neurotransmitter release by modulating several steps in the synaptic vesicle cycle, including synaptic vesicle clustering and docking, SNARE complex assembly, and homeostasis of synaptic vesicle pools. These functions have been ascribed to α-synuclein's interactions with the synaptic vesicle SNARE protein VAMP2/synaptobrevin-2, the synaptic vesicle-attached synapsins, and the synaptic vesicle membrane itself. How α-synuclein affects these processes, and whether disease is due to loss-of-function or gain-of-toxic-function of α-synuclein remains unclear. In this review, we provide an in-depth summary of the existing literature, discuss possible reasons for the discrepancies in the field, and propose a working model that reconciles the findings in the literature.
Collapse
Affiliation(s)
- Virginia Gao
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Neurology, New York Presbyterian/Weill Cornell Medicine, New York, NY, USA.
| | - Juan A Briano
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lauren E Komer
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA. https://www.twitter.com/lauren_komer
| | - Jacqueline Burré
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
41
|
Wang H, Chen G, Ahn EH, Xia Y, Kang SS, Liu X, Liu C, Han MH, Chen S, Ye K. C/EBPβ/AEP is age-dependently activated in Parkinson's disease and mediates α-synuclein in the gut and brain. NPJ Parkinsons Dis 2023; 9:1. [PMID: 36609384 PMCID: PMC9822984 DOI: 10.1038/s41531-022-00430-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/11/2022] [Indexed: 01/09/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder, and its pathologic hallmarks include extensive dopaminergic neuronal degeneration in the Substantia nigra associated with Lewy bodies, predominantly consisting of phosphorylated and truncated α-Synuclein (α-Syn). Asparagine endopeptidase (AEP) cleaves human α-Syn at N103 residue and promotes its aggregation, contributing to PD pathogenesis. However, how AEP mediates Lewy body pathologies during aging and elicits PD onset remains incompletely understood. Knockout of AEP or C/EBPβ from α-SNCA mice, and their chronic rotenone exposure models were used, and the mechanism of α-Syn from the gut that spread to the brain was observed. Here we report that C/EBPβ/AEP pathway, aggravated by oxidative stress, is age-dependently activated and cleaves α-Syn N103 and regulates Lewy body-like pathologies spreading from the gut into the brain in human α-SNCA transgenic mice. Deletion of C/EBPβ or AEP substantially diminished the oxidative stress, neuro-inflammation, and PD pathologies, attenuating motor dysfunctions in aged α-SNCA mice. Noticeably, PD pathologies initiate in the gut and progressively spread into the brain. Chronic gastric exposure to a low dose of rotenone initiates Lewy body-like pathologies in the gut that propagate into the brain in a C/EBPβ/AEP-dependent manner. Hence, our studies demonstrate that C/EBPβ/AEP pathway is critical for mediating Lewy body pathology progression in PD.
