1
|
Abdelnour C, Young CB, Shahid-Besanti M, Smith A, Wilson EN, Ramos Benitez J, Vossler H, Plastini MJ, Winer JR, Kerchner GA, Cholerton B, Andreasson KI, Henderson VW, Yutsis M, Montine TJ, Tian L, Mormino EC, Poston KL. Plasma pTau181 Reveals a Pathological Signature that Predicts Cognitive Outcomes in Lewy Body Disease. Ann Neurol 2024; 96:526-538. [PMID: 38888142 PMCID: PMC11324388 DOI: 10.1002/ana.27003] [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: 12/03/2023] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE To determine whether plasma phosphorylated-Tau181 (pTau181) could be used as a diagnostic biomarker of concurrent Alzheimer's disease neuropathologic change (ADNC) or amyloidosis alone, as well as a prognostic, monitoring, and susceptibility/risk biomarker for clinical outcomes in Lewy body disease (LBD). METHODS We studied 565 participants: 94 LBD with normal cognition, 83 LBD with abnormal cognition, 114 with Alzheimer's disease, and 274 cognitively normal. Plasma pTau181 levels were measured with the Lumipulse G platform. Diagnostic accuracy for concurrent ADNC and amyloidosis was assessed with Receiver Operating Characteristic curves in a subset of participants with CSF pTau181/Aβ42, and CSF Aβ42/Aβ40 or amyloid-β PET, respectively. Linear mixed effects models were used to examine the associations between baseline and longitudinal plasma pTau181 levels and clinical outcomes. RESULTS Plasma pTau181 predicted concurrent ADNC and amyloidosis in LBD with abnormal cognition with 87% and 72% accuracy, respectively. In LBD patients with abnormal cognition, higher baseline plasma pTau181 was associated with worse baseline MoCA and CDR-SB, as well as accelerated decline in CDR-SB. Additionally, in this group, rapid increases in plasma pTau181 over 3 years predicted a faster decline in CDR-SB and memory. In LBD patients with normal cognition, there was no association between baseline or longitudinal plasma pTau181 levels and clinical outcomes; however, elevated pTau181 at baseline increased the risk of conversion to cognitive impairment. INTERPRETATION Our findings suggest that plasma pTau181 is a promising biomarker for concurrent ADNC and amyloidosis in LBD. Furthermore, plasma pTau181 holds potential as a prognostic, monitoring, and susceptibility/risk biomarker, predicting disease progression in LBD. ANN NEUROL 2024;96:526-538.
Collapse
Affiliation(s)
- Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marian Shahid-Besanti
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alena Smith
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Javier Ramos Benitez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hillary Vossler
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie J Plastini
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey A Kerchner
- Pharma Research and Early Development, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Brenna Cholerton
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Maya Yutsis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lu Tian
- Department of Biomedical Data Science, School of Medicine, Department of Statistics, School of Humanities and Sciences, Stanford University, Stanford, CA, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
2
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
3
|
Cristiani CM, Scaramuzzino L, Quattrone A, Parrotta EI, Cuda G, Quattrone A. Serum Oligomeric α-Synuclein and p-tau181 in Progressive Supranuclear Palsy and Parkinson's Disease. Int J Mol Sci 2024; 25:6882. [PMID: 38999992 PMCID: PMC11241320 DOI: 10.3390/ijms25136882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Clinical differentiation of progressive supranuclear palsy (PSP) from Parkinson's disease (PD) is challenging due to overlapping phenotypes and the late onset of specific atypical signs. Therefore, easily assessable diagnostic biomarkers are highly needed. Since PD is a synucleopathy while PSP is a tauopathy, here, we investigated the clinical usefulness of serum oligomeric-α-synuclein (o-α-synuclein) and 181Thr-phosphorylated tau (p-tau181), which are considered as the most important pathological protein forms in distinguishing between these two parkinsonisms. We assessed serum o-α-synuclein and p-tau181 by ELISA and SIMOA, respectively, in 27 PSP patients, 43 PD patients, and 39 healthy controls (HC). Moreover, we evaluated the correlation between serum biomarkers and biological and clinical features of these subjects. We did not find any difference in serum concentrations of p-tau181 and o-α-synuclein nor in the o-α-synuclein/p-tau181 ratio between groups. However, we observed that serum p-tau181 positively correlated with age in HC and PD, while serum o-α-synuclein correlated positively with disease severity in PD and negatively with age in PSP. Finally, the o-α-synuclein/p-tau181 ratio showed a negative correlation with age in PD.
Collapse
Affiliation(s)
- Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Luana Scaramuzzino
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Andrea Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Elvira Immacolata Parrotta
- Institute of Molecular Biology, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Department of Clinical and Experimental Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Aldo Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| |
Collapse
|
4
|
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
|
5
|
Furlepa M, Zhang YP, Lobanova E, Kahanawita L, Vivacqua G, Williams-Gray CH, Klenerman D. Single-molecule characterization of salivary protein aggregates from Parkinson's disease patients: a pilot study. Brain Commun 2024; 6:fcae178. [PMID: 38863577 PMCID: PMC11166177 DOI: 10.1093/braincomms/fcae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 04/03/2024] [Accepted: 05/20/2024] [Indexed: 06/13/2024] Open
Abstract
Saliva is a convenient and accessible biofluid that has potential as a future diagnostic tool for Parkinson's disease. Candidate diagnostic tests for Parkinson's disease to date have predominantly focused on measurements of α-synuclein in CSF, but there is a need for accurate tests utilizing more easily accessible sample types. Prior studies utilizing saliva have used bulk measurements of salivary α-synuclein to provide diagnostic insight. Aggregate structure may influence the contribution of α-synuclein to disease pathology. Single-molecule approaches can characterize the structure of individual aggregates present in the biofluid and may, therefore, provide greater insight than bulk measurements. We have employed an antibody-based single-molecule pulldown assay to quantify salivary α-synuclein and amyloid-β peptide aggregate numbers and subsequently super-resolved captured aggregates using direct Stochastic Optical Reconstruction Microscopy to describe their morphological features. We show that the salivary α-synuclein aggregate/amyloid-β aggregate ratio is increased almost 2-fold in patients with Parkinson's disease (n = 20) compared with controls (n = 20, P < 0.05). Morphological information also provides insight, with saliva from patients with Parkinson's disease containing a greater proportion of larger and more fibrillar amyloid-β aggregates than control saliva (P < 0.05). Furthermore, the combination of count and morphology data provides greater diagnostic value than either measure alone, distinguishing between patients with Parkinson's disease (n = 17) and controls (n = 18) with a high degree of accuracy (area under the curve = 0.87, P < 0.001) and a larger dynamic range. We, therefore, demonstrate for the first time the application of highly sensitive single-molecule imaging techniques to saliva. In addition, we show that aggregates present within saliva retain relevant structural information, further expanding the potential utility of saliva-based diagnostic methods.
Collapse
Affiliation(s)
- Martin Furlepa
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Yu P Zhang
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at Cambridge, Cambridge CB2 0XY, UK
| | - Evgeniia Lobanova
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at Cambridge, Cambridge CB2 0XY, UK
| | - Lakmini Kahanawita
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Giorgio Vivacqua
- Microscopic and Ultrastructural Anatomy Research Unit-Integrated Research Centre (PRABB), Campus Biomedico University of Rome, 00128 Rome, Italy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | | | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
6
|
Liu C, Su Y, Ma X, Wei Y, Qiao R. How close are we to a breakthrough? The hunt for blood biomarkers in Parkinson's disease diagnosis. Eur J Neurosci 2024; 59:2563-2576. [PMID: 38379501 DOI: 10.1111/ejn.16290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/22/2024]
Abstract
Parkinson's disease (PD), being the second largest neurodegenerative disease, poses challenges in early detection, resulting in a lack of timely treatment options to effectively manage the disease. By the time clinical diagnosis becomes possible, more than 60% of dopamine neurons in the substantia nigra (SN) of patients have already degenerated. Therefore, early diagnosis or identification of warning signs is crucial for the prompt and timely beginning of the treatment. However, conducting invasive or complex diagnostic procedures on asymptomatic patients can be challenging, making routine blood tests a more feasible approach in such cases. Numerous studies have been conducted over an extended period to search for effective diagnostic biomarkers in blood samples. However, thus far, no highly effective biomarkers have been confirmed. Besides classical proteins like α-synuclein (α-syn), phosphorylated α-syn and oligomeric α-syn, other molecules involved in disease progression should also be given equal attention. In this review, we will not only discuss proposed biomarkers that are currently under investigation but also delve into the mechanisms underlying the disease, focusing on processes such as α-syn misfolding, intercellular transmission and the crossing of the blood-brain barrier (BBB). Our aim is to provide an updated overview of molecules based on these processes that may potentially serve as blood biomarkers.
Collapse
Affiliation(s)
- Cheng Liu
- Peking University Third Hospital, Beijing, China
| | - Yang Su
- Peking University Third Hospital, Beijing, China
| | - Xiaolong Ma
- Peking University Third Hospital, Beijing, China
| | - Yao Wei
- Peking University Third Hospital, Beijing, China
| | - Rui Qiao
- Peking University Third Hospital, Beijing, China
| |
Collapse
|
7
|
Vijiaratnam N, Foltynie T. How should we be using biomarkers in trials of disease modification in Parkinson's disease? Brain 2023; 146:4845-4869. [PMID: 37536279 PMCID: PMC10690028 DOI: 10.1093/brain/awad265] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson's disease has formed the backbone for a proposed staging system for incorporation in Parkinson's disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson's disease patients into trials (as distinct from patients with non-Parkinson's disease parkinsonism or non-Parkinson's disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson's disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer's disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson's disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson's disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson's disease.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
8
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
9
|
Wolff A, Schumacher NU, Pürner D, Machetanz G, Demleitner AF, Feneberg E, Hagemeier M, Lingor P. Parkinson's disease therapy: what lies ahead? J Neural Transm (Vienna) 2023; 130:793-820. [PMID: 37147404 PMCID: PMC10199869 DOI: 10.1007/s00702-023-02641-6] [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: 02/15/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023]
Abstract
The worldwide prevalence of Parkinson's disease (PD) has been constantly increasing in the last decades. With rising life expectancy, a longer disease duration in PD patients is observed, further increasing the need and socioeconomic importance of adequate PD treatment. Today, PD is exclusively treated symptomatically, mainly by dopaminergic stimulation, while efforts to modify disease progression could not yet be translated to the clinics. New formulations of approved drugs and treatment options of motor fluctuations in advanced stages accompanied by telehealth monitoring have improved PD patients care. In addition, continuous improvement in the understanding of PD disease mechanisms resulted in the identification of new pharmacological targets. Applying novel trial designs, targeting of pre-symptomatic disease stages, and the acknowledgment of PD heterogeneity raise hopes to overcome past failures in the development of drugs for disease modification. In this review, we address these recent developments and venture a glimpse into the future of PD therapy in the years to come.
