1
|
Akhtar A, Singh P, Admane N, Grover A. Salvianolic acid B prevents the amyloid transformation of A53T mutant of α-synuclein. Biophys Chem 2025; 318:107379. [PMID: 39693815 DOI: 10.1016/j.bpc.2024.107379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder involving the progressive loss of dopaminergic neurons in the substantia nigra pars compacta triggered by the accumulation of amyloid aggregates of α-synuclein protein. This study investigates the potential of Salvianolic Acid B (SalB), a water-soluble polyphenol derived from Salvia miltiorrhiza Bunge, in modulating the aggregation of the A53T mutant of α-synuclein (A53T Syn). This mutation is associated with rapid aggregation and a higher rate of protofibril formation in early-onset familial PD. Computational and experimental approaches demonstrated Sal-B effectively prevents the amyloid fibrillation of A53T Syn by interacting with the N-terminal region and NAC domain. Sal-B particularly associates with the KTKEGV motif and NACore segment of A53T Syn by hydrophobic and hydrogen bonding interactions. Replica exchange molecular dynamics (REMD) simulations indicated that Sal-B reduces intramolecular hydrogen bonding and structural transitions into β-sheet rich conformations, thereby lowering the aggregation propensity of A53T Syn. Systematic analysis conducted using biophysical techniques and high-end microscopy has demonstrated significant inhibition in the amyloid transformation of A53T Syn corroborated by a 92 % decrease in ThT maxima at 100 μM Sal-B concentration and microscopic techniques validated the absence of mature fibrillar amyloids. DLS data revealed heterogeneous particle sizes, supporting the formation of smaller unstructured aggregates. These findings underscore Sal-B as a promising therapeutic candidate for PD and related synucleinopathies, warranting further investigation in cellular and animal models to advance potential treatments and early intervention strategies.
Collapse
Affiliation(s)
- Almas Akhtar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Payal Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nikita Admane
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
2
|
Parveen S, Alam P, Orrù CD, Vascellari S, Hughson AG, Zou WQ, Beach TG, Serrano GE, Goldstein DS, Ghetti B, Cossu G, Pisano G, Pinna B, Caughey B. A same day α-synuclein RT-QuIC seed amplification assay for synucleinopathy biospecimens. NPJ BIOSENSING 2025; 2:8. [PMID: 39950033 PMCID: PMC11813799 DOI: 10.1038/s44328-024-00023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/13/2024] [Indexed: 02/16/2025]
Abstract
Parkinson's disease (PD), dementia with Lewy bodies (DLB), and other synucleinopathies are characterized by the accumulation of abnormal, self-propagating aggregates of α-synuclein. RT-QuIC or seed amplification assays are currently showing unprecedented diagnostic sensitivities and specificities for synucleinopathies even in prodromal phases years in advance of the onset of Parkinsonian signs or dementia. However, commonly used α-synuclein seed amplification assays take ≥48 h to perform as applied to patients' diagnostic biospecimens. Here, we report the development of a faster α-synuclein RT-QuIC assay that is as analytically sensitive as prior assays of this type, but can be completed in ≤12 h for brain, skin, and intestinal mucosa, with positive signals often arising in <5 h. CSF assays took a few hours longer. Our same-day α-synuclein RT-QuIC (sdRT-QuIC) assay should increase the practicality, cost-effectiveness, and throughput of measurements of pathological forms of α-synuclein for fundamental research, clinical diagnosis, and therapeutics development.
Collapse
Affiliation(s)
- Sabiha Parveen
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Parvez Alam
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Christina D. Orrù
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Sarah Vascellari
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Andrew G. Hughson
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Wen-Quan Zou
- Departments of Pathology and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH USA
- Institute of Neurology, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province China
| | | | | | - David S. Goldstein
- Autonomic Medicine Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN USA
| | - Giovanni Cossu
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Giada Pisano
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Beatrice Pinna
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Byron Caughey
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| |
Collapse
|
3
|
Ren G, Wang Y, Tian H, Zhang K, Zhang H, Liu X, Chen Z. Multiple system atrophy related neurogenic bladder: mechanism and treatment. Neurol Sci 2025:10.1007/s10072-025-08002-3. [PMID: 39875674 DOI: 10.1007/s10072-025-08002-3] [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: 11/07/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Multiple system atrophy (MSA) is a progressive neurodegenerative disease characterized by its aggressive nature. Its main clinical features include autonomic dysfunction, Parkinson's disease, and cerebellar ataxia. METHODS We conducted a comprehensive review of the existing literature, exploring studies and reports related to the mechanisms and treatment of multiple system atrophy related neurogenic bladder. Our aim is to provide a detailed and up-to-date overview of its underlying pathophysiology and current therapeutic strategies. RESULTS AND CONCLUSION Neurogenic bladder, a common manifestation of MSA, often goes untreated or mistreated, significantly affecting patients' quality of life. Early-stage bladder dysfunction is frequent in MSA patients and correlates with disease severity. The mechanisms of MSA related neurogenic bladder are related to the autonomic nervous system, somatic nerves, frontal cortex, brainstem, and sacral medulla center. Currently, treatment for MSA related neurogenic bladder is mainly symptomatic, and specific drugs are lacking. Further in-depth research is needed to develop more effective therapeutic options that improve patients' quality of life and reduce the risk of complications.
Collapse
Affiliation(s)
- Gengqing Ren
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- Tsinghua University, Beijing, 100084, China
| | - Hao Tian
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kaige Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Han Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoxu Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhigang Chen
- Neurology Department One, Dongfang Hospital, Beijing University of Chinese Medicine, No. 6, Fangxingyuan Community, Fangzhuang, Fengtai District, Beijing, 100078, People's Republic of China.
| |
Collapse
|
4
|
Bayati A, McPherson PS. Alpha-synuclein, autophagy-lysosomal pathway, and Lewy bodies: Mutations, propagation, aggregation, and the formation of inclusions. J Biol Chem 2024; 300:107742. [PMID: 39233232 PMCID: PMC11460475 DOI: 10.1016/j.jbc.2024.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
Research into the pathophysiology of Parkinson's disease (PD) is a fast-paced pursuit, with new findings about PD and other synucleinopathies being made each year. The involvement of various lysosomal proteins, such as TFEB, TMEM175, GBA, and LAMP1/2, marks the rising awareness about the importance of lysosomes in PD and other neurodegenerative disorders. This, along with recent developments regarding the involvement of microglia and the immune system in neurodegenerative diseases, has brought about a new era in neurodegeneration: the role of proinflammatory cytokines on the nervous system, and their downstream effects on mitochondria, lysosomal degradation, and autophagy. More effort is needed to understand the interplay between neuroimmunology and disease mechanisms, as many of the mechanisms remain enigmatic. α-synuclein, a key protein in PD and the main component of Lewy bodies, sits at the nexus between lysosomal degradation, autophagy, cellular stress, neuroimmunology, PD pathophysiology, and disease progression. This review revisits some fundamental knowledge about PD while capturing some of the latest trends in PD research, specifically as it relates to α-synuclein.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| |
Collapse
|
5
|
Sorrentino ZA, Riklan J, Lloyd GM, Lucke-Wold BP, Mampre D, Quintin S, Zakare-Fagbamila R, Still M, Chandra V, Foote KD, Giasson BI, Hilliard JD. Neuronal tissue collection from intra-cranial instruments used in deep brain stimulation surgery for Parkinson's disease with implications for study of alpha-synuclein. Sci Rep 2024; 14:21641. [PMID: 39284884 PMCID: PMC11405830 DOI: 10.1038/s41598-024-72542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
Alpha-synuclein (αSyn) forms pathologic aggregates in Parkinson's disease (PD) and is implicated in mechanisms underlying neurodegeneration. While pathologic αSyn has been extensively studied, there is currently no method to evaluate αSyn within the brains of living patients. Patients with PD are often treated with deep brain stimulation (DBS) surgery in which surgical instruments are in direct contact with neuronal tissue; herein, we describe a method by which tissue is collected from DBS surgical instruments in PD and essential tremor (ET) patients and demonstrate that αSyn is detected. 24 patients undergoing DBS surgery for PD (17 patients) or ET (7 patients) were enrolled; from patient samples, 81.2 ± 44.8 µg of protein (n = 15), on average, was collected from surgical instruments. Light microscopy revealed axons, capillaries, and blood cells as the primary components of purified tissue (n = 3). ELISA assay further confirmed the presence of neuronal and glial tissue in DBS samples (n = 4). Further analysis was conducted using western blot, demonstrating that multiple αSyn antibodies are reactive in PD (n = 5) and ET (n = 3) samples; truncated αSyn (1-125 αSyn) was significantly increased in PD (n = 5) compared to ET (n = 3), in which αSyn misfolding is not expected (0.64 ± 0.25 vs. 0.25 ± 0.12, P = 0.046), thus showing that multiple forms of αSyn can be detected from living PD patients with this method.
Collapse
Affiliation(s)
- Zachary A Sorrentino
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA.
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA.
