1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
Feng Z, Hou Y, Yu C, Li T, Fu H, Lv F, Li P. Mitophagy in perioperative neurocognitive disorder: mechanisms and therapeutic strategies. Eur J Med Res 2025; 30:270. [PMID: 40211418 PMCID: PMC11987364 DOI: 10.1186/s40001-025-02400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common neurological complication after surgery/anesthesia in elderly patients that affect postoperative outcome and long-term quality of life, which increases the cost of family and social resources. The pathological mechanism of PND is complex and not fully understood, and the methods of prevention and treatment of PND are very limited, so it is particularly important to analyze the mechanism of PND. Research indicates that mitochondrial dysfunction is pivotal in the initiation and progression of PND, although the precise mechanisms remain elusive and could involve disrupted mitophagy. We reviewed recent studies on the link between mitophagy and PND, highlighting the role of key proteins in abnormal mitophagy and discussing therapeutic strategies aimed at mitophagy regulation. This provides insights into the mechanisms underlying PND and potential therapeutic targets.
Collapse
Affiliation(s)
- Zhen Feng
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Yan Hou
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Chang Yu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Ting Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Haoyang Fu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| | - Ping Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
3
|
Mohan AA, Talwar P. MAM kinases: physiological roles, related diseases, and therapeutic perspectives-a systematic review. Cell Mol Biol Lett 2025; 30:35. [PMID: 40148800 PMCID: PMC11951743 DOI: 10.1186/s11658-025-00714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondria-associated membranes (MAMs) are tethering regions amid the membranes of the endoplasmic reticulum (ER) and mitochondria. They are a lipid raft-like structure occupied by various proteins that facilitates signal transduction between the two organelles. The MAM proteome participates in cellular functions such as calcium (Ca2+) homeostasis, lipid synthesis, ER stress, inflammation, autophagy, mitophagy, and apoptosis. The human kinome is a superfamily of homologous proteins consisting of 538 kinases. MAM-associated kinases participate in the aforementioned cellular functions and act as cell fate executors. Studies have proved the dysregulated kinase interactions in MAM as an etiology for various diseases including cancer, diabetes mellitus, neurodegenerative diseases, cardiovascular diseases (CVDs), and obesity. Several small kinase inhibitory molecules have been well explored as promising drug candidates in clinical trials with an accelerating impact in the field of precision medicine. This review narrates the physiological actions, pathophysiology, and therapeutic potential of MAM-associated kinases with recent updates in the field.
Collapse
Affiliation(s)
- A Anjana Mohan
- Apoptosis and Cell Survival Research Laboratory, 412G Pearl Research Park, Department of Biosciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Laboratory, 412G Pearl Research Park, Department of Biosciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
Vaughan DP, Real R, Jensen MT, Fumi RG, Hodgson M, Jabbari E, Lux D, Wu L, Warner TT, Jaunmuktane Z, Revesz T, Rowe JB, Rohrer J, Morris HR. Analysis of C9orf72 repeat length in progressive supranuclear palsy, corticobasal syndrome, corticobasal degeneration, and atypical parkinsonism. J Neurol 2025; 272:293. [PMID: 40138021 PMCID: PMC11947049 DOI: 10.1007/s00415-025-12990-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/09/2025] [Accepted: 02/15/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Pathogenic hexanucleotide repeat expansions in C9orf72 are the commonest genetic cause of frontotemporal dementia and/or amyotrophic lateral sclerosis. There is growing interest in intermediate repeat expansions in C9orf72 and their relationship to a wide range of neurological presentations, including Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, corticobasal degeneration, and corticobasal syndromes. AIMS To assess the prevalence of intermediate C9orf72 repeat expansions in a large cohort of prospectively-recruited patients clinically diagnosed with progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), and atypical parkinsonism (APS), compared with healthy controls. We also sought to replicate the association between C9orf72 repeat length and CBD in neuropathologically confirmed cases. METHODS 626 cases, including PSP (n = 366), CBS (n = 130), and APS (n = 53) from the PROSPECT study, and 77 cases with pathologically confirmed CBD were screened for intermediate repeat expansions in C9orf72 using repeat-primed PCR. These were compared to controls from the PROSPECT-M-UK study and from the 1958 Birth Cohort. RESULTS There was no difference in the mean or largest allele size in any affected patient group compared with controls. A higher proportion of our affected cohort had large C9orf72 repeat expansions compared to controls, but there was no difference when comparing the frequency of intermediate expansions between affected patients and controls. There was no relationship between repeat length and age at onset, level of disability, or survival. CONCLUSIONS Intermediate expansions in C9orf72 do not appear to be a genetic risk factor for PSP, CBS, CBD, or atypical parkinsonism. They are not associated with age at onset, disability, or survival in our study.
Collapse
Affiliation(s)
- David P Vaughan
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Marte Theilmann Jensen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Riona G Fumi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Megan Hodgson
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Danielle Lux
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Lesley Wu
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Zane Jaunmuktane
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK
- Queen Square Brain Bank, Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
| | - Tamas Revesz
- Queen Square Brain Bank, Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Jonathan Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK.
- Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
5
|
Daniel Estrella L, Trease AJ, Sheldon L, Roland NJ, Fox HS, Stauch KL. Tau association with synaptic mitochondria coincides with energetic dysfunction and excitatory synapse loss in the P301S tauopathy mouse model. Neurobiol Aging 2025; 147:163-175. [PMID: 39778459 DOI: 10.1016/j.neurobiolaging.2024.12.006] [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: 09/06/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Neurodegenerative Tauopathies are a part of several neurological disorders and aging-related diseases including, but not limited to, Alzheimer's Disease, Frontotemporal Dementia with Parkinsonism, and Chronic Traumatic Encephalopathy. The major hallmarks present in these conditions include Tau pathology (composed of hyperphosphorylated Tau tangles) and synaptic loss. in vivo studies linking Tau pathology and mitochondrial alterations at the synapse, an avenue that could lead to synaptic loss, remain predominantly scarce. For this reason, using 3-month-old wild-type and human mutant Tau P301S transgenic mice, we investigated the association of Tau with mitochondria, synaptosome bioenergetics, and characterized excitatory synaptic loss across hippocampal regions (Dentate Gyrus, perisomatic CA3, and perisomatic CA1) and in the parietal cortex. We found a significant loss of excitatory synapses in the parietal cortex and hippocampal Dentate Gyrus (DG) of Tau P301S mice. Furthermore, we found that Tau (total and disease-relevant phosphorylated Tau) associates with both the non-synaptic and synaptic mitochondria of Tau P301S mice and this coincided with synaptic mitochondrial dysfunction. The findings presented here suggest that Tau associates with mitochondria at the synapse, leading to synaptic mitochondrial dysfunction, and likely contributing to synaptic loss.
Collapse
Affiliation(s)
- L Daniel Estrella
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Andrew J Trease
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Lexi Sheldon
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Nashanthea J Roland
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Howard S Fox
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Kelly L Stauch
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA.
| |
Collapse
|
6
|
Schmid ET, Schinaman JM, Liu-Abramowicz N, Williams KS, Walker DW. Accumulation of F-actin drives brain aging and limits healthspan in Drosophila. Nat Commun 2024; 15:9238. [PMID: 39455560 PMCID: PMC11512044 DOI: 10.1038/s41467-024-53389-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The actin cytoskeleton is a key determinant of cell structure and homeostasis. However, possible tissue-specific changes to actin dynamics during aging, notably brain aging, are not understood. Here, we show that there is an age-related increase in filamentous actin (F-actin) in Drosophila brains, which is counteracted by prolongevity interventions. Critically, decreasing F-actin levels in aging neurons prevents age-onset cognitive decline and extends organismal healthspan. Mechanistically, we show that autophagy, a recycling process required for neuronal homeostasis, is disabled upon actin dysregulation in the aged brain. Remarkably, disrupting actin polymerization in aged animals with cytoskeletal drugs restores brain autophagy to youthful levels and reverses cellular hallmarks of brain aging. Finally, reducing F-actin levels in aging neurons slows brain aging and promotes healthspan in an autophagy-dependent manner. Our data identify excess actin polymerization as a hallmark of brain aging, which can be targeted to reverse brain aging phenotypes and prolong healthspan.
Collapse
Affiliation(s)
- Edward T Schmid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joseph M Schinaman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Naomi Liu-Abramowicz
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kylie S Williams
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - David W Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
7
|
Quintero-Espinosa DA, Jimenez-Del-Rio M, Velez-Pardo C. LRRK2 Kinase Inhibitor PF-06447475 Protects Drosophila melanogaster against Paraquat-Induced Locomotor Impairment, Life Span Reduction, and Oxidative Stress. Neurochem Res 2024; 49:2440-2452. [PMID: 38847910 PMCID: PMC11310290 DOI: 10.1007/s11064-024-04141-9] [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: 03/07/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 08/09/2024]
Abstract
Parkinson's disease (PD) is a complex multifactorial progressive neurodegenerative disease characterized by locomotor alteration due to the specific deterioration of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc). Mounting evidence shows that human LRRK2 (hLRRK2) kinase activity is involved in oxidative stress (OS)-induced neurodegeneration, suggesting LRRK2 inhibition as a potential therapeutic target. We report that the hLRRK2 inhibitor PF-06447475 (PF-475) prolonged lifespan, increased locomotor activity, maintained DAergic neuronal integrity, and reduced lipid peroxidation (LPO) in female Drosophila melanogaster flies chronically exposed to paraquat (PQ), a redox cycling compound, compared to flies treated with vehicle only. Since LRRK2 is an evolutionary conserved kinase, the present findings reinforce the idea that either reduction or inhibition of the LRRK2 kinase might decrease OS and locomotor alterations associated with PD. Our observations highlight the importance of uncovering the function of the hLRRK2 orthologue dLrrk2 in D. melanogaster as an excellent model for pharmacological screenings.
Collapse
Affiliation(s)
- Diana A Quintero-Espinosa
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| |
Collapse
|
8
|
Afsheen S, Rehman AS, Jamal A, Khan N, Parvez S. Understanding role of pesticides in development of Parkinson's disease: Insights from Drosophila and rodent models. Ageing Res Rev 2024; 98:102340. [PMID: 38759892 DOI: 10.1016/j.arr.2024.102340] [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: 03/10/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Parkinson's disease is a neurodegenerative illness linked to ageing, marked by the gradual decline of dopaminergic neurons in the midbrain. The exact aetiology of Parkinson's disease (PD) remains uncertain, with genetic predisposition and environmental variables playing significant roles in the disease's frequency. Epidemiological data indicates a possible connection between pesticide exposure and brain degeneration. Specific pesticides have been associated with important characteristics of Parkinson's disease, such as mitochondrial dysfunction, oxidative stress, and α-synuclein aggregation, which are crucial for the advancement of the disease. Recently, many animal models have been developed for Parkinson's disease study. Although these models do not perfectly replicate the disease's pathology, they provide valuable insights that improve our understanding of the condition and the limitations of current treatment methods. Drosophila, in particular, has been useful in studying Parkinson's disease induced by toxins or genetic factors. The review thoroughly analyses many animal models utilised in Parkinson's research, with an emphasis on issues including pesticides, genetic and epigenetic changes, proteasome failure, oxidative damage, α-synuclein inoculation, and mitochondrial dysfunction. The text highlights the important impact of pesticides on the onset of Parkinson's disease (PD) and stresses the need for more research on genetic and mechanistic alterations linked to the condition.