Collapse
Affiliation(s)
- Andreas Wolff
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Nicolas U Schumacher
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Dominik Pürner
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Gerrit Machetanz
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Antonia F Demleitner
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Emily Feneberg
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Maike Hagemeier
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Paul Lingor
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
10
|
Qamar MA, Rota S, Batzu L, Subramanian I, Falup-Pecurariu C, Titova N, Metta V, Murasan L, Odin P, Padmakumar C, Kukkle PL, Borgohain R, Kandadai RM, Goyal V, Chaudhuri KR. Chaudhuri's Dashboard of Vitals in Parkinson's syndrome: an unmet need underpinned by real life clinical tests. Front Neurol 2023; 14:1174698. [PMID: 37305739 PMCID: PMC10248458 DOI: 10.3389/fneur.2023.1174698] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
We have recently published the notion of the "vitals" of Parkinson's, a conglomeration of signs and symptoms, largely nonmotor, that must not be missed and yet often not considered in neurological consultations, with considerable societal and personal detrimental consequences. This "dashboard," termed the Chaudhuri's vitals of Parkinson's, are summarized as 5 key vital symptoms or signs and comprise of (a) motor, (b) nonmotor, (c) visual, gut, and oral health, (d) bone health and falls, and finally (e) comorbidities, comedication, and dopamine agonist side effects, such as impulse control disorders. Additionally, not addressing the vitals also may reflect inadequate management strategies, leading to worsening quality of life and diminished wellness, a new concept for people with Parkinson's. In this paper, we discuss possible, simple to use, and clinically relevant tests that can be used to monitor the status of these vitals, so that these can be incorporated into clinical practice. We also use the term Parkinson's syndrome to describe Parkinson's disease, as the term "disease" is now abandoned in many countries, such as the U.K., reflecting the heterogeneity of Parkinson's, which is now considered by many as a syndrome.
Collapse
Affiliation(s)
- Mubasher A. Qamar
- Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Division of Neuroscience, King’s College London, London, United Kingdom
- King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Silvia Rota
- Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Division of Neuroscience, King’s College London, London, United Kingdom
- King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Lucia Batzu
- Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Division of Neuroscience, King’s College London, London, United Kingdom
- King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Indu Subramanian
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Parkinson’s Disease Research, Education and Clinical Centers, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA, United States
| | - Cristian Falup-Pecurariu
- Faculty of Medicine, Transilvania University of Braşov, Brașov, Romania
- Department of Neurology, County Clinic Hospital, Brașov, Romania
| | - Nataliya Titova
- Department of Neurology, Neurosurgery and Medical Genetics, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov Russian National Research Medical University” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Neurodegenerative Diseases, Federal State Budgetary Institution “Federal Center of Brain Research and Neurotechnologies” of the Federal Medical Biological Agency, Moscow, Russia
| | - Vinod Metta
- Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Division of Neuroscience, King’s College London, London, United Kingdom
- King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Lulia Murasan
- Faculty of Medicine, Transilvania University of Braşov, Brașov, Romania
- Department of Neurology, County Clinic Hospital, Brașov, Romania
| | - Per Odin
- Department of Neurology, University Hospital, Lund, Sweden
| | | | - Prashanth L. Kukkle
- Center for Parkinson’s Disease and Movement Disorders, Manipal Hospital, Karnataka, India, Bangalore
- Parkinson’s Disease and Movement Disorders Clinic, Bangalore, Karnataka, India
| | - Rupam Borgohain
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rukmini Mridula Kandadai
- Department of Neurology, Nizam’s Institute of Medical Sciences, Autonomous University, Hyderabad, India
| | - Vinay Goyal
- Neurology Department, Medanta, Gurugram, India
| | - Kallo Ray Chaudhuri
- Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Division of Neuroscience, King’s College London, London, United Kingdom
- King’s College Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
11
|
Bougea A, Stefanis L. microRNA and circRNA in Parkinson's Disease and atypical parkinsonian syndromes. Adv Clin Chem 2023; 115:83-133. [PMID: 37673523 DOI: 10.1016/bs.acc.2023.03.002] [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: 03/30/2023]
Abstract
Multiple System Atrophy (MSA) and Progressive Supranuclear Palsy (PSP) are atypical parkinsonian syndromes (APS) with various clinical phenotypes and considerable clinical overlap with idiopathic Parkinson's disease (iPD). This disease heterogeneity makes ante-mortem diagnosis extremely challenging with up to 24% of patients misdiagnosed. Because diagnosis is predominantly clinical, there is great interest in identifying biomarkers for early diagnosis and differentiation of the different types of parkinsonism. Compared to protein biomarkers, microRNAs (miRNAs) and circularRNAs (circRNAs) are stable tissue-specific molecules that can be accurately measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). This chapter critically reviews miRNAs and circRNAs as diagnostic biomarkers and therapeutics to differentiate atypical parkinsonian disorders and their role in disease pathogenesis.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Leonidas Stefanis
- 1st Department of Neurology, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Huang SY, Chen SF, Cui M, Zhao M, Shen XN, Guo Y, Zhang YR, Zhang W, Wang HF, Huang YY, Cheng W, Zuo CT, Dong Q, Yu JT. Plasma Biomarkers and Positron Emission Tomography Tau Pathology in Progressive Supranuclear Palsy. Mov Disord 2023; 38:676-682. [PMID: 36781585 DOI: 10.1002/mds.29339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Development of disease-modifying therapeutic trials of progressive supranuclear palsy (PSP) urges the need for sensitive fluid biomarkers. OBJECTIVES The objectives of this study were to explore the utility of plasma biomarkers in the diagnosis, differential diagnosis, and assessment of disease severity, brain atrophy, and tau deposition in PSP. METHODS Plasma biomarkers were measured using a single-molecule array in a cohort composed of patients with PSP, Parkinson's disease (PD), multiple system atrophy with predominant parkinsonism (MSA-P), and healthy controls (HCs). RESULTS Plasma neurofilament light chain (NfL) outperformed other plasma makers (ie, glial fibrillary acidic protein [GFAP], phosphorylated-tau 181 [p-tau181], amyloid-β 1-40, amyloid-β 1-42) in identifying PSP from HC (area under the curve [AUC] = 0.904) and from MSA-P (AUC = 0.711). Plasma GFAP aided in distinguishing PSP from HC (AUC = 0.774) and from MSA-P (AUC = 0.832). It correlated with brainstem atrophy and higher regional tau accumulation. However, plasma p-tau181 neither helped in diagnosis nor was it associated with clinical or neuroimaging measures. CONCLUSIONS Plasma NfL and GFAP showed different values in differentiating PSP from HC or controls with other forms of neurodegenerative parkinsonism and detecting disease severity, brain atrophy, or tau deposition in PSP. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Shu-Fen Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Mei Cui
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Meng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xue-Ning Shen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Hui-Fu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yu-Yuan Huang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wei Cheng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| |
Collapse
|
13
|
Gonzalez-Robles C, Weil RS, van Wamelen D, Bartlett M, Burnell M, Clarke CS, Hu MT, Huxford B, Jha A, Lambert C, Lawton M, Mills G, Noyce A, Piccini P, Pushparatnam K, Rochester L, Siu C, Williams-Gray CH, Zeissler ML, Zetterberg H, Carroll CB, Foltynie T, Schrag A. Outcome Measures for Disease-Modifying Trials in Parkinson's Disease: Consensus Paper by the EJS ACT-PD Multi-Arm Multi-Stage Trial Initiative. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1011-1033. [PMID: 37545260 PMCID: PMC10578294 DOI: 10.3233/jpd-230051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/23/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Multi-arm, multi-stage (MAMS) platform trials can accelerate the identification of disease-modifying treatments for Parkinson's disease (PD) but there is no current consensus on the optimal outcome measures (OM) for this approach. OBJECTIVE To provide an up-to-date inventory of OM for disease-modifying PD trials, and a framework for future selection of OM for such trials. METHODS As part of the Edmond J Safra Accelerating Clinical Trials in Parkinson Disease (EJS ACT-PD) initiative, an expert group with Patient and Public Involvement and Engagement (PPIE) representatives' input reviewed and evaluated available evidence on OM for potential use in trials to delay progression of PD. Each OM was ranked based on aspects such as validity, sensitivity to change, participant burden and practicality for a multi-site trial. Review of evidence and expert opinion led to the present inventory. RESULTS An extensive inventory of OM was created, divided into: general, motor and non-motor scales, diaries and fluctuation questionnaires, cognitive, disability and health-related quality of life, capability, quantitative motor, wearable and digital, combined, resource use, imaging and wet biomarkers, and milestone-based. A framework for evaluation of OM is presented to update the inventory in the future. PPIE input highlighted the need for OM which reflect their experience of disease progression and are applicable to diverse populations and disease stages. CONCLUSION We present a range of OM, classified according to a transparent framework, to aid selection of OM for disease-modifying PD trials, whilst allowing for inclusion or re-classification of relevant OM as new evidence emerges.
Collapse
Affiliation(s)
| | | | | | | | - Matthew Burnell
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yu Y, Xia X, Meng X, Li D, Qin Q. Plasma Phosphorylated Tau181 and Amyloid-β42 in Dementia with Lewy Bodies Compared with Alzheimer's Disease and Cognitively Healthy People. J Alzheimers Dis 2023; 95:161-169. [PMID: 37482995 DOI: 10.3233/jad-230085] [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: 07/25/2023]
Abstract
BACKGROUND Increasing evidence illustrates the value of plasma biomarkers of Alzheimer's disease (AD) to screen for and identify dementia with Lewy bodies (DLB). However, confirmatory studies are needed to demonstrate the feasibility of these markers. OBJECTIVE To determine the feasibility of plasma tau phosphorylated at threonine 181 (p-tau181) and amyloid-β42 (Aβ42) as potential biomarkers to differentiate AD and DLB. METHODS We evaluated plasma samples from patients with DLB (n = 47) and AD (n = 55) and healthy controls (HCs, n = 30), using ELISAs to measure p-tau181 and Aβ42. Additionally, we examined neuropsychological assessment scores for participants. The plasma biomarkers were investigated for correlation with neuropsychological assessments and discriminant ability to identify DLB. RESULTS Plasma p-tau181 was significantly lower in DLB than in AD and HCs. Plasma Aβ42 was significantly higher in DLB than in AD but lower in DLB than in HCs. We found good correlations between plasma Aβ42 and neuropsychological scores in the whole cohort, while p-tau181 was associated with cognitive status in DLB. In the distinction between DLB and HCs, plasma p-tau181 and Aβ42 showed similar accuracy, while Aβ42 showed better accuracy than p-tau181 in discriminating DLB and AD. CONCLUSION In a single-center clinical cohort, we confirmed the high diagnostic value of plasma p-tau181 and Aβ42 for distinguishing patients with DLB from HCs. Plasma Aβ42 improved the differential diagnosis of DLB from AD.