Collapse
Affiliation(s)
- Hualong Wang
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China ,grid.452458.aDepartment of Neurology, The First Hospital of Hebei Medical University (Department of Neurology, Hebei Hospital of Xuanwu Hospital Capital Medical University), Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Neuromedical Technology Innovation Center of Hebei Province, Shijiazhuang, 050031 Hebei P. R. China ,grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Guiqin Chen
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.412632.00000 0004 1758 2270Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060 China
| | - Eun Hee Ahn
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.256753.00000 0004 0470 5964Department of Physiology, College of Medicine, Hallym University, Hallymdaehak-gil, Chuncheon-si, Gangwon-Do, 24252, South Korea
| | - Yiyuan Xia
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Seong Su Kang
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Xia Liu
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Chang Liu
- grid.458489.c0000 0001 0483 7922CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China ,grid.458489.c0000 0001 0483 7922Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
| | - Ming-Hu Han
- grid.458489.c0000 0001 0483 7922Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China
| | - Shengdi Chen
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Keqiang Ye
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.458489.c0000 0001 0483 7922Department of Biology, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China
| |
Collapse
|
42
|
Chopade P, Chopade N, Zhao Z, Mitragotri S, Liao R, Chandran Suja V. Alzheimer's and Parkinson's disease therapies in the clinic. Bioeng Transl Med 2023; 8:e10367. [PMID: 36684083 PMCID: PMC9842041 DOI: 10.1002/btm2.10367] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most prevalent neurodegenerative diseases, affecting millions and costing billions each year in the United States alone. Despite tremendous progress in developing therapeutics that manage the symptoms of these two diseases, the scientific community has yet to develop a treatment that effectively slows down, inhibits, or cures neurodegeneration. To gain a better understanding of the current therapeutic frontier for the treatment of AD and PD, we provide a review on past and present therapeutic strategies for these two major neurodegenerative disorders in the clinical trial process. We briefly recap currently US Food and Drug Administration-approved therapies, and then explore trends in clinical trials across the variables of therapy mechanism of disease intervention, administration route, use of delivery vehicle, and outcome measures, across the clinical phases over time for "Drug" and "Biologic" therapeutics. We then present the success rate of past clinical trials and analyze the intersections in therapeutic approaches for AD and PD, revealing the shift in clinical trials away from therapies targeting neurotransmitter systems that provide symptomatic relief, and towards anti-aggregation, anti-inflammatory, anti-oxidant, and regeneration strategies that aim to inhibit the root causes of disease progression. We also highlight the evolving distribution of the types of "Biologic" therapies investigated, and the slowly increasing yet still severe under-utilization of delivery vehicles for AD and PD therapeutics. We then briefly discuss novel preclinical strategies for treating AD and PD. Overall, this review aims to provide a succinct overview of the clinical landscape of AD and PD therapies to better understand the field's therapeutic strategy in the past and the field's evolution in approach to the present, to better inform how to effectively treat AD and PD in the future.
Collapse
Affiliation(s)
| | | | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of PharmacyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| | - Rick Liao
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| | - Vineeth Chandran Suja
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| |
Collapse
|
43
|
Kalra J. Crosslink between mutations in mitochondrial genes and brain disorders: implications for mitochondrial-targeted therapeutic interventions. Neural Regen Res 2023. [PMID: 35799515 PMCID: PMC9241418 DOI: 10.4103/1673-5374.343884] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
|
44
|
Coleman C, Martin I. Unraveling Parkinson's Disease Neurodegeneration: Does Aging Hold the Clues? JOURNAL OF PARKINSON'S DISEASE 2022; 12:2321-2338. [PMID: 36278358 PMCID: PMC9837701 DOI: 10.3233/jpd-223363] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Aging is the greatest risk factor for Parkinson's disease (PD), suggesting that mechanisms driving the aging process promote PD neurodegeneration. Several lines of evidence support a role for aging in PD. First, hallmarks of brain aging such as mitochondrial dysfunction and oxidative stress, loss of protein homeostasis, and neuroinflammation are centrally implicated in PD development. Second, mutations that cause monogenic PD are present from conception, yet typically only cause disease following a period of aging. Third, lifespan-extending genetic, dietary, or pharmacological interventions frequently attenuate PD-related neurodegeneration. These observations support a central role for aging in disease development and suggest that new discoveries in the biology of aging could be leveraged to elucidate novel mechanisms of PD pathophysiology. A recent rapid growth in our understanding of conserved molecular pathways that govern model organism lifespan and healthspan has highlighted a key role for metabolism and nutrient sensing pathways. Uncovering how metabolic pathways involving NAD+ consumption, insulin, and mTOR signaling link to the development of PD is underway and implicates metabolism in disease etiology. Here, we assess areas of convergence between nervous system aging and PD, evaluate the link between metabolism, aging, and PD and address the potential of metabolic interventions to slow or halt the onset of PD-related neurodegeneration drawing on evidence from cellular and animal models.