| | - Joshua Riklan
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
| | - Grace M Lloyd
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Brandon P Lucke-Wold
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - David Mampre
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Stephan Quintin
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rasheedat Zakare-Fagbamila
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Megan Still
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Vyshak Chandra
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kelly D Foote
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Benoit I Giasson
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Justin D Hilliard
- University of Florida College of Medicine, 1505 SW Archer Rd, Gainesville, FL, 32608, USA
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
6
|
Ying C, Zhang H, Wang T, Li Y, Mao W, Hu S, Zhao L, Cai Y. Plasma level of alpha-synuclein oligomers as a biomarker for isolated rapid eye movement sleep behavior disorder diagnosis and progression: a prospective cohort study. Front Neurol 2024; 15:1442173. [PMID: 39246606 PMCID: PMC11377258 DOI: 10.3389/fneur.2024.1442173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alpha-synuclein oligomers (o-α-syn) are pivotal in the pathogenesis of α-synucleinopathy. Isolated rapid eye movement (REM) sleep behavior disorder (iRBD) serves as an early indicator of the disease, offering insights into disease mechanisms and early intervention. Nevertheless, the diagnostic and predictive potential of o-α-syn in iRBD remains largely unexplored. This study aimed to evaluate the plasma levels of o-α-syn in patients and investigate their utility as biomarkers for diagnosis of and predicting phenoconversion in iRBD. Methods A total of 143 participants, including 77 polysomnography-confirmed iRBD patients and 66 normal controls (NC), were recruited for this longitudinal observational study. Baseline clinical assessments and plasma collection were conducted for all iRBD patients, with 72 of them undergoing regularly prospective follow-up assessments for parkinsonism or dementia. Plasma levels of o-α-syn were quantified using enzyme-linked immunosorbent assay, and were compared between groups using a general linear model adjusted for age and sex. The diagnostic performance of plasma o-α-syn in iRBD was evaluated by area under the receiver operating characteristic curve (AUC) with 95% CI. Cox regression analysis and Kaplan-Meier survival curves were employed to assess the predictive value of plasma o-α-syn for phenoconversion in iRBD. Results Plasma o-α-syn levels did not exhibit statistically significant differences among iRBD converter patients, iRBD nonconverter patients, and NC. The AUC for distinguishing NC from iRBD was 0.52 (95% CI: 0.42-0.62, p = 0.682). Spearman correlation analysis revealed a significant positive correlation between plasma o-α-syn levels and MOCA scores in the iRBD group (p < 0.001). Subgroup analyses indicated that iRBD patients with cognitive decline (p = 0.058) and depressive symptoms (p = 0.017) had notably lower o-α-syn levels compared to those without such symptoms. Over a median follow-up period of 5.83 years, 26 iRBD patients developed neurodegenerative synucleinopathies. Cox regression and Kaplan-Meier survival curve analyses indicated that plasma level of o-α-syn lacked a predictive value for disease conversion in iRBD patients. Conclusion Despite a potential role in the pathophysiology of iRBD, o-α-syn are not appropriate biomarkers for diagnosing or predicting disease progression. While this study offers insights into the pathogenesis of iRBD and neurodegenerative synucleinopathies, further large-scale longitudinal studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Chao Ying
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Hui Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ting Wang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yuan Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Mao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Songnian Hu
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Lifang Zhao
- Beijing Geriatric Medical Research Center, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Mazzotta GM, Conte C. Alpha Synuclein Toxicity and Non-Motor Parkinson's. Cells 2024; 13:1265. [PMID: 39120295 PMCID: PMC11311369 DOI: 10.3390/cells13151265] [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: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a common multisystem neurodegenerative disorder affecting 1% of the population over the age of 60 years. The main neuropathological features of PD are the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the presence of alpha synuclein (αSyn)-rich Lewy bodies both manifesting with classical motor signs. αSyn has emerged as a key protein in PD pathology as it can spread through synaptic networks to reach several anatomical regions of the body contributing to the appearance of non-motor symptoms (NMS) considered prevalent among individuals prior to PD diagnosis and persisting throughout the patient's life. NMS mainly includes loss of taste and smell, constipation, psychiatric disorders, dementia, impaired rapid eye movement (REM) sleep, urogenital dysfunction, and cardiovascular impairment. This review summarizes the more recent findings on the impact of αSyn deposits on several prodromal NMS and emphasizes the importance of early detection of αSyn toxic species in biofluids and peripheral biopsies as prospective biomarkers in PD.
Collapse
Affiliation(s)
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| |
Collapse
|
8
|
Li B, Xiao X, Bi M, Jiao Q, Chen X, Yan C, Du X, Jiang H. Modulating α-synuclein propagation and decomposition: Implications in Parkinson's disease therapy. Ageing Res Rev 2024; 98:102319. [PMID: 38719160 DOI: 10.1016/j.arr.2024.102319] [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: 04/03/2024] [Accepted: 04/27/2024] [Indexed: 05/14/2024]
Abstract
α-Synuclein (α-Syn) is closely related to the pathogenesis of Parkinson's disease (PD). Under pathological conditions, the conformation of α-syn changes and different forms of α-syn lead to neurotoxicity. According to Braak stages, α-syn can propagate in different brain regions, inducing neurodegeneration and corresponding clinical manifestations through abnormal aggregation of Lewy bodies (LBs) and lewy axons in different types of neurons in PD. So far, PD lacks early diagnosis biomarkers, and treatments are mainly targeted at some clinical symptoms. There is no effective therapy to delay the progression of PD. This review first summarized the role of α-syn in physiological and pathological states, and the relationship between α-syn and PD. Then, we focused on the origin, secretion, aggregation, propagation and degradation of α-syn as well as the important regulatory factors in these processes systematically. Finally, we reviewed some potential drug candidates for alleviating the abnormal aggregation of α-syn in order to provide valuable targets for the treatment of PD to cope with the occurrence and progression of this disease.
Collapse
Affiliation(s)
- Beining Li
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xue Xiao
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Mingxia Bi
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Qian Jiao
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xi Chen
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Chunling Yan
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China
| | - Xixun Du
- School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China.
| | - Hong Jiang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China; School of Basic Medicine, Medical College of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
9
|
Riegelman E, Xue KS, Wang JS, Tang L. Gut-Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson's Disease. Nutrients 2024; 16:2041. [PMID: 38999791 PMCID: PMC11243524 DOI: 10.3390/nu16132041] [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: 06/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
With the recognition of the importance of the gut-brain axis in Parkinson's disease (PD) etiology, there is increased interest in developing therapeutic strategies that target α-synuclein, the hallmark abhorrent protein of PD pathogenesis, which may originate in the gut. Research has demonstrated that inhibiting the aggregation, oligomerization, and fibrillation of α-synuclein are key strategies for disease modification. Polyphenols, which are rich in fruits and vegetables, are drawing attention for their potential role in this context. In this paper, we reviewed how polyphenols influence the composition and functional capabilities of the gut microbiota and how the resulting microbial metabolites of polyphenols may potentially enhance the modulation of α-synuclein aggregation. Understanding the interaction between polyphenols and gut microbiota and identifying which specific microbes may enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.
Collapse
Affiliation(s)
| | | | | | - Lili Tang
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, USA; (E.R.); (K.S.X.); (J.-S.W.)
| |
Collapse
|
10
|
Sorrentino Z, Riklan J, Lloyd G, Lucke-Wold B, Mampre D, Quintin S, Zakare-Fagbamila R, Still M, Chandra V, Foote K, Giasson B, Hilliard J. Analysis of alpha-synuclein harvested from intracranial instruments used in deep brain stimulation surgery for Parkinson's disease. RESEARCH SQUARE 2024:rs.3.rs-4369598. [PMID: 38826474 PMCID: PMC11142310 DOI: 10.21203/rs.3.rs-4369598/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alpha-synuclein (αSyn) forms pathologic aggregates in Parkinson's disease (PD) and is implicated in mechanisms underlying neurodegeneration. While pathologic αSyn has been extensively studied, there is currently no method to evaluate αSyn within the brains of living patients. Patients with PD are often treated with deep brain stimulation (DBS) surgery in which surgical instruments are in direct contact with neuronal tissue; herein, we describe a method by which tissue is purified from DBS surgical instruments in PD and essential tremor (ET) patients and demonstrate that αSyn is robustly detected. 24 patients undergoing DBS surgery for PD (17 patients) or ET (7 patients) were enrolled; from patient samples, 81.2 ± 44.8 μg protein (n=15) is able to be purified, with immunoblot assays specific for αSyn reactive in all tested samples. Light microscopy revealed axons and capillaries as the primary components of purified tissue (n=3). Further analysis was conducted using western blot, demonstrating that truncated αSyn (1-125 αSyn) was significantly increased in PD (n=5) compared to ET (n=3), in which αSyn misfolding is not expected (0.64 ± 0.25 vs. 0.25 ± 0.12, P = 0.046), thus showing that pathologic αSyn can be reliably purified from living PD patients with this method.
Collapse
|
11
|
Chatterjee D, D’Brant LY, Hiller BM, Marmion DJ, Sandoval IM, Luk KC, Manfredsson FP, Messer A, Kordower JH, Butler DC. ENGINEERED NANOBODIES WITH PROGRAMMABLE TARGET ANTIGEN PROTEOLYSIS (PTAP) FUSIONS REGULATE INTRACELLULAR ALPHA-SYNUCLEIN IN VITRO AND IN VIVO. RESEARCH SQUARE 2024:rs.3.rs-4088206. [PMID: 38585932 PMCID: PMC10996777 DOI: 10.21203/rs.3.rs-4088206/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Alpha-synuclein (αSyn) aggregation and the formation of Lewy pathology (LP) is a foundational pathophysiological phenomenon in synucleinopathies. Delivering therapeutic single-chain and single-domain antibodies that bind pathogenic targets can disrupt intracellular aggregation. The fusion of antibody fragments to a negatively-charged proteasomal targeting motif (PEST) creates bifunctional constructs that enhance both solubility and turnover. With sequence-specific point mutations of PEST sequences that modulate proteasomal degradation efficiency, we report the creation of Programmable Target Antigen Proteolysis (PTAP) technology that can provide graded control over the levels of target antigens. We have previously demonstrated our lead anti-αSyn intrabody, VH14-PEST, is capable of reducing the pathological burden of synucleinopathy in vitro and in vivo. Here, we report a family of fully humanized VH14-PTAP constructs for controllable, therapeutic targeting of intracellular α-Syn. In cells, we demonstrate successful target engagement and efficacy of VH14-hPEST intrabodies, and validate proof-of-principle in human cells using 3D human organoids derived from PD-patient induced pluripotent stem cells (iPSC). In two synuclein-based rat models, PTAP intrabodies attenuated nigral αSyn pathology, preserved nigrostriatal dopaminergic tone, and slowed the propagation of αSyn pathology. These data demonstrate the potency of intracellular αSyn targeting as a method to alleviate pathology and highlight the potential clinical utility of PTAP intrabodies.
Collapse
Affiliation(s)
- Diptaman Chatterjee
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Lianna Y. D’Brant
- Regenerative Research Foundation, Neural Stem Cell Institute, Rensselaer, NY 12144
| | - Benjamin M. Hiller
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - David J. Marmion
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Ivette M. Sandoval
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013
| | - Kelvin C. Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19147
| | - Fredric P. Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013
| | - Anne Messer
- Regenerative Research Foundation, Neural Stem Cell Institute, Rensselaer, NY 12144
- Department of Biomedical Sciences, University at Albany, Albany, NY 12208
| | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ 85281
| | - David C. Butler
- Regenerative Research Foundation, Neural Stem Cell Institute, Rensselaer, NY 12144
- Department of Biomedical Sciences, University at Albany, Albany, NY 12208
| |
Collapse
|
12
|
Jin H, Arase H. Neoself Antigens Presented on MHC Class II Molecules in Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:51-65. [PMID: 38467972 DOI: 10.1007/978-981-99-9781-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Major histocompatibility complex (MHC) class II molecules play a crucial role in immunity by presenting peptide antigens to helper T cells. Immune cells are generally tolerant to self-antigens. However, when self-tolerance is broken, immune cells attack normal tissues or cells, leading to the development of autoimmune diseases. Genome-wide association studies have shown that MHC class II is the gene most strongly associated with the risk of most autoimmune diseases. When misfolded self-antigens, called neoself antigens, are associated with MHC class II molecules in the endoplasmic reticulum, they are transported by the MHC class II molecules to the cell surface without being processed into peptides. Moreover, neoself antigens that are complexed with MHC class II molecules of autoimmune disease risk alleles exhibit distinct antigenicities compared to normal self-antigens, making them the primary targets of autoantibodies in various autoimmune diseases. Elucidation of the immunological functions of neoself antigens presented on MHC class II molecules is crucial for understanding the mechanism of autoimmune diseases.