Collapse
Affiliation(s)
- Yueyi Yu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinyi Xia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaosheng Meng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dan Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
15
|
Wang Z, Zheng Y, Cai H, Yang C, Li S, Lv H, Feng T, Yu Z. Aβ1-42-containing platelet-derived extracellular vesicle is associated with cognitive decline in Parkinson's disease. Front Aging Neurosci 2023; 15:1170663. [PMID: 37122378 PMCID: PMC10140302 DOI: 10.3389/fnagi.2023.1170663] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Background Cortical amyloid deposition is a common observation in Parkinson's disease dementia (PDD) patients. Aβ1-42 is linked to a more rapid progression of dementia. Platelets, which degranulate upon activation, are a primary source of Aβ. It has been repeatedly reported that peripheral extracellular vesicles (EVs) can partially reach the central nervous system. Thus, we speculate that activated platelet-derived Aβ1-42-containing EVs (PEV-Aβ1-42) play a crucial role in the cognitive decline of PD patients. Methods The study included 189 participants: 66 with non-dementia PD, 73 with PDD, and 50 healthy controls. All participants underwent blood collection and clinical assessments. Twenty PD patients underwent re-examination and repeated blood collection 14 months later. A nano-scale flow cytometry assay was used to detect PEVs and PEV-Aβ1-42 using fluorescence-labeled CD62P and Aβ1-42 antibodies. Results Parkinson's disease dementia patients had higher PEV-Aβ1-42 concentrations than healthy controls (p = 0.028). The ratio of PEV-Aβ1-42 to PEV was significantly higher in PDD patients compared to those in non-dementia PD and healthy controls (p PD-ND < 0.001, p HC = 0.041). The PEV-Aβ1-42/PEV ratio appears to influence the odds of developing dementia (OR = 1.76, p < 0.001). The change in the PEV-Aβ1-42/PEV ratio was also correlated with cognitive decline over 14 months (r = -0.447, p < 0.05). Conclusion The plasma PEV-Aβ1-42/PEV ratio may serve as a diagnostic and prognostic biomarker for PDD patients.
Collapse
Affiliation(s)
- Ziyu Wang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanchu Zheng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huihui Cai
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chen Yang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Siming Li
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Lv
- Clinical Diagnosis Department of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Zhenwei Yu, ; Tao Feng, , ; Hong Lv,
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- *Correspondence: Zhenwei Yu, ; Tao Feng, , ; Hong Lv,
| | - Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- *Correspondence: Zhenwei Yu, ; Tao Feng, , ; Hong Lv,
| |
Collapse
|
16
|
Chen WR, Chen JC, Chang SY, Chao CT, Wu YR, Chen CM, Chou C. Phosphorylated α-synuclein in diluted human serum as a biomarker for Parkinson's disease. Biomed J 2022; 45:914-922. [PMID: 34974168 PMCID: PMC9795354 DOI: 10.1016/j.bj.2021.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 12/17/2021] [Accepted: 12/26/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders, which characterized by increased pathological marker protein, α-synuclein (α-syn) and phosphorylated-Ser129-α-syn in the extracellular fluids. Current methods of measuring the p-Ser129-α-syn concentration in cerebrospinal fluid for PD are based on ELISA method, however, the amount of area under the curve (AUC) to predict PD is around 0.7-0.8. Higher confidence level of AUC in p-Ser129-α-syn quantification for the early diagnosis of PD would be essential. METHODS Detection of p-Ser129-α-syn in diluted human serum for diagnosis of PD was investigated by a modified paired surface plasma wave biosensor (PSPWB) using a quarter wave plate for better detection performance. The method combining an immunoassay and non-labeled technique measures the p-Ser129-α-syn level with high sensitivity and specificity. Ten patients with PD at early stage (Hohn & Yahr stage I and II) and 11 age-matched healthy control participants were recruited for measurement of serum p-Ser129-α-syn. RESULTS AUC of the p-Ser129-α-syn in diluted human serum was 0.92 and it shows that p-Ser129-α-syn in diluted human serum could be used as a sensitive biomarker for the diagnosis of PD in clinics. Results clearly show that the measured p-Ser129-α-syn concentration in diluted human serum displays a statistical significance between health control subjects and PD patients. CONCLUSIONS P-Ser129-α-syn has low abundance in human serum, high detection sensitivity and specificity are critical to the success of the diagnosis of PD in clinics. In this study, a modified PSPWB was developed that the limit of detection at 1 ng/mL for p-Ser129-α-syn (standard) spiked into diluted human serum of a healthy control was performed. This result shows that the modified PSPWB can be used as a platform for detecting p-Ser129-α-syn in diluted human serum as a potential biomarker for PD.
Collapse
Affiliation(s)
- Wei-Ru Chen
- PhD Program in Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Jin-Chung Chen
- Institute of Biomedical Science, Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Yi Chang
- PhD Program in Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan,The General Education Center, Ming Chi University of Technology, Taipei, Taiwan
| | - Chi-Tse Chao
- Graduate Institute of Electro-optical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan,College of Medicine, Chang-Gung University, Taoyuan, Taiwan,Corresponding author. Department of Neurology, Chang-Gung Memorial Hospital at Linkou, 5, Fusing St., Gueishan, Taoyuan 333 Taiwan.
| | - Chiung-Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital at Linkou, Taoyuan, Taiwan,College of Medicine, Chang-Gung University, Taoyuan, Taiwan,Corresponding author. Department of Neurology, Chang-Gung Memorial Hospital at Linkou, 5, Fusing St., Gueishan, Taoyuan 333 Taiwan.
| | - Chien Chou
- PhD Program in Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan,Graduate Institute of Electro-optical Engineering, Chang Gung University, Taoyuan, Taiwan,Corresponding author. PhD Program in Biomedical Engineering, Chang Gung University, 259, Wenhua 1st Rd., Gueishan, Taoyuan 333, Taiwan.
| |
Collapse
|
17
|
Batzu L, Rota S, Hye A, Heslegrave A, Trivedi D, Gibson LL, Farrell C, Zinzalias P, Rizos A, Zetterberg H, Chaudhuri KR, Aarsland D. Plasma p-tau181, neurofilament light chain and association with cognition in Parkinson’s disease. NPJ Parkinsons Dis 2022; 8:154. [DOI: 10.1038/s41531-022-00384-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/30/2022] [Indexed: 11/14/2022] Open
Abstract
AbstractEarly identification of cognitive impairment in Parkinson’s disease (PD) has important clinical and research implications. The aim of our study was to investigate the role of plasma tau phosphorylated at amino acid 181 (p-tau181) and plasma neurofilament light chain (NfL) as biomarkers of cognition in PD. Baseline concentrations of plasma p-tau181 and NfL were measured in a cohort of 136 patients with PD and 63 healthy controls (HC). Forty-seven PD patients were followed up for up to 2 years. Cross-sectional and longitudinal associations between baseline plasma biomarkers and cognitive progression were investigated using linear regression and linear mixed effects models. At baseline, plasma p-tau181 concentration was significantly higher in PD subjects compared with HC (p = 0.026). In PD patients, higher plasma NfL was associated with lower MMSE score at baseline, after adjusting for age, sex and education (p = 0.027). Baseline plasma NfL also predicted MMSE decline over time in the PD group (p = 0.020). No significant association between plasma p-tau181 concentration and baseline or longitudinal cognitive performance was found. While the role of p-tau181 as a diagnostic biomarker for PD and its relationship with cognition need further elucidation, plasma NfL may serve as a feasible, non-invasive biomarker of cognitive progression in PD.
Collapse
|
18
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
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: 19] [Impact Index Per Article: 9.5] [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
|
20
|
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: 10.5] [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
|
21
|
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
|
22
|
Bougea A. MicroRNA as Candidate Biomarkers in Atypical Parkinsonian Syndromes: Systematic Literature Review. Medicina (B Aires) 2022; 58:medicina58040483. [PMID: 35454322 PMCID: PMC9025474 DOI: 10.3390/medicina58040483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Background and Objectives: Multiple system atrophy (MSA) and progressive supranuclear palsy (PSP) are rare atypical parkinsonian syndromes, characterized by motor and cognitive symptoms. Their clinical diagnosis is challenging because there are no established biomarkers. Dysregulation of microRNAs (miRNAs/miRs) has been reported to serve an important role in neurodegenerative diseases. However, the miRNA profiles of MSA and PSP patients are rarely reported. The aim of this study was to critically review the role of miRNAs as diagnostic biomarkers to differentiate these atypical parkinsonian disorders and their role in disease pathogenesis. Materials and Methods: A systematic literature search of PubMed was conducted up to February 2022 according the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results: A total of 15 studies were analyzed. Three studies have shown that miR-9-3p, miR-19a, miR-19b, and miR-24 are potential biomarkers for MSA. In two studies, miR-132 was downregulated, whereas miR-147a and miR-518e were upregulated in the brain tissue of PSP patients. Conclusions: The potential of miRNA is still uncertain as a potential differential diagnostic marker to identify these disorders. Pre-analytical and analytical factors of included studies were important limitations to justify the introduction of miRNAs into clinical practice.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 72-74 Vassilisis Sofia's Avenue, 11528 Athens, Greece
| |
Collapse
|
23
|
Ren J, Pan C, Wang Y, Xue C, Lin H, Xu J, Wang H, Zhang W, Xu P, Chen Y, Liu W. Plasma α-synuclein and phosphorylated tau 181 as a diagnostic biomarker panel for de novo Parkinson's Disease. J Neurochem 2022; 161:506-515. [PMID: 35234288 PMCID: PMC9314946 DOI: 10.1111/jnc.15601] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022]
Abstract
The use of a diagnostic panel comprising multiple biomarkers has the potential to accurately diagnose Parkinson’s disease (PD). However, a panel consisting solely of plasma biomarkers to diagnose PD is not available. This study aimed to examine the diagnostic ability of plasma biomarker panels for de novo PD using novel digital ultrasensitive immunoassay technology. We recruited 45 patients with de novo PD and 20 healthy controls (HCs). The concentrations of plasma α‐synuclein (α‐syn), amyloid β‐42 (Aβ42), Aβ40, phosphorylated tau 181 (p‐tau181), neurofilament light (NFL), and glial fibrillary acidic protein (GFAP) were quantified using the ultrasensitive single molecule array (Simoa) platform. Patients with de novo PD had higher plasma levels of α‐syn and p‐tau181 than HCs, adjusting for age and sex. Plasma levels of α‐syn and p‐tau181 were positively correlated in de novo PD patients. Higher plasma α‐syn levels were significantly associated with worse Unified Parkinson’s Disease Rating Scale (UPDRS) Part III motor scores, modified Hoehn and Yahr (H‐Y) stages, and increased risk of PD with mild cognitive impairment (PD‐MCI). Higher plasma p‐tau181 concentrations were linked to worse H‐Y stages. The diagnostic panel using plasma α‐syn and p‐tau181, combined with age and sex, showed good performance in discriminating de novo PD patients from HCs (area under the curve = 0.806). These findings suggest that plasma α‐syn and p‐tau181 together may be a promising diagnostic biomarker panel for de novo PD patients.
