1
|
Flores-Ocampo V, Lim AWY, Ogonowski NS, García-Marín LM, Ong JS, Yeow D, Gonzaga-Jauregui C, Kumar KR, Rentería ME. Population-Specific Differences in Pathogenic Variants of Genes Associated with Monogenic Parkinson's Disease. Genes (Basel) 2025; 16:454. [PMID: 40282414 PMCID: PMC12027003 DOI: 10.3390/genes16040454] [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: 03/08/2025] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a genetically complex neurodegenerative disorder. Up to 15% of cases are considered monogenic. However, research on monogenic PD has largely focused on populations of European ancestry, leaving gaps in our understanding of genetic variability in other populations. This study addresses this gap by analysing the allele frequencies of pathogenic and likely pathogenic variants in known monogenic PD genes across eight global populations, using data from the gnomAD database. METHODS We compiled a list of 27 genes associated with Mendelian PD from the Online Mendelian Inheritance in Man (OMIM) database, and identified pathogenic and likely pathogenic variants using ClinVar. We then performed pairwise comparisons of allele frequencies across populations included in the gnomAD database. Variants with significant frequency differences were further assessed using in silico pathogenicity predictions. RESULTS We identified 81 variants across 17 genes with statistically significant allele frequency differences between at least two populations. Variants in GBA1 were the most prevalent among monogenic PD-related genes, followed by PLA2G6, ATP13A2, VPS13C, and PRKN. GBA1 exhibited the greatest variability in allele frequencies, particularly the NM_000157.4:c.1226A>G (p.Asn409Ser) variant. Additionally, we observed significant population-specific differences in PD-related variants, such as the NM_032409.3:c.1040T>C (p.Leu347Pro) variant in PINK1, which was most prevalent in East Asian populations. CONCLUSIONS Our findings reveal substantial population-specific differences in the allele frequencies of pathogenic variants linked to monogenic PD, emphasising the need for broader genetic studies beyond European populations. These insights have important implications for PD research, genetic screening, and understanding the pathogenesis of PD in diverse populations.
Collapse
Affiliation(s)
- Victor Flores-Ocampo
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.F.-O.); (A.W.-Y.L.); (N.S.O.); (L.M.G.-M.)
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla 76230, Querétaro, Mexico;
| | - Amanda Wei-Yin Lim
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.F.-O.); (A.W.-Y.L.); (N.S.O.); (L.M.G.-M.)
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
| | - Natalia S. Ogonowski
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.F.-O.); (A.W.-Y.L.); (N.S.O.); (L.M.G.-M.)
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Luis M. García-Marín
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.F.-O.); (A.W.-Y.L.); (N.S.O.); (L.M.G.-M.)
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jue-Sheng Ong
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
| | - Dennis Yeow
- Molecular Medicine Laboratory, Concord Repatriation General Hospital, Concord, NSW 2139, Australia or (D.Y.); (K.R.K.)
- Neurology Department, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia
- Translational Neurogenomics Group, Genomics and Inherited Disease Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Claudia Gonzaga-Jauregui
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla 76230, Querétaro, Mexico;
| | - Kishore R. Kumar
- Molecular Medicine Laboratory, Concord Repatriation General Hospital, Concord, NSW 2139, Australia or (D.Y.); (K.R.K.)
- Neurology Department, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia
- Translational Neurogenomics Group, Genomics and Inherited Disease Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2033, Australia
| | - Miguel E. Rentería
- Brain & Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.F.-O.); (A.W.-Y.L.); (N.S.O.); (L.M.G.-M.)
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
2
|
Nuermaimaiti M, Ishikawa KI, Oyama G, Nonaka R, Shiga T, Jo T, Tsunemi T, Nakamura R, Krüger R, Akamatsu W, Hattori N. Human induced pluripotent stem cell-derived dopaminergic neurons release alpha-synuclein through neuronal activity. Neurosci Res 2025; 212:105-114. [PMID: 39617169 DOI: 10.1016/j.neures.2024.11.007] [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: 07/23/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024]
Abstract
Lewy body diseases, including Parkinson's disease (PD), are characterized by the spread of alpha-synuclein (αSyn) between neurons across synapses, a process crucial for understanding their pathophysiology and developing effective treatments. In this study, we aimed to investigate the role of neuronal activity in releasing αSyn from human induced pluripotent stem cell-derived dopaminergic neurons. We examined human induced pluripotent stem cell-derived dopaminergic neurons, both healthy and those with the αSyn gene mutation associated with PD. We employed pharmacological agents and optogenetic techniques and demonstrated that increased neuronal activity, induced by bicuculline or optogenetic stimulation, significantly enhances αSyn release. However, suppression of neuronal activity with cyanquixaline reduces αSyn secretion. These findings underscore the pivotal role of neuronal activity in αSyn transmission between neurons, showing its potential impact on the spread of Lewy pathology in patients with neurodegenerative diseases like PD. Therefore, this study advances our understanding of PD and opens new avenues for therapeutic strategies to mitigate Lewy body disease progression.
Collapse
Affiliation(s)
- Maierdanjiang Nuermaimaiti
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Clinical Data of Parkinson's Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kei-Ichi Ishikawa
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan.
| | - Genko Oyama
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Risa Nonaka
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Laboratory of Molecular Biology, Faculty of Pharmacy, Juntendo University, Chiba, Japan
| | - Takahiro Shiga
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takayuki Jo
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Taiji Tsunemi
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Ryota Nakamura
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Clinical Data of Parkinson's Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
3
|
Ma Q, Tian JL, Lou Y, Guo R, Ma XR, Wu JB, Yang J, Tang BJ, Li S, Qiu M, Duan S, Zhao JW, Zhang J, Xu ZZ. Oligodendrocytes drive neuroinflammation and neurodegeneration in Parkinson's disease via the prosaposin-GPR37-IL-6 axis. Cell Rep 2025; 44:115266. [PMID: 39913287 DOI: 10.1016/j.celrep.2025.115266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease and is difficult to treat due to its elusive mechanisms. Recent studies have identified a striking association between oligodendrocytes and PD progression, yet how oligodendrocytes regulate the pathogenesis of PD is still unknown. Here, we show that G-protein-coupled receptor 37 (GPR37) is upregulated in oligodendrocytes of the substantia nigra and that prosaposin (PSAP) secretion is increased in parkinsonian mice. The released PSAP can induce interleukin (IL)-6 upregulation and secretion from oligodendrocytes via a GPR37-dependent pathway, resulting in enhanced neuroinflammation, dopamine neuron degeneration, and behavioral deficits. GPR37 deficiency in oligodendrocytes prevents neurodegeneration in multiple PD models. Finally, the hallmarks of the PSAP-GPR37-IL-6 axis are observed in patients with PD. Thus, our results reveal that dopaminergic neurons interact with oligodendrocytes via secreted PSAP, and our findings identify the PSAP-GPR37-IL-6 axis as a driver of PD pathogenesis and a potential therapeutic target that might alleviate PD progression in patients.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China.
| | - Jin-Lan Tian
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Yao Lou
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Ran Guo
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jian-Bin Wu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Yang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Bing-Jie Tang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Shumin Duan
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Zhang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Department of Pathology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China; National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou 310002, China
| | - Zhen-Zhong Xu
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China.
| |
Collapse
|
4
|
Yan Y, Cao L, Gu L, Xu C, Lu J, Lv D, Tian J, Yin X, Pu J, Zhang B, Zhao G. Analysis of Common Genetic Variation of Anxiety Disorders in Essential Tremor. J Mol Neurosci 2025; 75:14. [PMID: 39890685 DOI: 10.1007/s12031-024-02226-2] [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: 01/27/2024] [Accepted: 04/15/2024] [Indexed: 02/03/2025]
Abstract
The objective of this study is to explore the association of common genetic variation of anxiety disorders and essential tremor (ET). We genotyped 25 anxiety-specific risk variants in a cohort of 478 unrelated ET patients and 504 age and gender-matched healthy controls from eastern China using a MassARRAY system. The association between candidate variants and ET patients was evaluated using gene-based analysis. A total of 159 patients (33.3%) had anxiety. In genotypic analysis, rs708012 (in an intergenic region) in the dominant models was found to be significantly associated with ET (P < 0.001, OR = 0.605). In allelic analysis, the carriers of the C allele of NTRK2 rs1187280 (P = 0.027, OR = 0.626), T allele of TMEM106B rs3807866 (P = 0.030, OR = 1.287), and T allele of rs708012 (P < 0.001, OR = 0.679) occupy a larger proportion of ET patients compared with healthy controls. Anxiety-specific risk SNPs of TMEM106B rs3807866 increase the risk for ET, while two SNPs of NTRK2 rs1187280 and rs708012 show a protective role.
Collapse
Affiliation(s)
- Yaping Yan
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Lanxiao Cao
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang, China
| | - Luyan Gu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Congying Xu
- Department of Neurology, Jiaxing Second Hospital, Jiaxing, China
| | - Jinyu Lu
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang, China
| | - Dayao Lv
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang, China
| | - Jun Tian
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xinzhen Yin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Guohua Zhao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
5
|
Arya R, Haque AKMA, Shakya H, Billah MM, Parvin A, Rahman MM, Sakib KM, Faruquee HM, Kumar V, Kim JJ. Parkinson's Disease: Biomarkers for Diagnosis and Disease Progression. Int J Mol Sci 2024; 25:12379. [PMID: 39596444 PMCID: PMC11594627 DOI: 10.3390/ijms252212379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disease that causes both motor and nonmotor symptoms. While our understanding of putative mechanisms has advanced significantly, it remains challenging to verify biomarkers with sufficient evidence for regular clinical use. Clinical symptoms are the primary basis for diagnosing the disease, which can be mild in the early stages and overlap with other neurological disorders. As a result, clinical testing and medical records are mostly relied upon for diagnosis, posing substantial challenges during both the initial diagnosis and the continuous disease monitoring. Recent biochemical, neuroimaging, and genetic biomarkers have helped us understand the pathophysiology of Parkinson's disease. This comprehensive study focuses on these biomarkers, which were chosen based on their relevance, methodological excellence, and contribution to the field. Biochemical biomarkers, including α-synuclein and glial fibrillary acidic protein (GFAP), can predict disease severity and progression. The dopaminergic system is widely used as a neuroimaging biomarker to diagnose PD. Numerous genes and genome wide association study (GWAS) sites have been related to the development of PD. Recent research on the SNCA gene and leucine-rich repeat protein kinase 2 (LRRK2) has shown promising results. By evaluating current studies, this review intends to uncover gaps in biomarker validation and use, while also highlighting promising improvements. It emphasizes the need for dependable and reproducible indicators in improving PD diagnosis and prognosis. These biomarkers may open up new avenues for early diagnosis, disease progression tracking, and the development of personalized treatment programs.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - A. K. M. Ariful Haque
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh; (A.K.M.A.H.); (M.M.B.); (A.P.); (M.-M.R.); (H.M.F.)
| | - Hemlata Shakya
- Department of Biomedical Engineering, Shri G. S. Institute of Technology and Science, Indore 452003, India;
| | - Md. Masum Billah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh; (A.K.M.A.H.); (M.M.B.); (A.P.); (M.-M.R.); (H.M.F.)
| | - Anzana Parvin
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh; (A.K.M.A.H.); (M.M.B.); (A.P.); (M.-M.R.); (H.M.F.)
| | - Md-Mafizur Rahman
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh; (A.K.M.A.H.); (M.M.B.); (A.P.); (M.-M.R.); (H.M.F.)
| | - Khan Mohammad Sakib
- Department of Biology, Adamjee Cantonment College, Dhaka Cantonment, Dhaka 1206, Bangladesh;
| | - Hossain Md. Faruquee
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh; (A.K.M.A.H.); (M.M.B.); (A.P.); (M.-M.R.); (H.M.F.)
| | - Vijay Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
6
|
Qin F, Zhang M, Wang P, Dai Z, Li X, Li D, Jing L, Qi C, Fan H, Qin M, Li Y, Huang L, Wang T. Transcriptome analysis reveals the anti-Parkinson's activity of Mangiferin in zebrafish. Biomed Pharmacother 2024; 179:117387. [PMID: 39245002 DOI: 10.1016/j.biopha.2024.117387] [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: 05/24/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
As the global population ages, the incidence of Parkinson's Disease (PD) continues to rise, imposing significant social and economic burdens. Mangiferin (MGF), a polyphenolic, bioactive compound has been shown to play a role in the prevention and treatment of PD. This study investigates the neuroprotective effects of MGF in an MPTP-induced zebrafish model of PD through transcriptome analysis. Initially, optimal concentrations for modeling were determined using various MPTP and MGF combinations. The zebrafish were then divided into control, MPTP-treated, and MGF co-treated groups. Subsequent evaluations included hatching rates, mortality rates, growth and development conditions, spontaneous motor abilities, as well as measurements of enzymatic activities of SOD, CAT, and levels of GSH. Ultimately, the therapeutic efficacy of MGF on the PD model in zebrafish was assessed through transcriptome sequencing. The results demonstrated that MPTP treatment induced PD-associated symptoms in zebrafish, while MGF treatment significantly improved the motor abilities and survival rates of the PD model zebrafish, effectively reducing oxidative stress and ameliorating PD symptoms. Transcriptome sequencing further revealed that MGF may mitigate mitochondrial-related oxidative stress in PD zebrafish by modulating the expression of critical genes including lrrk2, vps35, atp13a, dnajc6, and uchl1. Differential gene expression analysis indicated that these genes are primarily involved in vital signaling pathways, such as neuroactive ligand-receptor interaction, and the calcium signaling pathway. In summary, our study provides robust scientific evidence supporting MGF as a potential therapeutic candidate for PD by preserving mitochondrial homeostasis and elucidating its mechanisms of action.
