1
|
Brooker SM, Gonzalez-Latapi P. Biomarkers in Parkinson's Disease. Neurol Clin 2025; 43:229-248. [PMID: 40185520 DOI: 10.1016/j.ncl.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Parkinson's disease (PD) is a leading cause of disability worldwide, and there is a pressing need to develop therapeutics to slow or halt disease progression. The identification of reliable biomarkers of PD at all stages of disease will be a critical step toward optimizing diagnosis and therapeutic development. For PD, biomarkers could serve multiple important functions. There have been significant advances in biomarker development in PD in recent years, and in this review, the authors summarize the current state of the PD biomarker field covering major advances in fluid, tissue, and neuroimaging biomarkers.
Collapse
Affiliation(s)
- Sarah M Brooker
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paulina Gonzalez-Latapi
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
2
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
3
|
Li R, Zhuang J. Diagnostic values of serum levels of α-synuclein, homocysteine and leucine-rich α2 glycoprotein for predicting cognitive impairment in Parkinson's disease. Acta Neurol Belg 2025; 125:435-444. [PMID: 39738968 DOI: 10.1007/s13760-024-02710-x] [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: 04/11/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND α-synuclein, homocysteine (Hcy) and leucine-rich α2-glycoprotein (LRG) have been shown to correlate to Parkinson's disease (PD). However, it remains unclear whether these factors are associated with the occurrence of cognitive impairment (CI) in PD patients. METHODS This study initially enrolled eligible PD patients without cognitive impairment. Blood samples were collected to measure serum levels of α-synuclein, Hcy, and LRG using enzyme-linked immunosorbent assay. After one year of treatment, patients were divided into CI group and non-CI groups based on their Montreal Cognitive Assessment (MoCA) scores. Baseline clinical characteristics and the levels of these three factors were compared between the two groups. Additionally, Receiver Operating Characteristic (ROC) analysis was used to assess the predictive value of these factors for the occurrence of CI in PD patients. RESULTS The study included 102 eligible PD patients without baseline CI, divided into 65 patients in the non-CI group and 37 patients in the CI group after one year, based on the MoCA scores. Serum levels of α-synuclein, Hcy, and LRG in the CI group were 0.42 ± 0.33 ng/mL, 19.85 ± 6.31 µmol/L, and 14.53 ± 5.11 ng/mL respectively, compared to 0.04 ± 0.03 ng/mL, 14.32 ± 5.25 µmol/L, and 11.67 ± 3.10 ng/mL in the non-CI group. Patients in the CI group had MoCA scores below 26, indicating cognitive impairment. ROC analysis revealed that α-synuclein, Hcy, and LRG levels effectively predicted the occurrence of CI in PD patients. CONCLUSIONS Serum levels of α-synuclein, Hcy and LRG were elevated in the CI group compared to the non-CI group, suggesting that these factors may serve as predictors of cognitive impairment in PD patients.
Collapse
Affiliation(s)
- Rongchuan Li
- Department of Neurology, Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian Campus), No.16 Jingguang Road, Luoshan Section, Jinjiang, 362200, Fujian, China
| | - Jinhong Zhuang
- Department of Neurology, Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian Campus), No.16 Jingguang Road, Luoshan Section, Jinjiang, 362200, Fujian, China.
| |
Collapse
|
4
|
de Mena L, Parés G, Garrido A, Pilco‐Janeta DF, Fernández M, Pérez J, Tolosa E, Cámara A, Valldeoriola F, Ezquerra M, Martí M, Fernández‐Santiago R. α-Synuclein Gene Hypomethylation in LRRK2 Parkinson's Disease Patients. Mov Disord 2025; 40:550-555. [PMID: 39711195 PMCID: PMC11926512 DOI: 10.1002/mds.30094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND α-Synuclein (SNCA) gene hypomethylation was reported in idiopathic Parkinson's disease (iPD). Based on a high clinical resemblance between iPD and leucine-rich repeat kinase 2 (LRRK2)-driven Parkinson's disease (L2PD), we investigated the epigenetic status of SNCA in an extensive LRRK2 clinical cohort from Spain. METHODS We assessed the methylation levels of 23 CpG sites in the SNCA promoter region using peripheral blood DNA from L2PD patients (n = 151), LRRK2 nonmanifesting carriers (n = 55), iPD patients (n = 115), and healthy control subjects (n = 154) (total: N = 475). RESULTS Compared with control subjects, we found significant SNCA hypomethylation in 11 of 23 CpGs in L2PD (48%), whereas 22 CpGs (96%) were hypomethylated in iPD. In line with a healthy status, asymptomatic mutation carriers had similar SNCA methylation profiles to control subjects. CONCLUSIONS This study shows for the first time that SNCA hypomethylation occurs in patients with L2PD. Further studies addressing SNCA methylation status in additional worldwide LRRK2 cohorts are warranted. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Lorena de Mena
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
| | - Guillem Parés
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
| | - Alicia Garrido
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Daniel F. Pilco‐Janeta
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Manel Fernández
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
| | - Jesica Pérez
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Eduardo Tolosa
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Ana Cámara
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Francesc Valldeoriola
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Mario Ezquerra
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - María‐José Martí
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | - Rubén Fernández‐Santiago
- Laboratory of Parkinson's and Other Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i SunyerBarcelonaSpain
- Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas CB06/05/0018‐ISCIIIBarcelonaSpain
| | | |
Collapse
|
5
|
Zhu Z, Cordato D, Chen R, Xu YH, Shulruf B, Chan DKY. Plasma alpha-synuclein predicts cognitive impairment in Parkinson's disease: a systematic review and meta-analysis. J Neurol 2025; 272:124. [PMID: 39812682 DOI: 10.1007/s00415-024-12871-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Alpha-synuclein (ɑ-syn) plays a key role in Parkinson's disease (PD) pathogenesis, but existing studies have found mixed results regarding the associations between plasma ɑ-syn and the development of cognitive impairment. We aim to clarify the potentially important relationship between ɑ-syn level in plasma and development of cognitive impairment in PD through systematic review and meta-analysis. METHODS A systematic search was conducted in the PubMed, Embase and Web of Science databases for studies reporting plasma ɑ-syn concentrations and cognitive impairment in PD. Effect directions were plotted to investigate methodological factors, and a meta-analysis was performed comparing PD patients with dementia (PDD) to PD patients with normal cognition (PDNC). RESULTS Twenty-five studies were identified for the systematic review, involving 1,888 PD patients. Studies using the clinical diagnostic Movement Disorder Society (MDS) criteria for PD with mild cognitive impairment and PDD found consistently positive associations with plasma ɑ-syn level. This was further supported by a meta-analysis which found a significant standardised mean difference (g = 1.770, 95% CI: 0.749-2.790, p < 0.001) between PDD and PDNC patients in 10 studies. Furthermore, studies using emerging immunomagnetic reduction or single-molecule array techniques to quantify ɑ-syn reported strong positive associations. In contrast, studies using enzyme-linked immunoassay and cognitive screening tests alone found mixed results. CONCLUSION There is an overall positive effect between plasma ɑ-syn levels and cognitive impairment in PD. As methodological factors can significantly affect associations, future studies should carefully select ɑ-syn immunoassays and utilise the MDS diagnostic criteria for cognitive impairment in PD.
Collapse
Affiliation(s)
- Ziyue Zhu
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Dennis Cordato
- Faculty of Medicine, University of New South Wales, Sydney, Australia
- Department of Neurology and Neurophysiology, Liverpool Hospital, Sydney, Australia
| | - Renfen Chen
- Central Sydney Immunology Laboratory, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Ying Hua Xu
- Faculty of Medicine, University of New South Wales, Sydney, Australia
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Sydney, NSW, 2200, Australia
| | - Boaz Shulruf
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Daniel Kam Yin Chan
- Faculty of Medicine, University of New South Wales, Sydney, Australia.
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Sydney, NSW, 2200, Australia.
| |
Collapse
|
6
|
Luo Z, Zhu Y, Zhu Y, Liu B, Li Y, Yin L, Liu J, Xu Z, Ren H, Yang X. Cognitive function in Parkinson's disease: associations with perivascular space in basal ganglia. Neurol Sci 2024; 45:5973-5981. [PMID: 39212793 DOI: 10.1007/s10072-024-07729-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cognitive impairment is one of the most common symptoms of Parkinson's disease (PD), and may be detectable through changes in neural features visualized by magnetic resonance imaging (MRI). Mild cognitive impairment is a transitional state between normal aging and dementia, and early recognition of Parkinson's disease with mild cognitive impairment (PD-MCI) can help improve the quality of life and treatment for patients. This study investigated the association of enlarged perivascular space (EPVS) and white matter hyperintensity (WMH) with PD-MCI. AIMS This study aimed to evaluate whether EPVS and WMH can be used as potential MRI markers for PD-MCI. METHODS This retrospective study involved 200 patients with PD who underwent cranial MRI in our hospital from April 2021 to April 2022. Patients were divided into those with no cognitive impairment (PD-NCI) or mild cognitive impairment. Uni- and multivariate logistic regression analyzed associations of EPVS, WMH, and clinicodemographic characteristics with cognitive decline. RESULTS Univariate regression identified severe EPVS in basal ganglia, severe WMH, older age, late-onset, male sex, low educational level, longer duration of disease, low triglycerides, low uric acid, and low scores on the Mini-mental State Exam as risk factors for PD-MCI. After adjusting for clinicodemographic risk factors in multivariate regression, low education level and EPVS in basal ganglia remained risk factors for cognitive impairment. CONCLUSIONS Severe EPVS in basal ganglia and poor education, but not WMH, are independent risk factors of PD-MCI. Our findings suggest that non-invasive detection of EPVS in basal ganglia by MRI may be a valuable early indicator of cognitive decline in PD patients.
Collapse
Affiliation(s)
- Zhenglong Luo
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Yangfan Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Yongyun Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Bin Liu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Yuxia Li
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Lei Yin
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Jie Liu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Zhong Xu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China.
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China.
| | - Xinglong Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China.
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan Province, P.R. China.
| |
Collapse
|
7
|
Scorziello A, Sirabella R, Sisalli MJ, Tufano M, Giaccio L, D’Apolito E, Castellano L, Annunziato L. Mitochondrial Dysfunction in Parkinson's Disease: A Contribution to Cognitive Impairment? Int J Mol Sci 2024; 25:11490. [PMID: 39519043 PMCID: PMC11546611 DOI: 10.3390/ijms252111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Among the non-motor symptoms associated with Parkinson's disease (PD), cognitive impairment is one of the most common and disabling. It can occur either early or late during the disease, and it is heterogeneous in terms of its clinical manifestations, such as Subjective Cognitive Dysfunction (SCD), Mild Cognitive Impairment (MCI), and Parkinson's Disease Dementia (PDD). The aim of the present review is to delve deeper into the molecular mechanisms underlying cognitive decline in PD. This is extremely important to delineate the guidelines for the differential diagnosis and prognosis of the dysfunction, to identify the molecular and neuronal mechanisms involved, and to plan therapeutic strategies that can halt cognitive impairment progression. Specifically, the present review will discuss the pathogenetic mechanisms involved in the progression of cognitive impairment in PD, with attention to mitochondria and their contribution to synaptic dysfunction and neuronal deterioration in the brain regions responsible for non-motor manifestations of the disease.
Collapse
Affiliation(s)
- Antonella Scorziello
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Rossana Sirabella
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Maria Josè Sisalli
- Department of Translational Medicine, Federico II University of Naples, 80138 Napoli, Italy;
| | - Michele Tufano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lucia Giaccio
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Elena D’Apolito
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lorenzo Castellano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | | |
Collapse
|
8
|
Mills KA, Phillips O, Mahajan A. Hope vs. Hype I: Spreading alpha-synuclein explains cognitive deficits in Parkinson disease. Parkinsonism Relat Disord 2024; 126:106042. [PMID: 38365523 DOI: 10.1016/j.parkreldis.2024.106042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
The Parkinson Study Group (PSG) gathered North American experts in Parkinson disease during the 9th Annual Symposium on "Shaping the Management of Parkinson Disease: Debating Current Controversies". Debaters were tasked with agree or disagree positions to a particular prompt. This is the first in three-part series of "Hype vs. Hope" debates involving current trends and advances in Parkinson disease. With the prompt of "Spreading alpha-synuclein explains cognitive deficits in Parkinson disease," Dr. Kelly Mills, MD, MHS was tasked with the "agree" stance and Dr. Abhimanyu Mahajan, MD, MHS was tasked with the "disagree" stance. The following point-of-view article is an adaptation of this debate.