Collapse
Affiliation(s)
- Saba Afsheen
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ahmed Shaney Rehman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Nazia Khan
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
9
|
Jing C, Zhong X, Min X, Xu H. The causal effects of intelligence and fluid intelligence on Parkinson's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1388795. [PMID: 38846742 PMCID: PMC11153853 DOI: 10.3389/fnagi.2024.1388795] [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: 02/20/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
Background Parkinson's disease (PD) is a chronic neurodegenerative disease that affects the central nervous system, primarily the motor nervous system, and occurs most often in older adults. A large number of studies have shown that high intelligence leads to an increased risk of PD. However, whether there is a causal relationship between intelligence on PD has not yet been reported. Methods In this study, Mendelian randomization (MR) analysis was performed with intelligence (ebi-a-GCST006250) and fluid intelligence score (ukb-b-5238) as exposure factors and PD (ieu-b-7) as an outcome, which the datasets were mined from the IEU OpenGWAS database. MR analysis was performed through 3 methods [MR Egger, weighted median, inverse variance weighted (IVW)], of which IVW was the primary method. In addition, the reliability of the results of the MR analysis was assessed via the heterogeneity test, the horizontal polytropy test, and Leave-One-Out (LOO). Finally, based on gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, the genes corresponding to intelligence and fluid intelligence score related to SNPs were enriched for functional features and pathways. Results The results of MR analysis suggested that elevated intelligence indicators can increase the risk of PD [p = 0.015, Odd Ratio (OR) = 1.316]. Meanwhile, fluid intelligence score was causally associated with the PD (p = 0.035), which was a risk factor (OR = 1.142). The reliability of the results of MR analysis was demonstrated by sensitivity analysis. Finally, the results of GO enrichment analysis for 87 genes corresponding to intelligence related SNPs mainly included regulation of synapse organization, developmental cell growth, etc. These genes were enriched in the synaptic vessel cycle, polycomb expressive complex in KEGG. Similarly, 44 genes corresponding to SNPs associated with fluid intelligence score were used for enrichment analysis. Based on the GO database, these genes were mainly enriched in regulation of developmental growth, negative regulation of neuron projection development, etc. In KEGG, 44 genes corresponding to SNPs associated with fluid intelligence score were enriched in signaling pathways including Alzheimer's disease, the cellular senescence, etc. Conclusion The causal relationships between intelligence and fluid intelligence scores, and PD were demonstrated through MR analysis, providing an important reference and evidence for the study of PD.
Collapse
Affiliation(s)
- Cong Jing
- Departments of Interventional Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaojiao Zhong
- Yilong County General Hospital (Ma’an Campus), Nanchong, Sichuan, China
| | - XuLi Min
- Departments of Interventional Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hao Xu
- Departments of Interventional Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
10
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. Genome Res 2024; 34:590-605. [PMID: 38599684 PMCID: PMC11146598 DOI: 10.1101/gr.278576.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein TAU (current and approved symbol is MAPT) cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human TAU in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas-mediated gene editing to model frontotemporal dementia caused by the TAU P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for Tau P251L display age-dependent neurodegeneration, display metabolic defects, and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies, we performed single-cell RNA sequencing on approximately 130,000 cells from brains of Tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified. Gene expression was also altered in glial cells, suggestive of non-cell-autonomous regulation. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell type-specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy that faithfully recapitulates the genetic context and phenotypic features of the human disease, and use the results of comprehensive single-cell sequencing analysis to outline pathways of neurotoxicity and highlight the potential role of non-cell-autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- 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, Maryland 20815, USA
| | - Vanitha Nithianandam
- 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, Maryland 20815, USA
| | - Rachel A Battaglia
- 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, Maryland 20815, USA
| | - Anthony Cicalo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Matthew J Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, Massachusetts 02139, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - 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, Maryland 20815, USA
| |
Collapse
|
11
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578624. [PMID: 38352559 PMCID: PMC10862891 DOI: 10.1101/2024.02.02.578624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein tau cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human tau in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease, but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas mediated gene editing to model frontotemporal dementia caused by the tau P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for tau P251L display age-dependent neurodegeneration, metabolic defects and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies we performed single-cell RNA sequencing on approximately 130,000 cells from brains of tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified and non-cell autonomously in glial cells. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell-type specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy, which faithfully recapitulates the genetic context and phenotypic features of the human disease and use the results of comprehensive single cell sequencing analysis to outline pathways of neurotoxicity and highlight the role of non-cell autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Rachel A. Battaglia
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Anthony Cicalo
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Matthew J. Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA 02139
| | - Xianjun Dong
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Genomics and Bioinformatics Hub, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
12
|
Giusto E, Maistrello L, Iannotta L, Giusti V, Iovino L, Bandopadhyay R, Antonini A, Bubacco L, Barresi R, Plotegher N, Greggio E, Civiero L. Prospective Role of PAK6 and 14-3-3γ as Biomarkers for Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:495-506. [PMID: 38640169 PMCID: PMC11091598 DOI: 10.3233/jpd-230402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/21/2024]
Abstract
Background Parkinson's disease is a progressive neurodegenerative disorder mainly distinguished by sporadic etiology, although a genetic component is also well established. Variants in the LRRK2 gene are associated with both familiar and sporadic disease. We have previously shown that PAK6 and 14-3-3γ protein interact with and regulate the activity of LRRK2. Objective The aim of this study is to quantify PAK6 and 14-3-3γ in plasma as reliable biomarkers for the diagnosis of both sporadic and LRRK2-linked Parkinson's disease. Methods After an initial quantification of PAK6 and 14-3-3γ expression by means of Western blot in post-mortem human brains, we verified the presence of the two proteins in plasma by using quantitative ELISA tests. We analyzed samples obtained from 39 healthy subjects, 40 patients with sporadic Parkinson's disease, 50 LRRK2-G2019S non-manifesting carriers and 31 patients with LRRK2-G2019S Parkinson's disease. Results The amount of PAK6 and 14-3-3γ is significantly different in patients with Parkinson's disease compared to healthy subjects. Moreover, the amount of PAK6 also varies with the presence of the G2019S mutation in the LRRK2 gene. Although the generalized linear models show a low association between the presence of Parkinson's disease and PAK6, the kinase could be added in a broader panel of biomarkers for the diagnosis of Parkinson's disease. Conclusions Changes of PAK6 and 14-3-3γ amount in plasma represent a shared readout for patients affected by sporadic and LRRK2-linked Parkinson's disease. Overall, they can contribute to the establishment of an extended panel of biomarkers for the diagnosis of Parkinson's disease.
Collapse
Affiliation(s)
| | | | - Lucia Iannotta
- Department of Biology, University of Padova, Padova, Italy
| | | | | | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
| | - Angelo Antonini
- Padova Neuroscience Center, University of Padova, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Laura Civiero
- IRCCS San Camillo Hospital, Venice, Italy
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Jiang Y, MacNeil LT. Simple model systems reveal conserved mechanisms of Alzheimer's disease and related tauopathies. Mol Neurodegener 2023; 18:82. [PMID: 37950311 PMCID: PMC10638731 DOI: 10.1186/s13024-023-00664-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023] Open
Abstract
The lack of effective therapies that slow the progression of Alzheimer's disease (AD) and related tauopathies highlights the need for a more comprehensive understanding of the fundamental cellular mechanisms underlying these diseases. Model organisms, including yeast, worms, and flies, provide simple systems with which to investigate the mechanisms of disease. The evolutionary conservation of cellular pathways regulating proteostasis and stress response in these organisms facilitates the study of genetic factors that contribute to, or protect against, neurodegeneration. Here, we review genetic modifiers of neurodegeneration and related cellular pathways identified in the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, and the fruit fly Drosophila melanogaster, focusing on models of AD and related tauopathies. We further address the potential of simple model systems to better understand the fundamental mechanisms that lead to AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuwei Jiang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
14
|
Hao S, Huang H, Ma RY, Zeng X, Duan CY. Multifaceted functions of Drp1 in hypoxia/ischemia-induced mitochondrial quality imbalance: from regulatory mechanism to targeted therapeutic strategy. Mil Med Res 2023; 10:46. [PMID: 37833768 PMCID: PMC10571487 DOI: 10.1186/s40779-023-00482-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Hypoxic-ischemic injury is a common pathological dysfunction in clinical settings. Mitochondria are sensitive organelles that are readily damaged following ischemia and hypoxia. Dynamin-related protein 1 (Drp1) regulates mitochondrial quality and cellular functions via its oligomeric changes and multiple modifications, which plays a role in mediating the induction of multiple organ damage during hypoxic-ischemic injury. However, there is active controversy and gaps in knowledge regarding the modification, protein interaction, and functions of Drp1, which both hinder and promote development of Drp1 as a novel therapeutic target. Here, we summarize recent findings on the oligomeric changes, modification types, and protein interactions of Drp1 in various hypoxic-ischemic diseases, as well as the Drp1-mediated regulation of mitochondrial quality and cell functions following ischemia and hypoxia. Additionally, potential clinical translation prospects for targeting Drp1 are discussed. This review provides new ideas and targets for proactive interventions on multiple organ damage induced by various hypoxic-ischemic diseases.
Collapse
Affiliation(s)
- Shuai Hao
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002 China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Rui-Yan Ma
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037 China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400010 China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| |
Collapse
|
15
|
Schmid ET, Schinaman JM, Williams KS, Walker DW. Accumulation of F-actin drives brain aging and limits healthspan in Drosophila. RESEARCH SQUARE 2023:rs.3.rs-3158290. [PMID: 37577708 PMCID: PMC10418561 DOI: 10.21203/rs.3.rs-3158290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The actin cytoskeleton is a key determinant of cell and tissue homeostasis. However, tissue-specific roles for actin dynamics in aging, notably brain aging, are not understood. Here, we show that there is an age-related increase in filamentous actin (F-actin) in Drosophila brains, which is counteracted by prolongevity interventions. Critically, modulating F-actin levels in aging neurons prevents age-onset cognitive decline and extends organismal healthspan. Mechanistically, we show that autophagy, a recycling process required for neuronal homeostasis, is disabled upon actin dysregulation in the aged brain. Remarkably, disrupting actin polymerization in aged animals with cytoskeletal drugs restores brain autophagy to youthful levels and reverses cellular hallmarks of brain aging. Finally, reducing F-actin levels in aging neurons slows brain aging and promotes healthspan in an autophagy-dependent manner. Our data identify excess actin polymerization as a hallmark of brain aging, which can be targeted to reverse brain aging phenotypes and prolong healthspan.