Collapse
Affiliation(s)
- Hui Jin
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan.
| |
Collapse
|
13
|
Li HY, Liu DS, Zhang YB, Rong H, Zhang XJ. The interaction between alpha-synuclein and mitochondrial dysfunction in Parkinson's disease. Biophys Chem 2023; 303:107122. [PMID: 37839353 DOI: 10.1016/j.bpc.2023.107122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/18/2023] [Accepted: 10/07/2023] [Indexed: 10/17/2023]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder with the hallmark of abnormal aggregates of alpha-synuclein (α-syn) in Lewy bodies (LBs) and Lewy neurites (LNs). Currently, pathogenic α-syn and mitochondrial dysfunction have been considered as prominent roles that give impetus to the PD onset. This review describes the α-syn pathology and mitochondrial alterations in PD, and focuses on how α-syn interacts with multiple aspects of mitochondrial homeostasis in the pathogenesis of PD.
Collapse
Affiliation(s)
- Hong-Yan Li
- Department of Basic Medical College, Heilongjiang University of Chinese Medicine, Haerbin 150000, PR China
| | - De-Shui Liu
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Ying-Bo Zhang
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Hua Rong
- Department of Pathology, Qiqihar Medical University, Qiqihar 161006, PR China
| | - Xiao-Jie Zhang
- Department of Basic Medical College, Heilongjiang University of Chinese Medicine, Haerbin 150000, PR China; Heilongjiang Nursing College, Haerbin 150000, PR China.
| |
Collapse
|
14
|
Nakamura R, Tomizawa I, Iwai A, Ikeda T, Hirayama K, Chiu YW, Suzuki T, Tarutani A, Mano T, Iwata A, Toda T, Sohma Y, Kanai M, Hori Y, Tomita T. Photo-oxygenation of histidine residue inhibits α-synuclein aggregation. FASEB J 2023; 37:e23311. [PMID: 37962096 DOI: 10.1096/fj.202301533r] [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: 07/28/2023] [Revised: 10/14/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023]
Abstract
Aggregation of α-synuclein (α-syn) into amyloid is the pathological hallmark of several neurodegenerative disorders, including Parkinson disease, dementia with Lewy bodies, and multiple system atrophy. It is widely accepted that α-syn aggregation is associated with neurodegeneration, although the mechanisms are not yet fully understood. Therefore, the inhibition of α-syn aggregation is a potential therapeutic approach against these diseases. This study used the photocatalyst for α-syn photo-oxygenation, which selectively adds oxygen atoms to fibrils. Our findings demonstrate that photo-oxygenation using this photocatalyst successfully inhibits α-syn aggregation, particularly by reducing its seeding ability. Notably, we also discovered that photo-oxygenation of the histidine at the 50th residue in α-syn aggregates is responsible for the inhibitory effect. These findings indicate that photo-oxygenation of the histidine residue in α-syn is a potential therapeutic strategy for synucleinopathies.
Collapse
Affiliation(s)
- Reito Nakamura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ikumi Tomizawa
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Atsushi Iwai
- Laboratory of Synthetic Organic Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ikeda
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kota Hirayama
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yung Wen Chiu
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takanobu Suzuki
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Airi Tarutani
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tatsuo Mano
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Atsushi Iwata
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Youhei Sohma
- Laboratory of Synthetic Organic Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Motomu Kanai
- Laboratory of Synthetic Organic Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Hori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Greco M, Munir A, Musarò D, Coppola C, Maffia M. Restoring autophagic function: a case for type 2 diabetes mellitus drug repurposing in Parkinson's disease. Front Neurosci 2023; 17:1244022. [PMID: 38027497 PMCID: PMC10654753 DOI: 10.3389/fnins.2023.1244022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Parkinson's disease (PD) is a predominantly idiopathic pathological condition characterized by protein aggregation phenomena, whose main component is alpha-synuclein. Although the main risk factor is ageing, numerous evidence points to the role of type 2 diabetes mellitus (T2DM) as an etiological factor. Systemic alterations classically associated with T2DM like insulin resistance and hyperglycemia modify biological processes such as autophagy and mitochondrial homeostasis. High glucose levels also compromise protein stability through the formation of advanced glycation end products, promoting protein aggregation processes. The ability of antidiabetic drugs to act on pathways impaired in both T2DM and PD suggests that they may represent a useful tool to counteract the neurodegeneration process. Several clinical studies now in advanced stages are looking for confirmation in this regard.
Collapse
Affiliation(s)
- Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | - Anas Munir
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Lecce, Italy
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | - Chiara Coppola
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| |
Collapse
|
16
|
Yang B, Yang Z, Liu H, Qi H. Dynamic modelling and tristability analysis of misfolded α-synuclein degraded via autophagy in Parkinson's disease. Biosystems 2023; 233:105036. [PMID: 37726073 DOI: 10.1016/j.biosystems.2023.105036] [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: 06/22/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023]
Abstract
The widely-accepted hallmark pathology of Parkinson's disease (PD) is the presence of Lewy bodies with characteristic abnormal aggregated α-synuclein (αSyn). Growing physiological evidence suggests that there is a pivotal role for the autophagy-lysosome pathway (ALP) in the clearance of misfolded αSyn (αSyn∗). This work establishes a mathematical model for αSyn∗ degradation through the ALP. Qualitative simulations are used to uncover the tristable behavior of αSyn∗, i.e., the lower, medium, and upper steady states, which correspond to the healthy, critical, and disease stages of PD, respectively. Time series and codimension-1 bifurcation analysis suggest that the system shows tristability dynamics. Furthermore, variations in the key parameters influence the tristable dynamic behavior, and the distribution of tristable regions is exhibited more comprehensively in codimension-2 bifurcation diagrams. In addition, robustness analysis demonstrates that tristability is a robust property of the system. These results may be valuable in therapeutic strategies for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Bojie Yang
- School of Mathematical Sciences and LMIB, Beihang University, Beijing, 100191, People's Republic of China
| | - Zhuoqin Yang
- School of Mathematical Sciences and LMIB, Beihang University, Beijing, 100191, People's Republic of China.
| | - Heng Liu
- School of Mathematical Sciences and LMIB, Beihang University, Beijing, 100191, People's Republic of China
| | - Hong Qi
- Complex Systems Research Center, Shanxi University, Taiyuan, 030006, People's Republic of China.
| |
Collapse
|
17
|
Liu Z, Lemus J, Smirnova IV, Liu W. Rehabilitation for non-motor symptoms for patients with Parkinson's disease from an α-synuclein perspective: a narrative review. EXPLORATION OF NEUROPROTECTIVE THERAPY 2023; 3:235-257. [PMID: 37920444 PMCID: PMC10621781 DOI: 10.37349/ent.2023.00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/22/2023] [Indexed: 11/04/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder affecting aged population around the world. PD is characterized by neuronal Lewy bodies present in the substantia nigra of the midbrain and the loss of dopaminergic neurons with various motor and non-motor symptoms associated with the disease. The protein α-synuclein has been extensively studied for its contribution to PD pathology, as α-synuclein aggregates form the major component of Lewy bodies, a hallmark of PD. In this narrative review, the authors first focus on a brief explanation of α-synuclein aggregation and circumstances under which aggregation can occur, then present a hypothesis for PD pathogenesis in the peripheral nervous system (PNS) and how PD can spread to the central nervous system from the PNS via the transport of α-synuclein aggregates. This article presents arguments both for and against this hypothesis. It also presents various non-pharmacological rehabilitation approaches and management techniques for both motor and non-motor symptoms of PD and the related pathology. This review seeks to examine a possible hypothesis of PD pathogenesis and points to a new research direction focus on rehabilitation therapy for patients with PD. As various non-motor symptoms of PD appear to occur earlier than motor symptoms, more focus on the treatment of non-motor symptoms as well as a better understanding of the biochemical mechanisms behind those non-motor symptoms may lead to better long-term outcomes for patients with PD.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Orthopedic Surgery and Sports Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jessica Lemus
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Irina V. Smirnova
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
18
|
Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab 2023; 74:101755. [PMID: 37329949 PMCID: PMC10359950 DOI: 10.1016/j.molmet.2023.101755] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Recently, the hallmarks of aging were updated to include dysbiosis, disabled macroautophagy, and chronic inflammation. In particular, the low-grade chronic inflammation during aging, without overt infection, is defined as "inflammaging," which is associated with increased morbidity and mortality in the aging population. Emerging evidence suggests a bidirectional and cyclical relationship between chronic inflammation and the development of age-related conditions, such as cardiovascular diseases, neurodegeneration, cancer, and frailty. How the crosstalk between chronic inflammation and other hallmarks of aging underlies biological mechanisms of aging and age-related disease is thus of particular interest to the current geroscience research. SCOPE OF REVIEW This review integrates the cellular and molecular mechanisms of age-associated chronic inflammation with the other eleven hallmarks of aging. Extra discussion is dedicated to the hallmark of "altered nutrient sensing," given the scope of Molecular Metabolism. The deregulation of hallmark processes during aging disrupts the delicate balance between pro-inflammatory and anti-inflammatory signaling, leading to a persistent inflammatory state. The resultant chronic inflammation, in turn, further aggravates the dysfunction of each hallmark, thereby driving the progression of aging and age-related diseases. MAIN CONCLUSIONS The crosstalk between chronic inflammation and other hallmarks of aging results in a vicious cycle that exacerbates the decline in cellular functions and promotes aging. Understanding this complex interplay will provide new insights into the mechanisms of aging and the development of potential anti-aging interventions. Given their interconnectedness and ability to accentuate the primary elements of aging, drivers of chronic inflammation may be an ideal target with high translational potential to address the pathological conditions associated with aging.