Collapse
Affiliation(s)
- Fengqing Qin
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China; College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China.
| | - Ming Zhang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Pei Wang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Ziru Dai
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Xi Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.
| | - Dongliang Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Lijun Jing
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Cen Qi
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Heliang Fan
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Mei Qin
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Ying Li
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Likun Huang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Tianci Wang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| |
Collapse
|
7
|
Date Y, Sasazawa Y, Kitagawa M, Gejima K, Suzuki A, Saya H, Kida Y, Imoto M, Itakura E, Hattori N, Saiki S. Novel autophagy inducers by accelerating lysosomal clustering against Parkinson's disease. eLife 2024; 13:e98649. [PMID: 38899618 PMCID: PMC11221835 DOI: 10.7554/elife.98649] [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: 04/16/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
The autophagy-lysosome pathway plays an indispensable role in the protein quality control by degrading abnormal organelles and proteins including α-synuclein (αSyn) associated with the pathogenesis of Parkinson's disease (PD). However, the activation of this pathway is mainly by targeting lysosomal enzymic activity. Here, we focused on the autophagosome-lysosome fusion process around the microtubule-organizing center (MTOC) regulated by lysosomal positioning. Through high-throughput chemical screening, we identified 6 out of 1200 clinically approved drugs enabling the lysosomes to accumulate around the MTOC with autophagy flux enhancement. We further demonstrated that these compounds induce the lysosomal clustering through a JIP4-TRPML1-dependent mechanism. Among them, the lysosomal-clustering compound albendazole promoted the autophagy-dependent degradation of Triton-X-insoluble, proteasome inhibitor-induced aggregates. In a cellular PD model, albendazole boosted insoluble αSyn degradation. Our results revealed that lysosomal clustering can facilitate the breakdown of protein aggregates, suggesting that lysosome-clustering compounds may offer a promising therapeutic strategy against neurodegenerative diseases characterized by the presence of aggregate-prone proteins.
Collapse
Affiliation(s)
- Yuki Date
- Department of Biology, Graduate School of Science and Engineering, Chiba University, Inage-kuChibaJapan
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
| | - Yukiko Sasazawa
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of MedicineTokyoJapan
- Division for Development of Autophagy Modulating Drugs, Juntendo University Faculty of MedicineTokyoJapan
| | - Mitsuhiro Kitagawa
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
| | - Kentaro Gejima
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
| | - Ayami Suzuki
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio UniversityTokyoJapan
- Division of Gene Regulation, Cancer Center, Fujita Health UniversityToyoakeJapan
| | - Yasuyuki Kida
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Masaya Imoto
- Division for Development of Autophagy Modulating Drugs, Juntendo University Faculty of MedicineTokyoJapan
| | - Eisuke Itakura
- Department of Biology, Graduate School of Science, Chiba University, Inage-kuChibaJapan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of MedicineTokyoJapan
- Division for Development of Autophagy Modulating Drugs, Juntendo University Faculty of MedicineTokyoJapan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain ScienceSaitamaJapan
| | - Shinji Saiki
- Department of Neurology, Juntendo University Faculty of MedicineTokyoJapan
- Division for Development of Autophagy Modulating Drugs, Juntendo University Faculty of MedicineTokyoJapan
- Department of Neurology, Institute of Medicine, University of TsukubaIbarakiJapan
| |
Collapse
|
8
|
Skrahin A, Horowitz M, Istaiti M, Skrahina V, Lukas J, Yahalom G, Cohen ME, Revel-Vilk S, Goker-Alpan O, Becker-Cohen M, Hassin-Baer S, Svenningsson P, Rolfs A, Zimran A. GBA1-Associated Parkinson's Disease Is a Distinct Entity. Int J Mol Sci 2024; 25:7102. [PMID: 39000225 PMCID: PMC11241486 DOI: 10.3390/ijms25137102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
GBA1-associated Parkinson's disease (GBA1-PD) is increasingly recognized as a distinct entity within the spectrum of parkinsonian disorders. This review explores the unique pathophysiological features, clinical progression, and genetic underpinnings that differentiate GBA1-PD from idiopathic Parkinson's disease (iPD). GBA1-PD typically presents with earlier onset and more rapid progression, with a poor response to standard PD medications. It is marked by pronounced cognitive impairment and a higher burden of non-motor symptoms compared to iPD. Additionally, patients with GBA1-PD often exhibit a broader distribution of Lewy bodies within the brain, accentuating neurodegenerative processes. The pathogenesis of GBA1-PD is closely associated with mutations in the GBA1 gene, which encodes the lysosomal enzyme beta-glucocerebrosidase (GCase). In this review, we discuss two mechanisms by which GBA1 mutations contribute to disease development: 'haploinsufficiency,' where a single functional gene copy fails to produce a sufficient amount of GCase, and 'gain of function,' where the mutated GCase acquires harmful properties that directly impact cellular mechanisms for alpha-synuclein degradation, leading to alpha-synuclein aggregation and neuronal cell damage. Continued research is advancing our understanding of how these mechanisms contribute to the development and progression of GBA1-PD, with the 'gain of function' mechanism appearing to be the most plausible. This review also explores the implications of GBA1 mutations for therapeutic strategies, highlighting the need for early diagnosis and targeted interventions. Currently, small molecular chaperones have shown the most promising clinical results compared to other agents. This synthesis of clinical, pathological, and molecular aspects underscores the assertion that GBA1-PD is a distinct clinical and pathobiological PD phenotype, necessitating specific management and research approaches to better understand and treat this debilitating condition.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, 6997801 Ramat Aviv, Israel
| | - Majdolen Istaiti
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
| | | | - Jan Lukas
- Translational Neurodegeneration Section Albrecht Kossel, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Gilad Yahalom
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Mikhal E. Cohen
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA
| | | | - Sharon Hassin-Baer
- Movement Disorders Institute, Department of Neurology, Chaim Sheba Medical Center, 5262101 Tel-Hashomer, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel-Aviv, Israel
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Basal and Clinical Neuroscience, King’s College London, London SE5 9RT, UK
| | - Arndt Rolfs
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Medical Faculty, University of Rostock, 18055 Rostock, Germany
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| |
Collapse
|
9
|
Sanluca C, Spagnolo P, Mancinelli R, De Bartolo MI, Fava M, Maccarrone M, Carotti S, Gaudio E, Leuti A, Vivacqua G. Interaction between α-Synuclein and Bioactive Lipids: Neurodegeneration, Disease Biomarkers and Emerging Therapies. Metabolites 2024; 14:352. [PMID: 39057675 PMCID: PMC11278689 DOI: 10.3390/metabo14070352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
The present review provides a comprehensive examination of the intricate dynamics between α-synuclein, a protein crucially involved in the pathogenesis of several neurodegenerative diseases, including Parkinson's disease and multiple system atrophy, and endogenously-produced bioactive lipids, which play a pivotal role in neuroinflammation and neurodegeneration. The interaction of α-synuclein with bioactive lipids is emerging as a critical factor in the development and progression of neurodegenerative and neuroinflammatory diseases, offering new insights into disease mechanisms and novel perspectives in the identification of potential biomarkers and therapeutic targets. We delve into the molecular pathways through which α-synuclein interacts with biological membranes and bioactive lipids, influencing the aggregation of α-synuclein and triggering neuroinflammatory responses, highlighting the potential of bioactive lipids as biomarkers for early disease detection and progression monitoring. Moreover, we explore innovative therapeutic strategies aimed at modulating the interaction between α-synuclein and bioactive lipids, including the development of small molecules and nutritional interventions. Finally, the review addresses the significance of the gut-to-brain axis in mediating the effects of bioactive lipids on α-synuclein pathology and discusses the role of altered gut lipid metabolism and microbiota composition in neuroinflammation and neurodegeneration. The present review aims to underscore the potential of targeting α-synuclein-lipid interactions as a multifaceted approach for the detection and treatment of neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Chiara Sanluca
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Paolo Spagnolo
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Romina Mancinelli
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | | | - Marina Fava
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Simone Carotti
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| | - Eugenio Gaudio
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | - Alessandro Leuti
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Giorgio Vivacqua
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| |
Collapse
|
10
|
Hattori N, Funayama M, Imai Y, Hatano T. Pathogenesis of Parkinson's disease: from hints from monogenic familial PD to biomarkers. J Neural Transm (Vienna) 2024; 131:709-719. [PMID: 38478097 DOI: 10.1007/s00702-024-02747-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 06/22/2024]
Abstract
Twenty-five years have passed since the causative gene for familial Parkinson's disease (PD), Parkin (now PRKN), was identified in 1998; PRKN is the most common causative gene in young-onset PD. Parkin encodes a ubiquitin-protein ligase, and Parkin is involved in mitophagy, a type of macroautophagy, in concert with PTEN-induced kinase 1 (PINK1). Both gene products are also involved in mitochondrial quality control. Among the many genetic PD-causing genes discovered, discovering PRKN as a cause of juvenile-onset PD has significantly impacted other neurodegenerative disorders. This is because the involvement of proteolytic systems has been suggested as a common mechanism in neurodegenerative diseases in which inclusion body formation is observed. The discovery of the participation of PRKN in PD has brought attention to the involvement of the proteolytic system in neurodegenerative diseases. Our research group has successfully isolated and identified CHCHD2, which is involved in the mitochondrial electron transfer system, and prosaposin (PSAP), which is involved in the lysosomal system, in this Parkin mechanism. Hereditary PD is undoubtedly an essential clue to solitary PD, and at least 25 or so genes and loci have been reported so far. This number of genes indicates that PD is a very diverse group of diseases. Currently, the diagnosis of PD is based on clinical symptoms and imaging studies. Although highly accurate diagnostic criteria have been published, early diagnosis is becoming increasingly important in treatment strategies for neurodegenerative diseases. Here, we also describe biomarkers that our group is working on.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan.
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan.
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1-Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
| | - Manabu Funayama
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Taku Hatano
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
11
|
Gathings A, Zaman V, Banik NL, Haque A. Insights into Calpain Activation and Rho-ROCK Signaling in Parkinson's Disease and Aging. Biomedicines 2024; 12:1074. [PMID: 38791036 PMCID: PMC11117523 DOI: 10.3390/biomedicines12051074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disease, has no cure, and current therapies are not effective at halting disease progression. The disease affects mid-brain dopaminergic neurons and, subsequently, the spinal cord, contributing to many debilitating symptoms associated with PD. The GTP-binding protein, Rho, plays a significant role in the cellular pathology of PD. The downstream effector of Rho, Rho-associated kinase (ROCK), plays multiple functions, including microglial activation and induction of inflammatory responses. Activated microglia have been implicated in the pathology of many neurodegenerative diseases, including PD, that initiate inflammatory responses, leading to neuron death. Calpain expression and activity is increased following glial activation, which triggers the Rho-ROCK pathway and induces inflammatory T cell activation and migration as well as mediates toxic α-synuclein (α-syn) aggregation and neuron death, indicating a pivotal role for calpain in the inflammatory and degenerative processes in PD. Increased calpain activity and Rho-ROCK activation may represent a new mechanism for increased oxidative damage in aging. This review will summarize calpain activation and the role of the Rho-ROCK pathway in oxidative stress and α-syn aggregation, their influence on the neurodegenerative process in PD and aging, and possible strategies and research directions for therapeutic intervention.