Collapse
Affiliation(s)
- Jingru Ren
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chenxi Pan
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yajie Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Xue
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Huixia Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jianxia Xu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Pingyi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong Chen
- Department of Laboratory, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Li Q, Li Z, Han X, Shen X, Wang F, Bai L, Li Z, Zhang R, Wang Y, Zhu X. A Panel of Plasma Biomarkers for Differential Diagnosis of Parkinsonian Syndromes. Front Neurosci 2022; 16:805953. [PMID: 35250451 PMCID: PMC8891509 DOI: 10.3389/fnins.2022.805953] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The aim of our study is to explore the most reliable panel of plasma biomarkers for differential diagnosis of parkinsonian syndromes (PDSs). We selected five kinds of neurodegenerative proteins in plasma: neurofilament light chain (NfL), α-synuclein (α-syn), total tau, β-amyloid 42 (Aβ42) and β-amyloid 40 (Aβ40), and investigated the diagnostic value of these biomarkers. Methods A total of 99 plasma samples from patients with Parkinson’s disease (PD), multiple system atrophy (MSA), progressive supranuclear palsy, and age-matched healthy controls (HCs) were enrolled in our study. Plasma NfL, α-syn, total tau, Aβ42, and Aβ40 levels were quantified by ultrasensitive single molecule array immunoassay. We used logistic regression analyses to examine diagnostic accuracy of these plasma biomarkers. Disease severity was assessed by the modified Hoehn and Yahr staging scale, Unified Parkinson’s Disease Rating Scale part III (UPDRS III), and the Mini-Mental State Examination (MMSE), and subsequently, correlation analysis was performed. Results A combination of α-syn, Aβ42, Aβ40, Aβ42/40, and NfL could achieve a best diagnostic value in differentiating PDSs from HC and PD from HC, with an AUC of 0.983 and 0.977, respectively. By adding NfL to measurements of α-syn or Aβ42 or Aβ40 or Aβ42/40, the best discriminating panel was formed in differentiating atypical parkinsonian disorder (APD) and HC, and the discriminatory potential could reach a sensitivity of 100% and specificity of 100% (AUC = 1.000). For further distinguishing PD from APD, we found a combination of NfL, Aβ42, and total tau was the most reliable panel with equally high diagnostic accuracy. With respect to differentiating the subtypes of APD from one another, our results revealed that measurement of NfL, total tau, Aβ42, Aβ40, and Aβ42/40 was the best discriminating panel. Correlation analysis suggests that plasma Aβ42 levels were positively correlated to UPDRS part III scores in MSA. In terms of cognitive function, there was a relationship between plasma Aβ42/40 level and MMSE scores in patients with APD. Conclusion In our study, various combinations of plasma biomarkers have great potentialities in identifying PDSs, with important clinical utility in improving diagnostic accuracy. Plasma NfL may have added value to a blood-based biomarker panel for differentiating PDSs.
Collapse
|
25
|
Scott GD, Arnold MR, Beach TG, Gibbons CH, Kanthasamy AG, Lebovitz RM, Lemstra AW, Shaw LM, Teunissen CE, Zetterberg H, Taylor AS, Graham TC, Boeve BF, Gomperts SN, Graff-Radford NR, Moussa C, Poston KL, Rosenthal LS, Sabbagh MN, Walsh RR, Weber MT, Armstrong MJ, Bang JA, Bozoki AC, Domoto-Reilly K, Duda JE, Fleisher JE, Galasko DR, Galvin JE, Goldman JG, Holden SK, Honig LS, Huddleston DE, Leverenz JB, Litvan I, Manning CA, Marder KS, Pantelyat AY, Pelak VS, Scharre DW, Sha SJ, Shill HA, Mari Z, Quinn JF, Irwin DJ. Fluid and Tissue Biomarkers of Lewy Body Dementia: Report of an LBDA Symposium. Front Neurol 2022; 12:805135. [PMID: 35173668 PMCID: PMC8841880 DOI: 10.3389/fneur.2021.805135] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
The Lewy Body Dementia Association (LBDA) held a virtual event, the LBDA Biofluid/Tissue Biomarker Symposium, on January 25, 2021, to present advances in biomarkers for Lewy body dementia (LBD), which includes dementia with Lewy bodies (DLBs) and Parkinson's disease dementia (PDD). The meeting featured eight internationally known scientists from Europe and the United States and attracted over 200 scientists and physicians from academic centers, the National Institutes of Health, and the pharmaceutical industry. Methods for confirming and quantifying the presence of Lewy body and Alzheimer's pathology and novel biomarkers were discussed.
Collapse
Affiliation(s)
- Gregory D. Scott
- Department of Pathology, Oregon Health and Science University, Portland, OR, United States
- Department of Pathology and Laboratory Services, VA Portland Medical Center, Portland, OR, United States
| | - Moriah R. Arnold
- Graduate Program in Biomedical Sciences, School of Medicine M.D./Ph.D. Program, Oregon Health and Science University, Portland, OR, United States
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology and Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Christopher H. Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Anumantha G. Kanthasamy
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | | | - Afina W. Lemstra
- Department of Neurology, Amsterdam University Medical Center (UMC), Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London (UCL) Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | | | - Todd C. Graham
- Lewy Body Dementia Association, Lilburn, GA, United States
| | - Bradley F. Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, MN, United States
| | - Stephen N. Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | | | - Charbel Moussa
- Department of Neurology, Georgetown University Medical Center, Washington DC, CA, United States
| | - Kathleen L. Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Liana S. Rosenthal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Ryan R. Walsh
- Barrow Neurological Institute and Muhammed Ali Parkinson Center, Phoenix, AZ, United States
| | - Miriam T. Weber
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Melissa J. Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jee A. Bang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bozoki
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
| | | | - John E. Duda
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jori E. Fleisher
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, United States
| | - Douglas R. Galasko
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - James E. Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jennifer G. Goldman
- Shirley Ryan Abilitylab and Department of Physical Medicine and Rehabilitation and Neurology, Parkinson's Disease and Movement Disorders, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha K. Holden
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lawrence S. Honig
- Columbia University Irving Medical Center, New York, NY, United States
| | - Daniel E. Huddleston
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Carol A. Manning
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Karen S. Marder
- Columbia University Irving Medical Center, New York, NY, United States
| | - Alexander Y. Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Victoria S. Pelak
- Departments of Neurology and Ophthalmology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Douglas W. Scharre
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sharon J. Sha
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Holly A. Shill
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Zoltan Mari
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Las Vegas, NV, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, VA Portland Medical Center, Portland, OR, United States
| | - David J. Irwin
- Department of Neurology, University of Pennsylvania Health System, Philadelphia, PA, United States
- Digital Neuropathology Laboratory, Philadelphia, PA, United States
- Lewy Body Disease Research Center of Excellence, Philadelphia, PA, United States
- Frontotemporal Degeneration Center, Philadelphia, PA, United States
| |
Collapse
|
26
|
Plasma phosphorylated-tau181 as a predictive biomarker for Alzheimer's amyloid, tau and FDG PET status. Transl Psychiatry 2021; 11:585. [PMID: 34775468 PMCID: PMC8590691 DOI: 10.1038/s41398-021-01709-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
Plasma phosphorylated-tau181 (p-tau181) showed the potential for Alzheimer's diagnosis and prognosis, but its role in detecting cerebral pathologies is unclear. We aimed to evaluate whether it could serve as a marker for Alzheimer's pathology in the brain. A total of 1189 participants with plasma p-tau181 and PET data of amyloid, tau or FDG PET were included from ADNI. Cross-sectional relationships of plasma p-tau181 with PET biomarkers were tested. Longitudinally, we further investigated whether different p-tau181 levels at baseline predicted different progression of Alzheimer's pathological changes in the brain. We found plasma p-tau181 significantly correlated with brain amyloid (Spearman ρ = 0.45, P < 0.0001), tau (0.25, P = 0.0003), and FDG PET uptakes (-0.37, P < 0.0001), and increased along the Alzheimer's continuum. Individually, plasma p-tau181 could detect abnormal amyloid, tau pathologies and hypometabolism in the brain, similar with or even better than clinical indicators. The diagnostic accuracy of plasma p-tau181 elevated significantly when combined with clinical information (AUC = 0.814 for amyloid PET, 0.773 for tau PET, and 0.708 for FDG PET). Relationships of plasma p-tau181 with brain pathologies were partly or entirely mediated by the corresponding CSF biomarkers. Besides, individuals with abnormal plasma p-tau181 level (>18.85 pg/ml) at baseline had a higher risk of pathological progression in brain amyloid (HR: 2.32, 95%CI 1.32-4.08) and FDG PET (3.21, 95%CI 2.06-5.01) status. Plasma p-tau181 may be a sensitive screening test for detecting brain pathologies, and serve as a predictive biomarker for Alzheimer's pathophysiology.