Collapse
Affiliation(s)
- Jordan J Baechle
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA
| | - Nan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - Priya Makhijani
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - David Furman
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina.
| | - Daniel A Winer
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Price DL, Khan A, Angers R, Cardenas A, Prato MK, Bani M, Bonhaus DW, Citron M, Biere AL. In vivo effects of the alpha-synuclein misfolding inhibitor minzasolmin supports clinical development in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:114. [PMID: 37460603 PMCID: PMC10352257 DOI: 10.1038/s41531-023-00552-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Direct targeting of alpha-synuclein (ASYN) has emerged as a disease-modifying strategy for Parkinson's disease and other synucleinopathies which is being approached using both small molecule compounds and ASYN-targeted biologics. Minzasolmin (UCB0599) is an orally bioavailable and brain-penetrant small molecule ASYN misfolding inhibitor in clinical development as a disease-modifying therapeutic for Parkinson's disease. Herein the results of preclinical evaluations of minzasolmin that formed the basis for subsequent clinical development are described. Pharmacokinetic evaluations of intraperitoneal 1 and 5 mg/kg minzasolmin in wildtype mice revealed parallel and dose-proportional exposures in brain and plasma. Three-month administration studies in the Line 61 transgenic mouse model of PD were conducted to measure ASYN pathology and other PD-relevant endpoints including markers of CNS inflammation, striatal DAT labeling and gait. Reductions in ASYN pathology were correlated with improved aspects of gait and balance, reductions in CNS inflammation marker abundance, and normalized striatal DAT levels. These findings provide support for human dose determinations and have informed the translational strategy for clinical trial design and biomarker selection for the ongoing clinical studies of minzasolmin in patients living with early-stage Parkinson's disease (ClinicalTrials.gov ID: NCT04658186; EudraCT Number 2020-003265).
Collapse
Affiliation(s)
| | - Asma Khan
- Neuropore Therapies, Inc., San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Maity A, Mondal A, Kundu S, Shome G, Misra R, Singh A, Pal U, Mandal AK, Bera K, Maiti NC. Naringenin-Functionalized Gold Nanoparticles and Their Role in α-Synuclein Stabilization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:7231-7248. [PMID: 37094111 DOI: 10.1021/acs.langmuir.2c03259] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Misfolding and self-assembly of several intrinsically disordered proteins into ordered β-sheet-rich amyloid aggregates emerged as hallmarks of several neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Here we show how the naringenin-embedded nanostructure effectively retards aggregation and fibril formation of α-synuclein, which is strongly associated with the pathology of Parkinson's-like diseases. Naringenin is a polyphenolic compound from a plant source, and in our current investigation, we reported the one-pot synthesis of naringenin-coated spherical and monophasic gold nanoparticles (NAR-AuNPs) under optimized conditions. The average hydrodynamic diameter of the produced nanoparticle was ∼24 nm and showed a distinct absorption band at 533 nm. The zeta potential of the nanocomposite was ∼-22 mV and indicated the presence of naringenin on the surface of nanoparticles. Core-level XPS spectrum analysis showed prominent peaks at 84.02 and 87.68 eV, suggesting the zero oxidation state of metal in the nanostructure. Additionally, the peaks at 86.14 and 89.76 eV were due to the Au-O bond, induced by the hydroxyl groups of the naringenin molecule. The FT-IR analysis further confirmed strong interactions of the molecule with the gold nanosurface via the phenolic oxygen group. The composite surface was found to interact with monomeric α-synuclein and caused a red shift in the nanoparticle absorption band by ∼5 nm. The binding affinity of the composite nanostructure toward α-synuclein was in the micromolar range (Ka∼ 5.02 × 106 M-1) and may produce a protein corona over the gold nanosurface. A circular dichroism study showed that the nanocomposite can arrest the conformational fluctuation of the protein and hindered its transformation into a compact cross-β-sheet conformation, a prerequisite for amyloid fibril formation. Furthermore, it was found that naringenin and its nanocomplex did not perturb the viability of neuronal cells. It thus appeared that engineering of the nanosurface with naringenin could be an alternative strategy in developing treatment approaches for Parkinson's and other diseases linked to protein conformation.
Collapse
Affiliation(s)
- Anupam Maity
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Animesh Mondal
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Shubham Kundu
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Gourav Shome
- Division of Molecular Medicine, Bose Institute, Kolkata 700091, India
| | - Rajdip Misra
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Aakriti Singh
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Uttam Pal
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Atin Kumar Mandal
- Division of Molecular Medicine, Bose Institute, Kolkata 700091, India
| | - Kaushik Bera
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Department of Chemistry, The Heritage School, 994 Chowbaga Road, Anandapur, East Kolkata Twp, Kolkata 700107, India
| | - Nakul C Maiti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
21
|
Kang YJ, Xue Y, Shin JH, Cho H. Human mini-brains for reconstituting central nervous system disorders. LAB ON A CHIP 2023; 23:964-981. [PMID: 36644973 DOI: 10.1039/d2lc00897a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neurological disorders in the central nervous system (CNS) are progressive and irreversible diseases leading to devastating impacts on patients' life as they cause cognitive impairment, dementia, and even loss of essential body functions. The development of effective medicines curing CNS disorders is, however, one of the most ambitious challenges due to the extremely complex functions and structures of the human brain. In this regard, there are unmet needs to develop simplified but physiopathologically-relevant brain models. Recent advances in the microfluidic techniques allow multicellular culture forming miniaturized 3D human brains by aligning parts of brain regions with specific cells serving suitable functions. In this review, we overview designs and strategies of microfluidics-based human mini-brains for reconstituting CNS disorders, particularly Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI), vascular dementia (VD), and environmental risk factor-driven dementia (ERFD). Afterward, the applications of the mini-brains in the area of medical science are introduced in terms of the clarification of pathogenic mechanisms and identification of promising biomarkers. We also present expanded model systems ranging from the CNS to CNS-connecting organ axes to study the entry pathways of pathological risk factors into the brain. Lastly, the advantages and potential challenges of current model systems are addressed with future perspectives.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yingqi Xue
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Hee Shin
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
22
|
Ozono T, Kimura Y, Suenaga T, Beck G, Jinno J, Aguirre C, Ikenaka K, Krainc D, Mochizuki H, Arase H. Extracellular transportation of α-synuclein by HLA class II molecules. Biochem Biophys Res Commun 2023; 644:25-33. [PMID: 36621149 DOI: 10.1016/j.bbrc.2022.12.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive accumulation of α-synuclein aggregates in form of Lewy bodies. Genome-wide association studies have revealed that human leukocyte antigen (HLA) class II is a PD-associated gene, although the mechanisms linking HLA class II and PD remain elusive. Here, we identified a novel function of HLA class II in the transport of intracellular α-synuclein to the outside of cells. HLA class II molecules and α-synuclein formed complexes and moved to the cell surface at various degrees among HLA-DR alleles. HLA-DR with a DRB5∗01:01 allele, a putative PD-risk allele, substantially translocated normal and conformationally abnormal α-synuclein to the cell surface and extracellular vesicles. α-Synuclein/HLA class II complexes were found in A2058 melanoma cells, which express intrinsic α-synuclein and HLA-DR with DRB5∗01:01. Our findings will expand our knowledge of unconventional HLA class II function from autoimmune diseases to neurodegenerative disorders, shedding light on the association between the GWAS-prioritized PD-risk gene HLA-DR and α-synuclein.
Collapse
Affiliation(s)
- Tatsuhiko Ozono
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory of Immunochemistry, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuyoshi Kimura
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tadahiro Suenaga
- Laboratory of Immunochemistry, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Immunology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Goichi Beck
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jyunki Jinno
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - César Aguirre
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hisashi Arase
- Laboratory of Immunochemistry, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
23
|
Sakakibara R, Sekiguchi Y, N Panicker J, Sekido N, Sugimoto H, Sugisaki Y, Shimizu A, Takahashi O, Ogata T, Sawai S, Tateno F, Aiba Y, Simeoni S. Female Urinary Retention Progressing to Possible Multiple System Atrophy-cerebellar Form after 12 Years. Intern Med 2022; 61:3599-3604. [PMID: 35569977 PMCID: PMC9790793 DOI: 10.2169/internalmedicine.8724-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We herein report a 73-year-old Japanese woman with possible multiple system atrophy-cerebellar form (MSA-C) who suffered from urinary retention (sacral autonomic disorder) for 12 years before exhibiting cerebellar ataxia. A peculiar combination of findings on urodynamics and sphincter electromyography (EMG), e.g. detrusor hyperactivity with impaired contraction (DHIC), detrusor-sphincter dyssynergia (DSD) and neurogenic sphincter EMG (upper and lower neuron-type autonomic dysfunction), seems to have been predictive of future development of MSA.
Collapse
Affiliation(s)
- Ryuji Sakakibara
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, Japan
| | - Yuki Sekiguchi
- Uro-gynecology, Female Medical Clinic 'Next Stage', Japan
| | - Jalesh N Panicker
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, UK
| | | | | | | | - Ayami Shimizu
- Clinical Physiology Unit, Sakura Medical Center, Toho University, Japan
| | - Osamu Takahashi
- Clinical Physiology Unit, Sakura Medical Center, Toho University, Japan
| | - Tsuyoshi Ogata
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, Japan
| | - Setsu Sawai
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, Japan
| | - Fuyuki Tateno
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, Japan
| | - Yosuke Aiba
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, Japan
| | - Sara Simeoni
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, UK
| |
Collapse
|
24
|
Quantitative super-resolution imaging of pathological aggregates reveals distinct toxicity profiles in different synucleinopathies. Proc Natl Acad Sci U S A 2022; 119:e2205591119. [PMID: 36206368 PMCID: PMC9573094 DOI: 10.1073/pnas.2205591119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Protein aggregation is a hallmark of major neurodegenerative disorders. Increasing data suggest that smaller aggregates cause higher toxic response than filamentous aggregates (fibrils). However, the size of small aggregates has challenged their detection within biologically relevant environments. Here, we report approaches to quantitatively super-resolve aggregates in live cells and ex vivo brain tissues. We show that Amytracker 630 (AT630), a commercial aggregate-activated fluorophore, has outstanding photophysical properties that enable super-resolution imaging of α-synuclein, tau, and amyloid-β aggregates, achieving ∼4 nm precision. Applying AT630 to AppNL-G-F mouse brain tissues or aggregates extracted from a Parkinson's disease donor, we demonstrate excellent agreement with antibodies specific for amyloid-β or α-synuclein, respectively, confirming the specificity of AT630. Subsequently, we use AT630 to reveal a linear relationship between α-synuclein aggregate size and cellular toxicity and discovered that aggregates smaller than 450 ± 60 nm (aggregate450nm) readily penetrated the plasma membrane. We determine aggregate450nm concentrations in six Parkinson's disease and dementia with Lewy bodies donor samples and show that aggregates in different synucleinopathies demonstrate distinct potency in toxicity. We further show that cell-penetrating aggregates are surrounded by proteasomes, which assemble into foci to gradually process aggregates. Our results suggest that the plasma membrane effectively filters out fibrils but is vulnerable to penetration by aggregates of 450 ± 60 nm. Together, our findings present an exciting strategy to determine specificity of aggregate toxicity within heterogeneous samples. Our approach to quantitatively measure these toxic aggregates in biological environments opens possibilities to molecular examinations of disease mechanisms under physiological conditions.