Collapse
Affiliation(s)
- Colin Coleman
- Department of Neurology, Jungers Center for Neurosciences, Oregon Health and Science University, Portland, OR, USA
| | - Ian Martin
- Department of Neurology, Jungers Center for Neurosciences, Oregon Health and Science University, Portland, OR, USA,Correspondence to: Ian Martin, Jungers Center for Neurosciences Research, Department of Neurology - Mail Code L623, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA. Tel.: +1 503 494 9140; E-mail:
| |
Collapse
|
45
|
Thomas Broome S, Castorina A. Systemic Rotenone Administration Causes Extra-Nigral Alterations in C57BL/6 Mice. Biomedicines 2022; 10:biomedicines10123174. [PMID: 36551930 PMCID: PMC9775048 DOI: 10.3390/biomedicines10123174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Systemic administration of rotenone replicates several pathogenic and behavioural features of Parkinson's disease (PD), some of which cannot be explained by deficits of the nigrostriatal pathway. In this study, we provide a comprehensive analysis of several neurochemical alterations triggered by systemic rotenone administration in the CNS of C57BL/6 mice. Mice injected with either 1, 3 or 10 mg/kg rotenone daily via intraperitoneal route for 21 days were assessed weekly for changes in locomotor and exploratory behaviour. Rotenone treatment caused significant locomotor and exploratory impairment at dosages of 3 or 10 mg/kg. Molecular analyses showed reductions of both TH and DAT expression in the midbrain, striatum and spinal cord, accompanied by altered expression of dopamine receptors and brain-derived neurotrophic factor (BDNF). Rotenone also triggered midbrain-restricted inflammatory responses with heightened expression of glial markers, which was not seen in extra-nigral regions. However, widespread alterations of mitochondrial function and increased signatures of oxidative stress were identified in both nigral and extra-nigral regions, along with disruptions of neuroprotective peptides, such as pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP) and activity-dependent neuroprotective protein (ADNP). Altogether, this study shows that systemic rotenone intoxication, similarly to PD, causes a series of neurochemical alterations that extend at multiple CNS levels, reinforcing the suitability of this pre-clinical model for the study extra-nigral defects of PD.
Collapse
|
46
|
Nadeem H, Zhou B, Goldman D, Romley J. Association between use of ß2-adrenergic receptor agonists and incidence of Parkinson's disease: Retrospective cohort analysis. PLoS One 2022; 17:e0276368. [PMID: 36441791 PMCID: PMC9704661 DOI: 10.1371/journal.pone.0276368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Previous observational studies assessing β2-agonist/-antagonist use on PD risk have yielded conflicting results. We evaluated the relationship between β2-agonist use and the incidence of Parkinson's disease in patients with chronic lung disease. METHODS We performed a retrospective cohort analysis on a 20% random sample abstracted from a traditional (fee-for-service) Medicare program in the United States. Inclusion criteria were individuals over 65 years old diagnosed with asthma, COPD, and/or bronchiectasis who were enrolled in a prescription drug (standalone Part D) plan over 2007-2010 and alive through 2014. The main outcome measure was a diagnosis of Parkinson's disease over the period 2011-2014, in relation to the number of 30-day-equivalent drug claims over 2007-2010. Logistic regression analysis was performed on a sample including 236,201 Medicare beneficiaries. RESULTS The sample was 68% female, 80% white, and on average 77 years old as of 2010. Compared to non-users, β2-agonist users were more likely to be younger (76.3y versus 78.0y), smokers (40.4% versus 31.1%) and asthmatic (62.4% versus 28.3%). The odds ratio for a β2-agonist claim on PD development was 0.986 (95% CI 0.977-0.995) after adjusting for demographics, smoking history, respiratory exacerbations, comorbidities, and other drug use. Risk reductions were larger for males than females (0.974 versus 0.994, P = 0.032), and for individuals with COPD compared to those with asthma (0.968 versus 0.998, P = 0.049). Reverse causality was addressed with a Cox analysis that allowed β2-agonist use to vary from medication initiation to disease onset. By the end of the follow-up period, β2-agonist use was shown to be associated with a true protective effect against PD onset. DISCUSSION β2-agonist use is associated with decreased risk of PD incidence. Further investigation, possibly including clinical trials, is warranted to strengthen the evidence base supporting clinical decision-makers looking to repurpose pharmaceuticals to prevent neurodegenerative disease onset.