Collapse
|
27
|
Li XY, Li W, Li X, Li XR, Sun L, Yang W, Cai Y, Chen Z, Wu J, Wang C, Yu S. Alterations of Erythrocytic Phosphorylated Alpha-Synuclein in Different Subtypes and Stages of Parkinson's Disease. Front Aging Neurosci 2021; 13:623977. [PMID: 34658833 PMCID: PMC8511781 DOI: 10.3389/fnagi.2021.623977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Serine 129-phosphorylated alpha-synuclein (pS-α-syn) is a major form of α-syn relevant to the pathogenesis of Parkinson's disease (PD), which has been recently detected in red blood cells (RBCs). However, alterations of RBC-derived pS-α-syn (pS-α-syn-RBC) in different subtypes and stages of PD remains to be investigated. In the present study, by using enzyme-linked immunosorbent assay (ELISA) to measure pS-α-syn-RBC, we demonstrated significantly higher levels of pS-α-syn-RBC in PD patients than in healthy controls. pS-α-syn-RBC separated the patients well from the controls, with a sensitivity of 93.39% (95% CI: 90.17-95.81%), a specificity of 93.11% (95% CI: 89.85-95.58%), and an area under the curve (AUC) of 0.96. Considering motor subtypes, the levels of pS-α-syn-RBC were significantly higher in late-onset than young-onset PD (p = 0.013) and in those with postural instability and gait difficulty than with tremor-dominant (TD) phenotype (p = 0.029). In addition, the levels of pS-α-syn-RBC were also different in non-motor subtypes, which were significantly lower in patients with cognitive impairment (p = 0.012) and olfactory loss (p = 0.004) than in those without such symptoms. Moreover, the levels of pS-α-syn-RBC in PD patients were positively correlated with disease duration and Hoehn & Yahr stages (H&Y) (p for trend =0.02 and <0.001) as well as UPDRS III (R 2 = 0.031, p = 0.0042) and MoCA scores (R 2 = 0.048, p = 0.0004). The results obtained suggest that pS-α-syn-RBC can be used as a potential biomarker for not only separating PD patients from healthy controls but also predicting the subtypes and stages of PD.
Collapse
Affiliation(s)
- Xu-Ying Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xu-Ran Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Linjuan Sun
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiwei Yang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhigang Chen
- Department of Neurology, Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Guangdong, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Lin WC, Lee PL, Lu CH, Lin CP, Chou KH. Linking Stage-Specific Plasma Biomarkers to Gray Matter Atrophy in Parkinson Disease. AJNR Am J Neuroradiol 2021; 42:1444-1451. [PMID: 34045303 DOI: 10.3174/ajnr.a7171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/17/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The shortcomings of synucleinopathy-based Parkinson disease staging highlight the need for systematic clinicopathologic elucidation and biomarkers. In this study, we investigated associations of proteinopathy and inflammation markers with changes in gray matter volume that accompany Parkinson disease progression. MATERIALS AND METHODS We prospectively enrolled 42 patients with idiopathic Parkinson disease, subdivided into early-/late-stage groups and 27 healthy controls. Parkinson disease severity and participants' functional and cognitive performance were evaluated. Peripheral plasma α-synuclein, β-amyloid42, and tau were quantified with immunomagnetic reduction assays, and nuclear DNA by polymerase chain reaction, and regional gray matter volumes were determined by MR imaging. Statistical tests identified stage-specific biomarkers and gray matter volume patterns in the early-stage Parkinson disease, late-stage Parkinson disease, and control groups. Correlations between gray matter volume atrophy, plasma biomarkers, Parkinson disease severity, and cognitive performance were analyzed. RESULTS Patients with Parkinson disease had significantly elevated α-synuclein, tau, and β-amyloid42 levels compared with controls; nuclear DNA levels were similar in early-stage Parkinson disease and controls, but higher in late-stage Parkinson disease (all P < .01). We identified 3 stage-specific gray matter volume atrophy patterns: 1) control > early-stage Parkinson disease = late-stage Parkinson disease: right midfrontal, left lingual, and fusiform gyri, left hippocampus, and cerebellum; 2) control > early-stage Parkinson disease > late-stage Parkinson disease: precentral, postcentral, parahippocampal, left superior-temporal, right temporal, right superior-frontal, and left cingulate gyri, occipital lobe, and bilateral parts of the cerebellum; 3) control = early-stage Parkinson disease > late-stage Parkinson disease: left midfrontal, superior-frontal and temporal, amygdala, and posterior cingulate gyri, caudate nucleus, and putamen. We discovered stage-specific correlations among proteinopathy, inflammation makers, topographic gray matter volume patterns, and cognitive performance that accompanied Parkinson disease progression. CONCLUSIONS Identifying associations linking peripheral plasma biomarkers, gray matter volume, and clinical status in Parkinson disease may facilitate earlier diagnosis and improve prognostic accuracy.
Collapse
Affiliation(s)
- W-C Lin
- From the Department of Diagnostic Radiology (W.-C.L.), Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - P-L Lee
- Institute of Neuroscience (P.-L.L., C.-P.L., K.-H.C.), National Yang-Ming University, Taipei, Taiwan
| | - C-H Lu
- Department of Neurology (C.-H.L.), Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - C-P Lin
- Institute of Neuroscience (P.-L.L., C.-P.L., K.-H.C.), National Yang-Ming University, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences (C.-P.L.), National Yang-Ming University, Taipei, Taiwan
- Brain Research Center (C.-P.L., K.-H.C.), National Yang-Ming University, Taipei, Taiwan
| | - K-H Chou
- Institute of Neuroscience (P.-L.L., C.-P.L., K.-H.C.), National Yang-Ming University, Taipei, Taiwan
- Brain Research Center (C.-P.L., K.-H.C.), National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
29
|
Ashton NJ, Leuzy A, Karikari TK, Mattsson-Carlgren N, Dodich A, Boccardi M, Corre J, Drzezga A, Nordberg A, Ossenkoppele R, Zetterberg H, Blennow K, Frisoni GB, Garibotto V, Hansson O. The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur J Nucl Med Mol Imaging 2021; 48:2140-2156. [PMID: 33677733 PMCID: PMC8175325 DOI: 10.1007/s00259-021-05253-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The development of blood biomarkers that reflect Alzheimer's disease (AD) pathophysiology (phosphorylated tau and amyloid-β) has offered potential as scalable tests for dementia differential diagnosis and early detection. In 2019, the Geneva AD Biomarker Roadmap Initiative included blood biomarkers in the systematic validation of AD biomarkers. METHODS A panel of experts convened in November 2019 at a two-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of blood biomarkers was assessed based on the Biomarker Roadmap methodology and discussed fully during the workshop which also evaluated cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers. RESULTS Plasma p-tau has shown analytical validity (phase 2 primary aim 1) and first evidence of clinical validity (phase 3 primary aim 1), whereas the maturity level for Aβ remains to be partially achieved. Full and partial achievement has been assigned to p-tau and Aβ, respectively, in their associations to ante-mortem measures (phase 2 secondary aim 2). However, only preliminary evidence exists for the influence of covariates, assay comparison and cut-off criteria. CONCLUSIONS Despite the relative infancy of blood biomarkers, in comparison to CSF biomarkers, much has already been achieved for phases 1 through 3 - with p-tau having greater success in detecting AD and predicting disease progression. However, sufficient data about the effect of covariates on the biomarker measurement is lacking. No phase 4 (real-world performance) or phase 5 (assessment of impact/cost) aim has been tested, thus not achieved.
Collapse
Affiliation(s)
- N J Ashton
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden.
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - A Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - T K Karikari
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
| | - N Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Center for Neurocognitive Rehabilitation (CeRiN), CIMeC, University of Trento, Trento, Italy
| | - M Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| | - J Corre
- Centre National de la Recherche Scientifique, Montpellier, France
| | - A Drzezga
- Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - R Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - H Zetterberg
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - K Blennow
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - G B Frisoni
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland
| | - O Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- UK Dementia Research Institute at UCL, London, UK.
- Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden.
| |
Collapse
|
30
|
Lin WC, Lu CH, Chiu PY, Yang SY. Plasma Total α-Synuclein and Neurofilament Light Chain: Clinical Validation for Discriminating Parkinson's Disease from Normal Control. Dement Geriatr Cogn Disord 2021; 49:401-409. [PMID: 33242863 DOI: 10.1159/000510325] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/15/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A previously published paper (referred to as the original cohort) showed that using a cutoff value of 116.1 fg/mL for the plasma total α-synuclein concentrations could discriminate Parkinson's disease (PD) patients from normal controls (NCs). In this study, another independent cohort (referred to as the validation cohort) was recruited to validate the agreement between the clinical diagnosis and the use of plasma total α-synuclein to identify PD patients. In addition to total α-synuclein, plasma neurofilament light chain (NfL) in the validation cohort was detected. METHODS Seventy PD patients and 33 NCs were enrolled in the validation cohort. A clinical diagnosis and the immunomagnetic reduction (IMR) assay for plasma total α-synuclein were performed for each participant. Thirty-three of 70 PD patients and 23 of 33 NCs were subjected to the plasma NfL assay via IMR. RESULTS The positive, negative, and overall percentages of agreement between the clinical diagnosis and plasma total α-synuclein diagnosis determined based on 116.1 fg/mL as the cutoff value were found to be 0.943, 0.818, and 0.903, respectively. The PD patients and NCs showed plasma NfL levels of 8.38 ± 4.19 pg/mL and 17.6 ± 7.95 pg/mL (p < 0.001), respectively. The cutoff value of the plasma NfL level used to differentiate PD patients from NCs was 12.8 pg/mL, with sensitivity and specificity values of 0.788 and 0.870, respectively. CONCLUSION The results demonstrate the usefulness of the plasma total α-synuclein concentration to discriminate PD patients from NCs and reveal the elevation of the plasma NfL level in PD patients.
Collapse
Affiliation(s)
- Wei-Che Lin
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Hsien Lu
- Department of Diagnostic Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital and MR-guided Focus Ultrasound Center, Chang Bin Show Chwan Memorial Hospital, Chunghwa, Taiwan
| | - Shieh-Yueh Yang
- MagQu Co., Ltd., New Taipei City, Taiwan, .,MagQu LLC, Surprise, Arizona, USA,
| |
Collapse
|
31
|
Amyloid related cerebral microbleed and plasma Aβ40 are associated with cognitive decline in Parkinson's disease. Sci Rep 2021; 11:7115. [PMID: 33782518 PMCID: PMC8007804 DOI: 10.1038/s41598-021-86617-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/25/2021] [Indexed: 02/01/2023] Open
Abstract
Cerebral microbleeds (MBs) have been found in patients with cognitive decline. We aimed to examine whether MBs are associated with motor or cognitive decline in patients with Parkinson’s disease (PD). We enrolled 135 PD patients and 34 healthy controls. All participants underwent brain MRI and plasma biomarker assays, including tau, Aβ42, Aβ40, and α-synuclein. PD with dementia (PDD) was operationally defined as Mini-Mental State Examination (MMSE) score < 26 and advanced motor stage was defined as Hoehn-Yahr stage ≥ 3 during “on” status. The association between MBs and disease severity was examined using multivariate logistic regression models. More lobar MBs were observed in PD patients than controls (20.7% vs. 3.3%, p = 0.031). PDD patients had more lobar MBs (33.3% vs. 15.6%, p = 0.034), more white matter hyperintensity (p = 0.021) and reduced hippocampal volume (p = 0.001) than PD with normal cognition. The presence of lobar MB (odds ratio = 2.83 [95% confidence interval 1.04–7.70], p = 0.042) and severe white matter hyperintensity (3.29 [1.21–8.96], p = 0.020) was independently associated with PDD after adjusting for vascular risk factors and other confounders. Furthermore, plasma Aβ40 levels were associated the MMSE score (p = 0.004) after adjusting for age and sex. Our findings demonstrated that lobar MBs, reduced hippocampal volume, and elevated plasma Aβ40 levels are associated with PDD.