Collapse
|
25
|
Therapeutic functions of astrocytes to treat α-synuclein pathology in Parkinson’s disease. Proc Natl Acad Sci U S A 2022; 119:e2110746119. [PMID: 35858361 PMCID: PMC9304026 DOI: 10.1073/pnas.2110746119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Intraneuronal inclusions of misfolded α-synuclein (α-syn) and prion-like spread of the pathologic α-syn contribute to progressive neuronal death in Parkinson’s disease (PD). Despite the pathologic significance, no efficient therapeutic intervention targeting α-synucleinopathy has been developed. In this study, we provide evidence that astrocytes, especially those cultured from the ventral midbrain (VM), show therapeutic potential to alleviate α-syn pathology in multiple in vitro and in vivo α-synucleinopathic models. Regulation of neuronal α-syn proteostasis underlies the therapeutic function of astrocytes. Specifically, VM-derived astrocytes inhibited neuronal α-syn aggregation and transmission in a paracrine manner by correcting not only intraneuronal oxidative and mitochondrial stresses but also extracellular inflammatory environments, in which α-syn proteins are prone to pathologic misfolding. The astrocyte-derived paracrine factors also promoted disassembly of extracellular α-syn aggregates. In addition to the aggregated form of α-syn, VM astrocytes reduced total α-syn protein loads both by actively scavenging extracellular α-syn fibrils and by a paracrine stimulation of neuronal autophagic clearance of α-syn. Transplantation of VM astrocytes into the midbrain of PD model mice alleviated α-syn pathology and protected the midbrain dopamine neurons from neurodegeneration. We further showed that cografting of VM astrocytes could be exploited in stem cell–based therapy for PD, in which host-to-graft transmission of α-syn pathology remains a critical concern for long-term cell therapeutic effects.
Collapse
|
26
|
Sidoroff V, Bower P, Stefanova N, Fanciulli A, Stankovic I, Poewe W, Seppi K, Wenning GK, Krismer F. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022? JOURNAL OF PARKINSON'S DISEASE 2022; 12:1369-1387. [PMID: 35491799 PMCID: PMC9398078 DOI: 10.3233/jpd-223183] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple system atrophy is a rapidly progressive and fatal neurodegenerative disorder. While numerous preclinical studies suggested efficacy of potentially disease modifying agents, none of those were proven to be effective in large-scale clinical trials. Three major strategies are currently pursued in preclinical and clinical studies attempting to slow down disease progression. These target α-synuclein, neuroinflammation, and restoration of neurotrophic support. This review provides a comprehensive overview on ongoing preclinical and clinical developments of disease modifying therapies. Furthermore, we will focus on potential shortcomings of previous studies that can be avoided to improve data quality in future studies of this rare disease.
Collapse
Affiliation(s)
- Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Pam Bower
- The Multiple System Atrophy Coalition, Inc., McLean, VA, USA
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Iva Stankovic
- Neurology Clinic, University Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krismer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Höllerhage M, Wolff A, Chakroun T, Evsyukov V, Duan L, Chua OWH, Tang Q, Koeglsperger T, Höglinger GU. Binding Stability of Antibody-α-Synuclein Complexes Predicts the Protective Efficacy of Anti-α-synuclein Antibodies. Mol Neurobiol 2022; 59:3980-3995. [PMID: 35460053 PMCID: PMC9167191 DOI: 10.1007/s12035-022-02824-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/01/2022] [Indexed: 01/01/2023]
Abstract
Spreading of alpha-synuclein (αSyn) may play an important role in Parkinson's disease and related synucleinopathies. Passive immunization with anti-αSyn antibodies is a promising method to slow down the spreading process and thereby the progression of synucleinopathies. Currently, it remains elusive which specific characteristics are essential to render therapeutic antibodies efficacious. Here, we established a neuronal co-culture model, in which αSyn species are being released from αSyn-overexpressing cells and induce toxicity in a priori healthy GFP-expressing cells. In this model, we investigated the protective efficacy of three anti-αSyn antibodies. Only two of these antibodies, one C-terminal and one N-terminal, protected from αSyn-induced toxicity by inhibiting the uptake of spreading-competent αSyn from the cell culture medium. Neither the binding epitope nor the affinity of the antibodies towards recombinant αSyn could explain differences in biological efficacy. However, both protective antibodies formed more stable antibody-αSyn complexes than the non-protective antibody. These findings indicate that the stability of antibody-αSyn complexes may be more important to confer protection than the binding epitope or affinity to recombinant αSyn.
Collapse
Affiliation(s)
- Matthias Höllerhage
- Department of Neurology, Hannover Medical School, Hannover, D-30625, Germany.
| | - Andreas Wolff
- Department of Neurology, Technical University of Munich (TUM), D-81675, Munich, Germany
| | - Tasnim Chakroun
- Department of Neurology, Technical University of Munich (TUM), D-81675, Munich, Germany
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), D-81377, Munich, Germany
| | - Valentin Evsyukov
- Department of Neurology, Hannover Medical School, Hannover, D-30625, Germany
| | - Linghan Duan
- Department of Neurology, Hannover Medical School, Hannover, D-30625, Germany
| | - Oscar Wing-Ho Chua
- Department of Neurology, Hannover Medical School, Hannover, D-30625, Germany
| | - Qilin Tang
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), D-81377, Munich, Germany
| | - Thomas Koeglsperger
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), D-81377, Munich, Germany
- Department of Neurology, Ludwig Maximilian University Munich, D-81377, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, Hannover Medical School, Hannover, D-30625, Germany
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), D-81377, Munich, Germany
| |
Collapse
|
28
|
Vidović M, Rikalovic MG. Alpha-Synuclein Aggregation Pathway in Parkinson's Disease: Current Status and Novel Therapeutic Approaches. Cells 2022; 11:cells11111732. [PMID: 35681426 PMCID: PMC9179656 DOI: 10.3390/cells11111732] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 01/27/2023] Open
Abstract
Following Alzheimer’s, Parkinson’s disease (PD) is the second-most common neurodegenerative disorder, sharing an unclear pathophysiology, a multifactorial profile, and massive social costs worldwide. Despite this, no disease-modifying therapy is available. PD is tightly associated with α-synuclein (α-Syn) deposits, which become organised into insoluble, amyloid fibrils. As a typical intrinsically disordered protein, α-Syn adopts a monomeric, random coil conformation in an aqueous solution, while its interaction with lipid membranes drives the transition of the molecule part into an α-helical structure. The central unstructured region of α-Syn is involved in fibril formation by converting to well-defined, β-sheet rich secondary structures. Presently, most therapeutic strategies against PD are focused on designing small molecules, peptides, and peptidomimetics that can directly target α-Syn and its aggregation pathway. Other approaches include gene silencing, cell transplantation, stimulation of intracellular clearance with autophagy promoters, and degradation pathways based on immunotherapy of amyloid fibrils. In the present review, we sum marise the current advances related to α-Syn aggregation/neurotoxicity. These findings present a valuable arsenal for the further development of efficient, nontoxic, and non-invasive therapeutic protocols for disease-modifying therapy that tackles disease onset and progression in the future.
Collapse
Affiliation(s)
- Marija Vidović
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
- Correspondence: ; Tel.: +38-16-4276-3221
| | - Milena G. Rikalovic
- Environment and Sustainable Development, Singidunum Univeristy, Danijelova 32, 11010 Belgrade, Serbia;
| |
Collapse
|
29
|
Glucocerebrosidase-associated Parkinson disease: Pathogenic mechanisms and potential drug treatments. Neurobiol Dis 2022; 166:105663. [DOI: 10.1016/j.nbd.2022.105663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/30/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
|
30
|
Prieto Huarcaya S, Drobny A, Marques ARA, Di Spiezio A, Dobert JP, Balta D, Werner C, Rizo T, Gallwitz L, Bub S, Stojkovska I, Belur NR, Fogh J, Mazzulli JR, Xiang W, Fulzele A, Dejung M, Sauer M, Winner B, Rose-John S, Arnold P, Saftig P, Zunke F. Recombinant pro-CTSD (cathepsin D) enhances SNCA/α-Synuclein degradation in α-Synucleinopathy models. Autophagy 2022; 18:1127-1151. [PMID: 35287553 PMCID: PMC9196656 DOI: 10.1080/15548627.2022.2045534] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by the abnormal intracellular accumulation of SNCA/α-synuclein. While the exact mechanisms underlying SNCA pathology are not fully understood, increasing evidence suggests the involvement of autophagy as well as lysosomal deficiencies. Because CTSD (cathepsin D) has been proposed to be the major lysosomal protease involved in SNCA degradation, its deficiency has been linked to the presence of insoluble SNCA conformers in the brain of mice and humans as well as to the transcellular transmission of SNCA aggregates. We here postulate that SNCA degradation can be enhanced by the application of the recombinant human proform of CTSD (rHsCTSD). Our results reveal that rHsCTSD is efficiently endocytosed by neuronal cells, correctly targeted to lysosomes and matured to an enzymatically active protease. In dopaminergic neurons derived from induced pluripotent stem cells (iPSC) of PD patients harboring the A53T mutation within the SNCA gene, we confirm the reduction of insoluble SNCA after treatment with rHsCTSD. Moreover, we demonstrate a decrease of pathological SNCA conformers in the brain and within primary neurons of a ctsd-deficient mouse model after dosing with rHsCTSD. Boosting lysosomal CTSD activity not only enhanced SNCA clearance in human and murine neurons as well as tissue, but also restored endo-lysosome and autophagy function. Our findings indicate that CTSD is critical for SNCA clearance and function. Thus, enzyme replacement strategies utilizing CTSD may also be of therapeutic interest for the treatment of PD and other synucleinopathies aiming to decrease the SNCA burden.Abbreviations: aa: amino acid; SNCA/α-synuclein: synuclein alpha; APP: amyloid beta precursor protein; BBB: blood brain barrier; BF: basal forebrain; CBB: Coomassie Brilliant Blue; CLN: neuronal ceroid lipofuscinosis; CNL10: neuronal ceroid lipofuscinosis type 10; Corr.: corrected; CTSD: cathepsin D; CTSB: cathepsin B; DA: dopaminergic; DA-iPSn: induced pluripotent stem cell-derived dopaminergic neurons; dox: doxycycline; ERT: enzyme replacement therapy; Fx: fornix, GBA/β-glucocerebrosidase: glucosylceramidase beta; h: hour; HC: hippocampus; HT: hypothalamus; i.c.: intracranially; IF: immunofluorescence; iPSC: induced pluripotent stem cell; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LSDs: lysosomal storage disorders; MAPT: microtubule associated protein tau; M6P: mannose-6-phosphate; M6PR: mannose-6-phosphate receptor; MB: midbrain; mCTSD: mature form of CTSD; neurofil.: neurofilament; PD: Parkinson disease; proCTSD: proform of CTSD; PRNP: prion protein; RFU: relative fluorescence units; rHsCTSD: recombinant human proCTSD; SAPC: Saposin C; SIM: structured illumination microscopy; T-insol: Triton-insoluble; T-sol: Triton-soluble; TEM: transmission electron microscopy, TH: tyrosine hydroxylase; Thal: thalamus.