Collapse
Affiliation(s)
- Hasan Nadeem
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Bo Zhou
- Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, California, United States of America
- School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
| | - Dana Goldman
- Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, California, United States of America
- School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
- Price School of Public Policy, University of Southern California, Los Angeles, California, United States of America
| | - John Romley
- Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, California, United States of America
- School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
- Price School of Public Policy, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Sriram S, Root K, Chacko K, Patel A, Lucke-Wold B. Surgical Management of Synucleinopathies. Biomedicines 2022; 10:2657. [PMID: 36289920 PMCID: PMC9599076 DOI: 10.3390/biomedicines10102657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 12/03/2022] Open
Abstract
Synucleinopathies represent a diverse set of pathologies with significant morbidity and mortality. In this review, we highlight the surgical management of three synucleinopathies: Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). After examining underlying molecular mechanisms and the medical management of these diseases, we explore the role of deep brain stimulation (DBS) in the treatment of synuclein pathophysiology. Further, we examine the utility of focused ultrasound (FUS) in the treatment of synucleinopathies such as PD, including its role in blood-brain barrier (BBB) opening for the delivery of novel drug therapeutics and gene therapy vectors. We also discuss other recent advances in the surgical management of MSA and DLB. Together, we give a diverse overview of current techniques in the neurosurgical management of these pathologies.
Collapse
Affiliation(s)
| | | | | | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
48
|
Gao F, Zhang H, Yang J, Cai M, Yang Q, Wang H, Xu Y, Chen H, Hu Y, He W, Zhang J. ATPase Thorase Deficiency Causes α-Synucleinopathy and Parkinson’s Disease-like Behavior. Cells 2022; 11:cells11192990. [PMID: 36230952 PMCID: PMC9563720 DOI: 10.3390/cells11192990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 11/29/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases and is pathologically characterized by α-synucleinopathy, which is harmful to dopaminergic neurons. However, the underlying mechanisms and pathogenesis of PD remain unclear. The AAA + ATPase Thorase was identified as being essential for neuroprotection and synaptic plasticity by regulating the AMPA receptor trafficking. Here, we found that conditional knockout of Thorase resulted in motor behaviors indicative of neurodegeneration. Genetic deletion of Thorase exacerbated phenotypes of α-synucleinopathy in a familial PD-like A53T mouse model, whereas overexpression of Thorase prevented α-syn accumulation in vivo. Biochemical and cell cultures studies presented here suggest that Thorase interacts with α-syn and regulates the degradation of ubiquitinated α-syn. Thorase deficiency promotes α-syn aggregation in primary cultured neurons. The discoveries in this study provide us with a further understanding of the pathogenesis of α-synucleinopathies including PD.
Collapse
Affiliation(s)
- Fei Gao
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Han Zhang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Jia Yang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Menghua Cai
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Qi Yang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Huaishan Wang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Yi Xu
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| | - Hui Chen
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yu Hu
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
| | - Wei He
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
- Correspondence: (W.H.); (J.Z.)
| | - Jianmin Zhang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Key Laboratory of T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
- Correspondence: (W.H.); (J.Z.)
| |
Collapse
|
49
|
Khot M, Sood A, Tryphena KP, Khan S, Srivastava S, Singh SB, Khatri DK. NLRP3 inflammasomes: A potential target to improve mitochondrial biogenesis in Parkinson's disease. Eur J Pharmacol 2022; 934:175300. [PMID: 36167151 DOI: 10.1016/j.ejphar.2022.175300] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative condition for which no approved treatment exists to prevent collective neuronal death. There is ample evidence that mitochondrial dysfunction, reactive oxygen species (ROS), and associated caspase activity underlie the pathology observed. Neurons rely on mitochondrial activity since they have such high energy consumption. Therefore, it is not surprising that mitochondrial alterations favour neuronal degeneration. In particular, mitochondrial dysregulation contributes to PD, based on the observation that mitochondrial toxins can cause parkinsonism in humans and animal models. Also, it is known that inflammatory cytokine-mediated neuroinflammation is the key pathogenic mechanism in neuronal loss. In recent years, the research has focussed on mitochondria being the platform for nucleotide-binding oligomerization domain-like receptors 3 (NLRP3) inflammasome activation. Mitochondrial dysfunction and NLRP3 activation are emerging as critical players in inducing and sustaining neuroinflammation. Moreover, mitochondrial-derived ROS and mitochondrial DNA (mtDNA) could serve as the priming signal for forming inflammasome complexes responsible for the activation, maturation, and release of pro-inflammatory cytokines, including interleukin-1(IL-1) and interleukin-18 (IL-18). The current review takes a more comprehensive approach to elucidating the link between mitochondrial dysfunction and aberrant NLRP3 activation in PD. In addition, we focus on some inhibitors of NLRP3 inflammatory pathways to alleviate the progression of PD.