Collapse
Affiliation(s)
- Edward T. Schmid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Joseph M. Schinaman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Kylie S. Williams
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
16
|
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
|
17
|
Varte V, Munkelwitz JW, Rincon-Limas DE. Insights from Drosophila on Aβ- and tau-induced mitochondrial dysfunction: mechanisms and tools. Front Neurosci 2023; 17:1184080. [PMID: 37139514 PMCID: PMC10150963 DOI: 10.3389/fnins.2023.1184080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia in older adults worldwide. Sadly, there are no disease-modifying therapies available for treatment due to the multifactorial complexity of the disease. AD is pathologically characterized by extracellular deposition of amyloid beta (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau. Increasing evidence suggest that Aβ also accumulates intracellularly, which may contribute to the pathological mitochondrial dysfunction observed in AD. According with the mitochondrial cascade hypothesis, mitochondrial dysfunction precedes clinical decline and thus targeting mitochondria may result in new therapeutic strategies. Unfortunately, the precise mechanisms connecting mitochondrial dysfunction with AD are largely unknown. In this review, we will discuss how the fruit fly Drosophila melanogaster is contributing to answer mechanistic questions in the field, from mitochondrial oxidative stress and calcium dysregulation to mitophagy and mitochondrial fusion and fission. In particular, we will highlight specific mitochondrial insults caused by Aβ and tau in transgenic flies and will also discuss a variety of genetic tools and sensors available to study mitochondrial biology in this flexible organism. Areas of opportunity and future directions will be also considered.
Collapse
Affiliation(s)
- Vanlalrinchhani Varte
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Jeremy W. Munkelwitz
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Diego E. Rincon-Limas
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
18
|
Ravinther AI, Dewadas HD, Tong SR, Foo CN, Lin YE, Chien CT, Lim YM. Molecular Pathways Involved in LRRK2-Linked Parkinson’s Disease: A Systematic Review. Int J Mol Sci 2022; 23:ijms231911744. [PMID: 36233046 PMCID: PMC9569706 DOI: 10.3390/ijms231911744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
Parkinson’s disease is one of the most common neurodegenerative diseases affecting the ageing population, with a prevalence that has doubled over the last 30 years. As the mechanism of the disease is not fully elucidated, the current treatments are unable to effectively prevent neurodegeneration. Studies have found that mutations in Leucine-rich-repeat-kinase 2 (LRRK2) are the most common cause of familial Parkinson’s disease (PD). Moreover, aberrant (higher) LRRK2 kinase activity has an influence in idiopathic PD as well. Hence, the aim of this review is to categorize and synthesize current information related to LRRK2-linked PD and present the factors associated with LRRK2 that can be targeted therapeutically. A systematic review was conducted using the databases PubMed, Medline, SCOPUS, SAGE, and Cochrane (January 2016 to July 2021). Search terms included “Parkinson’s disease”, “mechanism”, “LRRK2”, and synonyms in various combinations. The search yielded a total of 988 abstracts for initial review, 80 of which met the inclusion criteria. Here, we emphasize molecular mechanisms revealed in recent in vivo and in vitro studies. By consolidating the recent updates in the field of LRRK2-linked PD, researchers can further evaluate targets for therapeutic application.
Collapse
Affiliation(s)
- Ailyn Irvita Ravinther
- Centre for Cancer Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Hemaniswarri Dewi Dewadas
- Centre for Biomedical and Nutrition Research, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Perak, Malaysia
| | - Shi Ruo Tong
- Centre for Cancer Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
| | - Chai Nien Foo
- Centre for Cancer Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Department of Population Medicine, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
| | - Yu-En Lin
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Yang Mooi Lim
- Centre for Cancer Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Department of Pre-Clinical Sciences, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Correspondence:
| |
Collapse
|
19
|
Goel P, Chakrabarti S, Goel K, Bhutani K, Chopra T, Bali S. Neuronal cell death mechanisms in Alzheimer's disease: An insight. Front Mol Neurosci 2022; 15:937133. [PMID: 36090249 PMCID: PMC9454331 DOI: 10.3389/fnmol.2022.937133] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regulated cell death (RCD) is an ordered and tightly orchestrated set of changes/signaling events in both gene expression and protein activity and is responsible for normal development as well as maintenance of tissue homeostasis. Aberrant activation of this pathway results in cell death by various mechanisms including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. Such pathological changes in neurons alone or in combination have been observed in the pathogenesis of various neurodegenerative diseases including Alzheimer's disease (AD). Pathological hallmarks of AD focus primarily on the accumulation of two main protein markers: amyloid β peptides and abnormally phosphorylated tau proteins. These protein aggregates result in the formation of A-β plaques and neuro-fibrillary tangles (NFTs) and induce neuroinflammation and neurodegeneration over years to decades leading to a multitude of cognitive and behavioral deficits. Autopsy findings of AD reveal massive neuronal death manifested in the form of cortical volume shrinkage, reduction in sizes of gyri to up to 50% and an increase in the sizes of sulci. Multiple forms of cell death have been recorded in neurons from different studies conducted so far. However, understanding the mechanism/s of neuronal cell death in AD patients remains a mystery as the trigger that results in aberrant activation of RCD is unknown and because of the limited availability of dying neurons. This review attempts to elucidate the process of Regulated cell death, how it gets unregulated in response to different intra and extracellular stressors, various forms of unregulated cell death, their interplay and their role in pathogenesis of Alzheimer's Disease in both human and experimental models of AD. Further we plan to explore the correlation of both amyloid-beta and Tau with neuronal loss as seen in AD.
Collapse
Affiliation(s)
- Parul Goel
- Department of Biochemistry, Shri Atal Bihari Vajpayee Government Medical College Chhainsa, Faridabad, India
| | - Sasanka Chakrabarti
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Kapil Goel
- Department of Community Medicine and School of Public Health, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Karanpreet Bhutani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Tanya Chopra
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| | - Sharadendu Bali
- Department of Surgery, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar (Deemed to be University), Ambala, India
| |
Collapse
|
20
|
Tsujikawa K, Hamanaka K, Riku Y, Hattori Y, Hara N, Iguchi Y, Ishigaki S, Hashizume A, Miyatake S, Mitsuhashi S, Miyazaki Y, Kataoka M, Jiayi L, Yasui K, Kuru S, Koike H, Kobayashi K, Sahara N, Ozaki N, Yoshida M, Kakita A, Saito Y, Iwasaki Y, Miyashita A, Iwatsubo T, Japanese Alzheimer’s Disease Neuroimaging Initiative (J-ADNI), Ikeuchi T, Japanese Longitudinal Biomarker Study in PSP and CBD (JALPAC) Consortium, Miyata T, Sobue G, Matsumoto N, Sahashi K, Katsuno M. Actin-binding protein filamin-A drives tau aggregation and contributes to progressive supranuclear palsy pathology. SCIENCE ADVANCES 2022; 8:eabm5029. [PMID: 35613261 PMCID: PMC9132466 DOI: 10.1126/sciadv.abm5029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
While amyloid-β lies upstream of tau pathology in Alzheimer's disease, key drivers for other tauopathies, including progressive supranuclear palsy (PSP), are largely unknown. Various tau mutations are known to facilitate tau aggregation, but how the nonmutated tau, which most cases with PSP share, increases its propensity to aggregate in neurons and glial cells has remained elusive. Here, we identified genetic variations and protein abundance of filamin-A in the PSP brains without tau mutations. We provided in vivo biochemical evidence that increased filamin-A levels enhance the phosphorylation and insolubility of tau through interacting actin filaments. In addition, reduction of filamin-A corrected aberrant tau levels in the culture cells from PSP cases. Moreover, transgenic mice carrying human filamin-A recapitulated tau pathology in the neurons. Our data highlight that filamin-A promotes tau aggregation, providing a potential mechanism by which filamin-A contributes to PSP pathology.
Collapse
Affiliation(s)
- Koyo Tsujikawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neurology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
- Department of Neurology , National Hospital Organization Suzuka National Hospital, Suzuka, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yohei Iguchi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Japan
| | - Satomi Mitsuhashi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Genomic Function and Diversity, Medical Research Institute Tokyo Medical and Dental University, Tokyo, Japan
| | - Yu Miyazaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mayumi Kataoka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Li Jiayi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keizo Yasui
- Department of Neurology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Satoshi Kuru
- Department of Neurology , National Hospital Organization Suzuka National Hospital, Suzuka, Japan
| | - Haruki Koike
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuko Saito
- Department of Neurology and Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Yasushi Iwasaki
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Sobue
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Corresponding author.
| |
Collapse
|
21
|
Benarroch E. What Is the Role of Mitochondrial Fission in Neurologic Disease? Neurology 2022; 98:662-668. [PMID: 35437267 DOI: 10.1212/wnl.0000000000200233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
|
22
|
Cogo S, Ho FY, Tosoni E, Tomkins JE, Tessari I, Iannotta L, Montine TJ, Manzoni C, Lewis PA, Bubacco L, Chartier Harlin MC, Taymans JM, Kortholt A, Nichols J, Cendron L, Civiero L, Greggio E. The Roc domain of LRRK2 as a hub for protein-protein interactions: a focus on PAK6 and its impact on RAB phosphorylation. Brain Res 2022; 1778:147781. [PMID: 35016853 DOI: 10.1016/j.brainres.2022.147781] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has taken center stage in Parkinson's disease (PD) research as mutations cause familial PD and more common variants increase lifetime risk for disease. One unique feature in LRRK2 is the coexistence of GTPase/Roc (Ras of complex) and kinase catalytic functions, bridged by a COR (C-terminal Of Roc) platform for dimerization. Multiple PD mutations are located within the Roc/GTPase domain and concomitantly lead to defective GTPase activity and augmented kinase activity in cells, supporting a crosstalk between GTPase and kinase domains. In addition, biochemical and structural data highlight the importance of Roc as a molecular switch modulating LRRK2 monomer-to-dimer equilibrium and building the interface for interaction with binding partners. Here we review the effects of PD Roc mutations on LRRK2 function and discuss the importance of Roc as a hub for multiple molecular interactions relevant for the regulation of cytoskeletal dynamics and intracellular trafficking pathways. Among the well-characterized Roc interactors, we focused on the cytoskeletal-related kinase p21-activated kinase 6 (PAK6). We report the affinity between LRRK2-Roc and PAK6 measured by microscale thermophoresis (MST). We further show that PAK6 can modulate LRRK2-mediated phosphorylation of RAB substrates in the presence of LRRK2 wild-type (WT) or the PD G2019S kinase mutant but not when the PD Roc mutation R1441G is expressed. These findings support a mechanism whereby mutations in Roc might affect LRRK2 activity through impaired protein-protein interaction in the cell.