Collapse
Affiliation(s)
- Kelly A Mills
- Johns Hopkins Department of Neurology, Baltimore, MD600 N. Wolfe Street, Meyer 6-181D, 21287, United States.
| | - Oliver Phillips
- Geisel School of Medicine at Dartmouth, Hanover, NH, 18 Old Etna Road, 03756, United States.
| | - Abhimanyu Mahajan
- The University of Cincinnati James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, OH, 260 Stetson St., Suite 2300, Cincinnati, 45219, United States.
| |
Collapse
|
9
|
Ying C, Zhang H, Wang T, Li Y, Mao W, Hu S, Zhao L, Cai Y. Plasma level of alpha-synuclein oligomers as a biomarker for isolated rapid eye movement sleep behavior disorder diagnosis and progression: a prospective cohort study. Front Neurol 2024; 15:1442173. [PMID: 39246606 PMCID: PMC11377258 DOI: 10.3389/fneur.2024.1442173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alpha-synuclein oligomers (o-α-syn) are pivotal in the pathogenesis of α-synucleinopathy. Isolated rapid eye movement (REM) sleep behavior disorder (iRBD) serves as an early indicator of the disease, offering insights into disease mechanisms and early intervention. Nevertheless, the diagnostic and predictive potential of o-α-syn in iRBD remains largely unexplored. This study aimed to evaluate the plasma levels of o-α-syn in patients and investigate their utility as biomarkers for diagnosis of and predicting phenoconversion in iRBD. Methods A total of 143 participants, including 77 polysomnography-confirmed iRBD patients and 66 normal controls (NC), were recruited for this longitudinal observational study. Baseline clinical assessments and plasma collection were conducted for all iRBD patients, with 72 of them undergoing regularly prospective follow-up assessments for parkinsonism or dementia. Plasma levels of o-α-syn were quantified using enzyme-linked immunosorbent assay, and were compared between groups using a general linear model adjusted for age and sex. The diagnostic performance of plasma o-α-syn in iRBD was evaluated by area under the receiver operating characteristic curve (AUC) with 95% CI. Cox regression analysis and Kaplan-Meier survival curves were employed to assess the predictive value of plasma o-α-syn for phenoconversion in iRBD. Results Plasma o-α-syn levels did not exhibit statistically significant differences among iRBD converter patients, iRBD nonconverter patients, and NC. The AUC for distinguishing NC from iRBD was 0.52 (95% CI: 0.42-0.62, p = 0.682). Spearman correlation analysis revealed a significant positive correlation between plasma o-α-syn levels and MOCA scores in the iRBD group (p < 0.001). Subgroup analyses indicated that iRBD patients with cognitive decline (p = 0.058) and depressive symptoms (p = 0.017) had notably lower o-α-syn levels compared to those without such symptoms. Over a median follow-up period of 5.83 years, 26 iRBD patients developed neurodegenerative synucleinopathies. Cox regression and Kaplan-Meier survival curve analyses indicated that plasma level of o-α-syn lacked a predictive value for disease conversion in iRBD patients. Conclusion Despite a potential role in the pathophysiology of iRBD, o-α-syn are not appropriate biomarkers for diagnosing or predicting disease progression. While this study offers insights into the pathogenesis of iRBD and neurodegenerative synucleinopathies, further large-scale longitudinal studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Chao Ying
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Hui Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ting Wang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yuan Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Mao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Songnian Hu
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Lifang Zhao
- Beijing Geriatric Medical Research Center, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Department of Clinical Biobank and Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
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
|
11
|
Gorji A, Fathi Jouzdani A. Machine learning for predicting cognitive decline within five years in Parkinson's disease: Comparing cognitive assessment scales with DAT SPECT and clinical biomarkers. PLoS One 2024; 19:e0304355. [PMID: 39018311 PMCID: PMC11253925 DOI: 10.1371/journal.pone.0304355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 05/08/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is an age-related neurodegenerative condition characterized mostly by motor symptoms. Although a wide range of non-motor symptoms (NMS) are frequently experienced by PD patients. One of the important and common NMS is cognitive impairment, which is measured using different cognitive scales. Monitoring cognitive impairment and its decline in PD is essential for patient care and management. In this study, our goal is to identify the most effective cognitive scale in predicting cognitive decline over a 5-year timeframe initializing clinical biomarkers and DAT SPECT. METHODS Machine Learning has previously shown superior performance in image and clinical data classification and detection. In this study, we propose to use machine learning with different types of data, such as DAT SPECT and clinical biomarkers, to predict PD-CD based on various cognitive scales. We collected 330 DAT SPECT images and their clinical data in baseline, years 2,3,4, and 5 from Parkinson's Progression Markers Initiative (PPMI). We then designed a 3D Autoencoder to extract deep radiomic features (DF) from DAT SPECT images, and we then concatenated it with 17 clinical features (CF) to predict cognitive decline based on Montreal Cognitive Assessment (MoCA) and The Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS-I). RESULTS The utilization of MoCA as a cognitive decline scale yielded better performance in various years compared to MDS-UPDRS-I. In year 4, the application of the deep radiomic feature resulted in the highest achievement, with a cross-validation AUC of 89.28, utilizing the gradient boosting classifier. For the MDS-UPDRS-I scale, the highest achievement was obtained by utilizing the deep radiomic feature, resulting in a cross-validation AUC of 81.34 with the random forest classifier. CONCLUSIONS The study findings indicate that the MoCA scale may be a more effective predictor of cognitive decline within 5 years compared to MDS-UPDRS-I. Furthermore, deep radiomic features had better performance compared to sole clinical biomarkers or clinical and deep radiomic combined. These results suggest that using the MoCA score and deep radiomic features extracted from DAT SPECT could be a promising approach for identifying individuals at risk for cognitive decline in four years. Future research is needed to validate these findings and explore their utility in clinical practice.
Collapse
Affiliation(s)
- Arman Gorji
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Neuroscience and Artificial Intelligence Research Group (NAIRG), Hamadan University of Medical Sciences, Hamadan, Iran
- USERN Office, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Fathi Jouzdani
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Neuroscience and Artificial Intelligence Research Group (NAIRG), Hamadan University of Medical Sciences, Hamadan, Iran
- USERN Office, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
12
|
Shu H, Zhang P, Gu L. Alpha-synuclein in peripheral body fluid as a biomarker for Parkinson's disease. Acta Neurol Belg 2024; 124:831-842. [PMID: 38170418 DOI: 10.1007/s13760-023-02452-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE Whether alpha-synuclein in peripheral body fluids can be used for the diagnosis of Parkinson's disease (PD) remains in controversy. This study evaluates diagnostic potential of alpha-synuclein for PD in various peripheral body fluids using a meta-analysis approach. METHODS Studies published before October 2022 were searched in Web of Science and PubMed databases. The results were computed using the STATA 12.0 statistical software. RESULTS In plasma, PD patients exhibited elevated alpha-synuclein levels relative to healthy controls (HCs) [standard mean difference (SMD) = 0.78, 95% confidence interval (CI) = 0.42 to 1.15] with a sensitivity of 0.79 (95% CI: 0.64-0.89) and a specificity of 0.95 (95% CI: 0.90-0.98). Higher plasma alpha-synuclein levels were correlated with longer disease durations, higher Unified Parkinson's Disease Rating Scale motor scores, and higher Hoehn and Yahr stages in PD patients. Plasma neural-derived exosomal alpha-synuclein levels (SMD = 1.82, 95% CI = 0.30 to 3.35), ratio of plasma neural-derived exosomal alpha-synuclein to total alpha-synuclein (SMD = 1.26, 95% CI = 0.19 to 2.33), and erythrocytic alpha-synuclein levels were also increased in PD patients (SMD = 6.57, 95% CI = 3.55 to 9.58). In serum, there was no significant difference in alpha-synuclein levels between PD patients and HCs (SMD = 0.54, 95% CI = - 0.27 to 1.34). In saliva, reduced alpha-synuclein levels were observed in PD patients (SMD = - 0.85, 95% CI = - 1.67 to - 0.04). CONCLUSIONS Alpha-synuclein levels in plasma, plasma neural-derived exosome, erythrocyte, and saliva may serve as potential biomarkers for the diagnosis of PD.
Collapse
Affiliation(s)
- Hao Shu
- Department of Neurology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Pengcheng Zhang
- Institute of Environment and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300041, China
| | - Lihua Gu
- Department of Neurology, Tianjin Huanhu Hospital, No. 6 Jizhao Road, Tianjin, 300350, China.
| |
Collapse
|
13
|
Hatano T, Okuzumi A, Matsumoto G, Tsunemi T, Hattori N. α-Synuclein: A Promising Biomarker for Parkinson's Disease and Related Disorders. J Mov Disord 2024; 17:127-137. [PMID: 38589016 PMCID: PMC11082597 DOI: 10.14802/jmd.24075] [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: 03/22/2024] [Revised: 03/30/2024] [Accepted: 04/09/2024] [Indexed: 04/10/2024] Open
Abstract
Mutations in the SNCA gene, which encodes α-synuclein (α-syn), play a key role in the development of genetic Parkinson's disease (PD). α-Syn is a major component of Lewy bodies in PD and glial cytoplasmic inclusions in multiple system atrophy (MSA). Rapid eye movement sleep behavior disorder patients often progress to PD, dementia with Lewy bodies, or MSA, which are collectively known as α-synucleinopathies. The loss of dopaminergic neurons with Lewy bodies precedes motor dysfunction in these diseases, but the mechanisms of neurodegeneration due to α-syn aggregation are poorly understood. Monitoring α-syn aggregation in vivo could serve as a diagnostic biomarker and help elucidate pathogenesis, necessitating a simple and accurate detection method. Seed amplification assays (SAAs), such as real-time quaking-induced conversion and protein misfolding cyclic amplification, are used to detect small amounts of abnormally structured α-syn protofibrils, which are central to aggregation. These methods are promising for the early diagnosis of α-synucleinopathy. Differences in α-syn filament structures between α-synucleinopathies, as observed through transmission electron microscopy and cryo-electron microscopy, suggest their role in the pathogenesis of neurodegeneration. SAAs may differentiate between subtypes of α-synucleinopathy and other diseases. Efforts are also being made to identify α-syn from blood using various methods. This review introduces body fluid α-syn biomarkers based on pathogenic α-syn seeds, which are expected to redefine α-synucleinopathy diagnosis and staging, improving clinical research accuracy and facilitating biomarker development.
Collapse
Affiliation(s)
- Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Gen Matsumoto
- Department of Histology and Cell Biology, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Taiji Tsunemi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
14
|
Chopra A, Outeiro TF. Aggregation and beyond: alpha-synuclein-based biomarkers in synucleinopathies. Brain 2024; 147:81-90. [PMID: 37526295 DOI: 10.1093/brain/awad260] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023] Open
Abstract
Parkinson's disease is clinically known for the loss of dopaminergic neurons in the substantia nigra pars compacta and accumulation of intraneuronal cytoplasmic inclusions rich in alpha-synuclein called 'Lewy bodies' and 'Lewy neurites'. Together with dementia with Lewy bodies and multiple system atrophy, Parkinson's disease is part of a group of disorders called synucleinopathies. Currently, diagnosis of synucleinopathies is based on the clinical assessment which often takes place in advanced disease stages. While the causal role of alpha-synuclein aggregates in these disorders is still debatable, measuring the levels, types or seeding properties of different alpha-synuclein species hold great promise as biomarkers. Recent studies indicate significant differences in peptide, protein and RNA levels in blood samples from patients with Parkinson's disease. Seed amplification assays using CSF, blood, skin biopsy, olfactory swab samples show great promise for detecting synucleinopathies and even for discriminating between different synucleinopathies. Interestingly, small extracellular vesicles, such as exosomes, display differences in their cargoes in Parkinson's disease patients versus controls. In this update, we focus on alpha-synuclein aggregation and possible sources of disease-related species released in extracellular vesicles, which promise to revolutionize the diagnosis and the monitoring of disease progression.