Collapse
|
32
|
Li CH, Chen TF, Chiu MJ, Yen RF, Shih MC, Lin CH. Integrated 18F-T807 Tau PET, Structural MRI, and Plasma Tau in Tauopathy Neurodegenerative Disorders. Front Aging Neurosci 2021; 13:646440. [PMID: 33854426 PMCID: PMC8039308 DOI: 10.3389/fnagi.2021.646440] [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: 12/26/2020] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
Background and Objective: Tau-specific positron emission topography (PET) imaging enables in vivo assessment of Alzheimer's disease (AD). We aimed to investigate its performance in combination with plasma tau levels in patients with non-AD tauopathy. Methods: A total of 47 participants were enrolled, including 10 healthy controls, 16 with tauopathy parkinsonism syndromes (9 with corticobasal syndrome [CBS], 7 with progressive supranuclear palsy [PSP]), 9 with frontotemporal dementia (FTD), 4 with AD, and 8 with Parkinson's disease (PD). All participants underwent clinical assessments, 18F-T807 tau PET, brain MRI, and plasma tau assay. Results: The global cortical standard uptake value ratio (SUVR) of 18F-T807 PET was comparable between PD and control (p = 0.088). The cortical SUVR was significantly higher in AD group (p = 0.002) but was modestly increased in PSP group compared to the PD group (p = 0.044), especially in parietal and pallidal regions. Asymmetric 18F-T807 uptake at the pallidum was noted in patients with CBS and FTD. Cortical tau tracer uptake was associated with increased plasma total tau level (p = 0.016), especially in frontal and parietal regions. Regional tracer uptake was correlated with cortical thinning in patients with CBS and PSP (CBS: r = -0.092, p = 0.025; PSP: r = -0.114, p = 0.015). Conclusions: The 18F-T807 tau tracer uptake was only modestly increased in patients with PSP. Although the cortical tau tracer uptake correlated with regional cortical atrophy and plasma tau levels, a four-repeated tau-specific tracer is needed for future classifying tauopathy parkinsonism syndromes.
Collapse
Affiliation(s)
- Cheng-Hsuan Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University Biomedical Park Hospital, Hsinchu, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Psychology, National Taiwan University, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Chieh Shih
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
33
|
Ding X, Zhang S, Jiang L, Wang L, Li T, Lei P. Ultrasensitive assays for detection of plasma tau and phosphorylated tau 181 in Alzheimer's disease: a systematic review and meta-analysis. Transl Neurodegener 2021; 10:10. [PMID: 33712071 PMCID: PMC7953695 DOI: 10.1186/s40035-021-00234-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
A lack of convenient and reliable biomarkers for diagnosis and prognosis is a common challenge for neurodegenerative diseases such as Alzheimer's disease (AD). Recent advancement in ultrasensitive protein assays has allowed the quantification of tau and phosphorylated tau proteins in peripheral plasma. Here we identified 66 eligible studies reporting quantification of plasma tau and phosphorylated tau 181 (ptau181) using four ultrasensitive methods. Meta-analysis of these studies confirmed that the AD patients had significantly higher plasma tau and ptau181 levels compared with controls, and that the plasma tau and ptau181 could predict AD with high-accuracy area under curve of the Receiver Operating Characteristic. Therefore, plasma tau and plasma ptau181 can be considered as biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Xulong Ding
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Jiang
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
34
|
Campese N, Palermo G, Del Gamba C, Beatino MF, Galgani A, Belli E, Del Prete E, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Progress regarding the context-of-use of tau as biomarker of Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics 2021; 18:27-48. [PMID: 33545008 DOI: 10.1080/14789450.2021.1886929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Tau protein misfolding and accumulation in toxic species is a critical pathophysiological process of Alzheimer's disease (AD) and other neurodegenerative disorders (NDDs). Tau biomarkers, namely cerebrospinal fluid (CSF) total-tau (t-tau), 181-phosphorylated tau (p-tau), and tau-PET tracers, have been recently embedded in the diagnostic criteria for AD. Nevertheless, the role of tau as a diagnostic and prognostic biomarker for other NDDs remains controversial.Areas covered: We performed a systematical PubMed-based review of the most recent advances in tau-related biomarkers for NDDs. We focused on papers published from 2015 to 2020 assessing the diagnostic or prognostic value of each biomarker.Expert opinion: The assessment of tau biomarkers in alternative easily accessible matrices, through the development of ultrasensitive techniques, represents the most significant perspective for AD-biomarker research. In NDDs, novel tau isoforms (e.g. p-tau217) or proteolytic fragments (e.g. N-terminal fragments) may represent candidate diagnostic and prognostic biomarkers and may help monitoring disease progression. Protein misfolding amplification assays, allowing the identification of different tau strains (e.g. 3 R- vs. 4 R-tau) in CSF, may constitute a breakthrough for the in vivo stratification of NDDs. Tau-PET may help tracking the spatial-temporal evolution of tau pathophysiology in AD but its application outside the AD-spectrum deserves further studies.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Del Gamba
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| |
Collapse
|
35
|
Kang J, Kim JW, Heo H, Lee J, Park KY, Yoon JH, Chang J. Identification of BAG2 and Cathepsin D as Plasma Biomarkers for Parkinson's Disease. Clin Transl Sci 2020; 14:606-616. [PMID: 33202088 PMCID: PMC7993325 DOI: 10.1111/cts.12920] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/10/2020] [Indexed: 12/30/2022] Open
Abstract
The current diagnosis of Parkinson’s disease (PD) mostly relies on clinical rating scales related to motor dysfunction. Given that clinical symptoms of PD appear after significant neuronal cell death in the brain, it is required to identify accessible, objective, and quantifiable biomarkers for early diagnosis of PD. In this study, a total of 20 patients with idiopathic PD and 20 age‐matched patients with essential tremor according to the UK Brain Bank Criteria were consecutively enrolled to identify peripheral blood biomarkers for PD. Clinical data were obtained by clinical survey and assessment. Using albumin‐depleted and immunoglobulin G‐depleted plasma samples, we performed immunoblot analysis of seven autophagy‐related proteins and compared the levels of proteins to those of the control group. We also analyzed the correlation between the levels of candidate proteins and clinical characteristics. Finally, we validated our biomarker models using receiver operating characteristic curve analysis. We found that the levels of BCL2‐associated athanogene 2 (BAG2) and cathepsin D were significantly decreased in plasma of patients with PD (P = 0.009 and P = 0.0077, respectively). The level of BAG2 in patients with PD was significantly correlated with Cross‐Culture Smell Identification Test score, which indicates olfactory dysfunction. We found that our biomarker model distinguishes PD with 87.5% diagnostic accuracy (area under the curve (AUC) = 0.875, P < 0.0001). Our result suggests BAG2 and cathepsin D as candidates for early‐diagnosis plasma biomarkers for PD. We provide the possibility of plasma biomarkers related to the autophagy pathway, by which decreased levels of BAG2 and cathepsin D might lead to dysfunction of autophagy.
Collapse
Affiliation(s)
- Juhee Kang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jae Whan Kim
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Hansol Heo
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jihyun Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, Korea
| | - Kwan Yong Park
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jung Han Yoon
- Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Jaerak Chang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
36
|
Lin CH, Chiu SI, Chen TF, Jang JSR, Chiu MJ. Classifications of Neurodegenerative Disorders Using a Multiplex Blood Biomarkers-Based Machine Learning Model. Int J Mol Sci 2020; 21:ijms21186914. [PMID: 32967146 PMCID: PMC7555120 DOI: 10.3390/ijms21186914] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
Easily accessible biomarkers for Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), and related neurodegenerative disorders are urgently needed in an aging society to assist early-stage diagnoses. In this study, we aimed to develop machine learning algorithms using the multiplex blood-based biomarkers to identify patients with different neurodegenerative diseases. Plasma samples (n = 377) were obtained from healthy controls, patients with AD spectrum (including mild cognitive impairment (MCI)), PD spectrum with variable cognitive severity (including PD with dementia (PDD)), and FTD. We measured plasma levels of amyloid-beta 42 (Aβ42), Aβ40, total Tau, p-Tau181, and α-synuclein using an immunomagnetic reduction-based immunoassay. We observed increased levels of all biomarkers except Aβ40 in the AD group when compared to the MCI and controls. The plasma α-synuclein levels increased in PDD when compared to PD with normal cognition. We applied machine learning-based frameworks, including a linear discriminant analysis (LDA), for feature extraction and several classifiers, using features from these blood-based biomarkers to classify these neurodegenerative disorders. We found that the random forest (RF) was the best classifier to separate different dementia syndromes. Using RF, the established LDA model had an average accuracy of 76% when classifying AD, PD spectrum, and FTD. Moreover, we found 83% and 63% accuracies when differentiating the individual disease severity of subgroups in the AD and PD spectrum, respectively. The developed LDA model with the RF classifier can assist clinicians in distinguishing variable neurodegenerative disorders.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (C.-H.L.); (T.-F.C.)
| | - Shu-I Chiu
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei 10617, Taiwan; (S.-I.C.); (J.-S.R.J.)
- Department of Computer Science, National Chengchi University, Taipei 11605, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (C.-H.L.); (T.-F.C.)
| | - Jyh-Shing Roger Jang
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei 10617, Taiwan; (S.-I.C.); (J.-S.R.J.)
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (C.-H.L.); (T.-F.C.)