Collapse
Affiliation(s)
- Susanna Cogo
- Department of Biology, University of Padova, Italy.
| | - Franz Y Ho
- Department of Cell Biochemistry, University of Groningen, The Netherlands
| | - Elena Tosoni
- Department of Biology, University of Padova, Italy
| | | | | | | | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, USA
| | - Claudia Manzoni
- Department of Pharmacology, University College London School of Pharmacy, UK
| | - Patrick A Lewis
- Royal Veterinary College, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Luigi Bubacco
- Department of Biology, University of Padova, Italy; Centro Studi per la Neurodegenerazione CESNE, University of Padova, Italy
| | | | - Jean-Marc Taymans
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, The Netherlands
| | - Jeremy Nichols
- Department of Pathology, Stanford University School of Medicine, USA
| | | | - Laura Civiero
- Department of Biology, University of Padova, Italy; IRCCS San Camillo Hospital, Venice, Italy; Centro Studi per la Neurodegenerazione CESNE, University of Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Italy; Centro Studi per la Neurodegenerazione CESNE, University of Padova, Italy.
| |
Collapse
|
23
|
Neves PFR, Milanesi BB, Paz LV, de Miranda Monteiro VAC, Neves LT, da Veiga LC, da Silva RB, Sulzbach JH, Knijkik GP, de Revoredo Ribeiro EC, de Souza Silva EL, Vieira MQ, Bagatini PB, Wieck A, Mestriner RG, Xavier LL. Age-related tolerance to paraquat-induced parkinsonism in Drosophila melanogaster. Toxicol Lett 2022; 361:43-53. [DOI: 10.1016/j.toxlet.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 11/28/2022]
|
24
|
Wang J, Zhu S, Lu W, Li A, Zhou Y, Chen Y, Chen M, Qian C, Hu X, Zhang Y, Huang C. Varenicline improved laparotomy-induced cognitive impairment by restoring mitophagy in aged mice. Eur J Pharmacol 2022; 916:174524. [PMID: 34582844 DOI: 10.1016/j.ejphar.2021.174524] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 01/06/2023]
Abstract
Growing incidence of postoperative cognitive dysfunction (POCD) in the elderly populations after major surgery challenges us to provide stable and effective treatments. Mitochondria dysfunction is essential in the pathogenesis of aging and neurodegenerative diseases. It is hypothesized that varenicline improves cognitive impairment through restoring mitophagy and tau phosphorylation. Wild type C57BL/6 mice (male, 18-month-old) were subjected to laparotomy with or without chronic varenicline administration. Postoperative cognition and anxiety were determined by Morris water maze and elevated plus maze tests. Meanwhile, oxidative stress, mitochondria function, mitophagy and tau phosphorylation, as well as the correlation of PKR and STAT3 were characterized. In aged mice following laparotomy, persistent cognitive dysfunction in spatial learning and memory were indicated by longer escape latency and less crossing frequency in the target quadrant. Laparotomy also induced anxiety responses deficits. After postoperative 14 days, significant ROS accumulation and smaller mitochondria with impaired function were presented in the hippocampus. Simultaneously, there were abundant of neuronal apoptosis and translocation of tau phosphorylation in the mitochondria. Enhanced mitophagy and down regulated ChAT activity were distributed in the mice subjected to laparotomy. PKR signaling was activated and required for subcellular activation of STAT3 in the brain. After chronic varenicline administration (1 mg/kg/day), cognitive dysfunction, hippocampal oxidative stress, as well as fragile mitophagy were improved. Our results highlight that laparotomy caused cognitive impairment with persistent oxidative stress, mitochondria dysfunction and autophagy dysregulation. PKR/STAT3 maybe the potential mechanism, and perioperative varenicline treatment could be an efficient therapeutic strategy for POCD.
Collapse
Affiliation(s)
- Jianan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Shoufeng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Wenping Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Ao Li
- The Second Clinical Medical College of Anhui Medical University, Hefei City, Anhui Province, China
| | - Yuqi Zhou
- The Second Clinical Medical College of Anhui Medical University, Hefei City, Anhui Province, China
| | - Yihuan Chen
- The Second Clinical Medical College of Anhui Medical University, Hefei City, Anhui Province, China
| | - Ming Chen
- The Second Clinical Medical College of Anhui Medical University, Hefei City, Anhui Province, China
| | - Cheng Qian
- Center for Scientific Research of Anhui Medical University, Hefei City, Anhui Province, China
| | - Xianwen Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Ye Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Chunxia Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei City, Anhui Province, China; Scientific Research and Experiment Center of the Second Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| |
Collapse
|
25
|
Suzuki M, Sango K, Nagai Y. Roles of α-Synuclein and Disease-Associated Factors in Drosophila Models of Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23031519. [PMID: 35163450 PMCID: PMC8835920 DOI: 10.3390/ijms23031519] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
α-Synuclein (αSyn) plays a major role in the pathogenesis of Parkinson’s disease (PD), which is the second most common neurodegenerative disease after Alzheimer’s disease. The accumulation of αSyn is a pathological hallmark of PD, and mutations in the SNCA gene encoding αSyn cause familial forms of PD. Moreover, the ectopic expression of αSyn has been demonstrated to mimic several key aspects of PD in experimental model systems. Among the various model systems, Drosophila melanogaster has several advantages for modeling human neurodegenerative diseases. Drosophila has a well-defined nervous system, and numerous tools have been established for its genetic analyses. The rapid generation cycle and short lifespan of Drosophila renders them suitable for high-throughput analyses. PD model flies expressing αSyn have contributed to our understanding of the roles of various disease-associated factors, including genetic and nongenetic factors, in the pathogenesis of PD. In this review, we summarize the molecular pathomechanisms revealed to date using αSyn-expressing Drosophila models of PD, and discuss the possibilities of using these models to demonstrate the biological significance of disease-associated factors.
Collapse
Affiliation(s)
- Mari Suzuki
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan;
- Department of Neurotherapeutics, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- Correspondence: (M.S.); (Y.N.); Tel.: +81-5316-3100 (M.S.); +81-72-366-0221 (Y.N.)
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan;
| | - Yoshitaka Nagai
- Department of Neurotherapeutics, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- Department of Neurology, Faculty of Medicine, Kindai University, Osaka-Sayama 589-8511, Japan
- Correspondence: (M.S.); (Y.N.); Tel.: +81-5316-3100 (M.S.); +81-72-366-0221 (Y.N.)
| |
Collapse
|
26
|
Bharat V, Hsieh CH, Wang X. Mitochondrial Defects in Fibroblasts of Pathogenic MAPT Patients. Front Cell Dev Biol 2021; 9:765408. [PMID: 34805172 PMCID: PMC8595217 DOI: 10.3389/fcell.2021.765408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in MAPT gene cause multiple neurological disorders, including frontal temporal lobar degeneration and parkinsonism. Increasing evidence indicates impaired mitochondrial homeostasis and mitophagy in patients and disease models of pathogenic MAPT. Here, using MAPT patients' fibroblasts as a model, we report that disease-causing MAPT mutations compromise early events of mitophagy. By employing biochemical and mitochondrial assays we discover that upon mitochondrial depolarization, the recruitment of LRRK2 and Parkin to mitochondria and degradation of the outer mitochondrial membrane protein Miro1 are disrupted. Using high resolution electron microscopy, we reveal that the contact of mitochondrial membranes with ER and cytoskeleton tracks is dissociated following mitochondrial damage. This membrane dissociation is blocked by a pathogenic MAPT mutation. Furthermore, we provide evidence showing that tau protein, which is encoded by MAPT gene, interacts with Miro1 protein, and this interaction is abolished by pathogenic MAPT mutations. Lastly, treating fibroblasts of a MAPT patient with a small molecule promotes Miro1 degradation following depolarization. Altogether, our results show molecular defects in a peripheral tissue of patients and suggest that targeting mitochondrial quality control may have a broad application for future therapeutic intervention.
Collapse
|
27
|
Pan L, Meng L, He M, Zhang Z. Tau in the Pathophysiology of Parkinson's Disease. J Mol Neurosci 2021; 71:2179-2191. [PMID: 33459970 PMCID: PMC8585831 DOI: 10.1007/s12031-020-01776-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
The pathological hallmarks of Parkinson's disease (PD) are the progressive loss of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies (LBs) in remaining neurons. LBs primarily consist of aggregated α-Synuclein (α-Syn). However, accumulating evidence suggests that Tau, which is associated with tauopathies such as Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and argyrophilic grain disease, is also involved in the pathophysiology of PD. A genome-wide association study (GWAS) identified MAPT, the gene encoding the Tau protein, as a risk gene for PD. Autopsy of PD patients also revealed the colocalization of Tau and α-Syn in LBs. Experimental evidence has shown that Tau interacts with α-Syn and influences the pathology of α-Syn in PD. In this review, we discuss the structure and function of Tau and provide a summary of the current evidence supporting Tau's involvement as either an active or passive element in the pathophysiology of PD, which may provide novel targets for the early diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Mingyang He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
28
|
Olsen AL, Feany MB. Parkinson's disease risk genes act in glia to control neuronal α-synuclein toxicity. Neurobiol Dis 2021; 159:105482. [PMID: 34390834 PMCID: PMC8502212 DOI: 10.1016/j.nbd.2021.105482] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 11/18/2022] Open
Abstract
Idiopathic Parkinson's disease is the second most common neurodegenerative disease and is estimated to be approximately 30% heritable. Genome wide association studies have revealed numerous loci associated with risk of development of Parkinson's disease. The majority of genes identified in these studies are expressed in glia at either similar or greater levels than their expression in neurons, suggesting that glia may play a role in Parkinson's disease pathogenesis. The role of individual glial risk genes in Parkinson's disease development or progression is unknown, however. We hypothesized that some Parkinson's disease risk genes exert their effects through glia. We developed a Drosophila model of α-synucleinopathy in which we can independently manipulate gene expression in neurons and glia. Human wild type α-synuclein is expressed in all neurons, and these flies develop the hallmarks of Parkinson's disease, including motor impairment, death of dopaminergic and other neurons, and α-synuclein aggregation. In these flies, we performed a candidate genetic screen, using RNAi to knockdown 14 well-validated Parkinson's disease risk genes in glia and measuring the effect on locomotion in order to identify glial modifiers of the α-synuclein phenotype. We identified 4 modifiers: aux, Lrrk, Ric, and Vps13, orthologs of the human genes GAK, LRRK2, RIT2, and VPS13C, respectively. Knockdown of each gene exacerbated neurodegeneration as measured by total and dopaminergic neuron loss. Knockdown of each modifier also increased α-synuclein oligomerization. These results suggest that some Parkinson's disease risk genes exert their effects in glia and that glia can influence neuronal α-synuclein proteostasis in a non-cell-autonomous fashion. Further, this study provides proof of concept that our novel Drosophila α-synucleinopathy model can be used to study glial modifier genes, paving the way for future large unbiased screens to identify novel glial risk factors that contribute to PD risk and progression.