Collapse
Affiliation(s)
- Amy Gathings
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (A.G.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA
| | - Narendra L. Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (A.G.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (A.G.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA
| |
Collapse
|
12
|
Zhu L, Zhang X, Guan Y, Zhu Y, Zhou Q, Liu B, Ren H, Yang X. Meta-analysis of the association of prosaposin polymorphisms rs4747203 and rs885828 with risk of Parkinson's disease. Acta Neurol Belg 2024; 124:573-580. [PMID: 38206457 DOI: 10.1007/s13760-023-02446-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/27/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Previous research has established a connection between polymorphisms rs4747203 and rs885828 in the prosaposin (PSAP) gene and an increased risk of Parkinson's disease (PD). However, other studies have found no significant difference in risk compared to the general population. METHODS To evaluate the current evidence linking rs4747203 and rs885828 to PD risk, we conducted a comprehensive search of PubMed, the Web of Science, Embase, and the Cochrane Library for relevant studies up until May 2023. In addition, we analyzed data from the publicly available "PD Variant Browser". We performed a meta-analysis using Stata 17.0 to synthesize the findings from the selected studies. RESULTS Our meta-analysis, which included data from six published studies and the public database, revealed no significant association between PD risk and either rs4747203 [OR (95% CI) = 0.99 (0.93-1.05), I2 = 90.3%, P = 0.635] or rs885828 [OR (95% CI) = 1.01 (0.95-1.07), I2 = 90.7%, P = 0.773]. These results remained consistent when examining subgroups of individuals within or outside of Asia. CONCLUSION The available evidence does not support an association between the genotype at rs4747203 or rs885828 and the risk of PD.
Collapse
Affiliation(s)
- Liuhui Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
- Joint Institute of Smoking and Health, Kunming, 650106, Yunnan, China
| | - Xinyue Zhang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Ying Guan
- Joint Institute of Smoking and Health, Kunming, 650106, Yunnan, China
| | - Yongyun Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Qian Zhou
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Bin Liu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Xinglong Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China.
- Joint Institute of Smoking and Health, Kunming, 650106, Yunnan, China.
| |
Collapse
|
13
|
Vlachakis D, Tsilafakis K, Kostavasili I, Kossida S, Mavroidis M. Unraveling Desmin's Head Domain Structure and Function. Cells 2024; 13:603. [PMID: 38607042 PMCID: PMC11012097 DOI: 10.3390/cells13070603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Understanding the structure and function of intermediate filaments (IFs) is necessary in order to explain why more than 70 related IF genes have evolved in vertebrates while maintaining such dramatically tissue-specific expression. Desmin is a member of the large multigene family of IF proteins and is specifically expressed in myocytes. In an effort to elucidate its muscle-specific behavior, we have used a yeast two-hybrid system in order to identify desmin's head binding partners. We described a mitochondrial and a lysosomal protein, NADH ubiquinone oxidoreductase core subunit S2 (NDUFS2), and saposin D, respectively, as direct desmin binding partners. In silico analysis indicated that both interactions at the atomic level occur in a very similar way, by the formation of a three-helix bundle with hydrophobic interactions in the interdomain space and hydrogen bonds at R16 and S32 of the desmin head domain. The interactions, confirmed also by GST pull-down assays, indicating the necessity of the desmin head domain and, furthermore, point out its role in function of mitochondria and lysosomes, organelles which are disrupted in myopathies due to desmin head domain mutations.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Biotechnology Department, Agricultural University of Athens, 11855 Athens, Greece;
| | - Konstantinos Tsilafakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
- Biochemistry & Biotechnology Department, University of Thessaly, 41500 Larisa, Greece
| | - Ioanna Kostavasili
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| | - Sophia Kossida
- IMGT, The International ImMunoGeneTics Information System, National Center for Scientific Research (CNRS), Institute of Human Genetics (IGH), University of Montpellier (UM), 34090 Montpellier, France;
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| |
Collapse
|
14
|
Kuo MC, Chu YT, Su YA, Chen ML, Wu RM. Prosaposin variants in sporadic, familial, and early-onset Parkinson's disease: a Taiwanese case-control study and meta-analysis. Sci Rep 2024; 14:2225. [PMID: 38278831 PMCID: PMC10817969 DOI: 10.1038/s41598-024-51646-y] [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: 08/07/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
Polymorphisms in the PSAP gene, which encodes prosaposin and is involved in the lysosomal function, yielded conflicting results regarding the association with Parkinson's disease (PD). Therefore, this study aims to investigate the role of PSAP in familial PD (FPD), early onset PD (EOPD) with age at onset before 50 years old, and sporadic PD (SPD) among Taiwanese population, and summarize relevant studies via meta-analysis. By sequencing exon 1 to 14 in 183 FPD and 219 EOPD, two novel exonic variants were found in EOPD, including p.A146E (c.437C > A) on exon 5 and p.Y248C (c.743A > G) on exon 7. Furthermore, four previously reported intronic variants (rs142614739/rs74733861), rs749823, rs4747203 and rs885828) in intron 11 and 12 were analyzed in 485 SPD and 712 in-hospital controls, in addition to the aforementioned FPD and EOPD groups. The adjusted odd ratios (ORs) by age and sex, only rs142614739 was significantly associated with higher risk of EOPD (OR = 1.85, 95% CI = 1.33-2.58). The risk effect was further confirmed by the meta-analysis of the association between rs142614739 and the risk of PD in both common effect (OR = 1.29, 95% CI = 1.11-1.50) and random effect (OR = 1.29, 95% CI = 1.11-1.50). Our findings suggest that the PSAP rs142614739 variant is associated with the risk of EOPD. Further functional studies are warranted to elucidate the biochemical mechanisms.
Collapse
Affiliation(s)
- Ming-Che Kuo
- Department of Neurology, Centre for Parkinson and Movement Disorder, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Tsai Chu
- Department of Neurology, Centre for Parkinson and Movement Disorder, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Jinshan Branch, New Taipei City, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Yu-An Su
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Ling Chen
- Department of Neurology, Centre for Parkinson and Movement Disorder, National Taiwan University Hospital, Taipei, Taiwan
| | - Ruey-Meei Wu
- Department of Neurology, Centre for Parkinson and Movement Disorder, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Neurology, College of Medicine, National Taiwan University, No.7, Chung Shan South. Road, Taipei, 10002, Taiwan.
| |
Collapse
|
15
|
Mackie PM, Koshy J, Bhogade M, Hammoor T, Hachmeister W, Lloyd GM, Paterno G, Bolen M, Tansey MG, Giasson BI, Khoshbouei H. Complement C1q-dependent engulfment of alpha-synuclein induces ENS-resident macrophage exhaustion and accelerates Parkinson's-like gut pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563832. [PMID: 37961460 PMCID: PMC10634831 DOI: 10.1101/2023.10.24.563832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Deposition of misfolded α-synuclein (αsyn) in the enteric nervous system (ENS) is found in multiple neurodegenerative diseases. It is hypothesized that ENS synucleinopathy contributes to both the pathogenesis and non-motor morbidity in Parkinson's Disease (PD), but the cellular and molecular mechanisms that shape enteric histopathology and dysfunction are poorly understood. Here, we demonstrate that ENS-resident macrophages, which play a critical role in maintaining ENS homeostasis, initially respond to enteric neuronal αsyn pathology by upregulating machinery for complement-mediated engulfment. Pharmacologic depletion of ENS-macrophages or genetic deletion of C1q enhanced enteric neuropathology. Conversely, C1q deletion ameliorated gut dysfunction, indicating that complement partially mediates αsyn-induced gut dysfunction. Internalization of αsyn led to increased endo-lysosomal stress that resulted in macrophage exhaustion and temporally correlated with the progression of ENS pathology. These novel findings highlight the importance of enteric neuron-macrophage interactions in removing toxic protein aggregates that putatively shape the earliest stages of PD in the periphery.
Collapse
Affiliation(s)
- P M Mackie
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - J Koshy
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M Bhogade
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - T Hammoor
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - W Hachmeister
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| | - G M Lloyd
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - G Paterno
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M Bolen
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - M G Tansey
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, Florida, 32610
| | - B I Giasson
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine. Gainesville, FL, 32610
| | - H Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine. Gainesville, FL, 32610
| |
Collapse
|
16
|
Rafique S, Murtaza MA, Hafiz I, Ameer K, Qayyum MMN, Yaqub S, Mohamed Ahmed IA. Investigation of the antimicrobial, antioxidant, hemolytic, and thrombolytic activities of Camellia sinensis, Thymus vulgaris, and Zanthoxylum armatum ethanolic and methanolic extracts. Food Sci Nutr 2023; 11:6303-6311. [PMID: 37823136 PMCID: PMC10563746 DOI: 10.1002/fsn3.3569] [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: 01/31/2023] [Revised: 05/17/2023] [Accepted: 06/28/2023] [Indexed: 10/13/2023] Open
Abstract
Camellia sinensis is rich in antioxidants such as polyphenols; Thymus vulgaris contains bioactive compounds (flavonoids, terpenoids, and tannins) and Zanthoxylum armatum is primarily composed of volatile oils, amides, alkaloids, flavonoids, lignan, and coumarin. The antibacterial, antifungal, biofilm inhibition, antioxidant, hemolytic, and thrombolytic activities of Camellia sinensis, Thymus vulgaris, and Zanthoxylum armatum ethanol and methanol extracts at different concentrations (30%, 50%, and 80%) were determined. The antioxidant activity and content were measured as free radical scavenging assay (DPPH), total flavonoid content (TFC), and total phenolic content (TPC). Furthermore, hemolytic and thrombolytic analysis was carried out to determine toxicity. In antimicrobial assays, 80% methanol thyme extract showed highest (15.31 mm) antibacterial activity against Bacillus subtilis, and 80% ethanol green tea extract showed optimal antibacterial activity against Staphylococcus aureus. Ethanol 30% green tea extract resulted in highest (26.61 mm) antifungal activity against Aspergillus niger. The maximum (54.73%) biofilm inhibition was resulted by methanol 50% thyme extract for Escherichia coli. In antioxidant activity and content, methanol 50% green tea extract had highest (80.82%) antioxidant activity, whereas, ethanol 80% green tea extract had maximum (1474.55 mg CE/g DW) TFC and methanol 80% green tea extract had maximum (593.05 mg GAE/g) TPC. In toxicological assays, methanol 30% green tea extract had highest (25.28%) thrombolytic activity, and ethanol 80% tejphal extract had maximum (18.24%) hemolytic activity. This study has highlighted the significant antimicrobial, antioxidant, hemolytic, and thrombolytic activities of Camellia sinensis, Thymus vulgaris, and Zanthoxylum armatum extracts that could be beneficial to treat various diseases (cancer, diabetes, and respiratory diseases) and may be utilized as functional ingredient in the preparation of functional foods and drinks.