Collapse
Affiliation(s)
- Avika Chopra
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| |
Collapse
|
15
|
Yang Y, Kim WS, Michaelian JC, Lewis SJG, Phillips CL, D'Rozario AL, Chatterjee P, Martins RN, Grunstein R, Halliday GM, Naismith SL. Predicting neurodegeneration from sleep related biofluid changes. Neurobiol Dis 2024; 190:106369. [PMID: 38049012 DOI: 10.1016/j.nbd.2023.106369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
Sleep-wake disturbances are common in neurodegenerative diseases and may occur years before the clinical diagnosis, potentially either representing an early stage of the disease itself or acting as a pathophysiological driver. Therefore, discovering biomarkers that identify individuals with sleep-wake disturbances who are at risk of developing neurodegenerative diseases will allow early diagnosis and intervention. Given the association between sleep and neurodegeneration, the most frequently analyzed fluid biomarkers in people with sleep-wake disturbances to date include those directly associated with neurodegeneration itself, such as neurofilament light chain, phosphorylated tau, amyloid-beta and alpha-synuclein. Abnormalities in these biomarkers in patients with sleep-wake disturbances are considered as evidence of an underlying neurodegenerative process. Levels of hormonal sleep-related biomarkers such as melatonin, cortisol and orexin are often abnormal in patients with clinical neurodegenerative diseases, but their relationships with the more standard neurodegenerative biomarkers remain unclear. Similarly, it is unclear whether other chronobiological/circadian biomarkers, such as disrupted clock gene expression, are causal factors or a consequence of neurodegeneration. Current data would suggest that a combination of fluid biomarkers may identify sleep-wake disturbances that are most predictive for the risk of developing neurodegenerative disease with more optimal sensitivity and specificity.
Collapse
Affiliation(s)
- Yue Yang
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Woojin Scott Kim
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Johannes C Michaelian
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Simon J G Lewis
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; Parkinson's Disease Research Clinic, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia.
| | - Craig L Phillips
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Angela L D'Rozario
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia; CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia.
| | - Pratishtha Chatterjee
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Ralph N Martins
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia; School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, WA 6009, Australia.
| | - Ron Grunstein
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW 2109, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Glenda M Halliday
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Sharon L Naismith
- Healthy Brain Ageing Program, School of Psychology, Brain and Mind Centre & The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2050, Australia.
| |
Collapse
|
16
|
Camacho-Ordonez A, Cervantes-Arriaga A, Rodríguez-Violante M, Hernandez-Medrano AJ, Somilleda-Ventura SA, Pérez-Cano HJ, Nava-Castañeda Á, Guerrero-Berger O. Is there any correlation between alpha-synuclein levels in tears and retinal layer thickness in Parkinson's disease? Eur J Ophthalmol 2024; 34:252-259. [PMID: 37151018 DOI: 10.1177/11206721231173725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
PURPOSE To determine the total alpha-synuclein (αSyn) reflex tears and its association with retinal layers thickness in Parkinson's disease (PD). METHODS Fifty-two eyes of 26 PD subjects and 52 eyes of age-and sex-matched healthy controls were included. Total αSyn in reflex tears was quantified using a human total αSyn enzyme-linked immunosorbent assay (ELISA) kit. The retinal thickness was evaluated with spectral-domain optical coherence tomography. The Movement Disorder Society-Unified Parkinsońs Disease Rating Scale (MDS-UPDRS), Non-Motor Symptoms Scale (NMSS), and Montreal Cognitive Assessment (MoCA) were used to assess motor, non-motor, and cognition. RESULTS In PD, total αSyn levels were increased compared to control subjects [1.76pg/mL (IQR 1.74-1.80) vs 1.73pg/mL (IQR 1.70-1.77), p < 0.004]. The nerve fiber layer, ganglion cell layer, internal plexiform layer, inner nuclear layer, and outer nuclear layer were thinner in PD in comparison with controls (p < 0.05). The outer plexiform layer and retinal pigment epithelium were thicker in PD (p < 0.05). The total αSyn levels positively correlated with the central volume of the inner nuclear layer (r = 0.357, p = 0.009). CONCLUSION Total αSyn reflex tear levels were increased in subjects with PD compared to controls. PD patients showed significant thinning of the inner retinal layers and thickening of outer retinal layers in comparison with controls. Total αSyn levels positively correlate with the central volume of the inner nuclear layer in PD. The combination of these biomarkers might have a possible role as a diagnostic tool in PD subjects.
Collapse
Affiliation(s)
- Azyadeh Camacho-Ordonez
- Neuro-ophthalmology Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
- Anterior Segment Department, Fundacion Hospital Nuestra Señora de la Luz, IAP, Mexico City, Mexico
| | - Amin Cervantes-Arriaga
- Movement Disorder Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | | | | | | | - Hector J Pérez-Cano
- Biomedical Research Center, Fundacion Hospital Nuestra Señora de la Luz, IAP, Mexico City, Mexico
| | - Ángel Nava-Castañeda
- Oculoplastics Department, Instituto de Oftalmologia Fundacion Conde de Valenciana IAP, Mexico City, Mexico
| | - Oscar Guerrero-Berger
- Anterior Segment Department, Fundacion Hospital Nuestra Señora de la Luz, IAP, Mexico City, Mexico
| |
Collapse
|
17
|
Lee KH, Hsu MH, Chen HH, Yang SY. Analyzer-to-Analyzer Variations in Assaying Ultralow Concentrated Biomarkers Associated with Neurodegenerative Diseases Using Immunomagnetic Reduction. ACS MEASUREMENT SCIENCE AU 2023; 3:488-495. [PMID: 38145030 PMCID: PMC10740117 DOI: 10.1021/acsmeasuresciau.3c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 12/26/2023]
Abstract
By utilizing a high-temperature superconducting quantum interference device (high-Tc SQUID) magnetometer, an alternating current (AC) magnetosusceptometer, referred to as an analyzer, was developed for ultrasensitive immunoassays. The analyzer has been applied to assay biomarkers in human plasma associated with Alzheimer's disease (AD) and Parkinson's disease (PD). The involved assay methodology is the so-called immunomagnetic reduction (IMR). Such an analyzer has been approved for clinical use in Taiwan and Europe. The mass production of the analyzer is needed for clinical utilities. The issue of exploring analyzer-to-analyzer variations in the performances becomes critical. Unfortunately, there is no standard characterization to determine the variations in performances among analyzers. In this study, key characterizations, such as output signal stability, signal-to-noise ratio, measured concentrations of a control sample, etc., are proposed. In total, three analyzers are characterized in this work. The detected biomarkers include amyloid peptides, total tau protein, phosphorylated tau protein, and α-synuclein protein for AD and PD. Through one-way ANOVA for any of the characterizations among the three analyzers, it was found that there was no significant difference in any of these characterizations among the analyzers (p > 0.05). Furthermore, the three analyzers are applied to assay biomolecules for AD and PD in reference samples. High correlations (r > 0.8) in measured concentrations of any of these biomarkers in reference samples were obtained among the three analyzers. The results demonstrate that the proposed characterizations are feasible for achieving consistent performance among high-Tc SQUID-based AC magnetosusceptometers for assaying biomolecules.
Collapse
|
18
|
Bodily TA, Ramanathan A, Wei S, Karkisaval A, Bhatt N, Jerez C, Haque MA, Ramil A, Heda P, Wang Y, Kumar S, Leite M, Li T, Zhao J, Lal R. In pursuit of degenerative brain disease diagnosis: Dementia biomarkers detected by DNA aptamer-attached portable graphene biosensor. Proc Natl Acad Sci U S A 2023; 120:e2311565120. [PMID: 37956285 PMCID: PMC10666025 DOI: 10.1073/pnas.2311565120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 11/15/2023] Open
Abstract
Dementia is a brain disease which results in irreversible and progressive loss of cognition and motor activity. Despite global efforts, there is no simple and reliable diagnosis or treatment option. Current diagnosis involves indirect testing of commonly inaccessible biofluids and low-resolution brain imaging. We have developed a portable, wireless readout-based Graphene field-effect transistor (GFET) biosensor platform that can detect viruses, proteins, and small molecules with single-molecule sensitivity and specificity. We report the detection of three important amyloids, namely, Amyloid beta (Aβ), Tau (τ), and α-Synuclein (αS) using DNA aptamer nanoprobes. These amyloids were isolated, purified, and characterized from the autopsied brain tissues of Alzheimer's Disease (AD) and Parkinson's Disease (PD) patients. The limit of detection (LoD) of the sensor is 10 fM, 1-10 pM, 10-100 fM for Aβ, τ, and αS, respectively. Synthetic as well as autopsied brain-derived amyloids showed a statistically significant sensor response with respect to derived thresholds, confirming the ability to define diseased vs. nondiseased states. The detection of each amyloid was specific to their aptamers; Aβ, τ, and αS peptides when tested, respectively, with aptamers nonspecific to them showed statistically insignificant cross-reactivity. Thus, the aptamer-based GFET biosensor has high sensitivity and precision across a range of epidemiologically significant AD and PD variants. This portable diagnostic system would allow at-home and POC testing for neurodegenerative diseases globally.
Collapse
Affiliation(s)
| | - Anirudh Ramanathan
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Shanhong Wei
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Abhijith Karkisaval
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA92093
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Md Anzarul Haque
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX77555
| | - Armando Ramil
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Prachi Heda
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Yi Wang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Sanjeev Kumar
- Department of Computer Science, University of Illinois Urbana-Champaign, Champaign, IL61820
| | - Mikayla Leite
- Department of Bioengineering, University of California, San Diego, CA92093
| | - Tie Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai200050, China
| | - Ratnesh Lal
- Department of Bioengineering, University of California, San Diego, CA92093
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA92093
- Materials Science and Engineering Program, University of California, San Diego, CA92093
| |
Collapse
|
19
|
Grant H, Anderton R, Gasson N, Lawrence BJ. The gut microbiome and cognition in Parkinson's disease: a systematic review. Nutr Neurosci 2023; 26:932-941. [PMID: 35965446 DOI: 10.1080/1028415x.2022.2110189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
BACKGROUND The pathology underlying cognitive changes in people with Parkinson's disease (PD) is not well understood. In healthy older adults, gut microbiome composition has been associated with cognitive function. In people with PD, preliminary evidence suggests that cortical spreading of abnormal alpha-synuclein aggregates may be associated with cognitive impairment. As changes in the gut have been linked to PD onset and associated Lewy body pathology, an investigation of the gut microbiome and cognition in PD is warranted. OBJECTIVE To synthesise existing evidence on the relationship between the gut microbiome and cognitive function in PD. METHODS A systematic review was conducted to search for peer-reviewed articles and grey literature published to July 2021 across seven electronic databases (MEDLINE, EMBASE, PsycINFO, Scopus, Cochrane Library, ProQuest, and ProQuest Dissertations and Theses). English language articles reporting the relationship between cognition and the gut microbiome in human participants with PD were considered for inclusion. Results were qualitatively synthesised and evidence quality was assessed using the QualSyst tool for quantitative studies. RESULTS Five cross-sectional studies reporting the association between the gut microbiome and cognition in 395 participants with PD were included. Studies provided preliminary evidence of a relationship between cognition and gut microbiota within the Bacteroidetes and Firmicutes phyla, however, associations with specific genera were inconsistent across studies. CONCLUSIONS Some species of short-chain fatty acid-producing bacteria (e.g. acetate, butyrate, and propionate producers) appear to be reduced in participants with PD with cognitive impairment. More research with larger samples and more consistent methodology is needed to substantiate these findings.