Collapse
Affiliation(s)
- Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, United States of America; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, United States of America
| | - Mel B Feany
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, United States of America; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, United States of America.
| |
Collapse
|
29
|
Sarkar S, Bardai F, Olsen AL, Lohr KM, Zhang YY, Feany MB. Oligomerization of Lrrk controls actin severing and α-synuclein neurotoxicity in vivo. Mol Neurodegener 2021; 16:33. [PMID: 34030727 PMCID: PMC8142648 DOI: 10.1186/s13024-021-00454-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mutations in LRRK2 are the most common cause of familial Parkinson's disease and typically cause disease in the context of abnormal aggregation and deposition of α-synuclein within affected brain tissue. METHODS We combine genetic analysis of Lrrk-associated toxicity in a penetrant Drosophila model of wild type human α-synuclein neurotoxicity with biochemical analyses and modeling of LRRK2 toxicity in human neurons and transgenic mouse models. RESULTS We demonstrate that Lrrk and α-synuclein interact to promote neuronal degeneration through convergent effects on the actin cytoskeleton and downstream dysregulation of mitochondrial dynamics and function. We find specifically that monomers and dimers of Lrrk efficiently sever actin and promote normal actin dynamics in vivo. Oligomerization of Lrrk, which is promoted by dominant Parkinson's disease-causing mutations, reduces actin severing activity in vitro and promotes excess stabilization of F-actin in vivo. Importantly, a clinically protective Lrrk mutant reduces oligomerization and α-synuclein neurotoxicity. CONCLUSIONS Our findings provide a specific mechanistic link between two key molecules in the pathogenesis of Parkinson's disease, α-synuclein and LRRK2, and suggest potential new approaches for therapy development.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| | - Farah Bardai
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| | - Abby L. Olsen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| | - Kelly M. Lohr
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| | - Ying-Yi Zhang
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Massachusetts Boston, USA
| |
Collapse
|
30
|
Lai D, Alipanahi B, Fontanillas P, Schwantes-An TH, Aasly J, Alcalay RN, Beecham GW, Berg D, Bressman S, Brice A, Brockman K, Clark L, Cookson M, Das S, Van Deerlin V, Follett J, Farrer MJ, Trinh J, Gasser T, Goldwurm S, Gustavsson E, Klein C, Lang AE, Langston JW, Latourelle J, Lynch T, Marder K, Marras C, Martin ER, McLean CY, Mejia-Santana H, Molho E, Myers RH, Nuytemans K, Ozelius L, Payami H, Raymond D, Rogaeva E, Rogers MP, Ross OA, Samii A, Saunders-Pullman R, Schüle B, Schulte C, Scott WK, Tanner C, Tolosa E, Tomkins JE, Vilas D, Trojanowski JQ, Uitti R, Vance JM, Visanji NP, Wszolek ZK, Zabetian CP, Mirelman A, Giladi N, Orr Urtreger A, Cannon P, Fiske B, Foroud T. Genomewide Association Studies of LRRK2 Modifiers of Parkinson's Disease. Ann Neurol 2021; 90:76-88. [PMID: 33938021 PMCID: PMC8252519 DOI: 10.1002/ana.26094] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/03/2023]
Abstract
Objective The aim of this study was to search for genes/variants that modify the effect of LRRK2 mutations in terms of penetrance and age‐at‐onset of Parkinson's disease. Methods We performed the first genomewide association study of penetrance and age‐at‐onset of Parkinson's disease in LRRK2 mutation carriers (776 cases and 1,103 non‐cases at their last evaluation). Cox proportional hazard models and linear mixed models were used to identify modifiers of penetrance and age‐at‐onset of LRRK2 mutations, respectively. We also investigated whether a polygenic risk score derived from a published genomewide association study of Parkinson's disease was able to explain variability in penetrance and age‐at‐onset in LRRK2 mutation carriers. Results A variant located in the intronic region of CORO1C on chromosome 12 (rs77395454; p value = 2.5E‐08, beta = 1.27, SE = 0.23, risk allele: C) met genomewide significance for the penetrance model. Co‐immunoprecipitation analyses of LRRK2 and CORO1C supported an interaction between these 2 proteins. A region on chromosome 3, within a previously reported linkage peak for Parkinson's disease susceptibility, showed suggestive associations in both models (penetrance top variant: p value = 1.1E‐07; age‐at‐onset top variant: p value = 9.3E‐07). A polygenic risk score derived from publicly available Parkinson's disease summary statistics was a significant predictor of penetrance, but not of age‐at‐onset. Interpretation This study suggests that variants within or near CORO1C may modify the penetrance of LRRK2 mutations. In addition, common Parkinson's disease associated variants collectively increase the penetrance of LRRK2 mutations. ANN NEUROL 2021;90:82–94
Collapse
Affiliation(s)
- Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | | | | | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Jan Aasly
- Department of Neurology, St. Olavs Hospital, Trondheim, Norway
| | - Roy N Alcalay
- Department of Neurology, Columbia University, New York, NY
| | - Gary W Beecham
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany.,Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, Inserm, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kathrin Brockman
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Lorraine Clark
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Mark Cookson
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD
| | | | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jordan Follett
- Laboratory of Neurogenetics and Neuroscience, Fixel Institute for Neurological Diseases, McKnight Brain Institute, L5-101D, UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL
| | - Matthew J Farrer
- Laboratory of Neurogenetics and Neuroscience, Fixel Institute for Neurological Diseases, McKnight Brain Institute, L5-101D, UF Clinical and Translational Science Institute, University of Florida, Gainesville, FL
| | - Joanne Trinh
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Emil Gustavsson
- Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Anthony E Lang
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | - J William Langston
- Departments of Neurology, Neuroscience, and Pathology, Stanford University School of Medicine, Stanford, CA
| | | | - Timothy Lynch
- Dublin Neurological Institute at the Mater Misericordiae University Hospital, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Karen Marder
- Department of Neurology and Psychiatry, Taub Institute and Sergievsky Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Cory Y McLean
- 23andMe, Inc., Sunnyvale, CA.,Google LLC, Cambridge, MA
| | | | - Eric Molho
- Department of Neurology, Albany Medical College, Albany, NY
| | | | - Karen Nuytemans
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Laurie Ozelius
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Deborah Raymond
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Neurology, University of Toronto, Toronto, Canada
| | - Michael P Rogers
- Department of General Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Owen A Ross
- Departments of Neuroscience and Clinical Genomics, Mayo Clinic, Jacksonville, FL.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Ali Samii
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA
| | | | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Claudia Schulte
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - William K Scott
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Caroline Tanner
- University of California, San Francisco Veterans Affairs Health Care System, San Francisco, CA
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Hospital Clínic Universitari, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | | | - Dolores Vilas
- Parkinson Disease and Movement Disorders Unit, Hospital Clínic Universitari, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | -
- 23andMe, Inc., Sunnyvale, CA
| | - Ryan Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL
| | - Naomi P Visanji
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Canada
| | | | - Cyrus P Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA
| | - Anat Mirelman
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Nir Giladi
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Avi Orr Urtreger
- Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Brian Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
31
|
Chemical Stimulation of Rodent and Human Cortical Synaptosomes: Implications in Neurodegeneration. Cells 2021; 10:cells10051174. [PMID: 34065927 PMCID: PMC8151714 DOI: 10.3390/cells10051174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity events, including long-term potentiation (LTP), are often regarded as correlates of brain functions of memory and cognition. One of the central players in these plasticity-related phenomena is the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR). Increased levels of AMPARs on postsynaptic membranes thus constitute a biochemical measure of LTP. Isolated synaptic terminals (synaptosomes) are an excellent ex vivo tool to monitor synaptic physiology in healthy and diseased brains, particularly in human research. We herein describe three protocols for chemically-induced LTP (cLTP) in synaptosomes from both rodent and human brain tissues. Two of these chemical stimulation protocols are described for the first time in synaptosomes. A pharmacological block of synaptosomal actin dynamics confirmed the efficiency of the cLTP protocols. Furthermore, the study prototypically evaluated the deficiency of cLTP in cortical synaptosomes obtained from human cases of early-onset Alzheimer’s disease (EOAD) and frontotemporal lobar degeneration (FLTD), as well as an animal model that mimics FLTD.
Collapse
|
32
|
Abstract
Point mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease (PD) and are implicated in a significant proportion of apparently sporadic PD cases. Clinically, LRRK2-driven PD is indistinguishable from sporadic PD, making it an attractive genetic model for the much more common sporadic PD. In this review, we highlight recent advances in understanding LRRK2's subcellular functions using LRRK2-driven PD models, while also considering some of the limitations of these model systems. Recent developments of particular importance include new evidence of key LRRK2 functions in the endolysosomal system and LRRK2's regulation of and by Rab GTPases. Additionally, LRRK2's interaction with the cytoskeleton allowed elucidation of the LRRK2 structure and appears relevant to LRRK2 protein degradation and LRRK2 inhibitor therapies. We further discuss how LRRK2's interactions with other PD-driving genes, such as the VPS35, GBA1, and SNCA genes, may highlight cellular pathways more broadly disrupted in PD.