Collapse
Affiliation(s)
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), Nova Medical School, Nms, Nova University Lisbon, Lisboa, Portugal
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Denise Balta
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Christian Werner
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Tania Rizo
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Gallwitz
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Simon Bub
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Iva Stojkovska
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Nandkishore R Belur
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | | | - Joseph R Mazzulli
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| | - Amitkumar Fulzele
- Institute of Molecular Biology (IMB), Ackermannweg 4, Mainz, Germany
| | - Mario Dejung
- Institute of Molecular Biology (IMB), Ackermannweg 4, Mainz, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Am Hubland, Würzburg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (Fau), Erlangen, Germany
| |
Collapse
|
31
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
32
|
Mechanisms of enhanced aggregation and fibril formation of Parkinson's disease-related variants of α-synuclein. Sci Rep 2022; 12:6770. [PMID: 35474118 PMCID: PMC9043213 DOI: 10.1038/s41598-022-10789-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/08/2022] [Indexed: 12/18/2022] Open
Abstract
Aggregation of α-synuclein (α-syn) into amyloid fibrils is closely associated with Parkinson’s disease (PD). Familial mutations or posttranslational truncations in α-syn are known as risk factor for PD. Here, we examined the effects of the PD-related A30P or A53T point mutation and C-terminal 123–140 or 104–140 truncation on the aggregating property of α-syn based on the kinetic and thermodynamic analyses. Thioflavin T fluorescence measurements indicated that A53T, Δ123‒140, and Δ104–140 variants aggregated faster than WT α-syn, in which the A53T mutation markedly increases nucleation rate whereas the Δ123‒140 or Δ104‒140 truncation significantly increases both nucleation and fibril elongation rates. Ultracentrifugation and western blotting analyses demonstrated that these mutations or truncations promote the conversion of monomer to aggregated forms of α-syn. Analysis of the dependence of aggregation reaction of α-syn variants on the monomer concentration suggested that the A53T mutation enhances conversion of monomers to amyloid nuclei whereas the C-terminal truncations, especially the Δ104–140, enhance autocatalytic aggregation on existing fibrils. In addition, thermodynamic analysis of the kinetics of nucleation and fibril elongation of α-syn variants indicated that both nucleation and fibril elongation of WT α-syn are enthalpically and entropically unfavorable. Interestingly, the unfavorable activation enthalpy of nucleation greatly decreases for the A53T and becomes reversed in sign for the C-terminally truncated variants. Taken together, our results indicate that the A53T mutation and the C-terminal truncation enhance α-syn aggregation by reducing unfavorable activation enthalpy of nucleation, and the C-terminal truncation further triggers the autocatalytic fibril elongation on the fibril surfaces.
Collapse
|
33
|
α-Synuclein phosphorylation at serine 129 occurs after initial protein deposition and inhibits seeded fibril formation and toxicity. Proc Natl Acad Sci U S A 2022; 119:e2109617119. [PMID: 35353605 PMCID: PMC9169642 DOI: 10.1073/pnas.2109617119] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
SignificanceConverging evidence points to the build-up of phosphorylated α-synuclein (α-syn) at residue serine 129 (pS129) in Lewy body disease, suggesting its central role in the regulation of α-syn aggregation and neuronal degeneration. However, a comprehensive understanding of the role of α-syn phosphorylation at pS129 in α-synuclenopathies pathogenesis is still lacking. Herein, we study the phosphorylation incidence and its effect on α-syn aggregation propensity and cellular toxicity. Collectively, our data suggest that pS129 occurred subsequent to initial α-syn aggregation, lessened aggregation propensity, and attenuated cytotoxicity through diverse assays. Our findings highlight major implications for a better understanding of the role of a molecular modification on protein aggregation.
Collapse
|
34
|
Abstract
The notion that autoimmune responses to α-synuclein may be involved in the pathogenesis of this disorder stems from reports that mutations in α-synuclein or certain alleles of the major histocompatibility complex (MHC) are associated with the disease and that dopaminergic and norepinephrinergic neurons in the midbrain can present antigenic epitopes. Here, we discuss recent evidence that a defined set of peptides derived from α-synuclein act as antigenic epitopes displayed by specific MHC alleles and drive helper and cytotoxic T cell responses in patients with PD. Moreover, phosphorylated α-synuclein may activate T cell responses in a less restricted manner in PD. While the roles for the acquired immune system in disease pathogenesis remain unknown, preclinical animal models and in vitro studies indicate that T cells may interact with neurons and exert effects related to neuronal death and neuroprotection. These findings suggest that therapeutics that target T cells and ameliorate the incidence or disease severity of inflammatory bowel disorders or CNS autoimmune diseases such as multiple sclerosis may be useful in PD.
Collapse
|
35
|
Grosso Jasutkar H, Oh SE, Mouradian MM. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson's Disease. Pharmacol Rev 2022; 74:207-237. [PMID: 35017177 PMCID: PMC11034868 DOI: 10.1124/pharmrev.120.000133] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and the fastest growing neurologic disease in the world, yet no disease-modifying therapy is available for this disabling condition. Multiple lines of evidence implicate the protein α-synuclein (α-Syn) in the pathogenesis of PD, and as such, there is intense interest in targeting α-Syn for potential disease modification. α-Syn is also a key pathogenic protein in other synucleionpathies, most commonly dementia with Lewy bodies. Thus, therapeutics targeting this protein will have utility in these disorders as well. Here we discuss the various approaches that are being investigated to prevent and mitigate α-Syn toxicity in PD, including clearing its pathologic aggregates from the brain using immunization strategies, inhibiting its misfolding and aggregation, reducing its expression level, enhancing cellular clearance mechanisms, preventing its cell-to-cell transmission within the brain and perhaps from the periphery, and targeting other proteins associated with or implicated in PD that contribute to α-Syn toxicity. We also discuss the therapeutics in the pipeline that harness these strategies. Finally, we discuss the challenges and opportunities for the field in the discovery and development of therapeutics for disease modification in PD. SIGNIFICANCE STATEMENT: PD is the second most common neurodegenerative disorder, for which disease-modifying therapies remain a major unmet need. A large body of evidence points to α-synuclein as a key pathogenic protein in this disease as well as in dementia with Lewy bodies, making it of leading therapeutic interest. This review discusses the various approaches being investigated and progress made to date toward discovering and developing therapeutics that would slow and stop progression of these disabling diseases.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
36
|
Nishitsuji K, Uchimura K. Contribution of Sulfated Glycosaminoglycans to the Pathology of Amyloidosis. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2105.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | - Kenji Uchimura
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université de Lille
| |
Collapse
|
37
|
Nishitsuji K, Uchimura K. Contribution of Sulfated Glycosaminoglycans to the Pathology of Amyloidosis. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2105.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | - Kenji Uchimura
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université de Lille
| |
Collapse
|
38
|
Glycoconjugate journal special issue on: the glycobiology of Parkinson's disease. Glycoconj J 2021; 39:55-74. [PMID: 34757539 DOI: 10.1007/s10719-021-10024-w] [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: 06/26/2021] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects over 10 million aging people worldwide. This condition is characterized by the degeneration of dopaminergic neurons in the pars compacta region of the substantia nigra (SNpc) and by aggregation of proteins, commonly α-synuclein (SNCA). The formation of Lewy bodies that encapsulate aggregated proteins in lipid vesicles is a hallmark of PD. Glycosylation of proteins and neuroinflammation are involved in the pathogenesis. SNCA has many posttranslational modifications and interacts with components of membranes that affect aggregation. The large membrane lipid dolichol accumulates in the brain upon age and has a significant effect on membrane structure. The replacement of dopamine and dopaminergic neurons are at the forefront of therapeutic development. This review examines the role of membrane lipids, glycolipids, glycoproteins and dopamine in the aggregation of SNCA and development of PD. We discuss the SNCA-dopamine-neuromelanin-dolichol axis and the role of membranes in neuronal stem cells that could be a regenerative therapy for PD patients.
Collapse
|
39
|
Jellinger KA, Wenning GK, Stefanova N. Is Multiple System Atrophy a Prion-like Disorder? Int J Mol Sci 2021; 22:10093. [PMID: 34576255 PMCID: PMC8472631 DOI: 10.3390/ijms221810093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive, fatal neurodegenerative disease of uncertain aetiology that belongs to the family of α-synucleinopathies. It clinically presents with parkinsonism, cerebellar, autonomic, and motor impairment in variable combinations. Pathological hallmarks are fibrillary α-synuclein (αSyn)-rich glial cytoplasmic inclusions (GCIs) mainly involving oligodendroglia and to a lesser extent neurons, inducing a multisystem neurodegeneration, glial activation, and widespread demyelinization. The neuronal αSyn pathology of MSA has molecular properties different from Lewy bodies in Parkinson's disease (PD), both of which could serve as a pool of αSyn (prion) seeds that could initiate and drive the pathogenesis of synucleinopathies. The molecular cascade leading to the "prion-like" transfer of "strains" of aggregated αSyn contributing to the progression of the disease is poorly understood, while some presented evidence that MSA is a prion disease. However, this hypothesis is difficult to reconcile with postmortem analysis of human brains and the fact that MSA-like pathology was induced by intracerebral inoculation of human MSA brain homogenates only in homozygous mutant 53T mice, without production of disease-specific GCIs, or with replication of MSA prions in primary astrocyte cultures from transgenic mice expressing human αSyn. Whereas recent intrastriatal injection of Lewy body-derived or synthetic human αSyn fibrils induced PD-like pathology including neuronal αSyn aggregates in macaques, no such transmission of αSyn pathology in non-human primates by MSA brain lysate has been reported until now. Given the similarities between αSyn and prions, there is a considerable debate whether they should be referred to as "prions", "prion-like", "prionoids", or something else. Here, the findings supporting the proposed nature of αSyn as a prion and its self-propagation through seeding as well as the transmissibility of neurodegenerative disorders are discussed. The proof of disease causation rests on the concordance of scientific evidence, none of which has provided convincing evidence for the classification of MSA as a prion disease or its human transmission until now.