Collapse
Affiliation(s)
- Mayuri Khot
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Anika Sood
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Sabiya Khan
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India.
| |
Collapse
|
50
|
Mysiris DS, Vavougios GD, Karamichali E, Papoutsopoulou S, Stavrou VT, Papayianni E, Boutlas S, Mavridis T, Foka P, Zarogiannis SG, Gourgoulianis K, Xiromerisiou G. Post-COVID-19 Parkinsonism and Parkinson's Disease Pathogenesis: The Exosomal Cargo Hypothesis. Int J Mol Sci 2022; 23:9739. [PMID: 36077138 PMCID: PMC9456372 DOI: 10.3390/ijms23179739] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease after Alzheimer's disease, globally. Dopaminergic neuron degeneration in substantia nigra pars compacta and aggregation of misfolded alpha-synuclein are the PD hallmarks, accompanied by motor and non-motor symptoms. Several viruses have been linked to the appearance of a post-infection parkinsonian phenotype. Coronavirus disease 2019 (COVID-19), caused by emerging severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, has evolved from a novel pneumonia to a multifaceted syndrome with multiple clinical manifestations, among which neurological sequalae appear insidious and potentially long-lasting. Exosomes are extracellular nanovesicles bearing a complex cargo of active biomolecules and playing crucial roles in intercellular communication under pathophysiological conditions. Exosomes constitute a reliable route for misfolded protein transmission, contributing to PD pathogenesis and diagnosis. Herein, we summarize recent evidence suggesting that SARS-CoV-2 infection shares numerous clinical manifestations and inflammatory and molecular pathways with PD. We carry on hypothesizing that these similarities may be reflected in exosomal cargo modulated by the virus in correlation with disease severity. Travelling from the periphery to the brain, SARS-CoV-2-related exosomal cargo contains SARS-CoV-2 RNA, viral proteins, inflammatory mediators, and modified host proteins that could operate as promoters of neurodegenerative and neuroinflammatory cascades, potentially leading to a future parkinsonism and PD development.
Collapse
Affiliation(s)
| | - George D. Vavougios
- Department of Neurology, Faculty of Medicine, University of Cyprus, Lefkosia 1678, Cyprus
- Laboratory of Pulmonary Testing and Rehabilitation, Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece
| | - Eirini Karamichali
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Stamatia Papoutsopoulou
- Department of Biochemistry and Biotechnology, Faculty of Life Sciences, University of Thessaly, Mezourlo, 41500 Larissa, Greece
| | - Vasileios T. Stavrou
- Laboratory of Pulmonary Testing and Rehabilitation, Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece
| | - Eirini Papayianni
- Laboratory of Pulmonary Testing and Rehabilitation, Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece
| | - Stylianos Boutlas
- Laboratory of Pulmonary Testing and Rehabilitation, Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece
| | - Theodoros Mavridis
- 1st Neurology Department, Eginition Hospital, Medical School, National & Kapodistrian University of Athens, 11528 Athens, Greece
| | - Pelagia Foka
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Sotirios G. Zarogiannis
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Konstantinos Gourgoulianis
- Laboratory of Pulmonary Testing and Rehabilitation, Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece
| | - Georgia Xiromerisiou
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| |
Collapse
|