Collapse
Affiliation(s)
- Ahsan Usmani
- Department of Pathology, University of California, San Diego, San Diego, California, USA
| | - Farbod Shavarebi
- Department of Pathology, University of California, San Diego, San Diego, California, USA
| | - Annie Hiniker
- Department of Pathology, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
33
|
Onur TS, Laitman A, Zhao H, Keyho R, Kim H, Wang J, Mair M, Wang H, Li L, Perez A, de Haro M, Wan YW, Allen G, Lu B, Al-Ramahi I, Liu Z, Botas J. Downregulation of glial genes involved in synaptic function mitigates Huntington's disease pathogenesis. eLife 2021; 10:64564. [PMID: 33871358 PMCID: PMC8149125 DOI: 10.7554/elife.64564] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/19/2021] [Indexed: 01/01/2023] Open
Abstract
Most research on neurodegenerative diseases has focused on neurons, yet glia help form and maintain the synapses whose loss is so prominent in these conditions. To investigate the contributions of glia to Huntington's disease (HD), we profiled the gene expression alterations of Drosophila expressing human mutant Huntingtin (mHTT) in either glia or neurons and compared these changes to what is observed in HD human and HD mice striata. A large portion of conserved genes are concordantly dysregulated across the three species; we tested these genes in a high-throughput behavioral assay and found that downregulation of genes involved in synapse assembly mitigated pathogenesis and behavioral deficits. To our surprise, reducing dNRXN3 function in glia was sufficient to improve the phenotype of flies expressing mHTT in neurons, suggesting that mHTT's toxic effects in glia ramify throughout the brain. This supports a model in which dampening synaptic function is protective because it attenuates the excitotoxicity that characterizes HD. When a neuron dies, through injury or disease, the body loses all communication that passes through it. The brain compensates by rerouting the flow of information through other neurons in the network. Eventually, if the loss of neurons becomes too great, compensation becomes impossible. This process happens in Alzheimer's, Parkinson's, and Huntington's disease. In the case of Huntington's disease, the cause is mutation to a single gene known as huntingtin. The mutation is present in every cell in the body but causes particular damage to parts of the brain involved in mood, thinking and movement. Neurons and other cells respond to mutations in the huntingtin gene by turning the activities of other genes up or down, but it is not clear whether all of these changes contribute to the damage seen in Huntington's disease. In fact, it is possible that some of the changes are a result of the brain trying to protect itself. So far, most research on this subject has focused on neurons because the huntingtin gene plays a role in maintaining healthy neuronal connections. But, given that all cells carry the mutated gene, it is likely that other cells are also involved. The glia are a diverse group of cells that support the brain, providing care and sustenance to neurons. These cells have a known role in maintaining the connections between neurons and may also have play a role in either causing or correcting the damage seen in Huntington's disease. The aim of Onur et al. was to find out which genes are affected by having a mutant huntingtin gene in neurons or glia, and whether severity of Huntington’s disease improved or worsened when the activity of these genes changed. First, Onur et al. identified genes affected by mutant huntingtin by comparing healthy human brains to the brains of people with Huntington's disease. Repeating the same comparison in mice and fruit flies identified genes affected in the same way across all three species, revealing that, in Huntington's disease, the brain dials down glial cell genes involved in maintaining neuronal connections. To find out how these changes in gene activity affect disease severity and progression, Onur et al. manipulated the activity of each of the genes they had identified in fruit flies that carried mutant versions of huntingtin either in neurons, in glial cells or in both cell types. They then filmed the flies to see the effects of the manipulation on movement behaviors, which are affected by Huntington’s disease. This revealed that purposely lowering the activity of the glial genes involved in maintaining connections between neurons improved the symptoms of the disease, but only in flies who had mutant huntingtin in their glial cells. This indicates that the drop in activity of these genes observed in Huntington’s disease is the brain trying to protect itself. This work suggests that it is important to include glial cells in studies of neurological disorders. It also highlights the fact that changes in gene expression as a result of a disease are not always bad. Many alterations are compensatory, and try to either make up for or protect cells affected by the disease. Therefore, it may be important to consider whether drugs designed to treat a condition by changing levels of gene activity might undo some of the body's natural protection. Working out which changes drive disease and which changes are protective will be essential for designing effective treatments.
Collapse
Affiliation(s)
- Tarik Seref Onur
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States
| | - Andrew Laitman
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - He Zhao
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Ryan Keyho
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Hyemin Kim
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Jennifer Wang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Megan Mair
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States
| | - Huilan Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lifang Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Alma Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Maria de Haro
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Ying-Wooi Wan
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Genevera Allen
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Departments of Electrical & Computer Engineering, Statistics and Computer Science, Rice University, Houston, United States
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ismael Al-Ramahi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States
| |
Collapse
|
34
|
Changes in Drp1 Function and Mitochondrial Morphology Are Associated with the α-Synuclein Pathology in a Transgenic Mouse Model of Parkinson's Disease. Cells 2021; 10:cells10040885. [PMID: 33924585 PMCID: PMC8070398 DOI: 10.3390/cells10040885] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/16/2022] Open
Abstract
Alterations in mitochondrial function and morphology are associated with many human diseases, including cancer and neurodegenerative diseases. Mitochondrial impairment is linked to Parkinson's disease (PD) pathogenesis, and alterations in mitochondrial dynamics are seen in PD models. In particular, α-synuclein (αS) abnormalities are often associated with pathological changes to mitochondria. However, the relationship between αS pathology and mitochondrial dynamics remains poorly defined. Herein, we examined a mouse model of α-synucleinopathy for αS pathology-linked alterations in mitochondrial dynamics in vivo. We show that α-synucleinopathy in a transgenic (Tg) mouse model expressing familial PD-linked mutant A53T human αS (TgA53T) is associated with a decrease in Drp1 localization and activity in the mitochondria. In addition, we show that the loss of Drp1 function in the mitochondria is associated with two distinct phenotypes of enlarged neuronal mitochondria. Mitochondrial enlargement was only present in diseased animals and, apart from Drp1, other proteins involved in mitochondrial dynamics are unlikely to cause these changes, as their levels remained mostly unchanged. Further, the levels of Mfn1, a protein that facilitates mitochondrial fusion, was decreased nonspecifically with transgene expression. These results support the view that altered mitochondrial dynamics are a significant neuropathological factor in α-synucleinopathies.
Collapse
|
35
|
Sarkar S, Olsen AL, Sygnecka K, Lohr KM, Feany MB. α-synuclein impairs autophagosome maturation through abnormal actin stabilization. PLoS Genet 2021; 17:e1009359. [PMID: 33556113 PMCID: PMC7895402 DOI: 10.1371/journal.pgen.1009359] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/19/2021] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Vesicular trafficking defects, particularly those in the autophagolysosomal system, have been strongly implicated in the pathogenesis of Parkinson’s disease and related α-synucleinopathies. However, mechanisms mediating dysfunction of membrane trafficking remain incompletely understood. Using a Drosophila model of α-synuclein neurotoxicity with widespread and robust pathology, we find that human α-synuclein expression impairs autophagic flux in aging adult neurons. Genetic destabilization of the actin cytoskeleton rescues F-actin accumulation, promotes autophagosome clearance, normalizes the autophagolysosomal system, and rescues neurotoxicity in α-synuclein transgenic animals through an Arp2/3 dependent mechanism. Similarly, mitophagosomes accumulate in human α-synuclein-expressing neurons, and reversal of excessive actin stabilization promotes both clearance of these abnormal mitochondria-containing organelles and rescue of mitochondrial dysfunction. These results suggest that Arp2/3 dependent actin cytoskeleton stabilization mediates autophagic and mitophagic dysfunction and implicate failure of autophagosome maturation as a pathological mechanism in Parkinson’s disease and related α-synucleinopathies. Vesicle trafficking is a central cell biological pathway perturbed in Parkinson’s disease. Here we use a genetic approach to define an underlying mechanism by demonstrating that the key Parkinson’s disease protein α-synuclein impairs maturation of autophagosomes and mitophagosomes through Arp2/3 dependent excess stabilization of cellular actin networks.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Abby L. Olsen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Katja Sygnecka
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kelly M. Lohr
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mel B. Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- * E-mail:
| |
Collapse
|
36
|
Jabbari E, Koga S, Valentino RR, Reynolds RH, Ferrari R, Tan MMX, Rowe JB, Dalgard CL, Scholz SW, Dickson DW, Warner TT, Revesz T, Höglinger GU, Ross OA, Ryten M, Hardy J, Shoai M, Morris HR. Genetic determinants of survival in progressive supranuclear palsy: a genome-wide association study. Lancet Neurol 2021; 20:107-116. [PMID: 33341150 PMCID: PMC7116626 DOI: 10.1016/s1474-4422(20)30394-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The genetic basis of variation in the progression of primary tauopathies has not been determined. We aimed to identify genetic determinants of survival in progressive supranuclear palsy (PSP). METHODS In stage one of this two stage genome-wide association study (GWAS), we included individuals with PSP, diagnosed according to pathological and clinical criteria, from two separate cohorts: the 2011 PSP GWAS cohort, from brain banks based at the Mayo Clinic (Jacksonville, FL, USA) and in Munich (Germany), and the University College London PSP cohort, from brain banks and the PROSPECT study, a UK-wide longitudinal study of patients with atypical parkinsonian syndromes. Individuals were included if they had clinical data available on sex, age at motor symptom onset, disease duration (from motor symptom onset to death or to the date of censoring, Dec 1, 2019, if individuals were alive), and PSP phenotype (with reference to the 2017 Movement Disorder Society criteria). Genotype data were used to do a survival GWAS using a Cox proportional hazards model. In stage two, data from additional individuals from the Mayo Clinic brain bank, which were obtained after the 2011 PSP GWAS, were used for a pooled analysis. We assessed the expression quantitative trait loci (eQTL) profile of variants that passed genome-wide significance in our GWAS using the Functional Mapping and Annotation of GWAS platform, and did colocalisation analyses using the eQTLGen and PsychENCODE datasets. FINDINGS Data were collected and analysed between Aug 1, 2016, and Feb 1, 2020. Data were available for 1001 individuals of white European ancestry with PSP in stage one. We found a genome-wide significant association with survival at chromosome 12 (lead single nucleotide polymorphism rs2242367, p=7·5 × 10-10, hazard ratio 1·42 [95% CI 1·22-1·67]). rs2242367 was associated with survival in the individuals added in stage two (n=238; p=0·049, 1·22 [1·00-1·48]) and in the pooled analysis of both stages (n=1239; p=1·3 × 10-10, 1·37 [1·25-1·51]). An eQTL database screen revealed that rs2242367 is associated with increased expression of LRRK2 and two long intergenic non-coding RNAs (lncRNAs), LINC02555 and AC079630.4, in whole blood. Although we did not detect a colocalisation signal for LRRK2, analysis of the PSP survival signal and eQTLs for LINC02555 in the eQTLGen blood dataset revealed a posterior probability of hypothesis 4 of 0·77, suggesting colocalisation due to a single shared causal variant. INTERPRETATION Genetic variation at the LRRK2 locus was associated with survival in PSP. The mechanism of this association might be through a lncRNA-regulated effect on LRRK2 expression because LINC02555 has previously been shown to regulate LRRK2 expression. LRRK2 has been associated with sporadic and familial forms of Parkinson's disease, and our finding suggests a genetic overlap with PSP. Further functional studies will be important to assess the potential of LRRK2 modulation as a disease-modifying therapy for PSP and related tauopathies. FUNDING PSP Association, CBD Solutions, Medical Research Council (UK).