Collapse
Affiliation(s)
- Sobia Rafique
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | - Mian Anjum Murtaza
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | - Iram Hafiz
- Institute of ChemistryUniversity of SargodhaSargodhaPakistan
| | - Kashif Ameer
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | | | - Shazia Yaqub
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
- Punjab Food AuthorityLahorePakistan
| | - Isam A. Mohamed Ahmed
- Department of Food Science and Nutrition, College of Food and Agricultural SciencesKing Saud UniversityRiyadhSaudi Arabia
- Faculty of Agriculture, Department of Food Science and TechnologyUniversity of KhartoumShambatSudan
| |
Collapse
|
17
|
He Y, Kaya I, Shariatgorji R, Lundkvist J, Wahlberg LU, Nilsson A, Mamula D, Kehr J, Zareba-Paslawska J, Biverstål H, Chergui K, Zhang X, Andren PE, Svenningsson P. Prosaposin maintains lipid homeostasis in dopamine neurons and counteracts experimental parkinsonism in rodents. Nat Commun 2023; 14:5804. [PMID: 37726325 PMCID: PMC10509278 DOI: 10.1038/s41467-023-41539-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
Prosaposin (PSAP) modulates glycosphingolipid metabolism and variants have been linked to Parkinson's disease (PD). Here, we find altered PSAP levels in the plasma, CSF and post-mortem brain of PD patients. Altered plasma and CSF PSAP levels correlate with PD-related motor impairments. Dopaminergic PSAP-deficient (cPSAPDAT) mice display hypolocomotion and depression/anxiety-like symptoms with mildly impaired dopaminergic neurotransmission, while serotonergic PSAP-deficient (cPSAPSERT) mice behave normally. Spatial lipidomics revealed an accumulation of highly unsaturated and shortened lipids and reduction of sphingolipids throughout the brains of cPSAPDAT mice. The overexpression of α-synuclein via AAV lead to more severe dopaminergic degeneration and higher p-Ser129 α-synuclein levels in cPSAPDAT mice compared to WT mice. Overexpression of PSAP via AAV and encapsulated cell biodelivery protected against 6-OHDA and α-synuclein toxicity in wild-type rodents. Thus, these findings suggest PSAP may maintain dopaminergic lipid homeostasis, which is dysregulated in PD, and counteract experimental parkinsonism.
Collapse
Affiliation(s)
- Yachao He
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ibrahim Kaya
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Johan Lundkvist
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
| | - Lars U Wahlberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Dejan Mamula
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Section of Pharmacological Neurochemistry, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Justyna Zareba-Paslawska
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Biverstål
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Karima Chergui
- Laboratory of Molecular Neurophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per E Andren
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
18
|
Nakamura R, Nonaka R, Oyama G, Jo T, Kamo H, Nuermaimaiti M, Akamatsu W, Ishikawa KI, Hattori N. A defined method for differentiating human iPSCs into midbrain dopaminergic progenitors that safely restore motor deficits in Parkinson's disease. Front Neurosci 2023; 17:1202027. [PMID: 37502682 PMCID: PMC10368972 DOI: 10.3389/fnins.2023.1202027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/20/2023] [Indexed: 07/29/2023] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative condition that primarily affects motor functions; it is caused by the loss of midbrain dopaminergic (mDA) neurons. The therapeutic effects of transplanting human-induced pluripotent stem cell (iPSC)-derived mDA neural progenitor cells in animal PD models are known and are being evaluated in an ongoing clinical trial. However, However, improvements in the safety and efficiency of differentiation-inducing methods are crucial for providing a larger scale of cell therapy studies. This study aimed to investigate the usefulness of dopaminergic progenitor cells derived from human iPSCs by our previously reported method, which promotes differentiation and neuronal maturation by treating iPSCs with three inhibitors at the start of induction. Methods Healthy subject-derived iPS cells were induced into mDA progenitor cells by the CTraS-mediated method we previously reported, and their proprieties and dopaminergic differentiation efficiency were examined in vitro. Then, the induced mDA progenitors were transplanted into 6-hydroxydopamine-lesioned PD model mice, and their efficacy in improving motor function, cell viability, and differentiation ability in vivo was evaluated for 16 weeks. Results Approximately ≥80% of cells induced by this method without sorting expressed mDA progenitor markers and differentiated primarily into A9 dopaminergic neurons in vitro. After transplantation in 6-hydroxydopamine-lesioned PD model mice, more than 90% of the engrafted cells differentiated into the lineage of mDA neurons, and approximately 15% developed into mature mDA neurons without tumour formation. The grafted PD model mice also demonstrated significantly improved motor functions. Conclusion This study suggests that the differentiation protocol for the preparation of mDA progenitors is a promising option for cell therapy in patients with PD.
Collapse
Affiliation(s)
- Ryota Nakamura
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Risa Nonaka
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Clinical Data of Parkinson’s Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Genko Oyama
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Takayuki Jo
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Hikaru Kamo
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Maierdanjiang Nuermaimaiti
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Clinical Data of Parkinson’s Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kei-ichi Ishikawa
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Clinical Data of Parkinson’s Disease, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Research and Development for Organoids, School of Medicine, Juntendo University, Tokyo, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
19
|
Wu KJ, Hung TW, Wang YS, Chen YH, Bae EK, Yu SJ. Prosaposin PS18 reduces dopaminergic neurodegeneration in a 6-hydroxydopamine rat model of Parkinson's disease. Sci Rep 2023; 13:8148. [PMID: 37208379 DOI: 10.1038/s41598-023-35274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
Saposin and its precursor prosaposin are endogenous proteins with neurotrophic and anti-apoptotic properties. Prosaposin or its analog prosaposin-derived 18-mer peptide (PS18) reduced neuronal damage in hippocampus and apoptosis in stroke brain. Its role in Parkinson's disease (PD) has not been well characterized. This study aimed to examine the physiological role of PS18 in 6-hydroxydopamine (6-OHDA) cellular and animal models of PD. We found that PS18 significantly antagonized 6-OHDA -mediated dopaminergic neuronal loss and TUNEL in rat primary dopaminergic neuronal culture. In SH-SY5Y cells overexpressing the secreted ER calcium-monitoring proteins, we found that PS18 significantly reduced thapsigargin and 6-OHDA-mediated ER stress. The expression of prosaposin and the protective effect of PS18 were next examined in hemiparkinsonian rats. 6-OHDA was unilaterally administered to striatum. The expression of prosaposin was transiently upregulated in striatum on D3 (day 3) after lesioning and returned below the basal level on D29. The 6-OHDA-lesioned rats developed bradykinesia and an increase in methamphetamine-mediated rotation, which was antagonized by PS18. Brain tissues were collected for Western blot, immunohistochemistry, and qRTPCR analysis. Tyrosine hydroxylase immunoreactivity was significantly reduced while the expressions of PERK, ATF6, CHOP, and BiP were upregulated in the lesioned nigra; these responses were significantly antagonized by PS18. Taken together, our data support that PS18 is neuroprotective in cellular and animal models of PD. The mechanisms of protection may involve anti-ER stress.
Collapse
Affiliation(s)
- Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yun-Hsiang Chen
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan.
| |
Collapse
|
20
|
Muraleedharan A, Vanderperre B. The endo-lysosomal system in Parkinson's disease: expanding the horizon. J Mol Biol 2023:168140. [PMID: 37148997 DOI: 10.1016/j.jmb.2023.168140] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence is increasing with age. A wealth of genetic evidence indicates that the endo-lysosomal system is a major pathway driving PD pathogenesis with a growing number of genes encoding endo-lysosomal proteins identified as risk factors for PD, making it a promising target for therapeutic intervention. However, detailed knowledge and understanding of the molecular mechanisms linking these genes to the disease are available for only a handful of them (e.g. LRRK2, GBA1, VPS35). Taking on the challenge of studying poorly characterized genes and proteins can be daunting, due to the limited availability of tools and knowledge from previous literature. This review aims at providing a valuable source of molecular and cellular insights into the biology of lesser-studied PD-linked endo-lysosomal genes, to help and encourage researchers in filling the knowledge gap around these less popular genetic players. Specific endo-lysosomal pathways discussed range from endocytosis, sorting, and vesicular trafficking to the regulation of membrane lipids of these membrane-bound organelles and the specific enzymatic activities they contain. We also provide perspectives on future challenges that the community needs to tackle and propose approaches to move forward in our understanding of these poorly studied endo-lysosomal genes. This will help harness their potential in designing innovative and efficient treatments to ultimately re-establish neuronal homeostasis in PD but also other diseases involving endo-lysosomal dysfunction.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| | - Benoît Vanderperre
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| |
Collapse
|
21
|
Tong J, Guan X, Jiang S, Sun L. A saposin domain-containing protein of tongue sole Cynoglossus semilaevis: Antimicrobial activity and mechanism. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104633. [PMID: 36610645 DOI: 10.1016/j.dci.2023.104633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Prosaposin is a precursor that can be processed into four different saposins, designated as A, B, C, and D, which have multiple functions in mammals, including neuroprotection and immune modulation. The immune function of saposin in teleost remains largely unknown. In the present study, a saposin (SAP) domain-containing protein was identified in half-smooth tongue sole Cynoglossus semilaevis and named CsSDP. CsSDP harbors one SAP A domain and two SAP B domains. When expressed in HEK293T cells, CsSDP was specifically localized in the lysosome. When overexpressed in Escherichia coli, CsSDP markedly inhibited bacterial growth, and the inhibitory effect depended on two specific regions in the SAP A and SAP B domains. Two polypeptides (P32 and P30) derived from the above SAP A and B domains could bind to and inhibit the growth of both Gram-negative and Gram-positive bacteria. The ultrastructural analysis revealed that P32 and P30 killed target bacteria by disrupting the bacterial cell wall and inducing substantial release of cytoplasmic contents. These results shed new lights on the immune function of saposin domain-containing protein in teleost.
Collapse
Affiliation(s)
- Jiazhou Tong
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China; CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Xiaolu Guan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Shuai Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| |
Collapse
|
22
|
Bogetofte H, Ryan BJ, Jensen P, Schmidt SI, Vergoossen DLE, Barnkob MB, Kiani LN, Chughtai U, Heon-Roberts R, Caiazza MC, McGuinness W, Márquez-Gómez R, Vowles J, Bunn FS, Brandes J, Kilfeather P, Connor JP, Fernandes HJR, Caffrey TM, Meyer M, Cowley SA, Larsen MR, Wade-Martins R. Post-translational proteomics platform identifies neurite outgrowth impairments in Parkinson's disease GBA-N370S dopamine neurons. Cell Rep 2023; 42:112180. [PMID: 36870058 DOI: 10.1016/j.celrep.2023.112180] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Variants at the GBA locus, encoding glucocerebrosidase, are the strongest common genetic risk factor for Parkinson's disease (PD). To understand GBA-related disease mechanisms, we use a multi-part-enrichment proteomics and post-translational modification (PTM) workflow, identifying large numbers of dysregulated proteins and PTMs in heterozygous GBA-N370S PD patient induced pluripotent stem cell (iPSC) dopamine neurons. Alterations in glycosylation status show disturbances in the autophagy-lysosomal pathway, which concur with upstream perturbations in mammalian target of rapamycin (mTOR) activation in GBA-PD neurons. Several native and modified proteins encoded by PD-associated genes are dysregulated in GBA-PD neurons. Integrated pathway analysis reveals impaired neuritogenesis in GBA-PD neurons and identify tau as a key pathway mediator. Functional assays confirm neurite outgrowth deficits and identify impaired mitochondrial movement in GBA-PD neurons. Furthermore, pharmacological rescue of glucocerebrosidase activity in GBA-PD neurons improves the neurite outgrowth deficit. Overall, this study demonstrates the potential of PTMomics to elucidate neurodegeneration-associated pathways and potential drug targets in complex disease models.
Collapse
Affiliation(s)
- Helle Bogetofte
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Brent J Ryan
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Sissel I Schmidt
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dana L E Vergoossen
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Mike B Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lisa N Kiani
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Uroosa Chughtai
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Rachel Heon-Roberts
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - William McGuinness
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Ricardo Márquez-Gómez
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Fiona S Bunn
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Janine Brandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Peter Kilfeather
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jack P Connor
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Hugo J R Fernandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Tara M Caffrey
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
23
|
Gharbi T, Liu C, Khan H, Zhang Z, Yang GY, Tang Y. Hypoxic Preconditioned Neural Stem Cell-Derived Extracellular Vesicles Contain Distinct Protein Cargo from Their Normal Counterparts. Curr Issues Mol Biol 2023; 45:1982-1997. [PMID: 36975497 PMCID: PMC10047917 DOI: 10.3390/cimb45030127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Hypoxic preconditioning has been demonstrated to increase the resistance of neural stem cells (NSCs) to hypoxic conditions, as well as to improve their capacity for differentiation and neurogenesis. Extracellular vesicles (EVs) have recently emerged as critical mediators of cell–cell communication, but their role in this hypoxic conditioning is presently unknown. Here, we demonstrated that three hours of hypoxic preconditioning triggers significant neural stem cell EV release. Proteomic profiling of EVs from normal and hypoxic preconditioned neural stem cells identified 20 proteins that were upregulated and 22 proteins that were downregulated after hypoxic preconditioning. We also found an upregulation of some of these proteins by qPCR, thus indicating differences also at the transcript level within the EVs. Among the upregulated proteins are CNP, Cyfip1, CASK, and TUBB5, which are well known to exhibit significant beneficial effects on neural stem cells. Thus, our results not only show a significant difference of protein cargo in EVs consequent to hypoxic exposure, but identify several candidate proteins that might play a pivotal role in the cell-to-cell mediated communication underlying neuronal differentiation, protection, maturation, and survival following exposure to hypoxic conditions.