Collapse
Affiliation(s)
- Hayley Grant
- Discipline of Psychology, School of Population Health, Curtin University, Bentley, Australia
| | - Ryan Anderton
- Institute for Health Research, The University of Notre Dame Australia, Fremantle, Australia
| | - Natalie Gasson
- Discipline of Psychology, School of Population Health, Curtin University, Bentley, Australia
| | - Blake J Lawrence
- Discipline of Psychology, School of Population Health, Curtin University, Bentley, Australia
| |
Collapse
|
20
|
Kalia V, Baccarelli AA, Happel C, Hollander JA, Jukic AM, McAllister KA, Menon R, Merrick BA, Milosavljevic A, Ravichandran LV, Roth ME, Subramanian A, Tyson FL, Worth L, Shaughnessy DT. Seminar: Extracellular Vesicles as Mediators of Environmental Stress in Human Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:104201. [PMID: 37861803 PMCID: PMC10588739 DOI: 10.1289/ehp12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), membrane-bound particles containing a variety of RNA types, DNA, proteins, and other macromolecules, are now appreciated as an important means of communication between cells and tissues, both in normal cellular physiology and as a potential indicator of cellular stress, environmental exposures, and early disease pathogenesis. Extracellular signaling through EVs is a growing field of research for understanding fundamental mechanisms of health and disease and for the potential for biomarker discovery and therapy development. EVs are also known to play important roles in mediating the effects of exposure to environmental stress. OBJECTIVES This seminar addresses the application of new tools and approaches for EV research, developed in part through the National Institutes of Health (NIH) Extracellular RNA Communication Program, and reflects presentations and discussions from a workshop held 27-28 September 2021 by the National Institute of Environmental Health Sciences (NIEHS) and the National Center for Advancing Translational Sciences (NCATS) on "Extracellular Vesicles, Exosomes, and Cell-Cell Signaling in Response to Environmental Stress." The panel of experts discussed current research on EVs and environmental exposures, highlighted recent advances in EV isolation and characterization, and considered research gaps and opportunities toward identifying and characterizing the roles for EVs in environmentally related diseases, as well as the current challenges and opportunities in this field. DISCUSSION The authors discuss the application of new experimental models, particularly organ-on-chip (OOC) systems and in vitro approaches and how these have the potential to extend findings in population-based studies of EVs in exposure-related diseases. Given the complex challenges of identifying cell-specific EVs related to environmental exposures, as well as the general heterogeneity and variability in EVs in blood and other accessible biological samples, there is a critical need for rigorous reporting of experimental methods and validation studies. The authors note that these efforts, combined with cross-disciplinary approaches, would ensure that future research efforts in environmental health studies on EV biomarkers are rigorous and reproducible. https://doi.org/10.1289/EHP12980.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Christine Happel
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), U.S. Department of Health and Human Services (DHHS), Bethesda, Maryland, USA
| | - Jonathan A. Hollander
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Anne Marie Jukic
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Kimberly A. McAllister
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Bruce A. Merrick
- Division of Translational Toxicology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | | | - Lingamanaidu V. Ravichandran
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Matthew E. Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Anita Subramanian
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Frederick L. Tyson
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Leroy Worth
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Daniel T. Shaughnessy
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
21
|
Taneva SG, Todinova S, Andreeva T. Morphometric and Nanomechanical Screening of Peripheral Blood Cells with Atomic Force Microscopy for Label-Free Assessment of Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:14296. [PMID: 37762599 PMCID: PMC10531602 DOI: 10.3390/ijms241814296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegenerative disorders (NDDs) are complex, multifactorial disorders with significant social and economic impact in today's society. NDDs are predicted to become the second-most common cause of death in the next few decades due to an increase in life expectancy but also to a lack of early diagnosis and mainly symptomatic treatment. Despite recent advances in diagnostic and therapeutic methods, there are yet no reliable biomarkers identifying the complex pathways contributing to these pathologies. The development of new approaches for early diagnosis and new therapies, together with the identification of non-invasive and more cost-effective diagnostic biomarkers, is one of the main trends in NDD biomedical research. Here we summarize data on peripheral biomarkers, biofluids (cerebrospinal fluid and blood plasma), and peripheral blood cells (platelets (PLTs) and red blood cells (RBCs)), reported so far for the three most common NDDs-Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). PLTs and RBCs, beyond their primary physiological functions, are increasingly recognized as valuable sources of biomarkers for NDDs. Special attention is given to the morphological and nanomechanical signatures of PLTs and RBCs as biophysical markers for the three pathologies. Modifications of the surface nanostructure and morphometric and nanomechanical signatures of PLTs and RBCs from patients with AD, PD, and ALS have been revealed by atomic force microscopy (AFM). AFM is currently experiencing rapid and widespread adoption in biomedicine and clinical medicine, in particular for early diagnostics of various medical conditions. AFM is a unique instrument without an analog, allowing the generation of three-dimensional cell images with extremely high spatial resolution at near-atomic scale, which are complemented by insights into the mechanical properties of cells and subcellular structures. Data demonstrate that AFM can distinguish between the three pathologies and the normal, healthy state. The specific PLT and RBC signatures can serve as biomarkers in combination with the currently used diagnostic tools. We highlight the strong correlation of the morphological and nanomechanical signatures between RBCs and PLTs in PD, ALS, and AD.
Collapse
Affiliation(s)
- Stefka G. Taneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Tonya Andreeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
- Faculty of Life Sciences, Reutlingen University, Alteburgstraße 150, D-72762 Reutlingen, Germany
| |
Collapse
|
22
|
Zhang Q, Duan Q, Gao Y, He P, Huang R, Huang H, Li Y, Ma G, Zhang Y, Nie K, Wang L. Cerebral Microvascular Injury Induced by Lag3-Dependent α-Synuclein Fibril Endocytosis Exacerbates Cognitive Impairment in a Mouse Model of α-Synucleinopathies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301903. [PMID: 37381656 PMCID: PMC10477873 DOI: 10.1002/advs.202301903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/26/2023] [Indexed: 06/30/2023]
Abstract
The pathological accumulation of α-synuclein (α-Syn) and the transmission of misfolded α-Syn underlie α-synucleinopathies. Increased plasma α-Syn levels are associated with cognitive impairment in Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies, but it is still unknown whether the cognitive deficits in α-synucleinopathies have a common vascular pathological origin. Here, it is reported that combined injection of α-Syn preformed fibrils (PFFs) in the unilateral substantia nigra pars compacta, hippocampus, and cerebral cortex results in impaired spatial learning and memory abilities at 6 months post-injection and that this cognitive decline is related to cerebral microvascular injury. Moreover, insoluble α-Syn inclusions are found to form in primary mouse brain microvascular endothelial cells (BMVECs) through lymphocyte-activation gene 3 (Lag3)-dependent α-Syn PFFs endocytosis, causing poly(ADP-ribose)-driven cell death and reducing the expression of tight junction proteins in BMVECs. Knockout of Lag3 in vitro prevents α-Syn PFFs from entering BMVECs, thereby reducing the abovementioned response induced by α-Syn PFFs. Deletion of endothelial cell-specific Lag3 in vivo reverses the negative effects of α-Syn PFFs on cerebral microvessels and cognitive function. In short, this study reveals the effectiveness of targeting Lag3 to block the spread of α-Syn fibrils to endothelial cells in order to improve cognition.
Collapse
Affiliation(s)
- Qingxi Zhang
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510100China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Qingrui Duan
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yuyuan Gao
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Peikun He
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Rui Huang
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Haifeng Huang
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yanyi Li
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Guixian Ma
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yuhu Zhang
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Kun Nie
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Lijuan Wang
- Department of NeurologyGuangdong Neuroscience InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative DiseasesGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| |
Collapse
|
23
|
Leme MS, Sanches SGG, Carvallo RMM. Peripheral hearing in Parkinson's disease: a systematic review. Int J Audiol 2023; 62:805-813. [PMID: 35980314 DOI: 10.1080/14992027.2022.2109073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To investigate the implications of Parkinson's disease (PD) in the peripheral auditory system, a systematic survey of the scientific literature was conducted. DESIGN Systematic review. STUDY SAMPLE An electronic search of the non-gray literature in the last decade was conducted using the digital databases MEDLINE® (PubMed interface), LILACS® (Virtual Health Library), Web of Science® (CAPES publications portal), and SciELO®. Studies addressing peripheral auditory function as part of the range of nonmotor PD symptoms were selected for analysis. RESULTS Pure tone audiometry data suggested that sensorineural hearing loss was more severe in the PD population than in the control groups. The effects of PD on cochlear function were evidenced by a decrease in the levels of otoacoustic emissions. CONCLUSIONS Sensorineural hearing loss and cochlear impairment are more severe in the PD population than in the control groups. Additional studies are recommended to further understand the characteristics of the peripheral auditory system in PD patients, which constitutes an emerging subject in the scientific literature.
Collapse
Affiliation(s)
- Mariana S Leme
- Department of Physiotherapy, Speech Therapy & Audiology and Occupational Therapy, Faculdade de Medicina da Universidade de São Paulo-FMUSP, São Paulo, Brazil
| | - Seisse G G Sanches
- Department of Physiotherapy, Speech Therapy & Audiology and Occupational Therapy, Faculdade de Medicina da Universidade de São Paulo-FMUSP, São Paulo, Brazil
| | - Renata M M Carvallo
- Department of Physiotherapy, Speech Therapy & Audiology and Occupational Therapy, Faculdade de Medicina da Universidade de São Paulo-FMUSP, São Paulo, Brazil
| |
Collapse
|
24
|
Kawahata I, Sekimori T, Oizumi H, Takeda A, Fukunaga K. Using Fatty Acid-Binding Proteins as Potential Biomarkers to Discriminate between Parkinson's Disease and Dementia with Lewy Bodies: Exploration of a Novel Technique. Int J Mol Sci 2023; 24:13267. [PMID: 37686075 PMCID: PMC10487513 DOI: 10.3390/ijms241713267] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
An increase in the global aging population is leading to an increase in age-related conditions such as dementia and movement disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and dementia with Lewy bodies (DLB). The accurate prediction of risk factors associated with these disorders is crucial for early diagnosis and prevention. Biomarkers play a significant role in diagnosing and monitoring diseases. In neurodegenerative disorders like α-synucleinopathies, specific biomarkers can indicate the presence and progression of disease. We previously demonstrated the pathogenic impact of fatty acid-binding proteins (FABPs) in α-synucleinopathies. Therefore, this study investigated FABPs as potential biomarkers for Lewy body diseases. Plasma FABP levels were measured in patients with AD, PD, DLB, and mild cognitive impairment (MCI) and healthy controls. Plasma FABP3 was increased in all groups, while the levels of FABP5 and FABP7 tended to decrease in the AD group. Additionally, FABP2 levels were elevated in PD. A correlation analysis showed that higher FABP3 levels were associated with decreased cognitive function. The plasma concentrations of Tau, GFAP, NF-L, and UCHL1 correlated with cognitive decline. A scoring method was applied to discriminate between diseases, demonstrating high accuracy in distinguishing MCI vs. CN, AD vs. DLB, PD vs. DLB, and AD vs. PD. The study suggests that FABPs could serve as potential biomarkers for Lewy body diseases and aid in early disease detection and differentiation.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| | - Tomoki Sekimori
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| | - Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai 982-0805, Japan (A.T.)
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai 982-0805, Japan (A.T.)