Collapse
Affiliation(s)
- Edwin Jabbari
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK; Movement Disorders Centre, University College London Queen Square Institute of Neurology, London, UK.
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Regina H Reynolds
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
| | - Raffaele Ferrari
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK
| | - Manuela M X Tan
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK; Movement Disorders Centre, University College London Queen Square Institute of Neurology, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sonja W Scholz
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA; Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Thomas T Warner
- Reta Lila Weston Institute, University College London Queen Square Institute of Neurology, London, UK; Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, London, UK
| | - Tamas Revesz
- Reta Lila Weston Institute, University College London Queen Square Institute of Neurology, London, UK; Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, London, UK
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mina Ryten
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK; Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
| | - John Hardy
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK; Reta Lila Weston Institute, University College London Queen Square Institute of Neurology, London, UK; Dementia Research Institute at University College London, University College London Queen Square Institute of Neurology, London, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region, China
| | - Maryam Shoai
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK; Movement Disorders Centre, University College London Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
37
|
Sarkar S, Murphy MA, Dammer EB, Olsen AL, Rangaraju S, Fraenkel E, Feany MB. Comparative proteomic analysis highlights metabolic dysfunction in α-synucleinopathy. NPJ PARKINSONS DISEASE 2020; 6:40. [PMID: 33311497 PMCID: PMC7732845 DOI: 10.1038/s41531-020-00143-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022]
Abstract
The synaptic protein α-synuclein is linked through genetics and neuropathology to the pathogenesis of Parkinson’s disease and related disorders. However, the mechanisms by which α-synuclein influences disease onset and progression are incompletely understood. To identify pathogenic pathways and therapeutic targets we performed proteomic analysis in a highly penetrant new Drosophila model of α-synucleinopathy. We identified 476 significantly upregulated and 563 significantly downregulated proteins in heads from α-synucleinopathy model flies compared to controls. We then used multiple complementary analyses to identify and prioritize genes and pathways within the large set of differentially expressed proteins for functional studies. We performed Gene Ontology enrichment analysis, integrated our proteomic changes with human Parkinson’s disease genetic studies, and compared the α-synucleinopathy proteome with that of tauopathy model flies, which are relevant to Alzheimer’s disease and related disorders. These approaches identified GTP cyclohydrolase (GCH1) and folate metabolism as candidate mediators of α-synuclein neurotoxicity. In functional validation studies, we found that the knockdown of Drosophila Gch1 enhanced locomotor deficits in α-synuclein transgenic flies, while folate supplementation protected from α-synuclein toxicity. Our integrative analysis suggested that mitochondrial dysfunction was a common downstream mediator of neurodegeneration. Accordingly, Gch1 knockdown enhanced metabolic dysfunction in α-synuclein transgenic fly brains while folate supplementation partially normalized brain bioenergetics. Here we outline and implement an integrative approach to identify and validate potential therapeutic pathways using comparative proteomics and genetics and capitalizing on the facile genetic and pharmacological tools available in Drosophila.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A Murphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric B Dammer
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Sharma A, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Sahib S, Tian ZR, Buzoianu AD, Patnaik R, Wiklund L, Sharma HS. Mild traumatic brain injury exacerbates Parkinson's disease induced hemeoxygenase-2 expression and brain pathology: Neuroprotective effects of co-administration of TiO 2 nanowired mesenchymal stem cells and cerebrolysin. PROGRESS IN BRAIN RESEARCH 2020; 258:157-231. [PMID: 33223035 DOI: 10.1016/bs.pbr.2020.09.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mild traumatic brain injury (mTBI) is one of the leading predisposing factors in the development of Parkinson's disease (PD). Mild or moderate TBI induces rapid production of tau protein and alpha synuclein (ASNC) in the cerebrospinal fluid (CSF) and in several brain areas. Enhanced tau-phosphorylation and ASNC alters the molecular machinery of the brain leading to PD pathology. Recent evidences show upregulation of constitutive isoform of hemeoxygenase (HO-2) in PD patients that correlates well with the brain pathology. mTBI alone induces profound upregulation of HO-2 immunoreactivity. Thus, it would be interesting to explore whether mTBI exacerbates PD pathology in relation to tau, ASNC and HO-2 expression. In addition, whether neurotrophic factors and stem cells known to reduce brain pathology in TBI could induce neuroprotection in PD following mTBI. In this review role of mesenchymal stem cells (MSCs) and cerebrolysin (CBL), a well-balanced composition of several neurotrophic factors and active peptide fragments using nanowired delivery in PD following mTBI is discussed based on our own investigation. Our results show that mTBI induces concussion exacerbates PD pathology and nanowired delivery of MSCs and CBL induces superior neuroprotection. This could be due to reduction in tau, ASNC and HO-2 expression in PD following mTBI, not reported earlier. The functional significance of our findings in relation to clinical strategies is discussed.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Canto AM, Matos AHB, Godoi AB, Vieira AS, Aoyama BB, Rocha CS, Henning B, Carvalho BS, Pascoal VDB, Veiga DFT, Gilioli R, Cendes F, Lopes-Cendes I. Multi-omics analysis suggests enhanced epileptogenesis in the Cornu Ammonis 3 of the pilocarpine model of mesial temporal lobe epilepsy. Hippocampus 2020; 31:122-139. [PMID: 33037862 DOI: 10.1002/hipo.23268] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/04/2020] [Accepted: 09/26/2020] [Indexed: 12/11/2022]
Abstract
Mesial temporal lobe epilepsy (MTLE) is a chronic neurological disorder characterized by the occurrence of seizures, and histopathological abnormalities in the mesial temporal lobe structures, mainly hippocampal sclerosis (HS). We used a multi-omics approach to determine the profile of transcript and protein expression in the dorsal and ventral hippocampal dentate gyrus (DG) and Cornu Ammonis 3 (CA3) in an animal model of MTLE induced by pilocarpine. We performed label-free proteomics and RNAseq from laser-microdissected tissue isolated from pilocarpine-induced Wistar rats. We divided the DG and CA3 into dorsal and ventral areas and analyzed them separately. We performed a data integration analysis and evaluated enriched signaling pathways, as well as the integrated networks generated based on the gene ontology processes. Our results indicate differences in the transcriptomic and proteomic profiles among the DG and the CA3 subfields of the hippocampus. Moreover, our data suggest that epileptogenesis is enhanced in the CA3 region when compared to the DG, with most abnormalities in transcript and protein levels occurring in the CA3. Furthermore, our results show that the epileptogenesis in the pilocarpine model involves predominantly abnormal regulation of excitatory neuronal mechanisms mediated by N-methyl D-aspartate (NMDA) receptors, changes in the serotonin signaling, and neuronal activity controlled by calcium/calmodulin-dependent protein kinase (CaMK) regulation and leucine-rich repeat kinase 2 (LRRK2)/WNT signaling pathways.
Collapse
Affiliation(s)
- Amanda M Canto
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Alexandre H B Matos
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Alexandre B Godoi
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - André S Vieira
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Beatriz B Aoyama
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Cristiane S Rocha
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Barbara Henning
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Benilton S Carvalho
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Statistics, Institute of Mathematics, Statistics and Scientific Computing. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Vinicius D B Pascoal
- Department of Basic Sciences, Fluminense Federal University (UFF), Nova Friburgo, Rio de Janeiroz, Brazil
| | - Diogo F T Veiga
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Rovilson Gilioli
- Laboratory of Animal Quality Control, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Fernando Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Neurology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| |
Collapse
|
40
|
Insights into Disease-Associated Tau Impact on Mitochondria. Int J Mol Sci 2020; 21:ijms21176344. [PMID: 32882957 PMCID: PMC7503371 DOI: 10.3390/ijms21176344] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/23/2022] Open
Abstract
Abnormal tau protein aggregation in the brain is a hallmark of tauopathies, such as frontotemporal lobar degeneration and Alzheimer’s disease. Substantial evidence has been linking tau to neurodegeneration, but the underlying mechanisms have yet to be clearly identified. Mitochondria are paramount organelles in neurons, as they provide the main source of energy (adenosine triphosphate) to these highly energetic cells. Mitochondrial dysfunction was identified as an early event of neurodegenerative diseases occurring even before the cognitive deficits. Tau protein was shown to interact with mitochondrial proteins and to impair mitochondrial bioenergetics and dynamics, leading to neurotoxicity. In this review, we discuss in detail the different impacts of disease-associated tau protein on mitochondrial functions, including mitochondrial transport, network dynamics, mitophagy and bioenergetics. We also give new insights about the effects of abnormal tau protein on mitochondrial neurosteroidogenesis, as well as on the endoplasmic reticulum-mitochondria coupling. A better understanding of the pathomechanisms of abnormal tau-induced mitochondrial failure may help to identify new targets for therapeutic interventions.
Collapse
|
41
|
Ermogenous C, Green C, Jackson T, Ferguson M, Lord JM. Treating age-related multimorbidity: the drug discovery challenge. Drug Discov Today 2020; 25:1403-1415. [DOI: 10.1016/j.drudis.2020.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022]
|
42
|
Shah EJ, Gurdziel K, Ruden DM. Drosophila Exhibit Divergent Sex-Based Responses in Transcription and Motor Function After Traumatic Brain Injury. Front Neurol 2020; 11:511. [PMID: 32636795 PMCID: PMC7316956 DOI: 10.3389/fneur.2020.00511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
Every year, millions of people in the US suffer brain damage from mild to severe traumatic brain injuries (TBI) that result from a sudden impact to the head. Despite TBI being a leading cause of death and disability worldwide, sex differences that contribute to varied outcomes post-injury are not extensively studied and therefore, poorly understood. In this study, we aimed to explore biological sex as a variable influencing response to TBI using Drosophila melanogaster as a model, since flies have been shown to exhibit symptoms commonly seen in other mammalian models of TBI. After inflicting TBI using the high-impact trauma device, we isolated w1118 fly brains and assessed gene transcription changes in male and female flies at control and 1, 2, and 4 hr after TBI. Our results suggest that overall, Drosophila females show more gene transcript changes than males. Females also exhibit upregulated expression changes in immune response and mitochondrial genes across all time-points. In addition, we looked at the impact of injury on mitochondrial health and motor function in both sexes before and after injury. Although both sexes report similar changes in mitochondrial oxidation and negative geotaxis, locomotor activity appears to be more impaired in males than females. These data suggest that sex-differences not only influence the response to TBI but also contribute to varied outcomes post-injury.