Collapse
|
24
|
Funayama M, Nishioka K, Li Y, Hattori N. Molecular genetics of Parkinson's disease: Contributions and global trends. J Hum Genet 2023; 68:125-130. [PMID: 35821405 PMCID: PMC9968657 DOI: 10.1038/s10038-022-01058-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/31/2022] [Accepted: 06/20/2022] [Indexed: 01/01/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder primarily characterized by motor dysfunction. Aging is the greatest risk factor for developing PD. Recent molecular genetic studies have revealed that genetic factors, in addition to aging and environmental factors, play an important role in the development of the disorder. Studies of familial PD have identified approximately 20 different causative genes. PRKN is the most frequently detected causative gene in Japan. The PRKN gene is located at a common fragile site, and both copy number variants as well as single nucleotide variants are frequently detected. The location and variety of variant types makes an accurate genetic diagnosis difficult with conventional genetic testing. In sporadic PD, genome-wide association studies have revealed more than 200 genes that are potential drivers for the development of PD. Many of these studies have been conducted in Caucasian populations alone, which has limited the identification of all genetic risk factors for sporadic PD, particularly as genetic backgrounds vary widely by race. The Global Parkinson's Genetics Program is a global undertaking meant to address the issue of regional differences in genetic studies of PD.
Collapse
Affiliation(s)
- Manabu Funayama
- Research Institute of Disease of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Nobutaka Hattori
- Research Institute of Disease of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0106, Japan
| |
Collapse
|
25
|
Pan X, Dutta D, Lu S, Bellen HJ. Sphingolipids in neurodegenerative diseases. Front Neurosci 2023; 17:1137893. [PMID: 36875645 PMCID: PMC9978793 DOI: 10.3389/fnins.2023.1137893] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Neurodegenerative Diseases (NDDs) are a group of disorders that cause progressive deficits of neuronal function. Recent evidence argues that sphingolipid metabolism is affected in a surprisingly broad set of NDDs. These include some lysosomal storage diseases (LSDs), hereditary sensory and autonomous neuropathy (HSAN), hereditary spastic paraplegia (HSP), infantile neuroaxonal dystrophy (INAD), Friedreich's ataxia (FRDA), as well as some forms of amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Many of these diseases have been modeled in Drosophila melanogaster and are associated with elevated levels of ceramides. Similar changes have also been reported in vertebrate cells and mouse models. Here, we summarize studies using fly models and/or patient samples which demonstrate the nature of the defects in sphingolipid metabolism, the organelles that are implicated, the cell types that are initially affected, and potential therapeutics for these diseases.
Collapse
Affiliation(s)
- Xueyang Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
26
|
Miyajima T, Saito R, Yanagisawa H, Igarashi M, Wu C, Iwamoto T, Eto Y. Characterization of cellular phenotypes in neurons derived from induced pluripotent stem cells of male patients with Fabry disease. J Inherit Metab Dis 2023; 46:143-152. [PMID: 36220782 DOI: 10.1002/jimd.12567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 01/19/2023]
Abstract
Fabry disease (FD) is an X-linked inherited lysosomal metabolism disorder in which globotriaosylceramide (Gb3) accumulates in various organs resulting from a deficiency in alpha-galactosidase A. The clinical features of FD include progressive impairments of the renal, cardiac, and peripheral nervous systems. In addition, patients with FD often develop neuropsychiatric symptoms, such as depression and dementia, which are believed to be induced by the cellular injury of cerebrovascular and partially neuronal cells due to Gb3 accumulation. Although the analysis of autopsy brain tissue from patients with FD showed no accumulation of Gb3, abnormal deposits of Gb3 were found in the neurons of several brain areas, including the hippocampus. Therefore, in this study, we generated induced pluripotent stem cells (iPSCs) from patients with FD and differentiated them into neuronal cells to investigate pathological and biological changes in the neurons of FD. Neural stem cells (NSCs) and neurons were successfully differentiated from the iPSCs we generated; however, cellular damage and morphological changes were not found in these cells. Immunostaining revealed no Gb3 accumulation in NSCs and neurons. Transmission electron microscopy did not reveal any zebra body-like structures or inclusion bodies, which are characteristic of FD. These results indicated that neuronal cells derived from FD-iPSCs exhibited normal morphology and no Gb3 accumulation. It is likely that more in vivo environment-like cultures are needed for iPSC-derived neurons to reproduce disease-specific features.
Collapse
Affiliation(s)
- Takashi Miyajima
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
| | - Ryo Saito
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
| | - Hiroko Yanagisawa
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
| | - Miki Igarashi
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
| | - Chen Wu
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
| | - Takeo Iwamoto
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, Kawasaki, Japan
- The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Krohn L, Heilbron K, Blauwendraat C, Reynolds RH, Yu E, Senkevich K, Rudakou U, Estiar MA, Gustavsson EK, Brolin K, Ruskey JA, Freeman K, Asayesh F, Chia R, Arnulf I, Hu MTM, Montplaisir JY, Gagnon JF, Desautels A, Dauvilliers Y, Gigli GL, Valente M, Janes F, Bernardini A, Högl B, Stefani A, Ibrahim A, Šonka K, Kemlink D, Oertel W, Janzen A, Plazzi G, Biscarini F, Antelmi E, Figorilli M, Puligheddu M, Mollenhauer B, Trenkwalder C, Sixel-Döring F, Cochen De Cock V, Monaca CC, Heidbreder A, Ferini-Strambi L, Dijkstra F, Viaene M, Abril B, Boeve BF, Scholz SW, Ryten M, Bandres-Ciga S, Noyce A, Cannon P, Pihlstrøm L, Nalls MA, Singleton AB, Rouleau GA, Postuma RB, Gan-Or Z. Genome-wide association study of REM sleep behavior disorder identifies polygenic risk and brain expression effects. Nat Commun 2022; 13:7496. [PMID: 36470867 PMCID: PMC9722930 DOI: 10.1038/s41467-022-34732-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/03/2022] [Indexed: 12/11/2022] Open
Abstract
Rapid-eye movement (REM) sleep behavior disorder (RBD), enactment of dreams during REM sleep, is an early clinical symptom of alpha-synucleinopathies and defines a more severe subtype. The genetic background of RBD and its underlying mechanisms are not well understood. Here, we perform a genome-wide association study of RBD, identifying five RBD risk loci near SNCA, GBA, TMEM175, INPP5F, and SCARB2. Expression analyses highlight SNCA-AS1 and potentially SCARB2 differential expression in different brain regions in RBD, with SNCA-AS1 further supported by colocalization analyses. Polygenic risk score, pathway analysis, and genetic correlations provide further insights into RBD genetics, highlighting RBD as a unique alpha-synucleinopathy subpopulation that will allow future early intervention.
Collapse
Affiliation(s)
- Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | | | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Regina H Reynolds
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Konstantin Senkevich
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Mehrdad A Estiar
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Emil K Gustavsson
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Kajsa Brolin
- Lund University, Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund, Sweden
| | - Jennifer A Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Kathryn Freeman
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Farnaz Asayesh
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Ruth Chia
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Arnulf
- Sleep Disorders Unit, Pitié Salpêtrière Hospital, APHP-Sorbonne, Paris Brain Insitute and Sorbonne University, Paris, France
| | - Michele T M Hu
- Oxford Parkinson's Disease Centre (OPDC), University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacques Y Montplaisir
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Psychiatry, Université de Montréal, Montréal, QC, Canada
| | - Jean-François Gagnon
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
| | - Alex Desautels
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Yves Dauvilliers
- National Reference Center for Narcolepsy, Sleep Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, University of Montpellier, Institute Neuroscience Montpellier Inserm, Montpellier, France
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Francesco Janes
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Andrea Bernardini
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Birgit Högl
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Abubaker Ibrahim
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Karel Šonka
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - David Kemlink
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Wolfgang Oertel
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Annette Janzen
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Giuseppe Plazzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio-Emilia, Modena, Italy
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - Francesco Biscarini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Elena Antelmi
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Michela Figorilli
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Monica Puligheddu
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Friederike Sixel-Döring
- Department of Neurology, Philipps-University, Marburg, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | - Valérie Cochen De Cock
- Sleep and Neurology Unit, Beau Soleil Clinic, Montpellier, France
- EuroMov Digital Health in Motion, University of Montpellier IMT Mines Ales, Montpellier, France
| | - Christelle Charley Monaca
- University Lille North of France, Department of Clinical Neurophysiology and Sleep Center, CHU Lille, Lille, France
| | - Anna Heidbreder
- Institute of Sleep Medicine and Neuromuscular Disorders, University of Münster, Münster, Germany
| | - Luigi Ferini-Strambi
- Department of Neurological Sciences, Università Vita-Salute San Raffaele, Milan, Italy
| | - Femke Dijkstra
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Mineke Viaene
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium
| | - Beatriz Abril
- Sleep disorder Unit, Carémeau Hospital, University Hospital of Nîmes, Nîmes, France
| | | | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Alastair Noyce
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
- Department of Clinical and Movement Neurosciences, University College London, Institute of Neurology, London, UK
| | | | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ronald B Postuma
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Tanaka H, Tsuji D, Watanabe R, Ohnishi Y, Kitaguchi S, Nakae R, Teramoto H, Tsukimoto J, Horii Y, Itoh K. Aberrant autophagy in lysosomal storage disorders marked by a lysosomal SNARE protein shortage due to suppression of endocytosis. J Inherit Metab Dis 2022; 45:1191-1202. [PMID: 36102069 DOI: 10.1002/jimd.12558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022]
Abstract
Lysosomal storage disorders (LSDs) are inherited metabolic diseases caused by genetic defects in lysosomal enzymes or related factors. LSDs are associated with excessive accumulation of natural substrates in lysosomes leading to central nervous system and peripheral tissue damage. Abnormal autophagy is also involved in pathogenesis, although the underlying mechanisms remain unclear. We demonstrated that impairment of lysosome-autophagosome fusion is due to suppressed endocytosis in LSDs. The fusion was reduced in several LSD cells and the brains of LSD model mice, suggesting that the completion of autophagy is suppressed by the accumulation of substrates. In this brain, the expression of the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) proteins, VAMP8 and Syntaxin7, was decreased on the lysosomal surface but not intracellular. This aberrant autophagy preceded the development of pathological phenotypes in LSD-model mice. Furthermore, the enzyme deficiency leading to the substrate accumulation could suppress endocytosis, and the inhibited endocytosis decreased SNARE proteins localized on lysosomes. These findings suggest that the shortage of SNARE proteins on lysosomes is one of the reasons for the impairment of lysosome-autophagosome fusion in LSD cells. Defects in lysosomal enzyme activity suppress endocytosis and decrease the supply of intracellular SNARE proteins recruited to lysosomes. This shortage of lysosomal SNARE proteins impairs lysosome-autophagosome fusion in lysosomal storage disorders.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Daisuke Tsuji
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Ryosuke Watanabe
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Yukiya Ohnishi
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Shindai Kitaguchi
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Ryuto Nakae
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Hiromi Teramoto
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Jun Tsukimoto
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Yuto Horii
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
29
|
Kojima R, Zurbruegg M, Li T, Paslawski W, Zhang X, Svenningsson P. Prosaposin Reduces α-Synuclein in Cells and Saposin C Dislodges it from Glucosylceramide-enriched Lipid Membranes. J Mol Neurosci 2022; 72:2313-2325. [PMID: 36152140 PMCID: PMC9726671 DOI: 10.1007/s12031-022-02066-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/10/2022] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder affecting over 1% of the 65 + age population. Saposin C, a lysosomal protein required for the normal activity of glucocerebrosidase (GCase), may serve as a disease modifier in PD. Saposin C is cleaved from its precursor, Prosaposin (PSAP), which is secreted as an uncleaved protein and exerts neuroprotective effects. In this study, we aim to elucidate the neuroprotective roles of PSAP and saposin C in PD by evaluating their effects on α-synuclein accumulation in human neuroblastoma cells. Stable overexpression of PSAP reduced monomeric α-synuclein levels in SH-SY5Y cells, while PSAP knockdown by small interfering RNA led to the opposite effect, and those effects were independent of GCase activity. Autophagy flux was decreased by stable PSAP overexpression. Furthermore, a flow-through assay revealed that recombinant saposin C was able to detach α-synuclein from artificial glucosylceramide-enriched lipid membranes at the lysosomal pH. Taken together, our findings provide further evidence that PSAP and saposin C as key proteins involved in α-synuclein clearance by dislodging it from lipid membranes.