Collapse
Affiliation(s)
| | - Gregor K. Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| |
Collapse
|
40
|
All Roads Lead to Rome: Different Molecular Players Converge to Common Toxic Pathways in Neurodegeneration. Cells 2021; 10:cells10092438. [PMID: 34572087 PMCID: PMC8468417 DOI: 10.3390/cells10092438] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple neurodegenerative diseases (NDDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD) are being suggested to have common cellular and molecular pathological mechanisms, characterized mainly by protein misfolding and aggregation. These large inclusions, most likely, represent an end stage of a molecular cascade; however, the soluble misfolded proteins, which take part in earlier steps of this cascade, are the more toxic players. These pathological proteins, which characterize each specific disease, lead to the selective vulnerability of different neurons, likely resulting from a combination of different intracellular mechanisms, including mitochondrial dysfunction, ER stress, proteasome inhibition, excitotoxicity, oxidative damage, defects in nucleocytoplasmic transport, defective axonal transport and neuroinflammation. Damage within these neurons is enhanced by damage from the nonneuronal cells, via inflammatory processes that accelerate the progression of these diseases. In this review, while acknowledging the hallmark proteins which characterize the most common NDDs; we place specific focus on the common overlapping mechanisms leading to disease pathology despite these different molecular players and discuss how this convergence may occur, with the ultimate hope that therapies effective in one disease may successfully translate to another.
Collapse
|
41
|
Emmenegger M, De Cecco E, Hruska‐Plochan M, Eninger T, Schneider MM, Barth M, Tantardini E, de Rossi P, Bacioglu M, Langston RG, Kaganovich A, Bengoa‐Vergniory N, Gonzalez‐Guerra A, Avar M, Heinzer D, Reimann R, Häsler LM, Herling TW, Matharu NS, Landeck N, Luk K, Melki R, Kahle PJ, Hornemann S, Knowles TPJ, Cookson MR, Polymenidou M, Jucker M, Aguzzi A. LAG3 is not expressed in human and murine neurons and does not modulate α-synucleinopathies. EMBO Mol Med 2021; 13:e14745. [PMID: 34309222 PMCID: PMC8422075 DOI: 10.15252/emmm.202114745] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/09/2022] Open
Abstract
While the initial pathology of Parkinson's disease and other α-synucleinopathies is often confined to circumscribed brain regions, it can spread and progressively affect adjacent and distant brain locales. This process may be controlled by cellular receptors of α-synuclein fibrils, one of which was proposed to be the LAG3 immune checkpoint molecule. Here, we analysed the expression pattern of LAG3 in human and mouse brains. Using a variety of methods and model systems, we found no evidence for LAG3 expression by neurons. While we confirmed that LAG3 interacts with α-synuclein fibrils, the specificity of this interaction appears limited. Moreover, overexpression of LAG3 in cultured human neural cells did not cause any worsening of α-synuclein pathology ex vivo. The overall survival of A53T α-synuclein transgenic mice was unaffected by LAG3 depletion, and the seeded induction of α-synuclein lesions in hippocampal slice cultures was unaffected by LAG3 knockout. These data suggest that the proposed role of LAG3 in the spreading of α-synucleinopathies is not universally valid.
Collapse
Affiliation(s)
- Marc Emmenegger
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | - Elena De Cecco
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | | | - Timo Eninger
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Matthias M Schneider
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
| | - Melanie Barth
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Elena Tantardini
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Pierre de Rossi
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Mehtap Bacioglu
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Rebekah G Langston
- Cell Biology and Gene Expression SectionLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMDUSA
| | - Alice Kaganovich
- Cell Biology and Gene Expression SectionLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMDUSA
| | - Nora Bengoa‐Vergniory
- Department of Physiology, Anatomy and GeneticsOxford Parkinson’s Disease Center (OPDC)Oxford UniversityOxfordUK
| | | | - Merve Avar
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | - Daniel Heinzer
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | - Regina Reimann
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | - Lisa M Häsler
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Therese W Herling
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
| | - Naunehal S Matharu
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
| | - Natalie Landeck
- Cell Biology and Gene Expression SectionLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMDUSA
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine and Center for Neurodegenerative Disease ResearchUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Ronald Melki
- Laboratory of Neurodegenerative DiseasesCNRSInstitut François Jacob (MIRCen)CEAFontenay‐aux‐RosesFrance
| | - Philipp J Kahle
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of NeurodegenerationHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Simone Hornemann
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| | - Tuomas P J Knowles
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeCambridgeUK
- Cavendish LaboratoryDepartment of PhysicsUniversity of CambridgeCambridgeUK
| | - Mark R Cookson
- Cell Biology and Gene Expression SectionLaboratory of NeurogeneticsNational Institute on AgingNational Institutes of HealthBethesdaMDUSA
| | | | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Adriano Aguzzi
- Institute of NeuropathologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
42
|
Hass EW, Sorrentino ZA, Xia Y, Lloyd GM, Trojanowski JQ, Prokop S, Giasson BI. Disease-, region- and cell type specific diversity of α-synuclein carboxy terminal truncations in synucleinopathies. Acta Neuropathol Commun 2021; 9:146. [PMID: 34454615 PMCID: PMC8403399 DOI: 10.1186/s40478-021-01242-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/14/2022] Open
Abstract
Synucleinopathies, including Parkinson's disease (PD), Lewy body dementia (LBD), Alzheimer's disease with amygdala restricted Lewy bodies (AD/ALB), and multiple system atrophy (MSA) comprise a spectrum of neurodegenerative disorders characterized by the presence of distinct pathological α-synuclein (αSyn) inclusions. Experimental and pathological studies support the notion that αSyn aggregates contribute to cellular demise and dysfunction with disease progression associated with a prion-like spread of αSyn aggregates via conformational templating. The initiating event(s) and factors that contribute to diverse forms of synucleinopathies remain poorly understood. A major post-translational modification of αSyn associated with pathological inclusions is a diverse array of specific truncations within the carboxy terminal region. While these modifications have been shown experimentally to induce and promote αSyn aggregation, little is known about their disease-, region- and cell type specific distribution. To this end, we generated a series of monoclonal antibodies specific to neo-epitopes in αSyn truncated after residues 103, 115, 119, 122, 125, and 129. Immunocytochemical investigations using these new tools revealed striking differences in the αSyn truncation pattern between different synucleinopathies, brain regions and specific cellular populations. In LBD, neuronal inclusions in the substantia nigra and amygdala were positive for αSyn cleaved after residues 103, 119, 122, and 125, but not 115. In contrast, in the same patients' brain αSyn cleaved at residue 115, as well as 103, 119 and 122 were abundant in the dorsal motor nucleus of the vagus. In patients with AD/ALB, these modifications were only weakly or not detected in amygdala αSyn inclusions. αSyn truncated at residues 103, 115, 119, and 125 was readily present in MSA glial cytoplasmic inclusions, but 122 cleaved αSyn was only weakly or not present. Conversely, MSA neuronal pathology in the pontine nuclei was strongly reactive to the αSyn x-122 neo-epitope but did not display any reactivity for αSyn 103 cleavage. These studies demonstrate significant disease-, region- and cell type specific differences in carboxy terminal αSyn processing associated with pathological inclusions that likely contributes to their distinct strain-like prion properties and promotes the diversity displayed in the degrees of these insidious diseases.
Collapse
Affiliation(s)
- Ethan W Hass
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary A Sorrentino
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Grace M Lloyd
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, AD Center Core (ADCC), Center for Neurodegenerative Disease Research, PENN) School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
43
|
Gegg ME, Verona G, Schapira AHV. Glucocerebrosidase deficiency promotes release of α-synuclein fibrils from cultured neurons. Hum Mol Genet 2021; 29:1716-1728. [PMID: 32391886 PMCID: PMC7322566 DOI: 10.1093/hmg/ddaa085] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/17/2023] Open
Abstract
Mutations in the GBA gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase), are the most important genetic risk factor for Parkinson disease (PD). GCase activity is also decreased in sporadic PD brains and with normal ageing. Loss of GCase activity impairs the autophagy lysosomal pathway resulting in increased α-synuclein (α-syn) levels. Furthermore, elevated α-syn results in decreased GCase activity. Although the role of α-syn in PD remains unclear, evidence indicates that aggregated α-syn fibrils are a pathogenic species in PD, passing between neurons and inducing endogenous native α-syn to aggregate; spreading pathology through the brain. We have investigated if preformed α-syn fibrils (PFFs) impair GCase activity in mouse cortical neurons and differentiated dopaminergic cells, and whether GCase deficiency in these models increased the transfer of α-syn pathology to naïve cells. Neurons treated with PFFs induced endogenous α-syn to become insoluble and phosphorylated at Ser129 to a greater extent than monomeric α-syn-treatment. PFFs, but not monomeric α-syn, inhibited lysosomal GCase activity in these cells and induced the unfolded protein response. Neurons in which GCase was inhibited by conduritol β-epoxide did not increase the amount of insoluble monomeric α-syn or its phosphorylation status. Instead the release of α-syn fibrils from GCase deficient cells was significantly increased. Co-culture studies showed that the transfer of α-syn pathology to naïve cells was greater from GCase deficient cells. This study suggests that GCase deficiency increases the spread of α-syn pathology and likely contributes to the earlier age of onset and increased cognitive decline associated with GBA-PD.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| | - Guglielmo Verona
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
44
|
Ko DWK. Transcutaneous vagus nerve stimulation (tVNS) as a potential therapeutic application for neurodegenerative disorders - A focus on dysautonomia in Parkinson's disease. Auton Neurosci 2021; 235:102858. [PMID: 34365230 DOI: 10.1016/j.autneu.2021.102858] [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: 05/29/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/29/2022]
Abstract
The understandings of pathogenic processes in major neurodegenerative diseases has significantly advanced in recent years, with evidence showing pathological spread of intraneuronal proteinaceous inclusions as a fundamental factor. In Parkinson's disease (PD), the culprit protein has been identified as α-synuclein as the main component for mediating progressive neurodegeneration. With severe pathology evident in the autonomic nervous system prior to clinical manifestations of PD, pathogenic spread can occur from the peripheral nervous system through key nuclei, such as the anterior olfactory nucleus and dorsal motor nucleus of the glossopharyngeal and vagal nerves, gradually reaching the brainstem, midbrain and cerebral cortex. With this understanding and the proposed involvement of the vagus nerve in disease progression in PD, notably occurring prior to characterized clinical motor features, it raises intriguing questions as to whether vagal nerve pathology can be accurately detected, and importantly used as a reliable marker for determining early neurodegeneration. Along with this is the potential use of vagus nerve neuromodulation for treatment of early disease symptoms like dysautonomia, for modulating sympatho-vagal imbalances and easing severe comorbidities of the disease. In this article, we take a closer look at the pathogenic transmission processes in neurodegenerative disorders that impact the vagus nerve, and how vagus nerve neuromodulation can be potentially applied as a therapeutic approach for major neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel W K Ko
- Neuropix Company Ltd, Core F, Cyberport 3, 100 Cyberport Road, Hong Kong Special Administrative Region.