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| |
Collapse
|
25
|
Lamberty BG, Estrella LD, Mattingly JE, Emanuel K, Trease A, Totusek S, Sheldon L, George JW, Almikhlafi MA, Farmer T, Stauch KL. Parkinson's disease relevant pathological features are manifested in male Pink1/Parkin deficient rats. Brain Behav Immun Health 2023; 31:100656. [PMID: 37484197 PMCID: PMC10362548 DOI: 10.1016/j.bbih.2023.100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Animal disease models are important for neuroscience experimentation and in the study of neurodegenerative disorders. The major neurodegenerative disorder leading to motor impairments is Parkinson's disease (PD). The identification of hereditary forms of PD uncovered gene mutations and variants, such as loss-of-function mutations in PTEN-induced putative kinase 1 (Pink1) and the E3 ubiquitin ligase Parkin, two proteins involved in mitochondrial quality control, that could be harnessed to create animal models. However, to date, such models have not reproducibly recapitulated major aspects of the disease. Here, we describe the generation and phenotypic characterization of a combined Pink1/Parkin double knockout (dKO) rat, which reproducibly exhibits PD-relevant abnormalities, particularly in male animals. Motor dysfunction in Pink1/Parkin dKO rats was characterized by gait abnormalities and decreased rearing frequency, the latter of which was responsive to levodopa treatment. Pink1/Parkin dKO rats exhibited elevated plasma levels of neurofilament light chain and significant loss of tyrosine hydroxylase expression in the substantia nigra pars compacta (SNpc). Glial cell activation was also observed in the SNpc. Pink1/Parkin dKO rats showed elevated plasma and reduced cerebrospinal levels of alpha-synuclein as well as the presence of alpha-synuclein aggregates in the striatum. Further, the profile of circulating lymphocytes was altered, as elevated CD3+CD4+ T cells and reduced CD3+CD8+ T cells in Pink1/Parkin dKO rats were found. This coincided with mitochondrial dysfunction and infiltration of CD3+ T cells in the striatum. Altogether, the Pink1/Parkin dKO rats exhibited phenotypes similar to what is seen with PD patients, thus highlighting the suitability of this model for mechanistic studies of the role of Pink1 and Parkin in PD pathogenesis and as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kelly L. Stauch
- Corresponding author. Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
26
|
Coughlin DG, Irwin DJ. Fluid and Biopsy Based Biomarkers in Parkinson's Disease. Neurotherapeutics 2023; 20:932-954. [PMID: 37138160 PMCID: PMC10457253 DOI: 10.1007/s13311-023-01379-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Several advances in fluid and tissue-based biomarkers for use in Parkinson's disease (PD) and other synucleinopathies have been made in the last several years. While work continues on species of alpha-synuclein (aSyn) and other proteins which can be measured from spinal fluid and plasma samples, immunohistochemistry and immunofluorescence from peripheral tissue biopsies and alpha-synuclein seeding amplification assays (aSyn-SAA: including real-time quaking induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA)) now offer a crucial advancement in their ability to identify aSyn species in PD patients in a categorical fashion (i.e., of aSyn + vs aSyn -); to augment clinical diagnosis however, aSyn-specific assays that have quantitative relevance to pathological burden remain an unmet need. Alzheimer's disease (AD) co-pathology is commonly found postmortem in PD, especially in those who develop dementia, and dementia with Lewy bodies (DLB). Biofluid biomarkers for tau and amyloid beta species can detect AD co-pathology in PD and DLB, which does have relevance for prognosis, but further work is needed to understand the interplay of aSyn tau, amyloid beta, and other pathological changes to generate comprehensive biomarker profiles for patients in a manner translatable to clinical trial design and individualized therapies.
Collapse
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, 9444 Medical Center Drive, ECOB 03-021, MCC 0886, La Jolla, CA, 92037, USA.
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
27
|
Guttuso T, Shepherd R, Frick L, Feltri ML, Frerichs V, Ramanathan M, Zivadinov R, Bergsland N. Lithium's effects on therapeutic targets and MRI biomarkers in Parkinson's disease: A pilot clinical trial. IBRO Neurosci Rep 2023; 14:429-434. [PMID: 37215748 PMCID: PMC10196787 DOI: 10.1016/j.ibneur.2023.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Background Lithium has a wide range of neuroprotective actions, has been effective in Parkinson's disease (PD) animal models and may account for the decreased risk of PD in smokers. Methods This open-label pilot clinical trial randomized 16 PD patients to "high-dose" (n = 5, lithium carbonate titrated to achieve serum level of 0.4-0.5 mmol/L), "medium-dose" (n = 6, 45 mg/day lithium aspartate) or "low-dose" (n = 5, 15 mg/day lithium aspartate) lithium therapy for 24-weeks. Peripheral blood mononuclear cell (PBMC) mRNA expression of nuclear receptor-related-1 (Nurr1) and superoxide dismutase-1 (SOD1) were assessed by qPCR in addition to other PD therapeutic targets. Two patients from each group received multi-shell diffusion MRI scans to assess for free water (FW) changes in the dorsomedial nucleus of the thalamus and nucleus basalis of Meynert, which reflect cognitive decline in PD, and the posterior substantia nigra, which reflects motor decline in PD. Results Two of the six patients receiving medium-dose lithium therapy withdrew due to side effects. Medium-dose lithium therapy was associated with the greatest numerical increases in PBMC Nurr1 and SOD1 expression (679% and 127%, respectively). Also, medium-dose lithium therapy was the only dosage associated with mean numerical decreases in brain FW in all three regions of interest, which is the opposite of the known longitudinal FW changes in PD. Conclusion Medium-dose lithium aspartate therapy was associated with engagement of blood-based therapeutic targets and improvements in MRI disease-progression biomarkers but was poorly tolerated in 33% of patients. Further PD clinical research is merited examining lithium's tolerability, effects on biomarkers and potential disease-modifying effects.
Collapse
Affiliation(s)
- Thomas Guttuso
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Rachel Shepherd
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Luciana Frick
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - M. Laura Feltri
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Valerie Frerichs
- Department of Chemistry, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Robert Zivadinov
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Center for Biomedical Imaging, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Niels Bergsland
- Department of Neurology, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Center for Biomedical Imaging, Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy
| |
Collapse
|
28
|
Okuzumi A, Hatano T, Matsumoto G, Nojiri S, Ueno SI, Imamichi-Tatano Y, Kimura H, Kakuta S, Kondo A, Fukuhara T, Li Y, Funayama M, Saiki S, Taniguchi D, Tsunemi T, McIntyre D, Gérardy JJ, Mittelbronn M, Kruger R, Uchiyama Y, Nukina N, Hattori N. Propagative α-synuclein seeds as serum biomarkers for synucleinopathies. Nat Med 2023; 29:1448-1455. [PMID: 37248302 PMCID: PMC10287557 DOI: 10.1038/s41591-023-02358-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 04/21/2023] [Indexed: 05/31/2023]
Abstract
Abnormal α-synuclein aggregation is a key pathological feature of a group of neurodegenerative diseases known as synucleinopathies, which include Parkinson's disease (PD), dementia with Lewy bodies and multiple system atrophy (MSA). The pathogenic β-sheet seed conformation of α-synuclein is found in various tissues, suggesting potential as a biomarker, but few studies have been able to reliably detect these seeds in serum samples. In this study, we developed a modified assay system, called immunoprecipitation-based real-time quaking-induced conversion (IP/RT-QuIC), which enables the detection of pathogenic α-synuclein seeds in the serum of individuals with synucleinopathies. In our internal first and second cohorts, IP/RT-QuIC showed high diagnostic performance for differentiating PD versus controls (area under the curve (AUC): 0.96 (95% confidence interval (CI) 0.95-0.99)/AUC: 0.93 (95% CI 0.84-1.00)) and MSA versus controls (AUC: 0.64 (95% CI 0.49-0.79)/AUC: 0.73 (95% CI 0.49-0.98)). IP/RT-QuIC also showed high diagnostic performance in differentiating individuals with PD (AUC: 0.86 (95% CI 0.74-0.99)) and MSA (AUC: 0.80 (95% CI 0.65-0.97)) from controls in a blinded external cohort. Notably, amplified seeds maintained disease-specific properties, allowing the differentiation of samples from individuals with PD versus MSA. In summary, here we present a novel platform that may allow the detection of individuals with synucleinopathies using serum samples.
Collapse
Affiliation(s)
- Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Gen Matsumoto
- Department of Histology and Cell Biology, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Shin-Ichi Ueno
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Haruka Kimura
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takeshi Fukuhara
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Manabu Funayama
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Neurology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Daisuke Taniguchi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Taiji Tsunemi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Deborah McIntyre
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Jean-Jacques Gérardy
- Luxembourg National Center of Pathology (NCP), Laboratoire National de Santé (LNS); Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH); Luxembourg Centre of Neuropathology (LCNP), Luxembourg Centre for Systems Biomedicine (LCSB), Faculty of Science, Technology and Medicine (FSTM) and Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michel Mittelbronn
- Luxembourg National Center of Pathology (NCP), Laboratoire National de Santé (LNS); Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH); Luxembourg Centre of Neuropathology (LCNP), Luxembourg Centre for Systems Biomedicine (LCSB), Faculty of Science, Technology and Medicine (FSTM) and Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rejko Kruger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL); Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Strassen, Luxembourg
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan.
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
29
|
Zhang Q, Lin Z, He Y, Jiang J, Hu D. Mendelian Randomization Analysis Reveals No Causal Relationship Between Plasma α-Synuclein and Parkinson's Disease. Mol Neurobiol 2023; 60:2268-2276. [PMID: 36640248 DOI: 10.1007/s12035-023-03206-0] [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: 04/26/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
So far, the studies exploring plasma α-synuclein as a biomarker of Parkinson's disease (PD) have provided contradictory results. Here, we first employed the Mendelian randomization (MR) approach to elucidate their potential causal relationship. Five genetic instrumental variables of plasma α-synuclein were acquired from two publicly available datasets. Three independent genome-wide association studies of PD were used as outcome cohorts (PD cohorts 1, 2, and 3). Two-sample MR analyses were conducted using inverse-variance weighted (IVW), MR-Egger, weighted median, simple mode, and leave-one-out methods. Though the IVW approach demonstrated positive plasma α-synuclein effect on the PD risk in three outcome cohorts (OR = 1.134, 1.164, and 1.189, respectively), the P values were all larger than 0.05. The conclusions were robust under complementary sensitivity analyses. Our results did not support the causal relationship between plasma α-synuclein and PD.
Collapse
Affiliation(s)
- Qi Zhang
- The Department of Neurology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Zenan Lin
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan He
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Junhong Jiang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Di Hu
- Children's Hospital of Fudan University, No.399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
30
|
Bearoff F, Dhavale D, Kotzbauer P, Kortagere S. Aggregated alpha-synuclein transcriptionally activates pro-inflammatory canonical and non-canonical NF-κB signaling pathways in peripheral monocytic cells. Mol Immunol 2023; 154:1-10. [PMID: 36571978 PMCID: PMC9905308 DOI: 10.1016/j.molimm.2022.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by chronic neuroinflammation, loss of dopaminergic neurons in the substantia nigra, and in several cases accumulation of alpha-synuclein fibril (α-syn) containing Lewy-bodies (LBs). Peripheral inflammation may play a causal role in inducing and perpetuating neuroinflammation in PD and accumulation of fibrillar α-syn has been reported at several peripheral sites including the gut and liver. Peripheral fibrillar α-syn may induce activation of monocytes via recognition by toll-like receptors (TLRs) and stimulation of downstream NF-κB signaling; however, the specific mechanism by which this occurs is not defined. In this study we utilized the THP-1 monocytic cell line to model the peripheral transcriptional response to preformed fibrillar (PFF) α-syn. Compared to monomeric α-syn, PFF α-syn displays overt inflammatory gene upregulation and pathway activation including broad pan-TLR signaling pathway activation and increases in TNF and IL1B gene expression. Notably, the non-canonical NF-κB signaling pathway gene and PD genome wide association study (GWAS) candidate NFKB2 was upregulated. Additionally, non-canonical NF-κB activation-associated RANK and CD40 pathways were also upregulated. Transcriptional-phenotype analysis suggests PFFs induce transcriptional programs associated with differentiation of monocytes towards macrophages and osteoclasts via non-canonical NF-κB signaling as a potential mechanism in which myeloid/monocyte cells may contribute to peripheral inflammation and pathogenesis in PD.
Collapse
Affiliation(s)
- Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Dhruva Dhavale
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Paul Kotzbauer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, United States.
| |
Collapse
|
31
|
Chiu PY, Yang FC, Chiu MJ, Lin WC, Lu CH, Yang SY. Relevance of plasma biomarkers to pathologies in Alzheimer's disease, Parkinson's disease and frontotemporal dementia. Sci Rep 2022; 12:17919. [PMID: 36289355 PMCID: PMC9605966 DOI: 10.1038/s41598-022-22647-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/18/2022] [Indexed: 01/20/2023] Open
Abstract
Amyloid plaques and tau tangles are pathological hallmarks of Alzheimer's disease (AD). Parkinson's disease (PD) results from the accumulation of α-synuclein. TAR DNA-binding protein (TDP-43) and total tau protein (T-Tau) play roles in FTD pathology. All of the pathological evidence was found in the biopsy. However, it is impossible to perform stein examinations in clinical practice. Assays of biomarkers in plasma would be convenient. It would be better to investigate the combinations of various biomarkers in AD, PD and FTD. Ninety-one subjects without neurodegenerative diseases, 76 patients with amnesic mild cognitive impairment (aMCI) or AD dementia, combined as AD family, were enrolled. One hundred and nine PD patients with normal cognition (PD-NC) or dementia (PDD), combined as PD family, were enrolled. Twenty-five FTD patients were enrolled for assays of plasma amyloid β 1-40 (Aβ1-40), Aβ1-42, T-Tau, α-synuclein and TDP-43 using immunomagnetic reduction (IMR). The results show that Aβs and T-Tau are major domains in AD family. α-synuclein is highly dominant in PD family. FTD is closely associated with TDP-43 and T-Tau. The dominant plasma biomarkers in AD family, PD family and FTD are consistent with pathology. This implies that plasma biomarkers are promising for precise and differential assessments of AD, PD and FTD in clinical practice.