Collapse
Affiliation(s)
- Rika Kojima
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Mark Zurbruegg
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Tianyi Li
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 171 76, Stockholm, Sweden.
- Basic and Clinical Neuroscience, King's College London, London, UK.
| |
Collapse
|
30
|
Decreased Prosaposin and Progranulin in the Cingulate Cortex Are Associated with Schizophrenia Pathophysiology. Int J Mol Sci 2022; 23:ijms231912056. [PMID: 36233357 PMCID: PMC9570388 DOI: 10.3390/ijms231912056] [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: 08/29/2022] [Revised: 10/02/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Prosaposin (PSAP) and progranulin (PGRN) are two lysosomal proteins that interact and modulate the metabolism of lipids, particularly sphingolipids. Alterations in sphingolipid metabolism have been found in schizophrenia. Genetic associations of PSAP and PGRN with schizophrenia have been reported. To further clarify the role of PSAP and PGRN in schizophrenia, we examined PSAP and PGRN levels in postmortem cingulate cortex tissue from healthy controls along with patients who had suffered from schizophrenia, bipolar disorder, or major depressive disorder. We found that PSAP and PGRN levels are reduced specifically in schizophrenia patients. To understand the role of PSAP in the cingulate cortex, we used an AAV strategy to knock down PSAP in neurons located in this region. Neuronal PSAP knockdown led to the downregulation of neuronal PGRN levels and behavioral abnormalities. Cingulate-PSAP-deficient mice exhibited increased anxiety-like behavior and impaired prepulse inhibition, as well as intact locomotion, working memory, and a depression-like state. The behavioral changes were accompanied by increased early growth response protein 1 (EGR-1) and activity-dependent cytoskeleton-associated protein (ARC) levels in the sensorimotor cortex and hippocampus, regions implicated in circuitry dysfunction in schizophrenia. In conclusion, PSAP and PGRN downregulation in the cingulate cortex is associated with schizophrenia pathophysiology.
Collapse
|
31
|
Suzuki C, Yamaguchi J, Sanada T, Oliva Trejo JA, Kakuta S, Shibata M, Tanida I, Uchiyama Y. Lack of Cathepsin D in the central nervous system results in microglia and astrocyte activation and the accumulation of proteinopathy-related proteins. Sci Rep 2022; 12:11662. [PMID: 35804072 PMCID: PMC9270453 DOI: 10.1038/s41598-022-15805-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
Neuronal ceroid lipofuscinosis is one of many neurodegenerative storage diseases characterized by excessive accumulation of lipofuscins. CLN10 disease, an early infantile neuronal ceroid lipofuscinosis, is associated with a gene that encodes cathepsin D (CtsD), one of the major lysosomal proteases. Whole body CtsD-knockout mice show neurodegenerative phenotypes with the accumulation of lipofuscins in the brain and also show defects in other tissues including intestinal necrosis. To clarify the precise role of CtsD in the central nervous system (CNS), we generated a CNS-specific CtsD-knockout mouse (CtsD-CKO). CtsD-CKO mice were born normally but developed seizures and their growth stunted at around postnatal day 23 ± 1. CtsD-CKO did not exhibit apparent intestinal symptoms as those observed in whole body knockout. Histologically, autofluorescent materials were detected in several areas of the CtsD-CKO mouse's brain, including: thalamus, cerebral cortex, hippocampus, and cerebellum. Expression of ubiquitin and autophagy-associated proteins was also increased, suggesting that the autophagy-lysosome system was impaired. Microglia and astrocytes were activated in the CtsD-CKO thalamus, and inducible nitric oxide synthase (iNOS), an inflammation marker, was increased in the microglia. Interestingly, deposits of proteinopathy-related proteins, phosphorylated α-synuclein, and Tau protein were also increased in the thalamus of CtsD-CKO infant mice. Considering these results, we propose thatt the CtsD-CKO mouse is a useful mouse model to investigate the contribution of cathepsin D to the early phases of neurodegenerative diseases in relation to lipofuscins, proteinopathy-related proteins and activation of microglia and astrocytes.
Collapse
Affiliation(s)
- Chigure Suzuki
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Junji Yamaguchi
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Laboratory of Morphology and Image Analysis, Biomedical Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takahito Sanada
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Juan Alejandro Oliva Trejo
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Souichirou Kakuta
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Laboratory of Morphology and Image Analysis, Biomedical Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Masahiro Shibata
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima-shi, Kagoshima, 890-8544, Japan
| | - Isei Tanida
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
32
|
Xue J, Zhu Y, Wei L, Huang H, Li G, Huang W, Zhu H, Duan R. Loss of Drosophila NUS1 results in cholesterol accumulation and Parkinson's disease-related neurodegeneration. FASEB J 2022; 36:e22411. [PMID: 35695805 DOI: 10.1096/fj.202200212r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 11/11/2022]
Abstract
NgBR is the Nogo-B receptor, encoded by NUS1 gene. As NgBR contains a C-terminal domain that is similar to cis-isoprenyltransferase (cis-IPTase), NgBR was speculated to stabilize nascent Niemann-Pick type C 2 (NPC2) to facilitate cholesterol transport out of lysosomes. Mutations in the NUS1 were known as risk factors for Parkinson's disease (PD). In our previous study, it was shown that knockdown of Drosophila NUS1 orthologous gene tango14 causes decreased climbing ability, loss of dopaminergic neurons, and decreased dopamine contents. In this study, tango14 mutant flies were generated with a mutation in the C-terminal enzyme activity region using CRISPR/Cas9. Tango14 mutant showed a reduced lifespan with locomotive defects and cholesterol accumulation in Malpighian tubules and brains, especially in dopaminergic neurons. Multilamellar bodies were found in tango14 mutants using electron microscopy. Neurodegenerative-related brain vacuolization was also detected in tango14 knockdown flies in an age-dependent manner. In addition, tango14 knockdown increased α-synuclein (α-syn) neurotoxicity in α-syn-overexpressing flies, with decreased locomotive activities, dopamine contents, and the numbers of dopaminergic neurons in aging flies. Thus, these observations suggest a role of NUS1, the ortholog of tango14, in PD-related pathogenesis.
Collapse
Affiliation(s)
- Jin Xue
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Yingbao Zhu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Liyi Wei
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Hongjing Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Guangxu Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Wen Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Hua Zhu
- Department of Clinical Laboratory, Jilin Cancer Hospital, Jilin, China
| | - Ranhui Duan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China.,Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China
| |
Collapse
|
33
|
Koçoğlu C, Van Broeckhoven C, van der Zee J. How network-based approaches can complement gene identification studies in frontotemporal dementia. Trends Genet 2022; 38:944-955. [DOI: 10.1016/j.tig.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022]
|
34
|
Sosero YL, Yu E, Estiar MA, Krohn L, Mufti K, Rudakou U, Ruskey JA, Asayesh F, Laurent SB, Spiegelman D, Trempe JF, Quinnell TG, Oscroft N, Arnulf I, Montplaisir JY, Gagnon JF, Desautels A, Dauvilliers Y, Gigli GL, Valente M, Janes F, Bernardini A, Sonka K, Kemlink D, Oertel W, Janzen A, Plazzi G, Antelmi E, Biscarini F, Figorilli M, Puligheddu M, Mollenhauer B, Trenkwalder C, Sixel-Döring F, Cochen De Cock V, Monaca CC, Heidbreder A, Ferini-Strambi L, Dijkstra F, Viaene M, Abril B, Boeve BF, Postuma RB, Rouleau GA, Ibrahim A, Stefani A, Högl B, Hu MTM, Gan-Or Z. Rare PSAP Variants and Possible Interaction with GBA in REM Sleep Behavior Disorder. JOURNAL OF PARKINSON'S DISEASE 2022; 12:333-340. [PMID: 34690151 DOI: 10.3233/jpd-212867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND PSAP encodes saposin C, the co-activator of glucocerebrosidase, encoded by GBA. GBA mutations are associated with idiopathic/isolated REM sleep behavior disorder (iRBD), a prodromal stage of synucleinopathy. OBJECTIVE To examine the role of PSAP mutations in iRBD. METHODS We fully sequenced PSAP and performed Optimized Sequence Kernel Association Test in 1,113 iRBD patients and 2,324 controls. We identified loss-of-function (LoF) mutations, which are very rare in PSAP, in three iRBD patients and none in controls (uncorrected p = 0.018). RESULTS Two variants were stop mutations, p.Gln260Ter and p.Glu166Ter, and one was an in-frame deletion, p.332_333del. All three mutations have a deleterious effect on saposin C, based on in silico analysis. In addition, the two carriers of p.Glu166Ter and p.332_333del mutations also carried a GBA variant, p.Arg349Ter and p.Glu326Lys, respectively. The co-occurrence of these extremely rare PSAP LoF mutations in two (0.2%) GBA variant carriers in the iRBD cohort, is unlikely to occur by chance (estimated co-occurrence in the general population based on gnomAD data is 0.00035%). Although none of the three iRBD patients with PSAP LoF mutations have phenoconverted to an overt synucleinopathy at their last follow-up, all manifested initial signs suggestive of motor dysfunction, two were diagnosed with mild cognitive impairment and all showed prodromal clinical markers other than RBD. Their probability of prodromal PD, according to the Movement Disorder Society research criteria, was 98% or more. CONCLUSION These results suggest a possible role of PSAP variants in iRBD and potential genetic interaction with GBA, which requires additional studies.