Collapse
Affiliation(s)
- Ekta J Shah
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Katherine Gurdziel
- Office of the Vice President for Research, Wayne State University, Detroit, MI, United States
| | - Douglas M Ruden
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States.,Office of the Vice President for Research, Wayne State University, Detroit, MI, United States.,Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
43
|
Loeffler DA, Aasly JO, LeWitt PA, Coffey MP. What Have We Learned from Cerebrospinal Fluid Studies about Biomarkers for Detecting LRRK2 Parkinson's Disease Patients and Healthy Subjects with Parkinson's-Associated LRRK2 Mutations? JOURNAL OF PARKINSONS DISEASE 2020; 9:467-488. [PMID: 31322581 PMCID: PMC6700639 DOI: 10.3233/jpd-191630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common known cause of autosomal dominant Parkinson’s disease (PD) and sporadic PD (sPD). The clinical presentation of LRRK2 PD is similar to sPD, and except for genetic testing, no biochemical or imaging markers can differentiate LRRK2 PD from sPD. Discovery of such biomarkers could indicate neuropathological mechanisms that are unique to or increased in LRRK2 PD. This review discusses findings in 17 LRRK2 - related CSF studies found on PubMed. Most of these studies compared analyte concentrations between four diagnostic groups: LRRK2 PD patients, sPD patients, asymptomatic control subjects carrying PD-associated LRRK2 mutations (LRRK2 CTL), and healthy control subjects lacking LRRK2 mutations (CTL). Analytes examined in these studies included Aβ1-42, tau, α-synuclein, oxidative stress markers, autophagy-related proteins, pteridines, neurotransmitter metabolites, exosomal LRRK2 protein, RNA species, inflammatory cytokines, mitochondrial DNA (mtDNA), and intermediary metabolites. FINDINGS: Pteridines, α-synuclein, mtDNA, 5-hydroxyindolacetic acid, β-D-glucose, lamp2, interleukin-8, and vascular endothelial growth factor were suggested to differentiate LRRK2 PD from sPD patients; 8-hydroxy-2’-deoxyguanosine (8-OHdG), 8-isoprostane (8-ISO), 2-hydroxybutyrate, mtDNA, lamp2, and neopterin may differentiate between LRRK2 CTL and LRRK2 PD subjects; and soluble oligomeric α-synuclein, 8-OHdG, and 8-ISO might differentiate LRRK2 CTL from CTL subjects. CONCLUSIONS: The low numbers of investigations of each analyte, small sample sizes, and methodological differences limit conclusions that can be drawn from these studies. Further investigations are indicated to determine the validity of the analytes identified in these studies as possible biomarkers for LRRK2 PD patients and/or LRRK2 CTL subjects.
Collapse
Affiliation(s)
- David A Loeffler
- Department of Neurology, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| | - Jan O Aasly
- Department of Neurology, St. Olav's Hospital, Trondheim, Norway
| | - Peter A LeWitt
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mary P Coffey
- Department of Biostatistics, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
44
|
Zhou L, Yao M, Tian Z, Song Y, Sun Y, Ye J, Li G, Sng KS, Xu L, Cui X, Wang Y. Echinacoside attenuates inflammatory response in a rat model of cervical spondylotic myelopathy via inhibition of excessive mitochondrial fission. Free Radic Biol Med 2020; 152:697-714. [PMID: 32014501 DOI: 10.1016/j.freeradbiomed.2020.01.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/27/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022]
Abstract
Cervical spondylotic myelopathy (CSM) is a leading cause of spinal cord dysfunction with few treatment options. Although mitochondrial dynamics are linked to a wide range of pathological changes in neurodegenerative diseases, a connection between aberrant mitochondrial dynamics and CSM remains to be illuminated. In addition, mechanisms underlying the emerging anti-inflammatory and neuroprotective effects of echinacoside (ECH), the main active ingredient of Cistanche salsa, are poorly understood. We hypothesized that excessive mitochondrial fission plays a critical role in regulating inflammatory responses in CSM, and ECH might alleviate such responses by regulating mitochondrial dynamics. To this end, we assessed the effects of ECH and Mdivi-1, a selective inhibitor of dynamin-related protein (Drp1), in a rat model of chronic cervical cord compression and activated BV2 cells. Our results showed that rats with Mdivi-1 intervention had improved motor function compared with vehicle-treated rats. Indeed, Mdivi-1 treatment attenuated pro-inflammatory cytokine expression, as well as activation of the nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, nuclear transcription factor-κB (NF-κB), and Drp1 in lesions. Compared with vehicle-treated rats, compression sites of Mdivi-1-treated animals exhibited elongated mitochondrial morphologies and reduced reactive oxygen species (ROS). Similarly, ECH-treated rats exhibited neurological recovery and suppression of inflammatory response or related signals in the lesion area after treatment. Interestingly, ECH treatment partly reversed aberrant mitochondrial fragmentation and oxidative stress within the lesion area. In vitro data suggested that ECH suppressed activated microglia by modulating activation of the NLRP3 inflammasome and NF-κB signaling. Furthermore, we observed that ECH markedly inhibited Drp1 translocation onto mitochondria, whereby it regulated mitochondrial dynamics and ROS production, which act as regulators of NLRP3 inflammasome activation and NF-κB signaling. Thus, our findings reveal that mitochondrial dynamics modulate inflammatory responses during CSM. Moreover, ECH may attenuate neuroinflammation in rats subjected to chronic cervical cord compression by regulating Drp1-dependent mitochondrial fission and activation of downstream signaling.
Collapse
Affiliation(s)
- Longyun Zhou
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Rehabilitation Medicine College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zirui Tian
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongjia Song
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yueli Sun
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jie Ye
- Department of Orthopedics and Traumatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Gan Li
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Kim Sia Sng
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Leqin Xu
- Xiamen Hospital of Traditional Chinese Medicine, Fujian, 361009, China
| | - Xuejun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yongjun Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
45
|
Bolus H, Crocker K, Boekhoff-Falk G, Chtarbanova S. Modeling Neurodegenerative Disorders in Drosophila melanogaster. Int J Mol Sci 2020; 21:E3055. [PMID: 32357532 PMCID: PMC7246467 DOI: 10.3390/ijms21093055] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Drosophila melanogaster provides a powerful genetic model system in which to investigate the molecular mechanisms underlying neurodegenerative diseases. In this review, we discuss recent progress in Drosophila modeling Alzheimer's Disease, Parkinson's Disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's Disease, Ataxia Telangiectasia, and neurodegeneration related to mitochondrial dysfunction or traumatic brain injury. We close by discussing recent progress using Drosophila models of neural regeneration and how these are likely to provide critical insights into future treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Harris Bolus
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Kassi Crocker
- Genetics Graduate Training Program, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA;
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Grace Boekhoff-Falk
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | | |
Collapse
|
46
|
Venkatesh K, Mathew A, Koushika SP. Role of actin in organelle trafficking in neurons. Cytoskeleton (Hoboken) 2020; 77:97-109. [DOI: 10.1002/cm.21580] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/14/2019] [Accepted: 11/05/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Keertana Venkatesh
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| | - Amal Mathew
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| | - Sandhya P. Koushika
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| |
Collapse
|
47
|
Bertogliat MJ, Morris-Blanco KC, Vemuganti R. Epigenetic mechanisms of neurodegenerative diseases and acute brain injury. Neurochem Int 2020; 133:104642. [PMID: 31838024 PMCID: PMC8074401 DOI: 10.1016/j.neuint.2019.104642] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/25/2019] [Accepted: 12/09/2019] [Indexed: 12/22/2022]
Abstract
Epigenetic modifications are emerging as major players in the pathogenesis of neurodegenerative disorders and susceptibility to acute brain injury. DNA and histone modifications act together with non-coding RNAs to form a complex gene expression machinery that adapts the brain to environmental stressors and injury response. These modifications influence cell-level operations like neurogenesis and DNA repair to large, intricate processes such as brain patterning, memory formation, motor function and cognition. Thus, epigenetic imbalance has been shown to influence the progression of many neurological disorders independent of aberrations in the genetic code. This review aims to highlight ways in which epigenetics applies to several commonly researched neurodegenerative diseases and forms of acute brain injury as well as shed light on the benefits of epigenetics-based treatments.
Collapse
Affiliation(s)
- Mario J Bertogliat
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Kahlilia C Morris-Blanco
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
48
|
Geng J, Liu W, Gao J, Jiang C, Fan T, Sun Y, Qin Z, Xu Q, Guo W, Gao J. Andrographolide alleviates Parkinsonism in MPTP-PD mice via targeting mitochondrial fission mediated by dynamin-related protein 1. Br J Pharmacol 2019; 176:4574-4591. [PMID: 31389613 PMCID: PMC6932945 DOI: 10.1111/bph.14823] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 07/17/2019] [Accepted: 07/20/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Accumulating evidence indicates that mitochondrial dynamics play an important role in the progressive deterioration of dopaminergic neurons. Andrographolide has been found to exert neuroprotective effects in several models of neurological diseases. However, the mechanism of how andrographolide protects neurons in Parkinson's disease (PD) remains not fully understood. EXPERIMENTAL APPROACH Behavioural experiments were performed to examine the effect of andrographolide in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-PD mice. Mitochondrial mass and morphology were visualized using transmission electron microscopy (TEM). SH-SY5Y cells and primary mouse neurons were exposed to rotenone to mimic PD in vitro. Western blotting, co-immunoprecipitation and immunofluorescence were performed. The target protein of andrographolide was identified by biotin-andrographolide pulldown assay as well as drug affinity responsive target stability (DARTS), cellular thermal shift (CETSA), and surface plasmon resonance (SPR) assays. KEY RESULTS Andrographolide administration improved behavioural deficits and attenuated loss of dopaminergic neurons in MPTP-exposed mice and reduced cell death induced by rotenone in vitro. An increased mitochondrial mass, and decreased surface area were found in the striatum from MPTP-PD mice, as well as in rotenone-treated primary neurons and SH-SY5Y cells, while andrographolide treatment preserved mitochondrial mass and morphology. Dynamin-related protein 1 (DRP1) was identified as a target protein of andrographolide. Andrographolide bound to DRP1 and inhibited its GTPase activity, thereby preventing excessive mitochondria fission and neuronal damage in PD. CONCLUSIONS AND IMPLICATIONS Our findings suggest that andrographolide may protect neurons against rotenone- or MPTP-induced damage in vitro and in vivo through inhibiting mitochondrial fission.
Collapse
Affiliation(s)
- Ji Geng
- School of PharmacyJiangsu UniversityZhenjiangChina
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro‐Psycho‐Diseases, College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Chunhong Jiang
- State Key Laboratory of Innovative Nature Medicine and TCM InjectionsJiangxi Qingfeng Pharmaceutical Co., Ltd.GanzhouChina
| | - Ting Fan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Zheng‐Hong Qin
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro‐Psycho‐Diseases, College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Jing Gao
- School of PharmacyJiangsu UniversityZhenjiangChina
| |
Collapse
|