Collapse
Affiliation(s)
- Pai-Yi Chiu
- grid.452796.b0000 0004 0634 3637Department of Neurology, Show Chwan Memorial Hospital, Chunghwa, 500 Taiwan ,MR-Guided Focus Ultrasound Center, Chang Bin Shaw Chwan Memorial Hospital, Changhwa, 505 Taiwan
| | - Fu-Chi Yang
- grid.278244.f0000 0004 0638 9360Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114 Taiwan
| | - Ming-Jang Chiu
- grid.19188.390000 0004 0546 0241Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100 Taiwan ,grid.19188.390000 0004 0546 0241Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 100 Taiwan ,grid.19188.390000 0004 0546 0241Department of Psychology, National Taiwan University, Taipei, 106 Taiwan ,grid.19188.390000 0004 0546 0241Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 106 Taiwan
| | - Wei-Che Lin
- grid.145695.a0000 0004 1798 0922Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung, 833 Taiwan
| | - Cheng-Hsien Lu
- grid.145695.a0000 0004 1798 0922Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung, 833 Taiwan
| | | |
Collapse
|
32
|
Su Q, Ng WL, Goh SY, Gulam MY, Wang LF, Tan EK, Ahn M, Chao YX. Targeting the inflammasome in Parkinson's disease. Front Aging Neurosci 2022; 14:957705. [PMID: 36313019 PMCID: PMC9596750 DOI: 10.3389/fnagi.2022.957705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/20/2022] [Indexed: 02/15/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases in which neuroinflammation plays pivotal roles. An important mechanism of neuroinflammation is the NLRP3 inflammasome activation that has been implicated in PD pathogenesis. In this perspective, we will discuss the relationship of some key PD-associated proteins including α-synuclein and Parkin and their contribution to inflammasome activation. We will also review promising inhibitors of NLRP3 inflammasome pathway that have potential as novel PD therapeutics. Finally, we will provide a summary of current and potential in vitro and in vivo models that are available for therapeutic discovery and development.
Collapse
Affiliation(s)
- Qi Su
- Programme in Emerging Infectious Diseases, Duke–NUS Medical School, Singapore, Singapore
| | - Wei Lun Ng
- Programme in Emerging Infectious Diseases, Duke–NUS Medical School, Singapore, Singapore
| | - Suh Yee Goh
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Muhammad Yaaseen Gulam
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke–NUS Medical School, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders Program, Duke–NUS Medical School, Singapore, Singapore
| | - Matae Ahn
- Programme in Emerging Infectious Diseases, Duke–NUS Medical School, Singapore, Singapore
| | - Yin-Xia Chao
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders Program, Duke–NUS Medical School, Singapore, Singapore
| |
Collapse
|
33
|
Tönges L, Buhmann C, Klebe S, Klucken J, Kwon EH, Müller T, Pedrosa DJ, Schröter N, Riederer P, Lingor P. Blood-based biomarker in Parkinson's disease: potential for future applications in clinical research and practice. J Neural Transm (Vienna) 2022; 129:1201-1217. [PMID: 35428925 PMCID: PMC9463345 DOI: 10.1007/s00702-022-02498-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/27/2022] [Indexed: 12/12/2022]
Abstract
The clinical presentation of Parkinson's disease (PD) is both complex and heterogeneous, and its precise classification often requires an intensive work-up. The differential diagnosis, assessment of disease progression, evaluation of therapeutic responses, or identification of PD subtypes frequently remains uncertain from a clinical point of view. Various tissue- and fluid-based biomarkers are currently being investigated to improve the description of PD. From a clinician's perspective, signatures from blood that are relatively easy to obtain would have great potential for use in clinical practice if they fulfill the necessary requirements as PD biomarker. In this review article, we summarize the knowledge on blood-based PD biomarkers and present both a researcher's and a clinician's perspective on recent developments and potential future applications.
Collapse
Affiliation(s)
- Lars Tönges
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany.
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, 44801, Bochum, Nordrhein-Westfalen, Germany.
| | - Carsten Buhmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Stephan Klebe
- Department of Neurology, University Hospital Essen, 45147, Essen, Germany
| | - Jochen Klucken
- Department of Digital Medicine, University Luxembourg, LCSB, L-4367, Belval, Luxembourg
- Digital Medicine Research Group, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Centre Hospitalier de Luxembourg, Digital Medicine Research Clinic, L-1210, Luxembourg, Luxembourg
| | - Eun Hae Kwon
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany
| | - Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weissensee, 13088, Berlin, Germany
| | - David J Pedrosa
- Department of Neurology, Universitätsklinikum Gießen and Marburg, Marburg Site, 35043, Marburg, Germany
- Center of Mind, Brain and Behaviour (CMBB), Philipps-Universität Marburg, 35043, Marburg, Germany
| | - Nils Schröter
- Department of Neurology and Clinical Neuroscience, University of Freiburg, 79106, Freiburg, Germany
| | - Peter Riederer
- Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, 97080, Wuerzburg, Germany
- University of Southern Denmark Odense, 5000, Odense, Denmark
| | - Paul Lingor
- School of Medicine, Klinikum Rechts Der Isar, Department of Neurology, Technical University of Munich, 81675, München, Germany
| |
Collapse
|
34
|
Li CH, Chen TF, Peng PL, Lin CH. A task-specific cognitive domain decline is correlated with plasma and neuroimaging markers in patients with Parkinson’s disease. J Neurol 2022; 269:6530-6543. [DOI: 10.1007/s00415-022-11301-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
|
35
|
Giri B, Seamon M, Banerjee A, Chauhan S, Purohit S, Morgan J, Baban B, Wakade C. Emerging urinary alpha-synuclein and miRNA biomarkers in Parkinson's disease. Metab Brain Dis 2022; 37:1687-1696. [PMID: 33881722 DOI: 10.1007/s11011-021-00735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases after Alzheimer's disease (AD), afflicting adults above the age of sixty irrespective of gender, race, ethnicity, and social status. PD is characterized by motor dysfunctions, displaying resting tremor, rigidity, bradykinesia, and postural imbalance. Non-motor symptoms, including rapid eye movement (REM) behavior disorder, constipation, and loss of sense of smell, typically occur many years before the appearance of the PD motor symptoms that lead to a diagnosis. The loss of dopaminergic neurons in the substantia nigra, which leads to the motor symptoms seen in PD, is associated with the deposition of aggregated, misfolded α-Synuclein (α-Syn, SNCA) proteins forming Lewy Bodies. Additionally, dysregulation of miRNA (a short form of mRNA) may contribute to the developing pathophysiology in PD and other diseases such as cancer. Overexpression of α-Syn and miRNA in human samples has been found in PD, AD, and dementia. Therefore, evaluating these molecules in urine, present either in the free form or in association with extracellular vesicles of biological fluids, may lead to early biomarkers for clinical diagnosis. Collection of urine is non-invasive and thus beneficial, particularly in geriatric populations, for biomarker analysis. Considering the expression and function of α-Syn and miRNA, we predict that they can be used as early biomarkers in the diagnosis and prognosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Banabihari Giri
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA.
| | - Marissa Seamon
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
- Department of Neuroscience, Augusta University, Augusta, GA, USA
| | - Aditi Banerjee
- Brain Peds Division, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Sneha Chauhan
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Sharad Purohit
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
| | - John Morgan
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Parkinson's Foundation Center of Excellence, Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Babak Baban
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation (CERSI), Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Chandramohan Wakade
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
- Department of Neuroscience, Augusta University, Augusta, GA, USA
- Parkinson's Foundation Center of Excellence, Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| |
Collapse
|
36
|
Plasma arylsulfatase A levels are associated with cognitive function in Parkinson’s disease. Neurol Sci 2022; 43:4753-4759. [PMID: 35486332 PMCID: PMC9349122 DOI: 10.1007/s10072-022-06093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/22/2022] [Indexed: 12/03/2022]
Abstract
Background Arylsulfatase A (ARSA), a lysosomal enzyme, has been shown to inhibit the aggregation and propagation of α-synuclein (α-syn) through its molecular chaperone function. The relationship between ARSA levels and Parkinson’s disease (PD) in the Chinese Han population remains controversial, and few quantitative research studies have investigated the relationship between plasma ARSA levels and PD. Objectives The purpose of this study was to investigate the relationships between ARSA levels and cognitive function in PD patients and to evaluate the association of ARSA and α-syn levels with nonmotor symptoms. Methods Enzyme-linked immunosorbent assay (ELISA) was used to measure the plasma ARSA and α-syn levels in 50 healthy controls, 120 PD patients (61 PD patients with no cognitive impairment (PD-NCI) and 59 PD patients with cognitive impairment (PD-CI)). Motor symptoms and nonmotor symptoms (cognitive function, Unified Parkinson’s Disease Rating Scale (UPDRS) score, depression, anxiety, constipation, olfactory dysfunction, sleep disruption, and other symptoms) were assessed with the relevant scales. The Kruskal–Wallis H test was used for comparison between groups, and Pearson/Spearman analysis was used for correlation analysis. Results The plasma ARSA concentrations were lower in the PD-CI group than in the PD-NCI group. The plasma α-syn levels in the PD-CI group were higher than those in the healthy control group, and the plasma ARSA levels were correlated with the Mini-Mental State Examination (MMSE scores) and Hoehn and Yahr (H-Y) stage. Conclusion We used a quantitative assessment method to show that low plasma ARSA levels and high α-syn levels are related to cognitive impairment in PD patients. Plasma ARSA levels gradually decrease with PD progression.
Collapse
|
37
|
Is peripheral alpha synuclein a marker for gait velocity in Parkinson's disease? Neurosci Lett 2022; 786:136819. [PMID: 35905887 DOI: 10.1016/j.neulet.2022.136819] [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: 06/03/2022] [Revised: 07/15/2022] [Accepted: 07/24/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The extent of gait abnormality is non-uniform across motor phenotypes of Parkinson's disease (PD). The biological basis of this heterogeneity remains intriguing. Moreover, the relationship of gait impairment with various neurodegenerative protein markers in PD is not well established. OBJECTIVES Here, we aimed to explore the interplay between gait parameters and specific serum protein markers in PD. METHODS A total of 62 PD patients were consecutively recruited. Blood samples and gait data were acquired from 37 and 34 patients respectively. Two-dimensional spatio-temporal gait parameters were estimated using an electronic walkway (GAITRite®, CIR Systems Inc., USA). Serum phosphorylated alpha synuclein (p-Ser129-a-syn) and total a-syn levels were measured using commercially available ELISA kit. Data was analyzed using SPSS Version 20 (IBM). RESULTS We found that phosphorylated a-syn levels were significantly higher in PD patients with postural instability and gait difficulty compared to tremor dominant variant. Significant reduction in gait velocity was also observed with increasing levels of this pathological form of a-syn. Regression modelling showed that phosphorylated a-syn is an independent predictor of gait velocity. DISCUSSION Our findings indicate that concentrations of peripheral p-Ser129-a-syn but not total a-syn could be a potential contributor of gait impairment in PD. Further investigation on the systemic role of phosphorylated a-syn on gait would bridge the gap between central and peripheral mechanisms underlying phenotypic variability in PD.