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 100233, Taiwan
- Graduate Institue of Psychology, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 65339)
| |
Collapse
|
37
|
Karikari TK, Pascoal TA, Ashton NJ, Janelidze S, Benedet AL, Rodriguez JL, Chamoun M, Savard M, Kang MS, Therriault J, Schöll M, Massarweh G, Soucy JP, Höglund K, Brinkmalm G, Mattsson N, Palmqvist S, Gauthier S, Stomrud E, Zetterberg H, Hansson O, Rosa-Neto P, Blennow K. Blood phosphorylated tau 181 as a biomarker for Alzheimer's disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol 2020; 19:422-433. [PMID: 32333900 DOI: 10.1016/s1474-4422(20)30071-5] [Citation(s) in RCA: 651] [Impact Index Per Article: 162.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND CSF and PET biomarkers of amyloid β and tau accurately detect Alzheimer's disease pathology, but the invasiveness, high cost, and poor availability of these detection methods restrict their widespread use as clinical diagnostic tools. CSF tau phosphorylated at threonine 181 (p-tau181) is a highly specific biomarker for Alzheimer's disease pathology. We aimed to assess whether blood p-tau181 could be used as a biomarker for Alzheimer's disease and for prediction of cognitive decline and hippocampal atrophy. METHODS We developed and validated an ultrasensitive blood immunoassay for p-tau181. Assay performance was evaluated in four clinic-based prospective cohorts. The discovery cohort comprised patients with Alzheimer's disease and age-matched controls. Two validation cohorts (TRIAD and BioFINDER-2) included cognitively unimpaired older adults (mean age 63-69 years), participants with mild cognitive impairment (MCI), Alzheimer's disease, and frontotemporal dementia. In addition, TRIAD included healthy young adults (mean age 23 years) and BioFINDER-2 included patients with other neurodegenerative disorders. The primary care cohort, which recruited participants in Montreal, Canada, comprised control participants from the community without a diagnosis of a neurological condition and patients referred from primary care physicians of the Canadian National Health Service for specialist care. Concentrations of plasma p-tau181 were compared with established CSF and PET biomarkers and longitudinal measurements using Spearman correlation, area under the curve (AUC), and linear regression analyses. FINDINGS We studied 37 individuals in the discovery cohort, 226 in the first validation cohort (TRIAD), 763 in the second validation cohort (BioFINDER-2), and 105 in the primary care cohort (n=1131 individuals). In all cohorts, plasma p-tau181 showed gradual increases along the Alzheimer's disease continuum, from the lowest concentrations in amyloid β-negative young adults and cognitively unimpaired older adults, through higher concentrations in the amyloid β-positive cognitively unimpaired older adults and MCI groups, to the highest concentrations in the amyloid β-positive MCI and Alzheimer's disease groups (p<0·001, Alzheimer's disease vs all other groups). Plasma p-tau181 distinguished Alzheimer's disease dementia from amyloid β-negative young adults (AUC=99·40%) and cognitively unimpaired older adults (AUC=90·21-98·24% across cohorts), as well as other neurodegenerative disorders, including frontotemporal dementia (AUC=82·76-100% across cohorts), vascular dementia (AUC=92·13%), progressive supranuclear palsy or corticobasal syndrome (AUC=88·47%), and Parkinson's disease or multiple systems atrophy (AUC=81·90%). Plasma p-tau181 was associated with PET-measured cerebral tau (AUC=83·08-93·11% across cohorts) and amyloid β (AUC=76·14-88·09% across cohorts) pathologies, and 1-year cognitive decline (p=0·0015) and hippocampal atrophy (p=0·015). In the primary care cohort, plasma p-tau181 discriminated Alzheimer's disease from young adults (AUC=100%) and cognitively unimpaired older adults (AUC=84·44%), but not from MCI (AUC=55·00%). INTERPRETATION Blood p-tau181 can predict tau and amyloid β pathologies, differentiate Alzheimer's disease from other neurodegenerative disorders, and identify Alzheimer's disease across the clinical continuum. Blood p-tau181 could be used as a simple, accessible, and scalable test for screening and diagnosis of Alzheimer's disease. FUNDING Alzheimer Drug Discovery Foundation, European Research Council, Swedish Research Council, Swedish Alzheimer Foundation, Swedish Dementia Foundation, Alzheimer Society Research Program.
Collapse
Affiliation(s)
- Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada; Montreal Neurological Institute, Montreal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia, South London and Maudsley NHS Foundation Trust, London, UK
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Andréa Lessa Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada; Montreal Neurological Institute, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Institute of Neurology and UK Dementia Research Institute, University College London, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC, Canada; Montreal Neurological Institute, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
38
|
Chen NC, Chen HL, Li SH, Chang YH, Chen MH, Tsai NW, Yu CC, Yang SY, Lu CH, Lin WC. Plasma Levels of α-Synuclein, Aβ-40 and T-tau as Biomarkers to Predict Cognitive Impairment in Parkinson's Disease. Front Aging Neurosci 2020; 12:112. [PMID: 32410983 PMCID: PMC7198695 DOI: 10.3389/fnagi.2020.00112] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Objective In this study, we assessed plasma biomarkers to identify cognitive impairment in Parkinson’s disease (PD) patients by applying ultra-sensitive immunomagnetic reduction-based immunoassay (IMR). Methods The study enrolled 60 PD patients and 28 age- and sex-matched normal controls. Complete cognitive function assessments were performed on participants using the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating. PD patients with an MMSE score of ≦26 were defined as having cognitive impairment. Meanwhile, a 99mTc-TRODAT-1 scan was performed and plasma levels of Aβ-40, Aβ-42, T-tau, and α-synuclein were evaluated using IMR, subsequent correlation analyses were then performed. Results Compared with normal adults, PD patients have higher plasma levels of α-synuclein and T-tau, and a lower level of Aβ-40 (p < 0.05). Plasma levels of α-synuclein (r = −0.323, p = 0.002), Aβ-40 (r = 0.276, p = 0.01), and T-tau (r = −0.322, p = 0.002) are significantly correlated with MMSE scores. The TRODAT scan results, including visual inspection and quantification, revealed significant correlations between Aβ-40 and PD. Multiple regression analysis showed that the plasma levels of Aβ-40 (OR = 0.921, 95% CI = 0.879–0.962), α-synuclein (OR = 3.016, 95% CI = 1.703–5.339), and T-tau (OR = 1.069, 95% CI = 1.026–1.115) were independently associated with PD patients with cognitive impairment. The cutoff values for predicting cognitive deficits in PD patients were 45.101 pg/ml of Aβ-40, (Area under curve (AUC) = 0.791), 0.389 pg/ml of α-synuclein, (AUC = 0.790), and 30.555 pg/ml of T-tau (AUC = 0.726). Conclusion Plasma levels of α-synuclein, Aβ-40, and T-tau are potential biomarkers to detect cognitive impairment in PD patients.
Collapse
Affiliation(s)
- Nai-Ching Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsiu-Ling Chen
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shau-Hsuan Li
- Department of Oncology and Hematology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Hsiang Chang
- Department of Nuclear Medicine, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Meng-Hsiang Chen
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Nai-Wen Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chiun-Chieh Yu
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | - Cheng-Hsien Lu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Che Lin
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
39
|
Ashton NJ, Hye A, Rajkumar AP, Leuzy A, Snowden S, Suárez-Calvet M, Karikari TK, Schöll M, La Joie R, Rabinovici GD, Höglund K, Ballard C, Hortobágyi T, Svenningsson P, Blennow K, Zetterberg H, Aarsland D. An update on blood-based biomarkers for non-Alzheimer neurodegenerative disorders. Nat Rev Neurol 2020; 16:265-284. [PMID: 32322100 DOI: 10.1038/s41582-020-0348-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 01/11/2023]
Abstract
Cerebrospinal fluid analyses and neuroimaging can identify the underlying pathophysiology at the earliest stage of some neurodegenerative disorders, but do not have the scalability needed for population screening. Therefore, a blood-based marker for such pathophysiology would have greater utility in a primary care setting and in eligibility screening for clinical trials. Rapid advances in ultra-sensitive assays have enabled the levels of pathological proteins to be measured in blood samples, but research has been predominantly focused on Alzheimer disease (AD). Nonetheless, proteins that were identified as potential blood-based biomarkers for AD, for example, amyloid-β, tau, phosphorylated tau and neurofilament light chain, are likely to be relevant to other neurodegenerative disorders that involve similar pathological processes and could also be useful for the differential diagnosis of clinical symptoms. This Review outlines the neuropathological, clinical, molecular imaging and cerebrospinal fluid features of the most common neurodegenerative disorders outside the AD continuum and gives an overview of the current status of blood-based biomarkers for these disorders.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Abdul Hye
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Anto P Rajkumar
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK.,Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Stuart Snowden
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Suárez-Calvet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Catalonia, Spain.,Department of Neurology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Memory Research Unit, Lund University, Malmö, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Disease Research, Neurogeriatrics Division, Karolinska Institutet, Novum, Huddinge, Stockholm, Sweden
| | | | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Per Svenningsson
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK. .,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK. .,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| |
Collapse
|
40
|
From the prion-like propagation hypothesis to therapeutic strategies of anti-tau immunotherapy. Acta Neuropathol 2020; 139:3-25. [PMID: 31686182 PMCID: PMC6942016 DOI: 10.1007/s00401-019-02087-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/15/2022]
Abstract
The term “propagon” is used to define proteins that may transmit misfolding in vitro, in tissues or in organisms. Among propagons, misfolded tau is thought to be involved in the pathogenic mechanisms of various “tauopathies” that include Alzheimer's disease, progressive supranuclear palsy, and argyrophilic grain disease. Here, we review the available data in the literature and point out how the prion-like tau propagation has been extended from Alzheimer's disease to tauopathies. First, in Alzheimer’s disease, the progression of tau aggregation follows stereotypical anatomical stages which may be considered as spreading. The mechanisms of the propagation are now subject to intensive and controversial research. It has been shown that tau may be secreted in the interstitial fluid in an active manner as reflected by high and constant concentration of extracellular tau during Alzheimer’s pathology. Animal and cell models have been devised to mimic tau seeding and propagation, and despite their limitations, they have further supported to the prion-like propagation hypothesis. Finally, such new ways of thinking have led to different therapeutic strategies in anti-tau immunotherapy among tauopathies and have stimulated new clinical trials. However, it appears that the prion-like propagation hypothesis mainly relies on data obtained in Alzheimer’s disease. From this review, it appears that further studies are needed (1) to characterize extracellular tau species, (2) to find the right pathological tau species to target, (3) to follow in vivo tau pathology by brain imaging and biomarkers and (4) to interpret current clinical trial results aimed at reducing the progression of these pathologies. Such inputs will be essential to have a comprehensive view of these promising therapeutic strategies in tauopathies.