| |
Collapse
|
45
|
Beekes M. The Neural Gut-Brain Axis of Pathological Protein Aggregation in Parkinson's Disease and Its Counterpart in Peroral Prion Infections. Viruses 2021; 13:1394. [PMID: 34372600 PMCID: PMC8310171 DOI: 10.3390/v13071394] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
A neuropathological hallmark of Parkinson's disease (PD) is the cerebral deposition of abnormally aggregated α-synuclein (αSyn). PD-associated αSyn (αSynPD) aggregates are assumed to act, in a prion-like manner, as proteinaceous nuclei ("seeds") capable of self-templated propagation. Braak and colleagues put forward the idea of a neural gut-brain axis mediating the centripetal spread of αSynPD pathology from the enteric nervous system (ENS) to the brain in PD. This has sparked great interest and initiated passionate discussions both in support of and opposing the suggested hypothesis. A precedent for the spread of protein seeds or seeding from the gastro-intestinal (GI) tract to the central nervous system (CNS) had been previously revealed for pathological prion protein in peroral prion infections. This article scrutinizes the similarities and dissimilarities between the pathophysiological spread of disease-associated protein aggregation along the neural gut-brain axis in peroral prion infections and PD. On this basis, evidence supporting the proposed neural gut-brain axis in PD is concluded to be not as robust as that established for peroral prion infections. New tools for the ultrasensitive detection of αSynPD-associated seeding activity in archived or fresh human tissue samples such as real-time quaking induced conversion (RT-QuIC) or protein misfolding cyclic amplification (PMCA) assays can possibly help to address this deficit in the future.
Collapse
Affiliation(s)
- Michael Beekes
- Prion and Prionoid Research Unit, ZBS 6-Proteomics and Spectroscopy, ZBS-Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| |
Collapse
|
46
|
Zheng T, Zhang Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson's disease. Neurochem Int 2021; 148:105094. [PMID: 34097990 DOI: 10.1016/j.neuint.2021.105094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and abnormal aggregates of α-synuclein protein called Lewy bodies. To date, there is no drug that can definitely slow down or stop the progression of this disease. The discovery of the cell-to-cell transmission of pathologic α-synuclein seeds offers the possibility to explore novel treatment strategies to prevent the spread of α-synuclein, with the purpose of slowing down the progression of PD in its tracks. Although recent studies have made tremendous progress in understanding how α-synuclein spreads throughout the brain, neuroinflammation seems to play a crucial role in the development of α-synuclein pathology in PD. The activation of microglia, one of the hallmarks of the neuroinflammatory process, is suggested to influence the neuron-to-neuron transmission of α-synuclein. This review summarizes how activated microglia facilitate this process, and focuses on the following mechanisms including the activation of microglia in PD, the reduced ability of activated microglia to clear α-synuclein and increased migratory capacity of microglia in PD, as well as the cooperation between microglia and exosomes in mediating α-synuclein release and propagation. In conclusion, this article help collate information on microglia in-relation to PD.
Collapse
Affiliation(s)
- Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China
| | - Zhengxiang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
47
|
Shen L, Wang C, Chen L, Wong G. Dysregulation of MicroRNAs and PIWI-Interacting RNAs in a Caenorhabditis elegans Parkinson's Disease Model Overexpressing Human α-Synuclein and Influence of tdp-1. Front Neurosci 2021; 15:600462. [PMID: 33762903 PMCID: PMC7982545 DOI: 10.3389/fnins.2021.600462] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) and PIWI-interacting RNAs (piRNAs) regulate gene expression and biological processes through specific genetic and epigenetic mechanisms. Recent studies have described a dysregulation of small non-coding RNAs in Parkinson’s disease (PD) tissues but have been limited in scope. Here, we extend these studies by comparing the dysregulation of both miRNAs and piRNAs from transgenic Caenorhabditis elegans (C. elegans) nematodes overexpressing pan-neuronally human α-synuclein wild-type (WT) (HASNWT OX) or mutant (HASNA53T OX). We observed 32 miRNAs and 112 piRNAs dysregulated in HASNA53T OX compared with WT. Genetic crosses of HASNA53T OX PD animal models with tdp-1 null mutants, the C. elegans ortholog of TDP-43, an RNA-binding protein aggregated in frontal temporal lobar degeneration, improved their behavioral deficits and changed the number of dysregulated miRNAs to 11 and piRNAs to none. Neuronal function-related genes T28F4.5, C34F6.1, C05C10.3, camt-1, and F54D10.3 were predicted to be targeted by cel-miR-1018, cel-miR-355-5p (C34F6.1 and C05C10.3), cel-miR-800-3p, and 21ur-1581 accordingly. This study provides a molecular landscape of small non-coding RNA dysregulation in an animal model that provides insight into the epigenetic changes, molecular processes, and interactions that occur during PD-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Linjing Shen
- Centre for Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| | - Changliang Wang
- Centre for Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, China.,Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou, China
| | - Garry Wong
- Centre for Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
48
|
Asher DM, Belay E, Bigio E, Brandner S, Brubaker SA, Caughey B, Clark B, Damon I, Diamond M, Freund M, Hyman BT, Jucker M, Keene CD, Lieberman AP, Mackiewicz M, Montine TJ, Morgello S, Phelps C, Safar J, Schneider JA, Schonberger LB, Sigurdson C, Silverberg N, Trojanowski JQ, Frosch MP. Risk of Transmissibility From Neurodegenerative Disease-Associated Proteins: Experimental Knowns and Unknowns. J Neuropathol Exp Neurol 2021; 79:1141-1146. [PMID: 33000167 PMCID: PMC7577514 DOI: 10.1093/jnen/nlaa109] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent studies in animal models demonstrate that certain misfolded proteins associated with neurodegenerative diseases can support templated misfolding of cognate native proteins, to propagate across neural systems, and to therefore have some of the properties of classical prion diseases like Creutzfeldt-Jakob disease. The National Institute of Aging convened a meeting to discuss the implications of these observations for research priorities. A summary of the discussion is presented here, with a focus on limitations of current knowledge, highlighting areas that appear to require further investigation in order to guide scientific practice while minimizing potential exposure or risk in the laboratory setting. The committee concluded that, based on all currently available data, although neurodegenerative disease-associated aggregates of several different non-prion proteins can be propagated from humans to experimental animals, there is currently insufficient evidence to suggest more than a negligible risk, if any, of a direct infectious etiology for the human neurodegenerative disorders defined in part by these proteins. Given the importance of this question, the potential for noninvasive human transmission of proteopathic disorders is deserving of further investigation.
Collapse
Affiliation(s)
- David M Asher
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Ermias Belay
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Eileen Bigio
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology Queen Square, London
| | - Scott A Brubaker
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Brychan Clark
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Inger Damon
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Marc Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michelle Freund
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen and German Center for Neurodegenerative Diseases (DZNE), Tübingen
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Miroslaw Mackiewicz
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California
| | - Susan Morgello
- Departments of Neurology, Neuroscience, and Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Creighton Phelps
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Jiri Safar
- Departments of Pathology and Neurology, Case Western Reserve University, Cleveland, Ohio
| | - Julie A Schneider
- Department of Neurological Sciences, Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Lawrence B Schonberger
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christina Sigurdson
- Department of Pathology, University of California - San Diego, San Diego, California
| | - Nina Silverberg
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthew P Frosch
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, University of Washington, Seattle, Washington.,C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
49
|
Shen Y, Yu WB, Shen B, Dong H, Zhao J, Tang YL, Fan Y, Yang YF, Sun YM, Luo SS, Chen C, Liu FT, Wu JJ, Xiao BG, Yu H, Koprich JB, Huang ZL, Wang J. Propagated α-synucleinopathy recapitulates REM sleep behaviour disorder followed by parkinsonian phenotypes in mice. Brain 2021; 143:3374-3392. [PMID: 33170925 DOI: 10.1093/brain/awaa283] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/18/2020] [Accepted: 07/12/2020] [Indexed: 11/14/2022] Open
Abstract
Idiopathic rapid eye movement sleep behaviour disorder (RBD) is now recognized as an early manifestation of α-synucleinopathies. Increasing experimental studies demonstrate that manipulative lesion or inactivation of the neurons within the sublaterodorsal tegmental nucleus (also known as the subcoeruleus nucleus in humans) can induce RBD-like behaviours in animals. As current RBD animal models are not established on the basis of α-synucleinopathy, they do not represent the pathological substrate of idiopathic RBD and thus cannot model the phenoconversion to Parkinson's disease. The purpose of this study was therefore to establish an α-synucleinopathy-based RBD animal model with the potential to convert to parkinsonian disorder. To this end, we first determined the functional neuroanatomical location of the sublaterodorsal tegmental nucleus in wild-type C57BL/6J mice and then validated its function by recapitulating RBD-like behaviours based on this determined nucleus. Next, we injected preformed α-synuclein fibrils into the sublaterodorsal tegmental nucleus and performed regular polysomnographic recordings and parkinsonian behavioural and histopathological studies in these mice. As a result, we recapitulated RBD-like behaviours in the mice and further showed that the α-synucleinopathy and neuron degeneration identified within the sublaterodorsal tegmental nucleus acted as the neuropathological substrates. Subsequent parkinsonian behavioural studies indicated that the α-synucleinopathy-based RBD mouse model were not stationary, but could further progress to display parkinsonian locomotor dysfunction, depression-like disorder, olfactory dysfunction and gastrointestinal dysmotility. Corresponding to that, we determined α-synuclein pathology in the substantia nigra pars compacta, olfactory bulb, enteral neuroplexus and dorsal motor nucleus of vagus nerve, which could underlie the parkinsonian manifestations in mice. In conclusion, we established a novel α-synucleinopathy-based RBD mouse model and further demonstrated the phenoconversion of RBD to Parkinson's disease in this animal model.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wen-Bo Yu
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bo Shen
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hui Dong
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jue Zhao
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Lin Tang
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun Fan
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan-Fei Yang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Su-Shan Luo
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng-Tao Liu
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian-Jun Wu
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bao-Guo Xiao
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huan Yu
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - James B Koprich
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
- Krembil Institute, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Zhi-Li Huang
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|