Collapse
Affiliation(s)
- Yuri L Sosero
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Mehrdad A Estiar
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Kheireddin Mufti
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Jennifer A Ruskey
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Farnaz Asayesh
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Sandra B Laurent
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Dan Spiegelman
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Jean-François Trempe
- Department of Pharmacology & Therapeutics and Centre de Recherche en Biologie Structurale, McGill University, Montréal, Québec, Canada
| | | | | | - Isabelle Arnulf
- Sleep Disorders Unit, Sorbonne University, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jacques Y Montplaisir
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada.,Department of Psychiatry, Université de Montréal, Montréal, QC, Canada
| | - Jean-François Gagnon
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada.,Department of Psychology, Université du Québec à Montréal, Montréal, QC, Canada
| | - Alex Desautels
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada.,Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Yves Dauvilliers
- National Reference Centre for Orphan Diseases, Narcolepsy- Rare hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Institute for Neurosciences of Montpellier INM, Univ Montpellier, INSERM, Montpellier, France
| | - Gian Luigi Gigli
- Department of Neurosciences, Clinical Neurology Unit, University Hospital of Udine, Udine, Italy.,Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Mariarosaria Valente
- Department of Neurosciences, Clinical Neurology Unit, University Hospital of Udine, Udine, Italy.,Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Francesco Janes
- Department of Neurosciences, Clinical Neurology Unit, University Hospital of Udine, Udine, Italy
| | - Andrea Bernardini
- Department of Neurosciences, Clinical Neurology Unit, University Hospital of Udine, Udine, Italy
| | - Karel Sonka
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - David Kemlink
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Wolfgang Oertel
- Department of Neurology, Philipps University, Marburg, Germany
| | - Annette Janzen
- Department of Neurology, Philipps University, Marburg, Germany
| | - Giuseppe Plazzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - Elena Antelmi
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Neurosciences, Neurology Unit, Movement Disorders Division, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Francesco Biscarini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Michela Figorilli
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Monica Puligheddu
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Centre Göttingen, Göttingen, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Centre Göttingen, Göttingen, Germany
| | - Friederike Sixel-Döring
- Department of Neurology, Philipps University, Marburg, Germany.,Paracelsus-Elena-Klinik, Kassel, Germany
| | - Valérie Cochen De Cock
- Sleep and Neurology Unit, Beau Soleil Clinic, Montpellier, France.,EuroMov, University of Montpellier, Montpellier, France
| | - Christelle Charley Monaca
- Department of Clinical Neurophysiology and Sleep Center, University Lille North of France, CHU Lille, Lille, France
| | - Anna Heidbreder
- Department of Sleep Medicine and Neuromuscular Disorders, University of Münster, Münster, Germany
| | - Luigi Ferini-Strambi
- Department of Neurological Sciences, Università Vita-Salute San Raffaele, Milan, Italy
| | - Femke Dijkstra
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium.,Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium.,Department of Neurology, University Hospital Antwerp, Edegem, Antwerp, Belgium
| | - Mineke Viaene
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium.,Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium
| | - Beatriz Abril
- Sleep disorder Unit, Carémeau Hospital, University Hospital of Nîmes, France
| | | | - Ronald B Postuma
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.,Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Coeur de Montréal, Montréal, QC, Canada
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Abubaker Ibrahim
- Department of Neurology, Sleep Disorders Clinic, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Sleep Disorders Clinic, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Sleep Disorders Clinic, Medical University of Innsbruck, Innsbruck, Austria
| | - Michele T M Hu
- Department of Sleep Medicine and Neuromuscular Disorders, University of Münster, Münster, Germany.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
35
|
Udayar V, Chen Y, Sidransky E, Jagasia R. Lysosomal dysfunction in neurodegeneration: emerging concepts and methods. Trends Neurosci 2022; 45:184-199. [PMID: 35034773 PMCID: PMC8854344 DOI: 10.1016/j.tins.2021.12.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/23/2021] [Accepted: 12/12/2021] [Indexed: 02/06/2023]
Abstract
The understanding of lysosomes has come a long way since the initial discovery of their role in degrading cellular waste. The lysosome is now recognized as a highly dynamic organelle positioned at the crossroads of cell signaling, transcription, and metabolism. Underscoring its importance is the observation that, in addition to rare monogenic lysosomal storage disorders, genes regulating lysosomal function are implicated in common sporadic neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Developing therapies for these disorders is particularly challenging, largely due to gaps in knowledge of the underlying molecular and cellular processes. In this review, we discuss technological advances that have propelled deeper understanding of the lysosome in neurodegeneration, from elucidating the functions of lysosome-related disease risk variants at the level of the organelle, cell, and tissue, to the development of disease-specific biological models that recapitulate disease manifestations. Finally, we identify key questions to be addressed to successfully bridge the gap to the clinic.
Collapse
Affiliation(s)
- Vinod Udayar
- Roche Pharmaceutical Research and Early Development, Neuroscience and Rare Diseases Discovery & Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Ravi Jagasia
- Roche Pharmaceutical Research and Early Development, Neuroscience and Rare Diseases Discovery & Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| |
Collapse
|
36
|
Kim MJ, Jeong H, Krainc D. Lysosomal ceramides regulate cathepsin B-mediated processing of saposin C and glucocerebrosidase activity. Hum Mol Genet 2022; 31:2424-2437. [PMID: 35181782 PMCID: PMC9307309 DOI: 10.1093/hmg/ddac047] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Variants in multiple lysosomal enzymes increase Parkinson's disease (PD) risk, including the genes encoding glucocerebrosidase (GCase), acid sphingomyelinase (ASMase) and galactosylceramidase. Each of these enzymes generates ceramide by hydrolysis of sphingolipids in lysosomes, but the role of this common pathway in PD pathogenesis has not yet been explored. Variations in GBA1, the gene encoding GCase, are the most common genetic risk factor for PD. The lysosomal enzyme cathepsin B has recently been implicated as an important genetic modifier of disease penetrance in individuals harboring GBA1 variants, suggesting a mechanistic link between these enzymes. Here, we found that ceramide activates cathepsin B, and identified a novel role for cathepsin B in mediating prosaposin cleavage to form saposin C, the lysosomal coactivator of GCase. Interestingly, this pathway was disrupted in Parkin-linked PD models, and upon treatment with inhibitor of ASMase which resulted in decreased ceramide production. Conversely, increasing ceramide production by inhibiting acid ceramidase activity was sufficient to upregulate cathepsin B- and saposin C-mediated activation of GCase. These results highlight a mechanistic link between ceramide and cathepsin B in regulating GCase activity and suggest that targeting lysosomal ceramide or cathepsin B represents an important therapeutic strategy for activating GCase in PD and related disorders.
Collapse
Affiliation(s)
- Myung Jong Kim
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hyunkyung Jeong
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- To whom correspondence should be addressed. Tel/Fax: 312-503-3936;
| |
Collapse
|
37
|
Fine-mapping of intracranial aneurysm susceptibility based on a genome-wide association study. Sci Rep 2022; 12:2717. [PMID: 35177760 PMCID: PMC8854430 DOI: 10.1038/s41598-022-06755-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 02/04/2022] [Indexed: 12/22/2022] Open
Abstract
In addition to conventional genome-wide association studies (GWAS), a fine-mapping analysis is increasingly used to identify the genetic function of variants associated with disease susceptibilities. Here, we used a fine-mapping approach to evaluate candidate variants based on a previous GWAS involving patients with intracranial aneurysm (IA). A fine-mapping analysis was conducted based on the chromosomal data provided by a GWAS of 250 patients diagnosed with IA and 296 controls using posterior inclusion probability (PIP) and log10 transformed Bayes factor (log10BF). The narrow sense of heritability (h2) explained by each candidate variant was estimated. Subsequent gene expression and functional network analyses of candidate genes were used to calculate transcripts per million (TPM) values. Twenty single-nucleotide polymorphisms (SNPs) surpassed a genome-wide significance threshold for creditable evidence (log10BF > 6.1). Among them, four SNPs, rs75822236 (GBA; log10BF = 15.06), rs112859779 (TCF24; log10BF = 12.12), rs79134766 (OLFML2A; log10BF = 14.92), and rs371331393 (ARHGAP32; log10BF = 20.88) showed a completed PIP value in each chromosomal region, suggesting a higher probability of functional candidate variants associated with IA. On the contrary, these associations were not shown clearly under different replication sets. Our fine-mapping analysis suggested that four functional candidate variants of GBA, TCF24, OLFML2A, and ARHGAP32 were linked to IA susceptibility and pathogenesis. However, this approach could not completely replace replication sets based on large-scale data. Thus, caution is required when interpreting results of fine-mapping analysis.
Collapse
|
38
|
Okano H, Morimoto S. iPSC-based disease modeling and drug discovery in cardinal neurodegenerative disorders. Cell Stem Cell 2022; 29:189-208. [PMID: 35120619 DOI: 10.1016/j.stem.2022.01.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
It has been 15 years since the birth of human induced pluripotent stem cell (iPSC) technology in 2007, and the scope of its application has been expanding. In addition to the development of cell therapies using iPSC-derived cells, pathological analyses using disease-specific iPSCs and clinical trials to confirm the safety and efficacy of drugs developed using iPSCs are progressing. With the innovation of related technologies, iPSC applications are about to enter a new stage. This review outlines advances in iPSC modeling and therapeutic development for cardinal neurodegenerative diseases, such as amyotrophic lateral sclerosis, Parkinson's disease, and Alzheimer's disease.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-shi, Saitama 351-0198, Japan.
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
39
|
Al-Azzawi ZAM, Arfaie S, Gan-Or Z. GBA1 and The Immune System: A Potential Role in Parkinson's Disease? JOURNAL OF PARKINSON'S DISEASE 2022; 12:S53-S64. [PMID: 36057834 PMCID: PMC9535551 DOI: 10.3233/jpd-223423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
It is clear that the immune system and inflammation have a role in Parkinson's disease (PD), including sporadic PD and some genetic forms such as LRRK2-associated PD. One of the most important genes associated with PD is GBA1, as variants in this gene are found in 5-20% of PD patients in different populations worldwide. Biallelic variants in GBA1 may cause Gaucher disease, a lysosomal storage disorder with involvement of the immune system, and other lines of evidence link GBA1 to the immune system and inflammation. In this review, we discuss these different pieces of evidence and whether the interplay between GBA1 and the immune system may have a role in PD.
Collapse
Affiliation(s)
- Zaid A M Al-Azzawi
- Faculty of Medicine, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Saman Arfaie
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- The Neuro - Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Petese A, Cesaroni V, Cerri S, Blandini F. Are Lysosomes Potential Therapeutic Targets for Parkinson's Disease? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:642-655. [PMID: 34370650 DOI: 10.2174/1871527320666210809123630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/16/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
Parkinson´s Disease (PD) is the second most common neurodegenerative disorder, affecting ~2-3% of the population over 65 years old. In addition to progressive degeneration of nigrostriatal neurons, the histopathological feature of PD is the accumulation of misfolded α-synuclein protein in abnormal cytoplasmatic inclusions, known as Lewy Bodies (LBs). Recently, Genome-Wide Association Studies (GWAS) have indicated a clear association of variants within several lysosomal genes with risk for PD. Newly evolving data have been shedding light on the relationship between lysosomal dysfunction and alpha-synuclein aggregation. Defects in lysosomal enzymes could lead to the insufficient clearance of neurotoxic protein materials, possibly leading to selective degeneration of dopaminergic neurons. Specific modulation of lysosomal pathways and their components could be considered a novel opportunity for therapeutic intervention for PD. The purpose of this review is to illustrate lysosomal biology and describe the role of lysosomal dysfunction in PD pathogenesis. Finally, the most promising novel therapeutic approaches designed to modulate lysosomal activity, as a potential disease-modifying treatment for PD will be highlighted.
Collapse
Affiliation(s)
- Alessandro Petese
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Valentina Cesaroni
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Silvia Cerri
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
41
|
Zhao YW, Pan HX, Liu Z, Wang Y, Zeng Q, Fang ZH, Luo TF, Xu K, Wang Z, Zhou X, He R, Li B, Zhao G, Xu Q, Sun QY, Yan XX, Tan JQ, Li JC, Guo JF, Tang BS. The Association Between Lysosomal Storage Disorder Genes and Parkinson's Disease: A Large Cohort Study in Chinese Mainland Population. Front Aging Neurosci 2021; 13:749109. [PMID: 34867278 PMCID: PMC8634711 DOI: 10.3389/fnagi.2021.749109] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Recent years have witnessed an increasing number of studies indicating an essential role of the lysosomal dysfunction in Parkinson’s disease (PD) at the genetic, biochemical, and cellular pathway levels. In this study, we investigated the association between rare variants in lysosomal storage disorder (LSD) genes and Chinese mainland PD. Methods: We explored the association between rare variants of 69 LSD genes and PD in 3,879 patients and 2,931 controls from Parkinson’s Disease & Movement Disorders Multicenter Database and Collaborative Network in China (PD-MDCNC) using next-generation sequencing, which were analyzed by using the optimized sequence kernel association test. Results: We identified the significant burden of rare putative LSD gene variants in Chinese mainland patients with PD. This association was robust in familial or sporadic early-onset patients after excluding the GBA variants but not in sporadic late-onset patients. The burden analysis of variant sets in genes of LSD subgroups revealed a suggestive significant association between variant sets in genes of sphingolipidosis deficiency disorders and familial or sporadic early-onset patients. In contrast, variant sets in genes of sphingolipidoses, mucopolysaccharidoses, and post-translational modification defect disorders were suggestively associated with sporadic late-onset patients. Then, SMPD1 and other four novel genes (i.e., GUSB, CLN6, PPT1, and SCARB2) were suggestively associated with sporadic early-onset or familial patients, whereas GALNS and NAGA were suggestively associated with late-onset patients. Conclusion: Our findings supported the association between LSD genes and PD and revealed several novel risk genes in Chinese mainland patients with PD, which confirmed the importance of lysosomal mechanisms in PD pathogenesis. Moreover, we identified the genetic heterogeneity in early-onset and late-onset of patients with PD, which may provide valuable suggestions for the treatment.