Collapse
|
38
|
Increased Levels of Plasma Alzheimer’s Disease Biomarkers and Their Associations with Brain Structural Changes and Carotid Intima-Media Thickness in Cognitively Normal Obstructive Sleep Apnea Patients. Diagnostics (Basel) 2022; 12:diagnostics12071522. [PMID: 35885428 PMCID: PMC9324500 DOI: 10.3390/diagnostics12071522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Obstructive sleep apnea (OSA) has been linked to Alzheimer’s disease (AD) and amyloid deposition in the brain. OSA is further linked to the development of cardiovascular and cerebrovascular diseases. In this study, we analyzed the plasma levels of AD neuropathology biomarkers and their relationships with structural changes of the brain and atherosclerosis. Thirty OSA patients with normal cognition and 34 normal controls were enrolled. Cognitive functions were assessed by the Wechsler Adult Intelligence Scale third edition and Cognitive Ability Screening Instrument. Plasma Aβ-40, Aβ-42, and T-tau levels were assayed using immunomagnetic reduction. The carotid intima-media thickness was measured to assess the severity of atherosclerosis. Structural MR images of brain were acquired with voxel-based morphometric analysis of T1 structural images. The OSA patients exhibited significantly elevated plasma levels of Aβ-42 and T-tau, as well as increased gray matter volume in the right precuneus. Plasma T-tau level is associated with carotid intima-media thickness and gray matter volume of the precuneus. These findings may indicate early changes that precede clinically apparent cognitive impairment. The measurement of these biomarkers may aid in the early detection of OSA-associated morbidity and possible treatment planning for the prevention of irreversible neuronal damage and cognitive dysfunction.
Collapse
|
39
|
Acquasaliente L, Pontarollo G, Radu CM, Peterle D, Artusi I, Pagotto A, Uliana F, Negro A, Simioni P, De Filippis V. Exogenous human α-Synuclein acts in vitro as a mild platelet antiaggregant inhibiting α-thrombin-induced platelet activation. Sci Rep 2022; 12:9880. [PMID: 35701444 PMCID: PMC9198058 DOI: 10.1038/s41598-022-12886-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
α-Synuclein (αSyn) is a small disordered protein, highly conserved in vertebrates and involved in the pathogenesis of Parkinson’s disease (PD). Indeed, αSyn amyloid aggregates are present in the brain of patients with PD. Although the pathogenic role of αSyn is widely accepted, the physiological function of this protein remains elusive. Beyond the central nervous system, αSyn is expressed in hematopoietic tissue and blood, where platelets are a major cellular host of αSyn. Platelets play a key role in hemostasis and are potently activated by thrombin (αT) through the cleavage of protease-activated receptors. Furthermore, both αT and αSyn could be found in the same spatial environment, i.e. the platelet membrane, as αT binds to and activates platelets that can release αSyn from α-granules and microvesicles. Here, we investigated the possibility that exogenous αSyn could interfere with platelet activation induced by different agonists in vitro. Data obtained from distinct experimental techniques (i.e. multiple electrode aggregometry, rotational thromboelastometry, immunofluorescence microscopy, surface plasmon resonance, and steady-state fluorescence spectroscopy) on whole blood and platelet-rich plasma indicate that exogenous αSyn has mild platelet antiaggregating properties in vitro, acting as a negative regulator of αT-mediated platelet activation by preferentially inhibiting P-selectin expression on platelet surface. We have also shown that both exogenous and endogenous (i.e. cytoplasmic) αSyn preferentially bind to the outer surface of activated platelets. Starting from these findings, a coherent model of the antiplatelet function of αSyn is proposed.
Collapse
Affiliation(s)
- Laura Acquasaliente
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Giulia Pontarollo
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Center for Thrombosis and Hemostasis (CTH) University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Claudia Maria Radu
- Department of Women's & Children's Health, University of Padua, Padua, Italy.,Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy
| | - Daniele Peterle
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Ave. 02115, Boston, MA, USA
| | - Ilaria Artusi
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Anna Pagotto
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Federico Uliana
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Institute of Molecular Systems Biology, ETH Zurich, 8093, Zurich, Switzerland
| | - Alessandro Negro
- Department of Biomedical Sciences, University of Padua, viale G. Colombo 3, 35100, Padua, Italy.
| | - Paolo Simioni
- Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy.
| | - Vincenzo De Filippis
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy. .,Biotechnology Center, CRIBI, University of Padua, Padua, Italy.
| |
Collapse
|
40
|
Zubelzu M, Morera-Herreras T, Irastorza G, Gómez-Esteban JC, Murueta-Goyena A. Plasma and serum alpha-synuclein as a biomarker in Parkinson's disease: A meta-analysis. Parkinsonism Relat Disord 2022; 99:107-115. [PMID: 35717321 DOI: 10.1016/j.parkreldis.2022.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Reliable biomarkers for Parkinson's disease (PD) diagnosis are urgently needed. Alpha-synuclein (α-syn) and its proteoforms play a key role in PD pathology but in vivo measurements have raised conflicting results, and whether α-syn in blood could distinguish PD patients from healthy controls is still controversial. METHODS A systematic literature search yielded 35 eligible studies for meta-analysis reporting the concentration of total, oligomeric or phosphorylated α-syn in plasma and/or serum of PD patients and healthy controls. Standardized mean differences (SMD) were pooled using multivariate/multilevel linear mixed-effects models. Meta-regression analyses were conducted to investigate possible modifiers. RESULTS A meta-analysis of 32 articles involving 2683 PD patients and 1838 controls showed a significant overall effect of PD on total α-syn levels (SMD = 0.85, p = 0.004). Meta-regression showed that increased SMD of total α-syn in PD was significantly associated with lower age, shorter disease duration, mild motor impairment, and Immunomagnetic Reduction assay for protein quantification. In contrast, no significant differences were observed for oligomeric or phosphorylated α-syn between PD and controls but increased oligomeric α-syn was significantly associated with shorter disease duration. The heterogeneity among studies was high (>98%). CONCLUSIONS These findings suggest that increased total plasma/serum α-syn levels in PD primarily occur in early phases of the disease. The evidence obtained from a small number of studies measuring plasma/serum concentrations of oligomeric and phosphorylated species of α-syn shows no difference. The clinical applicability of measuring plasma or serum α-syn species for differentiating PD from healthy control warrants further studies with better clinical profiling of PD patients.
Collapse
Affiliation(s)
- Maider Zubelzu
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| | - Gorka Irastorza
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Juan Carlos Gómez-Esteban
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Department of Neurology, Cruces University Hospital, Osakidetza, Barakaldo, Bizkaia, Spain
| | - Ane Murueta-Goyena
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
41
|
Chen YT, Orimo S, Wei CY, Hung GU, Yang SY, Chiu PY. Synuclein Motor Dysfunction Composite Scale for the Discrimination of Dementia With Lewy Bodies From Alzheimer’s Disease. Front Aging Neurosci 2022; 14:920591. [PMID: 35663565 PMCID: PMC9161692 DOI: 10.3389/fnagi.2022.920591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background An abnormal increase of α-synuclein in the brain is the hallmark of dementia with Lewy bodies (DLB). However, the diagnostic power of plasma α-synuclein in DLB is not yet confirmed. Parkinsonism is highly associated with and is one of the core clinical features of DLB. We studied plasma α-synuclein and developed a novel tool that combined plasma α-synuclein level and Motor Dysfunction Questionnaire (MDQ), namely Synuclein Motor Dysfunction Composite Scale (SMDCS), for the clinical discrimination of DLB from Alzheimer’s disease (AD). Methods This cross-sectional study analyzed participants’ demographical data, plasma α-synuclein level, MDQ, structured clinical history questionnaire, neuropsychological and motor function tests, and neuroimaging studies. The power of plasma α-synuclein level, MDQ, and SMDCS for discriminating DLB from non-demented controls (NC) or AD were compared. Results Overall, 121 participants diagnosed as 58 DLB, 31 AD, and 31 NC were enrolled. Patients with DLB had significantly higher mean plasma α-synuclein level (0.24 ± 0.32 pg/ml) compared to the NC group (0.08 ± 0.05 pg/ml) and the AD group (0.08 ± 0.05 pg/ml). The DLB group demonstrated higher MDQ (2.95 ± 1.60) compared to the NC (0.42 ± 0.98) or AD (0.44 ± 0.99) groups. The sensitivity/specificity of plasma α-synuclein level, MDQ, and SMDCS for differentiating DLB from non-DLB were 0.80/0.64, 0.83/0.89, and 0.88/0.93, respectively. Conclusion Both plasma α-synuclein and MDQ were significantly higher in patients with DLB compared to the NC or AD groups. The novel SMDCS, significantly improved accuracy for the clinical differentiation of DLB from AD or NC.
Collapse
Affiliation(s)
- Ying-Tsung Chen
- Department of Psychiatry, Show Chwan Memorial Hospital, Changhua, Taiwan
- Department of Psychiatry, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Satoshi Orimo
- Department of Neurology, Kamiyoga Setagaya Street Clinic, Tokyo, Japan
| | - Cheng-Yu Wei
- Department of Exercise and Health Promotion, College of Education, Chinese Culture University, Taipei, Taiwan
| | - Guang-Uei Hung
- Department of Nuclear Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | | | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan
- Department of Applied Mathematics, Tunghai University, Taichung, Taiwan
- *Correspondence: Pai-Yi Chiu,
| |
Collapse
|
42
|
Zhao X, He H, Xiong X, Ye Q, Feng F, Zhou S, Chen W, Xia K, Qian S, Yang Y, Xie C. Lewy Body-Associated Proteins A-Synuclein (a-syn) as a Plasma-Based Biomarker for Parkinson’s Disease. Front Aging Neurosci 2022; 14:869797. [PMID: 35645787 PMCID: PMC9133668 DOI: 10.3389/fnagi.2022.869797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/12/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction To explore the combined diagnostic value of plasma Lewy body-associated proteins (p-Asyn at ser129, total α-syn, and oligomeric α-syn) for the diagnosis of PD versus healthy controls (HCs) and other PD syndromes (PDs), as well as clinical characteristics prediction. Methods This study included 145 participants: 79 patients with PD, 24 patients with PDs, and 42 HCs. A panel of plasma levels of p-Asyn, total α-syn, and oligomeric α-syn was measured by enzyme-linked immunosorbent assay (ELISA). The primary outcome was the discriminative accuracy of the combined three plasma biomarkers for PD. Results The mean age was 65.43 (SD, 7.467) in the control group, 64.49 (SD, 8.224) in participants with PD, and 69.25 (SD, 7.952) in PDs. The plasma Lewy body-associated protein levels were significantly higher in patients with PD than in age-matched HCs, However, there was no difference in patients with PD and PDs. Of note, a combination of plasma p-Asyn, total α-syn, and oligomeric α-syn was a better biomarker for discriminating PD from HCs, with an AUC of 0.8552 (p < 0.0001, 95%CI, 0.7635–0.9409), which was significantly higher than plasma p-Asyn (ΔAUC, 0.1797), total α-syn (ΔAUC, 0.0891) and oligomeric α-syn (ΔAUC, 0.1592) alone. Meanwhile, Lewy body-associated proteins had no connections between different motor stages and dementia performances. Conclusion Our results suggested that plasma Lewy body-associated proteins, may serve as a non-invasive biomarker to aid the diagnosis of PD from HCs. In addition, increased plasma Lewy body-associated proteins were not associated with the progression of motor and non-motor symptoms.
Collapse
Affiliation(s)
- Xuemiao Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haijun He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Xiong
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qianqian Ye
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feifei Feng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuoting Zhou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weian Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Xia
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuangjie Qian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Yunjun Yang,
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Wenzhou, China
- Institute of Aging, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Wenzhou, China
- Chenglong Xie,
| |
Collapse
|
43
|
Detection and assessment of alpha-synuclein in Parkinson disease. Neurochem Int 2022; 158:105358. [PMID: 35561817 DOI: 10.1016/j.neuint.2022.105358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/19/2022] [Accepted: 05/01/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE Different studies have reported varying alpha-synuclein values in the cerebrospinal fluid (CSF), serum, and plasma, making determination of the alpha-synuclein cutoff value for Parkinson's disease difficult and rendering identifying the cause of variation essential. METHOD We searched PubMed from inception to June 2021 and identified 76 eligible studies. Included studies reported data on total, phosphorylated, and oligomeric alpha-synuclein in the CSF, serum, or plasma from individuals with Parkinson's disease and healthy controls. The mean or median alpha-synuclein values from the included studies were summarized and categorized through laboratory assays to visualize potential trends. RESULTS The enzyme-linked immunosorbent assay (ELISA) is the most common assay used to determine alpha-synuclein concentrations. Less common assays include Luminex, single molecule arrays, electrochemiluminescence, and immunomagnetic reduction (IMR). IMR is a single-antibody and wash-free immunoassay designed for determining the extremely low concentration of bio-molecules. For patients with Parkinson's disease, the median or mean testing values ranged from 60.9 to 55,000 pg/mL in the CSF, 0.446 to 1,777,100 pg/mL in plasma, and 0.0292 to 38,200,000 pg/mL in serum. The antibody selection was diverse between studies. The tendency of distribution was more centralized among studies that used the same kit. Studies adopting specific antibodies or in-house assays contribute to the extreme values. Only a few studies on phosphorylated and oligomeric alpha-synuclein were included. CONCLUSION The type of assay and antibody selection in the laboratory played major roles in the alpha-synuclein variation. Studies that used the same assay and kit yielded relatively unanimous results. Furthermore, IMR may be a promising assay for plasma and serum alpha-synuclein quantification. A consensus on sample preparation and testing protocol unification is warranted in the future.