Collapse
|
41
|
Lin CH, Liu HC, Yang SY, Yang KC, Wu CC, Chiu MJ. Plasma pS129-α-Synuclein Is a Surrogate Biofluid Marker of Motor Severity and Progression in Parkinson's Disease. J Clin Med 2019; 8:jcm8101601. [PMID: 31623323 PMCID: PMC6832465 DOI: 10.3390/jcm8101601] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 09/29/2019] [Indexed: 01/11/2023] Open
Abstract
Phosphorylated α-synuclein accounts for more than 90% of α-synuclein found in Lewy bodies of Parkinson’s disease (PD). We aimed to examine whether plasma Ser129-phosphorylated α-synuclein (pS129-α-synuclein) is a surrogate marker of PD progression. This prospective study enrolled 170 participants (122 PD patients, 68 controls). We measured plasma levels of total and pS129-α-synuclein using immunomagnetic reduction-based immunoassay. PD patients received evaluations of motor and cognition at baseline and at a mean follow-up interval of three years. Changes in the Movement Disorder Society revision of the Unified Parkinson’s Disease Rating Scale motor score (MDS-UPDRS part III) and Mini-Mental State Examination (MMSE) score were used to assess motor and cognition progression. Our results showed that plasma levels of total and pS129-α-synuclein were significantly higher in PD patients than controls (total: 1302.3 ± 886.6 fg/mL vs. 77.8 ± 36.6 fg/mL, p < 0.001; pS129-α-synuclein: 12.9 ± 8.7 fg/mL vs. 0.8 ± 0.6 fg/mL, p < 0.001), as was the pS129-α-synuclein/total α-synuclein ratio (2.8 ± 1.1% vs. 1.1 ± 0.6%, p = 0.01). Among PD patients, pS129-α-synuclein levels were higher with advanced motor stage (p < 0.001) and correlated with MDS-UPDRS part III scores (r = 0.27, 95% CI: 0.09–0.43, p = 0.004). However, we found no remarkable difference between PD patients with and without dementia (p = 0.75). After a mean follow-up of 3.5 ± 2.1 years, PD patients with baseline pS129-α-synuclein > 8.5 fg/mL were at higher risk of motor symptom progression of at least 3 points in the MDS-UPDRS part III scores than those with pS129-α-synuclein < 8.5 fg/mL (p = 0.03, log rank test). In conclusion, our data suggest that plasma pS129-α-synuclein levels correlate with motor severity and progression, but not cognitive decline, in patients with PD.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Huei-Chun Liu
- MagQu Co., Ltd., Xindian District, New Taipei City 231, Taiwan.
| | - Shieh-Yueh Yang
- MagQu Co., Ltd., Xindian District, New Taipei City 231, Taiwan.
- MagQu LLC, 12425 W Bell Rd, C107, Surprise, AZ 85378, USA.
| | - Kai-Chien Yang
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Chau-Chung Wu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei 116, Taiwan.
- Graduate institute of Psychology, National Taiwan University, Taipei 116, Taiwan.
| |
Collapse
|
42
|
Rösler TW, Tayaranian Marvian A, Brendel M, Nykänen NP, Höllerhage M, Schwarz SC, Hopfner F, Koeglsperger T, Respondek G, Schweyer K, Levin J, Villemagne VL, Barthel H, Sabri O, Müller U, Meissner WG, Kovacs GG, Höglinger GU. Four-repeat tauopathies. Prog Neurobiol 2019; 180:101644. [PMID: 31238088 DOI: 10.1016/j.pneurobio.2019.101644] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 02/08/2023]
Abstract
Tau is a microtubule-associated protein with versatile functions in the dynamic assembly of the neuronal cytoskeleton. Four-repeat (4R-) tauopathies are a group of neurodegenerative diseases defined by cytoplasmic inclusions predominantly composed of tau protein isoforms with four microtubule-binding domains. Progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease or glial globular tauopathy belong to the group of 4R-tauopathies. The present review provides an introduction in the current concept of 4R-tauopathies, including an overview of the neuropathological and clinical spectrum of these diseases. It describes the genetic and environmental etiological factors, as well as the contemporary knowledge about the pathophysiological mechanisms, including post-translational modifications, aggregation and fragmentation of tau, as well as the role of protein degradation mechanisms. Furthermore, current theories about disease propagation are discussed, involving different extracellular tau species and their cellular release and uptake mechanisms. Finally, molecular diagnostic tools for 4R-tauopathies, including tau-PET and fluid biomarkers, and investigational therapeutic strategies are presented. In summary, we report on 4R-tauopathies as overarching disease concept based on a shared pathophysiological concept, and highlight the challenges and opportunities on the way towards a causal therapy.
Collapse
Affiliation(s)
- Thomas W Rösler
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Amir Tayaranian Marvian
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Matthias Brendel
- Dept. of Nuclear Medicine, University of Munich, 81377 Munich, Germany
| | - Niko-Petteri Nykänen
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Matthias Höllerhage
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Sigrid C Schwarz
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | | | - Thomas Koeglsperger
- Dept. of Neurology, University of Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Gesine Respondek
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Kerstin Schweyer
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Johannes Levin
- Dept. of Neurology, University of Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Victor L Villemagne
- Dept. of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, 3084, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; Dept. of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia
| | - Henryk Barthel
- Dept. of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Osama Sabri
- Dept. of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Ulrich Müller
- Institute for Human Genetics, University of Giessen, 35392 Giessen, Germany
| | - Wassilios G Meissner
- Service de Neurologie, CHU Bordeaux, 33000 Bordeaux, France; Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Dept. of Medicine, University of Otago, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, 1090 Vienna, Austria; Dept. of Laboratory Medicine and Pathobiology, University of Toronto, Laboratory Medicine Program, University Health Network, Toronto, Canada; Tanz Centre for Research in Neurodegenerative Disease, Krembil Brain Institute, Toronto, Canada
| | - Günter U Höglinger
- Dept. of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Dept. of Neurology, Technical University of Munich, School of Medicine, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany; Dept. of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
43
|
Maclin JMA, Wang T, Xiao S. Biomarkers for the diagnosis of Alzheimer's disease, dementia Lewy body, frontotemporal dementia and vascular dementia. Gen Psychiatr 2019; 32:e100054. [PMID: 31179427 PMCID: PMC6551430 DOI: 10.1136/gpsych-2019-100054] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/21/2022] Open
Abstract
Background Dementia is a chronic brain disorder classified by four distinct diseases that impact cognition and mental degeneration. Each subgroup exhibits similar brain deficiencies and mutations. This review will focus on four dementia subgroups: Alzheimer’s disease, vascular dementia, frontotemporal dementia and dementia Lewy body. Aim The aim of this systematic review is to create a concise overview of unique similarities within dementia used to locate and identify new biomarker methods in diagnosing dementia. Methods 123 300 articles published after 2010 were identified from PubMed, JSTOR, WorldCat Online Computer Library and PALNI (Private Academic Library Network of Indiana) using the following search items (in title or abstract): ‘Neurodegenerative Diseases’ OR ‘Biomarkers’ OR ‘Alzheimer’s Disease’ OR ‘Frontal Temporal Lobe Dementia’ OR ‘Vascular Dementia’ OR ‘Dementia Lewy Body’ OR ‘Cerebral Spinal Fluid’ OR ‘Mental Cognitive Impairment’. 47 studies were included in the qualitative synthesis. Results Evidence suggested neuroimaging with amyloid positron emission tomography (PET) scanning and newly found PET tracers to be more effective in diagnosing Alzheimer’s and amnesiac mental cognitive impairment than carbon-11 Pittsburgh compound-B radioisotope tracer. Newly created methods to make PET scans more accurate and practical in clinical settings signify a major shift in diagnosing dementia and neurodegenerative diseases. Conclusion Vast improvements in neuroimaging techniques have led to newly discovered biomarkers and diagnostics. Neuroimaging with amyloid PET scanning surpasses what had been considered the dominant method of neuroimaging and MRI. Newly created methods to make PET scans more accurate and practical in clinical settings signify a major shift in diagnosing dementia pathology. Continued research and studies must be conducted to improve current findings and streamline methods to further subcategorise neurodegenerative disorders and diagnosis.
Collapse
Affiliation(s)
- Joshua Marvin Anthony Maclin
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China.,Department of Neuroscience, Earlham College, Richmond, Indiana, USA
| | - Tao Wang
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China.,Department of Neuroscience, Earlham College, Richmond, Indiana, USA
| | - Shifu Xiao
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China.,Department of Neuroscience, Earlham College, Richmond, Indiana, USA
| |
Collapse
|
44
|
Bougea A, Stefanis L, Paraskevas GP, Emmanouilidou E, Vekrelis K, Kapaki E. Plasma alpha-synuclein levels in patients with Parkinson's disease: a systematic review and meta-analysis. Neurol Sci 2019; 40:929-938. [PMID: 30715632 DOI: 10.1007/s10072-019-03738-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To date, there are no definitive biomarkers for diagnose Parkinson's disease (PD). The detection of α-synuclein (α-Syn) in plasma of PD patients has yielded promising but inconclusive results. To determine the performance of α-Syn as a diagnostic biomarker of PD, we used a meta-analysis. METHODS We identified 173 studies through a systematic literature review. From those, only studies reporting data on total α-Syn levels were included in the meta-analysis (10 publications, 1302 participants). Quality of studies was assessed by Newcastle-Ottawa scale. RESULTS The α-Syn levels were significantly higher in PD patients than healthy controls (standardized mean difference [SMD] = 0.778, 95% confidence interval = 0.284 to 1.272, p = 0.002). Similar results were found after omitting any individual study from meta-analysis, with SMD ranges from 0.318 (95% CI = 0.064 to 0.572, p = 0.014) to 0.914 (95% CI = 0.349 to 1.480, p = 0.002). According to meta-regression analysis, increased mean patients age (slope = - 0.232, 95% CI = - 0.456 to - 0.008, p = 0.042), increased total number of participants (slope = - 0.007, 95% CI = - 0.013 to - 0.0004, p = 0.038), and increased percentage of males (slope = - 6.444, 95% CI = - 10.841 to - 2.047, p = 0.004) were associated with decreased SMD of α-Syn levels across studies. We did not find any significant association between the SMD in α-Syn levels and disease duration, disease severity, and quality of studies. Most of studies applied ELISA assays. CONCLUSION Total plasma α-Syn levels were higher in PD patients than controls. Analytical factors were important limitations.
Collapse
Affiliation(s)
- Anastasia Bougea
- Neurochemistry laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, 72-74 Vasilissis Sofias Avenue, 11528, Athens, Greece.
- Neuroscience laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Leonidas Stefanis
- Neuroscience laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - George P Paraskevas
- Neurochemistry laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, 72-74 Vasilissis Sofias Avenue, 11528, Athens, Greece
| | - Evangelia Emmanouilidou
- Neuroscience laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Kostas Vekrelis
- Neuroscience laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Elisabeth Kapaki
- Neurochemistry laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, 72-74 Vasilissis Sofias Avenue, 11528, Athens, Greece
| |
Collapse
|