Collapse
Affiliation(s)
- Yu-Wen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong-Xu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yige Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Huan Fang
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Teng-Fei Luo
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Kun Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Ying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xin-Xiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie-Qiong Tan
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
42
|
Gharbi N, Røise D, Førre JE, Edson AJ, Hushagen HA, Tronci V, Frøyset AK, Fladmark KE. Reintroduction of DJ-1 in Müller Cells Inhibits Retinal Degeneration in the DJ-1 Deficient Retina. Antioxidants (Basel) 2021; 10:1862. [PMID: 34942966 PMCID: PMC8698414 DOI: 10.3390/antiox10121862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
The eye is continuously under oxidative stress due to high metabolic activity and reactive oxygen species generated by daily light exposure. The redox-sensitive protein DJ-1 has proven to be essential in order to protect retina and retinal pigment epithelium (RPE) from oxidative-stress-induced degeneration. Here, we analyzed the specific role of Müller cell DJ-1 in the adult zebrafish retina by re-establishing Müller-cell-specific DJ-1 expression in a DJ-1 knockout retina. Loss of DJ-1 resulted in an age-dependent retinal degeneration, including loss of cells in the ganglion cell layer, retinal thinning, photoreceptor disorganization and RPE cell dysfunction. The degenerative phenotype induced by the absence of DJ-1 was inhibited by solely expressing DJ-1 in Müller cells. The protective effect was dependent upon the cysteine-106 residue of DJ-1, which has been shown to be an oxidative sensor of DJ-1. In a label-free proteomics analysis of isolated retinas, we identified proteins differentially expressed after DJ-1 knockout, but with restored levels after Müller cell DJ-1 re-insertion. Our data show that Müller cell DJ-1 has a major role in protecting the retina from age-dependent oxidative stress.
Collapse
Affiliation(s)
- Naouel Gharbi
- Integrative Fish Biology Group (IFB), NORCE Norwegian Research Center AS, N-5020 Bergen, Norway; (N.G.); (V.T.)
| | - Dagne Røise
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Jorunn-Elise Førre
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Amanda J. Edson
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Helena A. Hushagen
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Valentina Tronci
- Integrative Fish Biology Group (IFB), NORCE Norwegian Research Center AS, N-5020 Bergen, Norway; (N.G.); (V.T.)
| | - Ann-Kristin Frøyset
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Kari E. Fladmark
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| |
Collapse
|
43
|
Identification of Potential Core Genes in Parkinson's Disease Using Bioinformatics Analysis. PARKINSON'S DISEASE 2021; 2021:1690341. [PMID: 34580608 PMCID: PMC8464436 DOI: 10.1155/2021/1690341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/21/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Purpose This study aimed to explore new core genes related to the occurrence of Parkinson's disease (PD) and core genes that can lead to the progression of PD. Methods The expression profile data of GSE42966, which contained six substantia nigra tissues isolated from normal individuals and nine substantia nigra tissues isolated from patients with PD, were obtained from Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified, followed by functional enrichment analysis and protein-protein interaction (PPI) network construction. We then identified 10 hub genes and analyzed their expression in different Braak stages. Results A total of 773 DEGs were identified that were significantly enriched in metabolic pathways. Ten hub genes were identified through the PPI network, namely, GNG3, MAPK1, FPR1, ATP5B, GNG2, PRKACA, HRAS, HSPA8, PSAP, and GABBR2. The expression of HRAS was different in patients with PD with Braak stages 3 and 4. Conclusion These 10 hub genes and the metabolic pathways they are enriched in may be involved in the pathogenesis of PD. HRAS may have potential value in predicting the progression of PD.
Collapse
|
44
|
Imbimbo BP, Ippati S, Watling M, Balducci C. A critical appraisal of tau-targeting therapies for primary and secondary tauopathies. Alzheimers Dement 2021; 18:1008-1037. [PMID: 34533272 DOI: 10.1002/alz.12453] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Primary tauopathies are neurological disorders in which tau protein deposition is the predominant pathological feature. Alzheimer's disease is a secondary tauopathy with tau forming hyperphosphorylated insoluble aggregates. Tau pathology can propagate from region to region in the brain, while alterations in tau processing may impair tau physiological functions. METHODS We reviewed literature on tau biology and anti-tau drugs using PubMed, meeting abstracts, and ClnicalTrials.gov. RESULTS The past 15 years have seen >30 drugs interfering with tau aggregation, processing, and accumulation reaching the clinic. Initial results with tau aggregation inhibitors and anti-tau monoclonal antibodies have not shown clinical efficacy. DISCUSSION The reasons for these clinical failures are unclear but could be linked to the clearing of physiological forms of tau by non-specific drugs. Research is now concentrating efforts on developing reliable translational animal models and selective compounds targeting specific tau epitopes, neurotoxic tau aggregates, and post-translational tau modifications.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Milan, Italy
| |
Collapse
|
45
|
Mendsaikhan A, Tooyama I, Serrano GE, Beach TG, Walker DG. Loss of Lysosomal Proteins Progranulin and Prosaposin Associated with Increased Neurofibrillary Tangle Development in Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:741-753. [PMID: 34374777 PMCID: PMC8433593 DOI: 10.1093/jnen/nlab056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disease causing cognitive decline in the aging population. To develop disease-modifying treatments, understanding the mechanisms behind the pathology is important, which should include observations using human brain samples. We reported previously on the association of lysosomal proteins progranulin (PGRN) and prosaposin (PSAP) with amyloid plaques in non-demented aged control and AD brains. In this study, we investigated the possible involvement of PGRN and PSAP in tangle formation using human brain tissue sections of non-demented aged control subjects and AD cases and compared with cases of frontotemporal dementia with granulin (GRN) mutations. The study revealed that decreased amounts of PGRN and PSAP proteins were detected even in immature neurofibrillary tangles, while colocalization was still evident in adjacent neurons in all cases. Results suggest that neuronal loss of PGRN preceded loss of PSAP as tangles developed and matured. The GRN mutation cases exhibited almost complete absence of PGRN in most neurons, while PSAP signal was preserved. Although based on correlative data, we suggest that reduced levels of PGRN and PSAP and their interaction in neurons might predispose to accumulation of p-Tau protein.
Collapse
Affiliation(s)
- Anarmaa Mendsaikhan
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona (GES, TGB)
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona (GES, TGB)
| | - Douglas G Walker
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
- School of Life Sciences and Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona (DGW)
| |
Collapse
|
46
|
Lin ZH, Zhang BR. Striking While the Iron is Hot: The Role of Prosaposin (PSAP) in Parkinson's Disease. Mov Disord 2021; 36:2224. [PMID: 34480373 DOI: 10.1002/mds.28781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Zhi-Hao Lin
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bao-Rong Zhang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
47
|
Tsuboi K, Tai T, Yamashita R, Ali H, Watanabe T, Uyama T, Okamoto Y, Kitakaze K, Takenouchi Y, Go S, Rahman IAS, Houchi H, Tanaka T, Okamoto Y, Tokumura A, Matsuda J, Ueda N. Involvement of acid ceramidase in the degradation of bioactive N-acylethanolamines. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158972. [PMID: 34033896 DOI: 10.1016/j.bbalip.2021.158972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 05/01/2021] [Accepted: 05/18/2021] [Indexed: 11/25/2022]
Abstract
Bioactive N-acylethanolamines (NAEs) include palmitoylethanolamide, oleoylethanolamide, and anandamide, which exert anti-inflammatory, anorexic, and cannabimimetic actions, respectively. The degradation of NAEs has been attributed to two hydrolases, fatty acid amide hydrolase and NAE acid amidase (NAAA). Acid ceramidase (AC) is a lysosomal enzyme that hydrolyzes ceramide (N-acylsphingosine), which resembles NAAA in structure and function. In the present study, we examined the role of AC in the degradation of NAEs. First, we demonstrated that purified recombinant human AC can hydrolyze various NAEs with lauroylethanolamide (C12:0-NAE) as the most reactive NAE substrate. We then used HEK293 cells metabolically labeled with [14C]ethanolamine, and revealed that overexpressed AC lowered the levels of 14C-labeled NAE. As analyzed with liquid chromatography-tandem mass spectrometry, AC overexpression decreased the amounts of different NAE species. Furthermore, suppression of endogenous AC in LNCaP prostate cells by siRNA increased the levels of various NAEs. Lastly, tissue homogenates from mice genetically lacking saposin D, a presumable activator protein of AC, showed much lower hydrolyzing activity for NAE as well as ceramide than the homogenates from wild-type mice. These results demonstrate the ability of AC to hydrolyze NAEs and suggest its physiological role as a third NAE hydrolase.
Collapse
Affiliation(s)
- Kazuhito Tsuboi
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | - Tatsuya Tai
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan; Department of Pharmacy, Kagawa University Hospital, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Ryouhei Yamashita
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hanif Ali
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Takashi Watanabe
- Department of Pathophysiology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Toru Uyama
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Yoko Okamoto
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Keisuke Kitakaze
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Yasuhiro Takenouchi
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Shinji Go
- Department of Pathophysiology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Iffat Ara Sonia Rahman
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Hitoshi Houchi
- Department of Pharmacy, Kagawa University Hospital, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan; Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa 769-2193, Japan
| | - Tamotsu Tanaka
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan; Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Yasuo Okamoto
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Akira Tokumura
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan; Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, Hiroshima 731-0153, Japan
| | - Junko Matsuda
- Department of Pathophysiology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| |
Collapse
|
48
|
Pan HX, Wang YG, Zhao YW, Zeng Q, Wang Z, Fang ZH, Zhang Y, Zhou X, He RC, Xu Q, Sun QY, Tan JQ, Yan XX, Li JC, Tang BS, Guo JF. Evaluating the role of ARSA in Chinese patients with Parkinson's disease. Neurobiol Aging 2021; 109:269-272. [PMID: 34531044 DOI: 10.1016/j.neurobiolaging.2021.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
Recent studies have suggested ARSA, a gene responsible for metachromatic leukodystrophy, could be a genetic modifier of Parkinson's disease (PD) pathogenesis, acting as a molecular chaperone for α-synuclein. To elucidate the role of ARSA variants in PD, we did a comprehensive analysis of ARSA variants by performing next-generation sequencing on 477 PD families, 1440 sporadic early-onset PD patients and 1962 sporadic late-onset PD patients and 2636 controls from Chinese mainland, as well as the association between ARSA variants and cognitive function of PD patients. We identified 2 familial PD following autosomal dominant inherence carrying rare variants of ARSA, but they had limited clinical significance. We detected a total of 81 coding variants of ARSA in our subjects but none of the identified variants were associated with either susceptibility or cognitive performance of PD, while loss-of-function variants showed slightly increased burden in late-onset PD (0.25% vs. 0%, p = 0.08). Our results suggested ARSA may not play important roles in PD of Chinese population.
Collapse
Affiliation(s)
- Hong-Xu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Ge Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Wen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng-Huan Fang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yi Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xun Zhou
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Run-Cheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi-Ying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie-Qiong Tan
- Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xin-Xiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Centre for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| |
Collapse
|
49
|
Oji Y, Hatano T, Funayama M, Hattori N. Reply: PSAP variants in Parkinson's disease: a large cohort study in Chinese mainland population. Brain 2021; 144:e26. [PMID: 33844829 DOI: 10.1093/brain/awaa393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yutaka Oji
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Manabu Funayama
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Zhao YW, Pan HX, Zeng Q, Fang ZH, Liu ZH, Wang Y, Jiang L, He R, Zhou X, Zhou YJ, Xu Q, Sun QY, Li B, Zhao G, Yang Y, Hu YC, Chen YS, Du J, Lei LF, Zhang HN, Wang CY, Yan XX, Shen L, Jiang H, Li JC, Tang BS, Tan JQ, Guo JF. PSAP variants in Parkinson's disease: a large cohort study in Chinese mainland population. Brain 2021; 144:e25. [PMID: 33793763 DOI: 10.1093/brain/awaa391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yu-Wen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong-Xu Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng-Huan Fang
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhen-Hua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yige Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xun Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang-Jie Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi-Ying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Cen Hu
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Se Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li-Fang Lei
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hai-Nan Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chun-Yu Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-Xiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Jie-Qiong Tan
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| |
Collapse
|