Collapse
|
44
|
Ardizzone A, Bova V, Casili G, Filippone A, Campolo M, Lanza M, Esposito E, Paterniti I. SUN11602, a bFGF mimetic, modulated neuroinflammation, apoptosis and calcium-binding proteins in an in vivo model of MPTP-induced nigrostriatal degeneration. J Neuroinflammation 2022; 19:107. [PMID: 35526035 PMCID: PMC9080217 DOI: 10.1186/s12974-022-02457-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Parkinson’s disease (PD) is the second most frequent neurodegenerative disease. PD etiopathogenesis is multifactorial and not yet fully known, however, the scientific world advised the establishment of neuroinflammation among the possible risk factors. In this field, basic fibroblast growth factor/fibroblast growth factor receptor-1 (bFGF/FGFR1) could be a promising way to treat CNS-mediated inflammation; unfortunately, the use of bFGF as therapeutic agent is limited by its side effects. The novel synthetic compound SUN11602 exhibited neuroprotective activities like bFGF. With this perspective, this study aimed to evaluate the effect of SUN11602 administration in a murine model of MPTP-induced dopaminergic degeneration. Methods Specifically, nigrostriatal degeneration was induced by intraperitoneal injection of MPTP (80 mg/kg). SUN11602 (1 mg/kg, 2.5 mg/kg, and 5 mg/kg) was administered daily by oral gavage starting from 24 h after the first administration of MPTP. Mice were killed 7 days after MPTP induction. Results The results obtained showed that SUN11602 administration significantly reduced the alteration of PD hallmarks, attenuating the neuroinflammatory state via modulation of glial activation, NF-κB pathway, and cytokine overexpression. Furthermore, we demonstrated that SUN11602 treatment rebalanced Ca2+ overload in neurons by regulating Ca2+-binding proteins while inhibiting the apoptotic cascade. Conclusion Therefore, in the light of these findings, SUN11602 could be considered a valuable pharmacological strategy for PD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02457-3.
Collapse
Affiliation(s)
- Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Valentina Bova
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy.
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| |
Collapse
|
45
|
Barba L, Paolini Paoletti F, Bellomo G, Gaetani L, Halbgebauer S, Oeckl P, Otto M, Parnetti L. Alpha and Beta Synucleins: From Pathophysiology to Clinical Application as Biomarkers. Mov Disord 2022; 37:669-683. [PMID: 35122299 PMCID: PMC9303453 DOI: 10.1002/mds.28941] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
The synuclein family includes three neuronal proteins, named α‐synuclein, β‐synuclein, and γ‐synuclein, that have peculiar structural features. α‐synuclein is largely known for being a key protein in the pathophysiology of Parkinson's disease (PD) and other synucleinopathies, namely, dementia with Lewy bodies and multisystem atrophy. The role of β‐synuclein and γ‐synuclein is less well understood in terms of physiological functions and potential contribution to human diseases. α‐synuclein has been investigated extensively in both cerebrospinal fluid (CSF) and blood as a potential biomarker for synucleinopathies. Recently, great attention has been also paid to β‐synuclein, whose CSF and blood levels seem to reflect synaptic damage and neurodegeneration independent of the presence of synucleinopathy. In this review, we aim to provide an overview on the pathophysiological roles of the synucleins. Because γ‐synuclein has been poorly investigated in the field of synucleinopathy and its pathophysiological roles are far from being clear, we focus on the interactions between α‐synuclein and β‐synuclein in PD. We also discuss the role of α‐synuclein and β‐synuclein as potential biomarkers to improve the diagnostic characterization of synucleinopathies, thus highlighting their potential application in clinical trials for disease‐modifying therapies. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Lorenzo Barba
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
- Department of Neurology University of Ulm Ulm Germany
- Department of Neurology Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | - Federico Paolini Paoletti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Giovanni Bellomo
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Lorenzo Gaetani
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | | | - Patrick Oeckl
- Department of Neurology University of Ulm Ulm Germany
- German Center for Neurodegenerative Disorders Ulm (DZNE e. V.) Ulm Germany
| | - Markus Otto
- Department of Neurology University of Ulm Ulm Germany
- Department of Neurology Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | - Lucilla Parnetti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| |
Collapse
|
46
|
Alterations of Sphingolipid and Phospholipid Pathways and Ornithine Level in the Plasma as Biomarkers of Parkinson's Disease. Cells 2022; 11:cells11030395. [PMID: 35159203 PMCID: PMC8834036 DOI: 10.3390/cells11030395] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/07/2022] [Accepted: 01/22/2022] [Indexed: 02/01/2023] Open
Abstract
The biomarkers of Parkinson’s disease (PD) remain to be investigated. This work aimed to identify blood biomarkers for PD using targeted metabolomics analysis. We quantified the plasma levels of 255 metabolites in 92 PD patients and 60 healthy controls (HC). PD patients were sub-grouped into early (Hoehn–Yahr stage ≤ 2, n = 72) and advanced (Hoehn–Yahr stage > 2, n = 20) stages. Fifty-nine phospholipids, 3 fatty acids, 3 amino acids, and 7 biogenic amines, demonstrated significant alterations in PD patients. Six of them, dihydro sphingomyelin (SM) 24:0, 22:0, 20:0, phosphatidylethanolamine-plasmalogen (PEp) 38:6, and phosphatidylcholine 38:5 and 36:6, demonstrated lowest levels in PD patients in the advanced stage, followed by those in the early stage and HC. By contrast, the level of ornithine was highest in PD patients at the advanced stage, followed by those at the early stage and HC. These biomarker candidates demonstrated significant correlations with scores of motor disability, cognitive dysfunction, depression, and quality of daily life. The support vector machine algorithm using α-synuclein, dihydro SM 24:0, and PEp 38:6 demonstrated good ability to separate PD from HC (AUC: 0.820). This metabolomic analysis demonstrates new plasma biomarker candidates for PD and supports their role in participating PD pathogenesis and monitoring disease progression.
Collapse
|
47
|
Siddique Y. Neurodegenerative Disorders and the Current State, Pathophysiology, and Management of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:574-595. [PMID: 34477534 DOI: 10.2174/1871527320666210903101841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/14/2020] [Accepted: 02/13/2021] [Indexed: 06/13/2023]
Abstract
In the last few decades, major knowledge has been gained about pathophysiological aspects and molecular pathways behind Parkinson's Disease (PD). Based on neurotoxicological studies and postmortem investigations, there is a general concept of how environmental toxicants (neurotoxins, pesticides, insecticides) and genetic factors (genetic mutations in PD-associated proteins) cause depletion of dopamine from substantia nigra pars compacta region of the midbrain and modulate cellular processes leading to the pathogenesis of PD. α-Synuclein, a neuronal protein accumulation in oligomeric form, called protofibrils, is associated with cellular dysfunction and neuronal death, thus possibly contributing to PD propagation. With advances made in identifying loci that contribute to PD, molecular pathways involved in disease pathogenesis are now clear, and introducing therapeutic strategy at the right time may delay the progression. Biomarkers for PD have helped monitor PD progression; therefore, personalized therapeutic strategies can be facilitated. In order to further improve PD diagnostic and prognostic accuracy, independent validation of biomarkers is required.
Collapse
Affiliation(s)
- Yasir Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| |
Collapse
|
48
|
Park DG, Kang J, An YS, Chang J, Yoon JH. Association of plasma α-synuclein with cardiac 123I-MIBG scintigraphy in early Parkinson’s disease. Neurosci Lett 2022; 770:136399. [DOI: 10.1016/j.neulet.2021.136399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/06/2021] [Accepted: 12/13/2021] [Indexed: 01/28/2023]
|
49
|
Chang KH, Liu KC, Lai CS, Yang SY, Chen CM. Assessing Plasma Levels of α-Synuclein and Neurofilament Light Chain by Different Blood Preparation Methods. Front Aging Neurosci 2021; 13:759182. [PMID: 34858161 PMCID: PMC8630588 DOI: 10.3389/fnagi.2021.759182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
The potential biomarkers of Parkinson’s disease are α-synuclein and neurofilament light chain (NFL). However, inconsistent preanalytical preparation of plasma could lead to variations in levels of these biomarkers. Different types of potassium salts of EDTA and different centrifugation temperatures during plasma preparation may affect the results of α-synuclein and NFL measurements. In this study, we prepared plasma from eight patients with Parkinson’s disease (PD) and seven healthy controls (HCs) by using di- and tri-potassium (K2- and K3-) EDTA tubes and recruited a separated cohort with 42 PD patients and 40 HCs for plasma samples prepared from whole blood by centrifugation at room temperature and 4°C, respectively, in K2-EDTA tubes. The plasma levels of α-synuclein and NFL in K2- and K3-EDTA were similar. However, the levels of α-synuclein in the plasma prepared at 4°C (101.57 ± 43.43 fg/ml) were significantly lower compared with those at room temperature (181.23 ± 196.31 fg/ml, P < 0.001). Room temperature preparation demonstrated elevated plasma levels of α-synuclein in PD patients (256.6 ± 50.2 fg/ml) compared with the HCs (102.1 ± 0.66 fg/ml, P < 0.001), whereas this increase in PD was not present by preparation at 4°C. Both plasma preparations at room temperature and 4°C demonstrated consistent results of NFL, which are increased in PD patients compared with HCs. Our findings confirmed that K2- and K3-EDTA tubes were interchangeable for analyzing plasma levels of α-synuclein and NFL. Centrifugation at 4°C during plasma preparation generates considerable reduction and variation of α-synuclein level that might hinder the detection of α-synuclein level changes in PD.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Kou-Chen Liu
- Department of Electronic Engineering, Artificial Intelligence and Green Technology Research Center, Chang Gung University, Taoyuan, Taiwan.,Division of Pediatric Infectious Disease, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chao-Sung Lai
- Department of Electronic Engineering, Artificial Intelligence and Green Technology Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | | | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
50
|
Simon C, Soga T, Okano HJ, Parhar I. α-Synuclein-mediated neurodegeneration in Dementia with Lewy bodies: the pathobiology of a paradox. Cell Biosci 2021; 11:196. [PMID: 34798911 PMCID: PMC8605528 DOI: 10.1186/s13578-021-00709-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is epitomized by the pathognomonic manifestation of α-synuclein-laden Lewy bodies within selectively vulnerable neurons in the brain. By virtue of prion-like inheritance, the α-synuclein protein inexorably undergoes extensive conformational metamorphoses and culminate in the form of fibrillar polymorphs, instigating calamitous damage to the brain's neuropsychological networks. This epiphenomenon is nebulous, however, by lingering uncertainty over the quasi "pathogenic" behavior of α-synuclein conformers in DLB pathobiology. Despite numerous attempts, a monolithic "α-synuclein" paradigm that is able to untangle the enigma enshrouding the clinicopathological spectrum of DLB has failed to emanate. In this article, we review conceptual frameworks of α-synuclein dependent cell-autonomous and non-autonomous mechanisms that are likely to facilitate the transneuronal spread of degeneration through the neuraxis. In particular, we describe how the progressive demise of susceptible neurons may evolve from cellular derangements perpetrated by α-synuclein misfolding and aggregation. Where pertinent, we show how these bona fide mechanisms may mutually accentuate α-synuclein-mediated neurodegeneration in the DLB brain.
Collapse
Affiliation(s)
- Christopher Simon
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Tomoko Soga
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|